High-Throughput Screen to Identify Small Molecule Inhibitors of the Canonical Wnt Signaling Pathway

Size: px
Start display at page:

Download "High-Throughput Screen to Identify Small Molecule Inhibitors of the Canonical Wnt Signaling Pathway"

Transcription

1 High-Throughput Screen to Identify Small Molecule Inhibitors of the Canonical Wnt Signaling Pathway by Stephen J Perusini A thesis submitted in conformity with the requirements for the degree of Masters of Science Department of Biochemistry University of Toronto Copyright by Stephen J. Perusini 2008

2 High-Throughput Screen to Identify Small Molecule Inhibitors of the Canonical Wnt Signaling Pathway Abstract Stephen J, Perusini Masters of Science Department of Biochemistry University of Toronto 2008 Wnt signaling is important in human development and disease, thus dysregulated β-catenin constitutes an attractive target for drug intervention. The few functional inhibitors currently available target transcriptional activation, therefore, identifying novel upstream modulators would be of tremendous importance to elucidating the mechanisms involved in regulating β- catenin activity. To achieve this, I developed a high-throughput screen to assess β-catenin stability in mammalian cells using a luciferase tagged β-catenin molecule. This assay was used to screen three chemical libraries to identify small molecule modulators of the pathway. Identified inhibitors/activators of the pathway were investigated via secondary assays. The most promising inhibitor, 21H7, significantly attenuated activated β-catenin signaling in colon cancer cells, decreasing β-catenin stability. The inhibitory effects of 21H7 and a structurally similar compound were shown to not only inhibit Wnt target gene expression in colon cancer cells, but also prostate cancer lines. Thus, 21H7 represents an attractive lead compound for further study. ii

3 Acknowledgments I wish to thank my supervisor, Dr. Liliana Attisano for allowing me to join her team, for her guidance throughout my graduate studies and the various revisions of this thesis. My thanks and appreciation also goes to my thesis committee members, Dr. Craig Smibert and Dr. Aaron Schimmer, and collaborators Alessandro Datti and Jeffrey Wrana. I am greatly indebted to my peers in the Attisano lab, for their expertise, kindness, and most of all, for creating an enjoyable atmosphere with worthwhile memories. iii

4 Table of Contents Acknowledgments... ii Table of Contents... iii List of Tables... vii List of Figures... viii 1 Introduction Wnt signaling Wnt genes and proteins The Wnt extracellular environment Wnt signal transduction The canonical branch of the Wnt pathway Additional factors involved in mediating a Wnt response Wnt signaling in development Invertebrates Xenopus development Mammalian development Wnt signaling in human disease Wnt signaling in Cancer Other human diseases and Wnt signaling Small molecule screening Drug discovery Small molecule libraries Chemical genomics Known small molecule modulators of the Wnt pathway Rationale for project...22 iv

5 2 Materials and methods Reagents Cell lines and maintenance Production of Wnt3A ligand in conditioned media Constructs, transcriptional reporter assays and immunoblotting High-throughput assays Real time quantitative reverse transcription-pcr analysis Results High throughput assay development Beta-catenin nuclear translocation assay Firefly luciferase β-catenin stabilization assay (Ff-luc) Ff-luc assay optimization Pilot high-throughput Ff-luc assay run Final screening results and data analysis Comparison of various statistical hit-selection methods Summary of results from the Lopac library Summary of results from the Prestwick library Summary of results from the Maybridge library Total assay hits from the entire HTP screen Investigating selected hits via secondary analyses Assessing the reproducibility of effects Effects of hits on Wnt transcriptional activity and assessing pathway specificity Analysis of 21H7, a selective Wnt pathway inhibitor Compound 21H7 preferentially inhibits Wnt responsive Topflash activity over the non-responsive Fopflash H7 blocks Wnt-induced β-catenin stabilization v

6 H7 inhibits Wnt dependent gene expression in colon cancer cell lines Expression of endogenous Wnt target genes is inhibited by 21H7 in CRC cell lines H7 inhibits expression of endogenous Wnt target genes in prostate cancer cell lines Wnt signaling is inhibited by 21H7 at the level of β-catenin Structurally related compounds mimic the effect of 21H Discussion References Appendices.92 vi

7 List of Tables Table 1: Known Small molecules modulators of the Wnt pathway 21 Table 2: Pilot screen hits indicate a reproducible Ff-luc hit rate.40 Table 3: Summary of results from all compounds screened 54 Table 4: Summary of secondary assay results for twenty-two selected inhibitors and random control vii

8 List of Figures Figure 1: Overview of the canonical Wnt signaling pathway 4 Figure 2: Known Chemical inhibitors of the Wnt signaling pathway..19 Figure 3: The β-catenin Nuclear translocation assay displays high variability...30 Figure 4: Development of the β-catenin-firefly-luciferase (Ff-luc) assay 33 Figure 5: The Ff-Luc assay Figure 6: Validation of the Ff-luc screening protocol 37 Figure 7: Validating the pipetting accuracy and chemiluminescent readout of the robotics platform..38 Figure 8: Comparison of different statistical hit-selection methods...44 Figure 9: Lopac library screening results Figure 10: Prestwick library screening results...49 Figure 11: Maybridge library screening results 51 Figure 12: Re-testing of select compounds using the Ff-luc assay Figure 13: Compound 21H7 preferentially inhibits the Wnt pathway...58 Figure 14: 21H7 preferentially inhibits the transcriptional activity of Wnt3A treated cells...60 Figure 15: Compound 21H7 inhibits the Wnt3A induced stabilization of endogenous β-catenin...62 Figure 16: Differential suppression of Wnt-dependent gene expression in colon cancer cell lines harboring activating mutations in the Wnt pathway by 21H Figure 17: Wnt target gene expression is inhibited by 21H7 in colorectal cancer (CRC) cell lines irrespective of the differing Wnt pathway..66 Figure 18: 21H7 inhibits Wnt target gene expression in prostate cancer cells Figure 19: Epistatic analysis indicates 21H7 effects Wnt-dependent signaling at the level of β-catenin 71 Figure 20: Screened Maybridge compounds with significant structural similarity to 21H7 73 Figure 21: Compound 21A8, the molecule of greatest structural similarity to 21H7 inhibits the expression of Wnt target genes in colon cancer cell lines 74 viii

9 1 1 Introduction 1.1 Wnt signaling Wnt genes and proteins Wnt genes, of which there are 19 in humans and mouse comprise a family of highly conserved secreted glycoproteins that regulate processes such as cell motility and differentiation during embryonic development by inducing diverse signaling cascades to effect cell growth and survival (Clevers, 2006; Logan and Nusse, 2004). The Wnt proteins are found in species ranging in complexity from sea anemone to humans, and effect embryo development by acting locally and at distances in a morphogenic manner. Due to the diverse roles of Wnts, they have typically been identified via sequence similarity and not functional properties. All possess a highly conserved spacing of 22 cysteine residues, several highly charged amino-acid with conserved positioning, a signal sequence for secretion and multiple potential sites of glycosylation (Miller, 2002). Post-translational processing such as glycosylation and lipid modification plays a significant role in Wnt functionality either during secretion from the ligand producing cell or signal transduction at the receiving cell. Loss of particular glycosylation sites has been shown to hinder the ability of Wnt3A and Wnt5A producing cells to efficiently secrete the ligand, however, the ability of Wnts to bind their receptors and transduce signals are unaffected (Coudreuse and Korswagen, 2007). Conversely, palmitoylation of Wnt ligands is crucial to ligand receptor binding and bears no effect on secretion (Kikuchi et al., 2007). Therefore, many Wnt modifications are essential for proper secretion profiles and initiating a response in signal receiving cells in development and beyond The Wnt extracellular environment Once secreted, the extracellular milieu allows Wnts to interact with a multitude of additional Wnt signaling co-factors that modulate the effect of Wnts in a variety of ways (Kikuchi et al.,

10 2 2007). Genetically identified factors such as secreted frizzled related protein (SFRP) and Wntinhibitory factor (WIF-1) were shown to bind Wnts, sequestering them from the Wnt receptor Frizzled (Fz) and thus antagonizing Wnt signaling (Kim et al., 2005). However, not all effectors which bind Wnts are antagonistic. R-spondin binds Wnts and was recently demonstrated to bind both Fz and its co-receptor LRP6 to synergistically activate the Wnt pathway (Kim et al., 2005; Nam et al., 2006). Additional factors such as the Dikkopf (Dkk) family of proteins antagonize the Wnt pathway not by binding Wnts, but through binding to the LRP co-receptor and blocking receptor availability (Mao et al., 2001). Norrin is not a Wnt ligand as those previously described, but was shown to activate the Wnt signaling pathway by independently binding select Fz isoforms and thus effects Wnt signaling without modulating ligand activity (Planutis et al., 2007). Consequently, the Wnt pathway extracellular environment is undoubtedly complex and influences Wnt signaling through a combination of agonists and antagonists vying for the ability to bind both ligands and their receptors Wnt signal transduction The various Wnt ligands induce one of three distinct but interconnected signaling pathways; canonical, planar cell polarity, and the Ca 2+ pathway. The canonical pathway is by far the best understood of the three and it transduces signals via a distinct signaling cascade. The other two Wnt pathways are far less understood. Both the Wnt/Ca 2+ and non-canonical pathways transduce signals through the Frizzled (Fz) receptors, but diverge afterwards. The non-canonical or planar cell polarity pathway involves the activation of RhoA and Jun Kinase to regulate spatial and temporal cytoskeletal rearrangements, cell migration and tissue polarity during embryogenesis and afterwards (Adler, 2002). It is believed that the group of non-canonical Wnts initiate signaling via binding the Frizzled receptors without co-receptor involvement and transduce signal through Disheveled (Dvl) proteins, activating the two independent and parallel small GTPase pathways, Rac and Rho. The Wnt/Ca 2+ pathway is best understood via its effect on intracellular Ca 2+ levels, subsequent activation of Protein Kinase C and Calmodulin kinase and principally affects cell movement (Kikuchi et al., 2007). Wnt associated Frizzled activation is thought to affect Ca 2+ levels through

11 3 the activation of heterotrimeric G-proteins, resulting in the production of DAG and inositol triphosphate second messengers (Slusarski et al., 1997) The canonical branch of the Wnt pathway By far the best understood of the Wnt pathways is canonical Wnt signaling which influences cell fate through the activation of the LEF/TCF family of transcription factors. The central component of the canonical Wnt signaling pathway is β-catenin. In the absence of Wnt ligand, its cytoplasmic levels are kept low via the sequential phosphorylation by Casein Kinase 1 (CK1) and GSK3β (Glycogen synthase kinase-3) on β-catenin. This phosphorylation permits it to be recognized by the E3 ubiquitin ligase, βtrcp, and undergo continuous proteosome-mediated degradation (Fig. 1) (Lustig and Behrens, 2003). This constitutive destruction of β-catenin is interrupted as the Wnt pathway is activated once canonical Wnt ligands bind the Frizzled (Fz) and LRP5/6 (low density lipoprotein related) co-receptors (Tamai et al., 2004). Current knowledge indicates LRP functions solely in the canonical Wnt pathway and is dispensable for the others (Wehrli 2000). These receptors signal to both Axin and the Disheveled (Dvl) proteins, both key intracellular components of the Wnt pathway. By a yet unknown mechanism, receptor activation allows Dvl to promote the dissolution of a multi-component cytoplasmic complex (Destruction complex) that phosphorylates β-catenin (Wharton, 2003). Furthermore, receptor activation recruits Axin, a member of the destruction complex, to LRP receptors phosphorylated on both conserved Ser/Thr residues and the PPPSP motif (Tamai et al., 2004). In a Wnt induced manner, CK1γ phosphorylates the Ser/Thr cluster regions of LRP5/6 while GSK3β subsequently phosphorylates the PPPSP motif, allowing for Axin recruitment to the plasma membrane and away from a multi-component destruction complex (Liu et al., 2002; Tamai et al., 2004). This event, in combination with Dvl s association with this multicomponent scaffolding/destruction complex containing Axin, GSK3β, the APC (Adenomatous Polyposis Coli) tumor suppressor further promote complex dissociation, thereby preventing β- catenin from being phosphorylated by both CK1 and GSK3β. In a non-wnt stimulated state, the destruction complex is believed to contain Axin as the primary scaffolding molecule from which, CK1 (Liu et al., 2002), GSK3β (Dajani et al., 2003), and APC

12 4 Figure 1. Overview of the canonical Wnt signaling pathway. In the basal state, defined by an absence of Wnt signal, β-catenin is held within a multicomponent complex. Complex formation facilitates the sequential phosphorylation of β-catenin by CK1α and GSK3β on conserved serine and threonine residues. Phosphorylation on its amino terminus allows β-trcp, an E3 ligase, to bind β-catenin and promote ubiquitin-mediated proteosomal degradation resulting in low levels of β-catenin. In the presence of Wnt ligand, the cell surface interaction between Wnt and its two co-receptors Frizzled and LRP5/6 promotes the formation of a Frizzled- Dishevelled complex and the phosporylation of LRP by CK1γ,ε and GSK3β. These modifications allow for Axin to bind the LRP intracellular domain. Axin is sequestered away from the destruction complex, to the cell surface, resulting in the dissociation of the multicomponent complex. β-catenin is freed, is not efficiently modified by CK1α and GSK3β and therefore goes unrecognized by the E3 ligase allowing for β-catenin levels to accumulate and β-catenin enters the nucleus. Once in the nucleus, β-catenin interacts with members of the TCF/LEF transcription factor family displacing transcriptional repressors and recruiting a host of co-activators to promote the expression of a variety of proliferative and cell-fate determining genes

13 5 Figure 1

14 6 (Kimelman and Xu, 2006; Spink et al., 2000) bind. β-catenin enters the complex by binding Axin through the conserved armadillo repeats and the 15 amino acid repeats on APC. These associations are thought to position the N-terminus β-catenin adjacent to the two kinases in the complex, enabling CK1 to phosphorylate β-catenin at Ser45 (Hagen and Vidal-Puig, 2002). This primary modification serves as a primer, permitting GSK3β to sequentially phosphorylate β-catenin at adjacent Thr 41, Ser 37 and Ser 33 (Amit et al., 2002). Upon Wnt stimulation, the complex dissociates in the manner described above and β-catenin is freed from these interactors and therefore is not efficiently modified by CK1 and GSK3β. Upon release from the scaffolding complex, the non-phosphorylated form of β-catenin goes unrecognized by the proteosome destruction complex. Thus, free β-catenin is stabilized and accumulates in the cytosol, allowing its translocation to the nucleus where it binds to transcription factors of the TCF/LEF (T cell-specific transcription factor/lymphoid enhancerbinding factor) family. This interaction transiently converts TCF factor repression into transcriptional activation thereby inducing the expression of a variety of downstream genes, many of which have been implicated in cancer (Logan and Nusse, 2004). Canonical Wnt target genes include cell cycle and anti-apoptotic factors such as survivin (Zhang et al., 2001a), Cyclin D1 (Tetsu and McCormick, 1999), C-myc (He et al., 1998), in addition to a variety of secreted growth factors including VEGF (Zhang et al., 2001b) and WISP (Xu et al., 2000). Furthermore, Wnt signaling promotes the expression of Wnt pathway component Axin2 (Jho et al., 2002), resulting in negative feedback Additional factors involved in mediating a Wnt response Like the Wnt extracellular environment, there are many co-factors outside of the core Wntpathway components which modulate intracellular Wnt signal transduction. In fact, there are currently 32 purported intracellular modulators outside of the core canonical pathway components (The Wnt homepage, For instance, members of the protein phosphatase family PP2A and C, are key components of canonical Wnt signaling and are often found within the multi-component destruction complex (Lustig and Behrens, 2003; Seeling et al., 1999). Their roles, however remain unclear,

15 7 obfuscated by the ability of PP2A to bind both Axin and APC while acting as both a positive and negative regulator of canonical Wnt signaling, in a yet unknown manner (Seeling et al., 1999; Willert et al., 1999). PP2C has been shown to interact with Dvl2 though its PDZ domain and as a result destabilizes Axin (Strovel et al., 2000). Other major cytoplasmic modulators such as Frodo and Dapper possess 90% sequence identity, yet have been shown to inversely effect canonical signaling. Required for proper development, both bind Dvl, but on different domains, Dapper is a negative regulator, while Frodo has been shown to positively regulate Wnt signaling by mechanisms still unknown (Wharton, 2003). Once in the nucleus, a variety of factors directly influence β-catenin/tcf/lef activity. ICAT, for example, binds β-catenin and is thought to displace it from its transcriptional partners TCF/LEF and CREB binding protein (Reifenberger et al., 2002). Furthermore, commonly seen splice variants of TCF/LEF are known to act as dominant-negatives lacking either β-catenin or DNA binding domains (Hoppler and Kavanagh, 2007). Furthermore, factors such as the TCF/LEF co-repressor Groucho, competes with β-catenin for TCF/Lef binding. All of these effectors contribute to the complexity of Wnt signal transduction and transcriptional activation. 1.2 Wnt signaling in development Invertebrates Wnt signaling is a crucial factor during embryonic development in organisms of varied complexity. Wnt-β-catenin signaling is a key regulator of germ layer formation in invertebrate species such as nematodes, sea urchina and cnidarians, in which factors stored in oocytes directly activate canonical Wnt signaling in a spatially restricted manner specifying germ layers and defining body axis patterns (Marikawa, 2006). This role of germ layer specification is most evident in C. elegans, where the Wnt3 homologue mom-2 plays a critical role in establishing embryonic polarity and inducing germ layer formation. After the second division of the zygote, in a four-cell-staged embryo, one of the cells (P2) becomes polarized and adjacent daughter cells which from there-on divide separately give rise to the endoderm or mesodermal lineages. In the

16 8 absence of Wnt signaling, both daughter cells adopt the mesodermal cell fate and genetic screens have linked mom2 as the essential P2 cell factor responsible for the polarization (Rocheleau et al., 1997). Furthermore, activity of a Lef-1 homologue in the polarized daughter cells is critical for the cells to adopt differing fates (Lin et al., 1995) Xenopus development Like the invertebrates, frog body axes are defined in the first few cellular divisions, establishing the three germ layers, endoderm, ectoderm and mesoderm in the polarizing oocyte. Major body axis organization is largely coordinated via a region of the developing embryo called the Spemann organizer. This organizer consists of a small group of cells in the vertebrate embryo which possess inductive morphogenic properties that establish the frog body plan (Garcia- Fernandez et al., 2007). Wnt signaling plays a crucial role in the development of the organizer, as activated β-catenin is present on the dorsal side of the blastula at the same time and location the organizer develops. Injection of murine Wnt1 mrna into early Xenopus blastomeres resulted in the induction of dorsal mesoderm formation and a duplicate body axis (McMahon and Moon, 1989). This quintessential Wnt experiment demonstrated a significant role for β-catenin signaling in vertebrate body plan formation. Moreover, it demonstrated the highly conserved nature of the canonical Wnt signaling pathway whereby murine Wnt1 was able to effect frog development. Equally important, Wnt signaling was shown to play a critical role in specifying germ layer and body plan in invertebrates and vertebrates alike. Subsequent studies revealed that identical axis duplications could be induced by injecting any of the canonical Wnt ligands and a wide variety of downstream Wnt pathway activating components including β-catenin, Dvl, and dominant-negative forms of GSK3β (Clevers, 2006; Wodarz and Nusse, 1998) Mammalian development Although Wnt signaling is responsible for establishing the dorso-ventral body axis in lower vertebrates, regulates germ layer formation and specifies body axes amongst invertebrate species, data on higher vertebrates is not as well established. The Wnt influence in mammalian development is significantly more complex and less delineated, however, significant insights

17 9 have been garnered from mouse studies. Wnts have been shown to play critical roles during various stages of development. Compared to other species, mammalian development is significantly different; the initial events of germ layer and body axis formation in nonmammalian species are secondary events in mammalian development. The first event in mammals is placenta development allowing for interacting with the mothers uterus, body plan and axis formation occur post implantation (Marikawa, 2006). A variety of studies have investigated the role of canonical Wnts during pre-implantation development using loss-offunction, gain-of-function, and overexpression analysis and none have reported any abnormalities until post implantation (Haegel et al., 1995; Kemler et al., 2004). Therefore, canonical Wnt signaling solely plays its role post implantation, despite a variety of ligands and receptors being expresses pre-implantation (Hamatani et al., 2004; Wang et al., 2004). The initial event of germ-layer formation occurs around E4.5, with the formation of the primitive endoderm in the developing blastocyst, defining the dorso-ventral (D-V) axis of the embryo. The role for Wnt in this event is unclear. β-catenin-null embryos develop normal primitive endoderm (Haegel et al., 1995), however, in vitro models suggest up-regulation of β-catenin signaling induces primitive endoderm formation (Liu et al., 1999). The first clear role for Wnt/β-catenin signaling appears to be at E5.5 in patterning the visceral endoderm and promoting its migration by regulating the expression of secreted molecules controlling the anterior movement of the visceral endoderm (Huelsken et al., 2000). After the formation of the endoderm, epiblast cells move opposite the anterior visceral ectoderm and undergo epithelial-mesenchymal transformation (gastrulation) to generate the primitive streak. It is from this structure that cells give rise to endoderm and mesoderm. Similar to Xenopus development, non-phosphoryated β-catenin accumulation is present in the region of epiblast cells where the primitive streak emerges during the onset of gastrulation (Marikawa, 2006; Mohamed et al., 2004). Furthermore, reporter constructs indicate TCF-mediated transcription is spatially and temporally activated during gastrulation (Mohamed et al., 2004). Also, several Wnt pathway components have demonstrated indispensability for both primitive streak and eventual axis formation (Liu et al., 1999), while excessive Wnt signal causes expansion and disorganization of the streak in mouse embryos (Kemler et al., 2004). The data

18 10 indicates a critical role for canonical Wnt signaling in the events leading up to and encompassing mouse gastrulation. After gastrulation, the anterior visceral endoderm (AVE) and the anterior visceral mesoderm (AME) which derive from the anterior primitive streak play critical roles in the formation of anterior structures. Interestingly, both of these regions express high levels of Wnt antagonists such as DKK, SFRP and CER1 (Biben et al., 1998; Glinka et al., 1998). Consequently, ectopic expression of canonical Wnts has been shown to suppress the formation of anterior structures, along with inducing partial axis duplication in mice. These experiments indicate critical roles for AME and AVE factors in suppressing local Wnt signaling for normal anterior structure development (Marikawa, 2006). Further, anterior involvement implicates canonical Wnts as inducers of the neural crest, promoting its formation, closure and dispersal throughout the embryo that eventually gives rise to most of the nervous system and the cranial skeleton (LaBonne, 2002). In addition to the early events in embryonic development, canonical Wnt signaling is required for early stages of mammalian osteoblastogenesis (Day et al., 2005), late stages of brain (Thomas and Capecchi, 1990), kidney (Herzlinger et al., 1994), tooth, and mammary gland development by exerting its influence upon proliferation, cell migration and differentiation similar to events surrounding gastrulation. It is through combinations of ligand expression patterns, agonists and antagonists vying for the ability to bind both ligands and their receptors and in combination with a variety of other embryonic factors, that Wnts contribute to the precise temporal and spatial patterning of the developing mouse embryo 1.3 Wnt signaling in human disease Developmentally important signaling pathways like that of Wnts, which coordinate diverse processes such as cell migration, proliferation, adhesion and death, play significant roles not only in development but also in tissue homeostasis (Clevers, 2006). When a critically important pathway such as Wnt becomes dysregulated, it predictably leads to a variety of pathological conditions. These conditions range from various types of cancers (Polakis, 2000), to multiple diseases related to unbalanced tissue homeostasis (Krishnan et al., 2006). The actions of these

19 11 diseases are mediated mostly through the canonical branch of the pathway and the number of Wnt-induced proliferative and or anti-apoptotic genes Wnt signaling in Cancer The best studied of diseases involving Wnt signaling relate to its role in a variety of cancers (Barker and Clevers, 2006; Giles RH, 2003; Polakis, 2000). Most evident are the implications within the intestinal epithelium in colorectal cancer (CRC) where mutations of key pathway components appear very early in adenoma development in approximately 90% of human CRC s. Mutational hot-spots located in APC account for 80% of inherited and random cases of colon cancer, while mutations in β-catenin are seen in 9% of such cases. Additional Wnt pathway components are mutated at a lesser frequency, yet all result in the stabilization of β-catenin, promoting constitutive activation of canonical Wnt signaling (Barker and Clevers, 2006; Giles RH, 2003; Polakis, 2000). Familial adenomatous polyposis (FAP) is an autosomal, inheritable disease predisposing patients to develop thousands of colon polyps typically progressing to carcinoma by early to mid adulthood. The majority of FAP associated mutations reflect heterozygous truncations of the APC gene and are associated with a subsequent loss of heterozygosity (Galiatsatos and Foulkes, 2006). This phenotype is mimicked in the Apc min mutant mouse heterozygous for a truncated form of APC, in which mice readily develop both polyps and colon carcinomas (Moser et al., 1995). Canonical Wnt signaling is required for maintaining epithelial homeostasis within the intestinal tract. The colon is comprised of both villi (protrusions) and invaginations (crypts). The basal crypt cells are highly proliferative cells that give rise to multiple cell types lining the intestinal tract. Canonical Wnts exert a proliferative effect on these crypt progenitor cells through the regulation of Wnt target genes c-myc and CyclinD1 (Gregorieff and Clevers, 2005). Both factors are highly overexpressed in cases of both FAP and colon cancer. Studies aimed at blocking their expression in CRC cell lines have indicated their cell proliferative effect can be mitigated and cellular differentiation programs induced via overexpression dntcf4 (van de Wetering et al., 2002). The precise mechanism by which Wnt signaling regulates the proliferative homeostasis of these cells remains a mystery. In the colon, however, it is believed that adenoma formation is promoted through Wnt induced unabated expansion of progenitor

20 12 cells which somehow retain their crypt phenotype, yet continue to expand outside the intestinal invaginations (Gregorieff and Clevers, 2005). In addition to the multiple examples of activating mutations of Wnt signal transduction components, recently, a variety of epigenetic regulatory mechanisms have been noted in cases of colon cancer. A number of studies have reported hypermethylated promoters of secreted Wnt ligand genes such as WIF1 (Taniguchi et al., 2005), DKK1 (Aguilera et al., 2006) and SFRP s (Suzuki et al., 2004) in multiple colon cancer cell lines and patients. These factors all act as inhibitors of Wnt signaling in the extracellular matrix where they either sequester Wnt ligands via direct binding or prevent Wnts from binding their target receptor. In effect, hypermethylation lessens the ability of these factors to quell an already hyperactive Wnt signal in many instances of colon cancer. A common theme within the Wnt field surrounds the related effects of Wnt in cancer and cellular or tissue self-renewal. While this theme is best characterized in the intestine, it is also evident in hair follicle tumours, and in cases of leukemia. Hair follicle tumours are the first reported case of a Wnt inactivating mutation seen to be highly correlative with a tumor (Takeda et al., 2006). A significant percentage of follicular sebaceous tumours harbour a Lef1 mutation rendering it unable to efficiently bind β-catenin. The end result is a set of early progenitor cells being directed towards the wrong cellular fate in the absence of sufficient Wnt signal. Obfuscating the picture in follicular tumours are the cases in which activated Wnt signaling is seen to play a significant role in promoting tumor development (Lo Celso et al., 2004). This disparity implies a more complex role for β-catenin/wnt signaling in the determination or balance of follicular cell fate. This balance between cell-fate and cancer seen in the colon and hair follicles is also seen in leukemias, whereby hyperactive Wnt signaling is believed to enhance the self-renewal activity of CML granulocyte-macrophage progenitors leading to increased leukemic potential (Jamieson et al., 2004). Moreover, similar instances of activated canonical Wnt pathways are seen and implicated as effectors in cases of breast, lung, prostate, ovarian, thyroid and a variety of additional cancers (Lustig and Behrens, 2003; Mazieres et al., 2005; Nusse and Varmus, 1982; Tekmal and Keshava, 1997; Yardy and Brewster, 2005).

21 Other human diseases and Wnt signaling In addition to cancer, canonical Wnt signaling has been implicated in diseases as far ranging as Alzheimer s, bipolar disorders, skin diseases, cardiovascular diseases, type two diabetes and a limb development disorder (Holmen et al., 2004; Johnson and Rajamannan, 2006; Luo et al., 2007). However, the best documented case outside of cancer is that relating to skeletal disorders. This crucial role for canonical Wnt signaling has only been documented in the last decade, as Wnts play an essential role in post-natal bone tissue homeostasis. In adult life, bone homeostasis is maintained by the balanced activities of osteoblasts, which produce bone matrix and their cellular counterpart osteoclasts, which resorb the same matrix. Therefore, bone mass is determined by the tightly coupled relative activities of these two cell types (Bennett et al., 2005). Loss-of-function mutations of LRP5, the canonical Wnt co-receptor, are associated with osteoporosis-pseudogloma syndrome, which is characterized by a loss of bone density, hence skeletal fragility (Krishnan et al., 2006). Conversely, gain-of function mutations in the LRP5 N-terminal domain which reduces its affinity for the Wnt antagonist DKK1 confers a high bone mass phenotype (Boyden LM, 2002). The bone density phenotype in LRP5 null mice is exacerbated by the heterozygous loss of LRP6, adding credence to the notion that the entire LRP family of receptors critically influence bone mass through canonical Wnt signaling (Holmen et al., 2004). While the molecular mechanisms by which Wnts influence bone mass are not fully understood, GSK3β inhibition was seen to stimulate precursor cells to differentiate into osteoblasts (Bennett et al., 2005). Additional studies have demonstrated stabilized β-catenin and Wnts 3, 1 and 10b, all canonical Wnts, induce osteoblastogenesis strongly indicating Wnts stimulate bone formation via directing cells towards an osteoblastic cell fate (Krishnan et al., 2006). 1.4 Small molecule screening Sequencing of the human genome has provided researchers with a multitude of new biological targets to study based on the approximately 25,000 genes likely to encode millions of protein products. Of these proteins, fewer than 1000 are currently targeted by small molecules (Drews,

22 ), and are predominantly pharmaceutical targets. The popularity of small molecule screening is likely linked to the abundance of information provided by the genomics revolution. New screening technologies and improved small molecule libraries have the potential to accelerate the transition of genomic data into biological or therapeutic significance (Austin, 2003). Used as a complement to traditional research, small molecules have a number of advantages; they target a gene product rather than mrna or a genetic locus, can be agonistic or antagonistic, and they can selectively affect their target for a defined period in vivo and in vitro. With virtually limitless structural diversity, there are conceivably specific agonists and antagonists for every protein. Therefore, by screening chemical libraries against a variety of assays, it is possible to identify specific effectors and develop small molecules as pharmaceuticals and as research tools. Conversely, it is also possible to screen chemicals with known molecular properties to dissect the function of novel genes and cellular pathways Drug discovery The process of drug discovery involves biological target identification, assay development, and high-throughput screening followed by identifying the mechanism of action and hit-to-lead validation. Most molecules in the drug discovery process fail due to lack of potency, toxicity or unsound biological targeting (Caldwell, 2007). The benefits, however, of being able to screen a quality drug target against thousands of potential chemical compounds are invaluable and have increased with the availability of drug-like compound libraries and robotics capabilities (Goodnow, 2001). High-throughput screening, therefore, has become commonplace in industry and increasingly so in academia as a means of drug discovery and as a tool to address key biological questions. Much like biological research itself, these screens take one of three distinct approaches; cellbased, in vitro, and in silico. All possess distinct benefits and limitations. For example, in vitro screens are predominantly used when a specific molecule of interest is directly targeted. In vitro assays provide researchers with direct readouts of activity or function of the drug target, and can be easier than cell-based screens. With an in vitro approach, however, it is not a certainty

23 15 whether similar effects will be duplicated in vivo, nor whether the compound is cytotoxic, or has multiple off target effects. Cell-based screens bypass two of these issues. With the proper controls, cytotoxic small molecules can be easily identified or screened for (Zaman, 2004) and in vivo effect are not in question. However, cell-based screening is not without problems. False positive rates are significantly higher, due to off target effects even in the best designed screen (Zaman, 2004). However, the biggest advantage of cell based screens is the ability to perform pathway and phenotypic screens. These screens allow researchers to address greater biological questions through a broader range of applications. The third approach, in silico screening, also has broad applications. In contrast to both in vitro and cell-based screening, in silico screens draws information from previous screens and literature to make computer generated predictions about compound effects on a particular protein. Structural modeling is used to generate inhibitors/activators of enzymatic activity or of protein-protein interactions. In silico screens, however, are limited to proteins with solved structures or with known small molecule effectors as a starting point (Shan et al., 2005). Regardless of screen type, of principal screening importance is identifying a proper target from the onset. Particularly more so when screening for anti-cancer drugs, as their properties are often toxic to normal tissues as well as the cancerous target cell population. Coupled with cancer being a diverse disease due to environmental factors as well as patient genetic variability, a screening target molecule or pathway must be very sound from the onset. This does not rule out the possibility of targeting a component or pathway essential to general cell survival, because the host of secondary issues these targets bring with them that can be satiated through appropriate dosing and directed drug distribution (Kamb et al., 2007). Furthermore, using a cell-based assay for drug screening can be advantageous in weeding out compounds with more general cytotoxic effects. An equally important part of the screening process is the development of a high-throughput readout to measure the activity of the target molecule or pathway. The recent advances made in cell-based screening technology including automated systems for cell manipulation, small molecule delivery tools, signal detection reagents and apparatus, along with better chemical libraries have all made cell-based small molecule implementation increasingly more reliable and viable.

24 16 The most difficult and time consuming aspect of the drug discovery process is not the screen itself, but the subsequent work validating the identified hits. The first goal is turning screened hits into lead chemical compounds (Goodnow, 2001). Small molecule leads need not only have a verified effect, but their cellular targets, and mechanism of action need to be identified in order to proceed to optimization. Target identification needs to be determined in phenotypic and or pathway screens, as opposed to targeted enzymatic screens in which the target is already known. While genomic and proteomic tools have been in place for a long time in yeast to more easily identify target molecules, these tools are not fully adaptable to mammalian cells (Luesch, 2006). Therefore other than coupling a small molecule screen with a genomic screen (sirna), a large amount of secondary work is necessary for target and/or mechanistic identification. After target identification, lead compounds typically go through an optimization stage in which their structure is refined through medicinal chemistry to produce a clinically viable candidate lead (Goodnow, 2001). From there, the long process of enabling their entry into animal and then human studies is started, with the vast majority of leads failing due to lack of potency, toxicity or specificity (Goodnow, 2001). It is precisely due to this high failure rate that a screens need to be designed for a sound druggable target, and hit validation and optimization be thorough and comprehensive Small molecule libraries As important as it is to design the proper screen against a well researched target, hits will only generate quality lead compounds if the chemical library contains compounds with biological activity. Medicinal chemistry has traditionally favored particular scaffolds from which drugs are derived. Therefore, molecules in a screening collection typically adhere to conventional rules for drug-like properties, taking into consideration solubility, H-bond donating and accepting groups, and size restrictions (Lipinski, 1997). The advantage of these chemical properties is considerable, as they not only predict drug activity, but also demonstrate adequate physiological absorption, distribution, metabolism, and excretion profiles (Yu and Adedoyin, 2003). Therefore, they are ideal compounds for post-screening purposes. The measure of chemical space is equally as important as the aforementioned properties. A chemical library designed to randomly populate chemical space is not of significant biological

25 17 use if chemical and biological space does not overlap (McMillan and Kahn, 2005). Therefore, compound libraries such as Maybridge (Thermo Fisher scientific), which are designed using core structures common to medicinal chemistry as scaffolds, from which diversity and variability of chemical space is explored, are ideal for drug screens. Moreover, a library of this type is best suited to identify novel modulators of a cellular target while also adhering to typical drug-like properties. Libraries such as the LOPAC (Sigma) and Prestwick (Prestwick chemicals), which consist of biologically active compounds and off-patent drugs, are not likely to produce selective inhibitors of a particular pathway of interest owing to their activity towards other cellular targets. However, these libraries are of chemical genomic interest, and can identify interactions or commonalities between different signaling pathways Chemical genomics In parallel with the drug discovery process is the emerging field of chemical genomics. Forward chemical genetic screens are identical to drug based screens, but differ in their end goals. While drug discovery aims to generate lead medicinal compounds, chemical genomics aims to identify a small molecule which can produce a particular phenotype (Lokey, 2003). In a manner very similar to forward genetics, chemical genomics seeks compounds possessing properties able to mimic a particular genetic mutation by altering the effects of one or more gene products (Bellows and Tyers, 2004). In many situations, the molecular targets or phenotypes studied through chemical genomics screens are the ones also of interest for drug discovery. At the core of excitement surrounding chemical genomics, are its wide-ranging application. Once a screen has been used to identify a chemical modulator, and its specificity of action upon a molecular target is verified, the small molecule can then be used as a tool to dissect the biology of the molecular target, its interactions, and its in vivo role. Moreover, the simplicity of use gives it an advantage over the myriad of genetic tools that can be used in parallel Known small molecule modulators of the Wnt pathway Dysregulated Wnt signaling was first linked to colon cancer in the late 1990 s (Korinek et al., 1997), and since then, a significant amount of research from both private and public sectors has

26 18 investigated that link. Therefore, as expected, studies of purported small molecule antagonists of canonical Wnt signaling have emerged (Fig. 2 and Table 1). The first inhibitor reported was nitric oxide donating aspirin compounds already in drug testing as Cox2 inhibitors (Nath et al., 2003). Subsequently, additional compounds such as hydrogen peroxide (Shin et al., 2004), Green tea compounds (Kim et al., 2006) and Quercetin (Park et al., 2005) were also reported as inhibitors. However, a mechanism for the activity of these and most other inhibitory compounds remains largely undefined. The largest class of compounds demonstrating an antagonistic effect on canonical Wnt signaling is the NSAIDS. Included in this category are Aspirin, the NO-donating forms of Aspirin, and sulindac sulfide. All have long been used for the treatment of pain and inflammation. The mechanisms by which they affect Wnt signaling are varied. Aspirin is believed to inhibit PP2A activity (Bos et al., 2006), while NO-donating aspirin have been suggested to disrupt the β- catenin/tcf complex in the nucleus (Nath et al., 2003), and Sulindac was shown to inhibit nuclear β-catenin accumulation in FAP patient colonic cells through an undetermined mechanism (Boon et al., 2004; Koornstra et al., 2005). While these compounds have demonstrated an inhibitory effect upon the canonical Wnt pathway, the effects were seen at high concentrations; moreover, they are not specific to Wnt signaling. In addition to reports of already studied compounds, three groups have used small molecule screens in an attempt to identify novel compounds inhibiting canonical Wnt signaling. The first of these reports characterized the compound ICG-001, identified in a cell-based screen using a Wnt responsive transcriptional reporter, Topflash, as assay readout. This synthetic compound demonstrated an inhibitory effect on Wnt transcriptional activity through its binding to the β- catenin transcriptional partner CREB Binding Protein (Emami KH, 2004). A more directed approach was taken by Leproucelet et al., in which they screened a chemical library in an ELISA-based high throughput screen for inhibitors of β-catenin-tcf interaction (Lepourcelet et al., 2004). The screen identified six inhibitors with IC-50 values in the low μm range, all of which demonstrated in vivo effects on canonical Wnt signaling. The third screen was accomplished in silico, using a structure-based approach. Utilizing the crystal structure of Dvl binding protein Dapper, Shan et al. ran a virtual screen of 250,000 compounds to identify small

27 19 Figure 2. Known Chemical inhibitors of the Wnt signaling pathway. Small molecules with antagonistic Wnt pathway activity are displayed at their molecular targets. Four of the six known inhibitors are downstream effectors of the pathway acting at the transcription level. Additional modulators, including agonists are listed in Table 1.

28 20 molecules which would bind Dvl and interrupt its association with the Frizzled receptor (Shan et al., 2005). One such molecule was identified and demonstrated an in vivo effect upon Wnt signaling, however, its effects were only seen at a very high concentration.

29 21 Table 1. Known small molecules modulators of the Wnt pathway. A number of smallmolecules have demonstrated the ability to effect β-catenin mediated Wnt signaling. While some molecules were identified through high-throughput screening as indicated above, most have been identified by alternative methods. All known Wnt pathway modulators are reported along with the associated IC50 value (mm) and the molecular target, if identified. ND indicates no data is available.

30 Rationale for project Due to the importance of Wnt signaling in human disease, dysregulated β-catenin constitutes an attractive target for drug intervention. However, in addition to anti-cancer therapy, the Wnt pathway plays a critical role in animal development. The few functional inhibitors in existence are downstream effectors of transcriptional activation. Therefore, identification of novel upstream small molecule inhibitors/activators would be of tremendous importance as a research tool to elucidate the mechanisms involved in regulating β-catenin stability, and thus Wnt signaling. My research endeavor therefore centered on the development and implementation of a highthroughput screen to assess the status of β-catenin activity in mammalian cells. Using a luciferase tagged β-catenin molecule to detect its stabilization, we screened the Maybridge, LOPAC and Prestwick chemical libraries to identify small molecule modulators of canonical Wnt signaling. Compounds identified as inhibitors or activators of the pathway were subsequently investigated via β-catenin stabilization assay, and pathways specific reporter assay, to assess repeatability and pathway specificity. The effects of the most promising inhibitory compound was tested in colon cancer cell lines and was shown to significantly attenuate activated β-catenin signaling via quantitative PCR analysis. Furthermore, through dissection of the Wnt signaling pathway, we were bale to pinpoint the effect of the compound upon β-catenin stability, through a currently undefined mechanism. Lastly, the inhibitory effects of this compound and ones of significant structural similarity were shown to not only inhibit Wnt target gene expression in colon cancer cell lines, but also effect prostate cancer models.

31 23 2 Materials and methods 2.1 Reagents The Lopac 1280, Prestwick and Maybridge libraries housed at the S.M.A.R.T Robotics Facility at the Samuel Lunenfeld Research Institutes ( were purchased from Lopac Chemicals, Prestwick Chemicals and Maybridge Chemicals respectively. Aliquots were stored at C in 96 and 384 well storage plates (Costar, Corning). Individual compounds used in secondary analyses were obtained from Ryan Scientific, dissolved in DMSO (Sigma) and stored in small 50 mm aliquots at C. 2.2 Cell lines and maintenance All cell lines were maintained in media according to American Type Culture Collection guidelines ( supplemented with 10% FBS (Hyclone), incubated at 37 0 C with 5% CO 2 and went without additional additives unless otherwise indicated. Briefly, SW480, SW620, LS1034, LS174T and HepG2 cells were cultured in alpha-mem. HepG2 cells also received non-essential amino acid supplementation (Gibco). The prostate derived cell lines Du145 and PC3 were maintained in regular MEM. Colo205 cells were sustained in RPMI media, while L-cells, HEK293 and HEK293T cells were cultured in DMEM. Wnt3A expressing L-cells (Labbe E, 2000) were maintained in DMEM containing 0.4 mg/ml G418 (Gibco). The Flag-β-catenin-Luciferase stable cell lines were generated by transfecting HEK293 cells with pcagip-flag-β-catenin-ff-luciferase (see below) using calcium phosphate transfection (see below) in DMEM. Cells stably expressing the fusion protein were selected with 1.5 μg/ml puromycin (Sigma) by ring cloning after one week. Early passages were frozen down and expanded when needed.

32 Production of Wnt3A ligand in conditioned media Mouse fibroblast L cells stably expressing the murinewnt3a gene were generated by Dr. Etienne Labbe using the ppgk-neo-wnt3a construct (Shibamoto et al., 1998). Cells were plated in 10 cm dishes and allowed to grow to 70% confluence, after which, the media was changed for ligand collection. Both control and Wnt3A conditioned media was collected from cells cultured for five days in DMEM supplemented with 0.2% fetal bovine serum. After collection, the media was filtered through a 0.22 μm bottle top filter (Corning) and stored at 4 o C for up to four weeks without significant loss of activity. Confirmation of ligand activity was performed on each new batch using Topflash transcriptional reporter assays to assess Wnt transcriptional activity and immunoblot analysis to detect stabilization of endogenous β-catenin in L-cells (see below). 2.4 Constructs, transcriptional reporter assays and immunoblotting Topflash and Fopflash reporter constructs were originally obtained from the Vogelstein Lab and are the same as those previously used in our lab (Labbe E, 2000). IBRE-lux (Benchabane and Wrana, 2003) and 3TP-luc (Wrana et al., 1992) reporters were obtained from the Wrana lab, and the NFκB reporter is from Upstate Biotechnology. Wnt pathway components, Dvl2, β-catenin, Lef1 and LRP6 all possess a C-terminal triple-flag-epitope-tag and were cloned into the pcmv5 vector by Dr. Bryan Miller. The Flag(N-terminal)-β-catenin-firefly-luciferase(C terminal) fusion construct was excised from a pcmv5b construct generated by Dr. Letamendia and cloned into the ClaI and BglII sites of the pcagip vector, which contains a Puromycin resistance cassette used to generate stable cell lines. For transcriptional reporter assays, cells were transiently transfected with a reporter plasmid specific to the Wnt, TGF-β, BMP or NFκB pathways, pcmvβ-gal, and any indicated constructs. To induce the luciferase reporters, cells were treated overnight with the combinations of ligand and compound as indicated. Luciferase activity in cell lysates was measured using the luciferase assay system (Promega) in the EG&G Berthold microplate luminometer and normalized to β-gal levels. HEK293T and HepG2 cells were transiently transfected using calcium-phosphate DNA precipitation. Briefly, cells plated in 24-well dishes were given a fresh complete medium change

33 25 2 hr prior to transformation. DNA (3.0 μg/6 wells) was added to 15 μl of 2.5 M CaCl 2 mixed with 150μl of 2 HBS (280 mm NaCl, 50 mm HEPES, and 1.5 mm sodium phosphate [ph 7.05]) and incubated at room temperature for 20 min. The precipitate was added to cells, which were incubated overnight (Labbe et al., 1998). Colo205, SW620 and SW480 cells were also transfected with a total of 0.5μg cdna per individual well of a 24 well plate, using 1 μl Lipofectmine (Invitrogen) according to standard protocols. After six hours, the medium was changed for overnight incubation. The following day, cells were treated with compounds and the assay was performed as described above. To determine endogenous β-catenin protein levels, lysates were prepared from cells plated in 12 well plates, treated with combination of Wnt and/or compound. Proteins from cell lysates were separated by SDS PAGE and transferred to nitrocellulose membranes. β-catenin was detected using the anti-β-catenin antibody (1:10,000, BD Transduction laboratories) and HRP conjugated secondary antibody via chemiluminesence as recommended by the manufacturer (ECL kit, Amersham). Actin levels were detected using an anti-actin (1:2000, Sigma) antibody. In the manual Ff-luc assay, stable 7 cells generated to express the flag-β-catenin-fireflyluciferase fusion construct (see above for details) were manually plated into 48-well plates (Greiner-Bio-One) at a density of 2500 cells/well and incubated for 24 hours. To the cells, media containing the compounds was added. After a one hour pre-incubation with compound, Ligand or control media was added and incubated for an additional 15 hours. Media was removed, cells were lysed and luciferase activity measured using the luciferase assay system (Promega). 2.5 High-throughput assays For the β-catenin nuclear translocation assay, NIH3T3 cells (5000/well) plated in 96-well flatbottom plates (Costar, Corning) were treated with control or Wnt (50/50) conditioned media. After a sixteen hour incubation, cells were washed twice in PBS prior to being fixed in 4% paraformaldehyde (in PBS) for 10 minutes, washed twice, permeabilized with 0.5% triton X-100 (90 seconds), and washed an additional two times (PBS). Cells were prepared for immunofluorescence by incubating with the mouse monoclonal anti-β-catenin antibody (1:1000,

34 26 BD Transduction Laboratories) for one hour, followed by a FITC conjugated secondary antibody (Jackson ImmunoResearch) and Hoesct nuclear staining for one hour. A Cellomics Arrayscan II HCS imager (Cellomics) was used to image cells per well from each 96-well plate. The Arrayscan V3.5 software captured images using a 10x lens and an XF100 filter. The molecular translocation application on the Arrayscan was configured to define the nuclear (Hoechst stained) and cytoplasmic cellular regions and to quantify the FITC fluorescent intensity difference of β-catenin in these compartments defined as (mean nuclear intensity-cytoplasmic intensity). These values were calculated for each cell with averages generated for individual wells. For the automated Ff-luc assay, stable 7 cells previously generated to express the flag-β-cateninfirefly-luciferase fusion construct (see above for details) were manually plated into 96-well plates (Greiner-Bio-One) at a density of 1250 cells/well and incubated for 24 hours at 37 0 C plus 5% CO 2. One hour prior to initiating the run, the medium was replaced with 50 μl of a starvation media containing 0.2% FBS via manual pipettor. All plate handling thereafter and reagent additions were performed by an integrated arm robotics platform (CRS robotic arm controlled by the Polara software, Thermo Electron). Chemical aliquots from the libraries were added to cells for a final assay concentration of approximately 1.2 μm and a final DMSO concentration of 0.2% (compound solvent) via the Multimek automated pipettor (Beckman Coulter) pinning apparatus. Control wells, treated with solvent alone were in the first and last columns of each plate for control purposes. Following a one hour pre-incubation with the compound at 2x final assay concentrations, 50μl of Wnt or control ligand was added to each well by the multidrop dispensor (Thermo Electron). After a 16 hour compound treatment, 50μl of a luciferase substrate/lysis reagent (Promega Bright-Glo) was directly added to each well via the multidrop dispensor and shaken for 10 minutes. Luciferase activity in each well was measured by the CLIPR (Molecular Devices), a high-throughput luminometer system set to quantify the luciferase activity for all wells on a plate in a 30 second read. The run was performed in duplicate, once with compound treatment and Wnt3A ligand, followed by compound and control media treatment. For consistency, Wnt-stimulated and unstimulated runs were executed together with the same pool of reagents.

35 Real time quantitative reverse transcription-pcr analysis Total cellular RNA was isolated from cells cultured in 35mm dishes (BD-Falcon) using RNeasy Mini Kits (Qiagen) according to standard procedures. Briefly, media was aspirated and cells washed with PBS, cells were then lysed and harvested with a cell scraper. RNA was then isolated according to kit instructions. RNA samples were quantified on a spectrophotometer (Eppendorf) and 6μg of each was treated with 3 units of DNase1 (Fermentas), and reverse transcribed using random hexamers (2 μl of 100 μm stock) and 400 units of RevertAid H Minus M-MuLV Reverse Transcriptase (Fermentas). Real-time PCR was performed using the SYBR Green PCR master mix (Applied Biosystems) on the ABI Prism 7000 or 7900 sequence detection system (Applied Biosystems). Primer pairs used at 100 nmol/l were designed and previously reported by Dr. Etienne Labbe (Labbe et al., 2007), and validated against glygeraldehyde-3- phosphate dehydrogenase (GAPDH) by comparative standard curve analysis. Relative quantitation was calculated by the ΔΔC t method normalized to GAPDH levels (docs.appliedbiosystems.com/pebiodocs/ pdf).

36 28 3 Results 3.1 High throughput assay development Development of a high-throughput robotics-based screen requires an assay with high reproducibility, efficiency and the appropriate sensitivity to chemical compounds. To identify small molecule inhibitors of the canonical Wnt pathway we developed and tested two assays to evaluate Wnt signaling. These included an immunoflourescence-based microscopy assay directed at quantifying β-catenin nuclear translocation and a luciferase-tagged β-catenin molecule to measure β-catenin stabilization Beta-catenin nuclear translocation assay Activation of the Wnt pathway induces the nuclear translocation of β-catenin (Polakis, 2000), thus the first assay tested was an immunofluorescent-based assay to measure the nuclear translocation of β-catenin in Wnt stimulated and untreated samples. For this assay, Wntresponsive NIH3T3 cells seeded at a density of 12,000 cells per well were plated in a 96-well dish and treated with Wnt3A containing cell culture media. For this and all other assays, Wnt3A containing media was generated by collecting media from mouse L cells stably expressing and secreting the Wnt3A ligand, with the appropriate control media from non-wnt3a transfected cells collected in parallel for the assay. In brief, the Articulated arm robotics system at the Samuel Lunenfeld Research Institute was used for ligand treatment, fixing, washing and staining of endogenous β-catenin using an anti-β-catenin primary and FITC-conjugated secondary antibody. A Cellomics Arrayscan II apparatus was used to visualize and quantify multiple wells in 96 well plates. The Arrayscan V3.5 molecular translocation software is configured to define the nuclear (Hoechst stained) and cytoplasmic cellular regions and to quantify the fluorescent intensity difference of β-catenin in these compartments. By counting cells per well, β- catenin nuclear accumulation was quantified in response to Wnt stimulation in the presence or absence of small molecule inhibitors.

37 29 The trial runs showed a clear difference in nuclear localization between the treated and untreated samples (Fig. 3), however, the variability was consistently too high for high-throughput screening (HTS) purposes. The Z-values, which are a statistical function indicative of the feasibility of an assay for HTS ranged from , which is below the threshold of 0.4 considered to be a feasible assay. Use of higher ligand concentrations or other cell lines did not improve the Z-values (data not shown). In summary, variability in the treated samples from multiple tests was consistently too high for a viable screen, even when run in duplicate, and was not further pursued.

38 30 Figure 3: The β-catenin nuclear translocation assay displays high variability. NIH3T3 cells were treated with Wnt3A or control media, fixed, washed and stained with a b-catenin antibody on the robotics platform. Nuclear and cytoplasmic subcellular regions were defined through staining patterns and the difference in β-catenin fluorescent intensity between these compartments was quantified using the Cellomics Arrayscan II apparatus. A) Raw data collected during a run from all six plates is plotted collectively, and indicates considerable variability amongst the Wnt treated samples. B) Data from the best (plate 1) and worst (plate 4) plates in the run are plotted with their associated Z-factor values. Z-factor values, which are a measure of assay viability are substantially under the threshold of 0.4 considered to be a viable assay C) Z-factor equation, which accounts for the dynamic range between control and experimental values, and the variability within the data set. Four parameters are needed to calculate Z-factor; the mean (m) and standard deviation (s) of both the control (c) and sample values (s).

39 31 Figure 3

40 Firefly luciferase β-catenin stabilization assay (Ff-luc) As the stabilization of β-catenin is the key event in dysregulated Wnt signaling, I next developed a method to measure β-catenin protein levels by tagging β-catenin with a firefly luciferase protein (Fig. 4A). The luciferase tag was fused C-terminally to human β-catenin in the pcagip plasmid under the control of a β-actin promoter. Multiple clones in two mammalian cell lines (HEK 293 and L s) stably expressing the fusion protein were isolated via puromycin selection and were tested for Wnt induced increases of luciferase signal, indicative of β-catenin stabilization. Most of the HEK 293 cell clones displayed roughly a two-fold increase in luciferase activity in response to Wnt treatment (Fig. 4B), while those obtained from L cells were less responsive (data not shown). Subsequent testing showed that HEK293 clone number 7 gave the most reproducible results, and a signal sufficiently high to be amenable for HTP detection and was selected for high-throughput screening. The cell density that yielded maximal β-catenin stabilization was next determined by seeding a varying number of cells (from ) in each well of a 96-well dish. The following day, media was aspirated, cells lysed, and the lysates were then measured for luciferase activity. A density dependent decrease in fold activation was observed (Fig. 4C). Thus, seeding of 1250 cells per well, roughly corresponding to 25% confluence, achieved a reproducible two-fold activation and was easily detectable with the CLIPR, a CCD camera used to measure the luciferase signal on the robotics platform. Further testing indicated that DMSO did not affect the viability of the cells, nor did it affect the readout (Fig. 4D) Ff-luc assay optimization Due to the limited (two-fold) dynamic range of the Ff-Luc assay, it was necessary to conduct a series of manual and then robotic runs to optimize and validate the HTP protocol. Thus a number of seemingly modest details of the assays were optimized to achieve best results. First, I tested a variety of different plate types and brands settling on Greiner Bio-One plates which allowed for easy robotics handling, permitted high cell adherence and the deep wells minimized glow contamination from adjacent wells. We varied the reading time on the CLIPR from seconds and established that 30 second reads were in the linear range of detection and gave a

41 33 Figure 4: Development of the β-catenin-firefly-luciferase (Ff-luc) stabilization assay. A) Schematic of the Ff-luc assay, which allows for high-throughput detection of b-catenin stability in human cell lines. B) In the presence and absence of Wnt3A, β-catenin levels were determined by measuring the luciferase signal of whole cell lysates in HEK293 clones stably expressing Ff-Luc-β-catenin. Most clones displayed a Wnt3A induced stabilization β-catenin. Clone 7 was selected for screening because it reproducibly displayed a robust Wnt3A stabilization and possessed a high signal intensity. C) The Ff-luc assay is influence by seeding density. Clone 7 cells were plated in 96-well dishes at the indicated cell densities and assayed for luciferase signal. A density of 1250 cells/well yielded a repeatable two-fold Wnt induction and a signal intensity high enough for detection on the robotics platform. D) Using the Ff-luc assay as readout, DMSO has no effect on assay signal or cell viability at concentration comparable to proposed screening conditions.

42 34 Figure 4

43 35 sufficiently high signal. In addition the ideal volumes of media, ligand and luciferase/lysis reagent were determined taking into consideration, cost, ease of use and reproducibility. The final established assay protocol involves manual plating of 1250 cells/well in a 96 well format, followed by a 24 hour incubation. One hour prior to initiating the run, the medium is replaced with 50 μl of media containing 0.2% FBS. Compounds are added by pinning DMSO soluble compounds to a final concentration of approximately 1.2 μm and then incubated for one hour prior to the addition of 50 μl of Wnt or control ligand overnight. On day three, 50μl of a luciferase substrate/lysis reagent (Promega Bright-Glo) is directly added to cells, the plate is incubated in a shaker for 10 minutes at room temperature and luciferase activity quantified using a CCD camera. The readout indicates Wnt induced stabilization of the β-catenin fusion protein (see Fig. 5). On each plate, the two outer columns are used for control purposes, with cells in one column treated with Wnt ligand and the other with control media, while the inner 10 columns are all treated with individual compounds. Prior to undertaking a HTS, it is essential to validate assay performance. However, in the case of Wnt signaling, upstream inhibitors were not available. As an alternative, we used an inhibitor of GSK3β, Indirubin-3 -monoxime (I3M, Sigma), which activates Wnt signaling, for proof of principle experiments (Polychronopoulos et al., 2004). I first confirmed compound activity in manual assays. I3M stabilized the β-catenin-luciferase fusion construct in a dose dependent manner in both Wnt treated and untreated samples (Fig. 6A). The stabilizing effect on β-catenin observed using the Ff-Luc assay was mimicked when endogenous β-catenin was examined by immunoblotting in a mouse fibroblast cell line (L cells) (Fig. 6B). To test for reliability and reproducibility of the HTP platform, we added I3M to a final concentration of 5 μm, which elicits signal stabilization roughly equivalent to Wnt3A treatment. Robotic runs showed minimal variability within I3M treated wells across plates, indicating accurate and reliable compound addition by the robotic platform (Fig. 7). Therefore, not only was it possible to identify chemical modulation of the pathway in the assay, the robotics protocol readout and compound addition steps were functioning as expected.

44 36 Figure 5: The Ff-luc assay. A) HEK293 cells (Clone 7 cells) stably expressing Firefly Luciferase tagged β-catenin were used to indentify compounds affecting b-catenin stability in the Ff-luc assay. B) Step-by-step flowchart of the Ff-luc assay. C) Raw data from plate 21 is shown for both the Wnt-treated and unstimulated runs. H7 denotes an inhibitory hit identified from the screen.

45 37 Figure 6: Validation of the Ff-luc screening protocol. A) Using the Ff-luc assay as readout, Wnt pathway agonist Indirubin-3'-monoxime (I3M, Sigma), a GSK3β inhibitor, was used to validate the screening protocol. Here, the small molecule I3M stabilizes the Ff-luc-β-catenin reporter in a dose-dependent manner. B) I3M is also shown to stabilize endogenous levels of β- catenin in mouse L-cells as determined by immunoblotting in a similar dose-dependent manner.

46 38 Figure 7: Validating the pipetting accuracy and chemiluminescent readout of the robotics platform. The robotics platform was programmed to add a 120 nl aliquot of concentrated Indirubin-3'-monoxime (I3M) using the Multimek pinning tool into alternating wells of four 96- well plates in the Ff-luc assay. The desired final concentration of (5 µm) is known to stabilize b- catenin similar to Wnt3A treatment. The Ff-luc assay was performed entirely on the robotics platform, and averages from the four different treatments across all four plates is displayed. The data from the robotic compound addition indicates low variability and consistency across the assay.

47 Pilot high-throughput Ff-luc assay run After having established a screening protocol, our next goal was to identify the ideal compound concentration critical for screening purposes. Identifying the proper screening concentration allows us to balance the search for compounds with high specificity at a low concentration while minimizing false negatives. Typical concentrations used in cell based screens range from 1-10 μm, therefore to determine optimal concentrations for our assay we first screened through the first 800 compounds (10 plates) of the Maybridge library at roughly 1.2 and 7 μm. Analysis of the 10-plate screen revealed that higher chemical concentration significantly increased variability in the assay. Average Z -values for the first 10 plates screened at the lower concentration was compared to for the same 800 compounds screened at the high dose. Therefore, all future screens were conducted using the lower (1.2 μm) concentrations, we expect this would enhance identification of more potent compounds. We next conducted a pilot screen using the first 4000 compounds of the Maybridge chemical library to assess the hit rate and reproducibility of the assay. For this, runs with and without Wnt ligand were performed together, while a replicate Wnt treated run was executed on a separate day. Results from the initial run (Wnt3A #1) of the first 4000 compounds (Table 2) showed 37 individual activating hits of the pathway, 25 of which were duplicated in the replicate Wnt treated run (Wnt3A #2), and 24 of these activators were also detected in the control run (Wo). The high degree of correlation amongst the three runs suggests a high rate of reproducibility for the screen. A similarly pattern of reproducibility was seem among inhibitory compounds. We identified fewer inhibitors with a total of 9 from the first Wnt3A screen; 5 duplicated in the second stimulated run with 4 being detected in all three runs. Furthermore, the 4 inhibitors common to all runs can be further broken down into those displaying an increased inhibitory effect in the Wnt treated samples, and those with similar levels of inhibition in both treated and untreated runs. One compound, 21H07, fit the profile of preferential Wnt inhibition. Using B-score analysis, the hit rate for activators from the initial run (Wnt3A #1) was 0.93%. Inhibitors, which were the more desired lead compound, had a lower rate of 0.23%. Based on these hit rates we predicted a total of 465 activating and 115 inhibiting lead compounds from the entire 50,000 molecule Maybridge library if we were simply to screen the library with one stimulated and one unstimulated run. Because the majority of the hits were duplicated in the

48 40 Table 2: Pilot screen hits indicate a reproducible Ff-luc hit rate. The pilot Ff-luc screen was performed on the first 4000 Maybridge compounds to assess the hit rate and reproducibility of the assay. For this, two duplicate Wnt treated runs were performed on separate days, while a single unstimulated run was run in conjunction with the first stimulated one. Hits (both activators and inhibitors) were defined by those points surpassing the 3 standard deviation threshold above or below the mean. The associated B-score for each compound is listed.

49 41

50 42 second Wnt treated run and considering screening costs, we concluded that a single stimulated run combined with an unstimulated run would be sufficient for the final screening procedure. Moreover, the 46 chemical modulators identified in the initial stimulated run (37 activators, 9 inhibitors), 30 were duplicated in the second run and 28 of these were identified in the unstimulated run. Consequently, a large increase in false positives is not expected to be very pronounced from using single runs, therefore, all subsequent runs were performed without a duplicate Wnt treated run. 3.3 Final screening results and data analysis Three diverse sets of compound libraries were available for our screen, the Maybridge, Lopac and Prestwick chemical sets. The Maybridge chemical library is a 50,000 molecule diverse library set, whose compounds possess typical drug-like properties. The Prestwick set (Prestwick chemicals) includes an 1120 molecule set of largely off-patent drugs and the 1280 molecule Lopac set contains drug compounds with some known molecular targets and properties Comparison of various statistical hit-selection methods As there is only a two-fold dynamic range in the Ff-Luc assay, the data analysis method would be critical to our hit selection process. To establish the most appropriate data analysis method, we focused on the Lopac library results due to its small size and the known properties of many of its compounds. The data from stimulated runs was analyzed using three distinct statistical methods; percent activity, fold over median and B-score. The percent activity measure is the simplest method, in which experimental data points (compound treated wells) are compared to control wells within the same plate. The data is converted into a percentage value, whereby the value of 100 indicates no effect and activators or inhibitors will show an increase or decrease relative to 100. The fold over median method is a measure of the compound treated wells with respect to each other on an individual plate, and does not take the control wells of the plate into consideration. The B-score is a more complex function which takes into consideration the experimental points (compound treated), yet also factors in plate location by relating the data

51 43 points to adjacent wells within a plate, thereby correcting for possible plate trends in a run (Brideau et al., 2003). The three data analysis methods for the Lopac library returned roughly the same number of hits for the Wnt3A treated run (Fig. 8). Fold over median (using a three standard deviation cutoff) identified 4 activators and 26 inhibitors, percent activity (also using three SD) showed 4 activators and 23 inhibitors, while B-score revealed 2 activators and 21 inhibitors. Notably, all the effectors, both β-catenin stabilizers and destabilizers indicated by the B-score method were hits in the other methods as well. Both fold over median and percent activity measures identified some hits that did not appear in the other methods. This analysis revealed two important conclusions. First, B-score appeared to be the most reliable of the methods as miscellaneous hits were apparently weeded out and secondly that irrespective of which statistical method is used to determine hits, most of the same hits are identified amongst all methods. This analysis confirmed that even though the dynamic range of the Ff-Luc assay is not very large, it is in fact a viable screen and variability within and between plates is best analyzed using the B-score method Summary of results from the Lopac library As reported above, B-score analysis of the Wnt stimulated run identified 4 compounds which stabilized the β-catenin-ff-luc construct and hence were possible Wnt pathway agonists, and 23 molecules that destabilized β-catenin and might work as Wnt antagonists (all listed in Table 3). Additionally, the unstimulated run displayed 15 inhibitors and 6 activators. Twelve of these inhibitors are common to both stimulated and unstimulated runs; therefore, 11 inhibitors were detected uniquely in the Wnt treated run (Fig. 9). These are the compounds that appear from screening results to possess a preferential inhibitory effect in the presence of Wnt treatment and are more likely to be Wnt pathway antagonists as opposed to general effectors of β-catenin stability. Interestingly, the inhibitory hit rate on average was about 4 fold higher that the activators.

52 44 Figure 8: Comparison of different statistical hit-selection methods. Fold over median, B- score and percent activity measurements were used to analyze raw data from Lopac library screen. A) Data from the Lopac library is depicted for each method of analysis, Fold over median, B-score and percent activity. The upper and lower lines mark the three standard deviation threshold we defined as a hit in the high-throughput screen. B) Summary of Lopac hits using the different methods of analysis, and those common amongst all three statistical methods of analysis.

53 45 Figure 8

54 46 Figure 9: Lopac library screening results. A) Summary of total hits and hit rate identified through B-score analysis of the 1280 molecules screened from the Lopac chemical library. Hits were defined by those points surpassing the 3 standard deviation threshold above or below the mean. B) Raw data plotted from B-score analysis of the entire screened Lopac run is depicted. Lines mark the 3 standard deviation threshold above and below the mean (middle line).

55 47 Figure 9

56 Summary of results from the Prestwick library The largely off-patent drug containing Prestwick library displayed a pattern similar to that of the Lopac compounds, where the vast majority of effecter molecules had a destabilizing effect on β- catenin. The inhibitory hit rate for the combined (stimulated and unstimulated) runs was 2.37%. Twenty seven destabilizers were identified in the Wnt treated run and 26 in the untreated run, with the vast majority (21) common to both (Fig. 10A and B). With only five activators identified in the Wnt3A treated run and three from the unstimulated run, it is interesting to note that only two are common to both screens, three are unique to stimulated while one is unique to the non-wnt treated run Summary of results from the Maybridge library Hits identified from the Maybridge chemical library would be particularly interesting as any inhibitors or activators identified would be novel and undocumented, unlike the Lopac and Prestwick sets. Of the entire 50,000 Maybridge set, 130 plates corresponding to compounds were screened. Based on the hit rates of activators (0.93%) and inhibitors (0.23%) from the initial 4000 Maybridge compound pilot screen, we predicted a total of 97 activating and 24 inhibiting lead compounds from the screened of the Maybridge set. The activator hit rate for the entire 10,400 compounds screened adhered roughly to the expected rates culminating in an overall activator hit rate of 1.52% or 158 compounds for the stimulated run. The unstimulated run displayed a higher activator hit rate of 188 compounds, 77 of which were unique to the unstimulated run. In contrast, the inhibitory hit rate increased significantly from the predicted 0.23% to 0.6%, consisting of 63 inhibitory compounds in the stimulated run and 0.44% or 46 compounds in the unstimulated run, with 40 hits unique to the stimulated run (Fig. 11 A and B). Of note, the Maybridge library displayed an approximately 2.5 fold higher ratio of activating versus inhibitory hits, an inverse of the pattern seen with the Prestwick and Lopac Libraries.

57 49 Figure 10: Prestwick library screening results. A) Summary of total hits and hit rate identified through B-score analysis of the 1120 molecules screened from the Prestwick chemical library. Hits were defined by those points surpassing the 3 standard deviation threshold above or below the mean. B) Raw data plotted from B-score analysis of the entire screened Prestwick run is depicted. Lines mark the 3 standard deviation threshold above and below the mean (middle line).

58 50 Figure 10

59 51 Figure 11: Maybridge library screening results. A) Summary of total hits and hit rate identified through B-score analysis from molecules screened from the Maybridge chemical library. Hits were defined by those points surpassing the 3 standard deviation threshold above or below the mean. B) Raw data plotted from B-score analysis of the entire screened Maybridge set run is depicted. Lines mark the 3 standard deviation threshold above and below the mean (middle line)

60 52 Figure 11

61 Total assay hits from the entire HTP screen Analysis of Wnt stimulated runs from all three libraries screened identified 120 inhibitors for follow up work. Of these, 57 were solely detected in the Wnt stimulated runs and an additional five of these displayed preferential inhibition upon Wnt stimulation. Similarly, there were a significant number of activators identified, 194 from the unstimulated runs alone. Eighty of these were detected only in the non-wnt treated run and are thus of more interest as activators. Therefore, there are a significant number of compounds which are worthy targets for secondary analysis (Table 3). 3.4 Investigating selected hits via secondary analyses Assessing the reproducibility of effects From the 13,080 screened compounds, we selected nine from Maybridge, three from Prestwick, and one from Lopac that inhibited β-catenin stability for further analysis. Nine randomly selected compounds were also used as comparative controls. Lead compound selection was based on a variety of criteria. Some were chosen for preferential inhibitory activity in the presence of Wnt stimulation, where others displayed similar affects in both ligand-treated and untreated screens. While all were hits based on statistical analyses, some compounds displayed potent destabilizing properties while others displayed more moderate effects. Compounds were first analyzed using a manual version of the Ff-Luc assay to assess the reproducibility of hit compounds using the same stable clone 7 cell line but in a 48 well format. Luciferase activity was measured subsequent to media removal and cell lysis, rather than using a homogeneous lysis as in the robotics protocol. Results from secondary analyses are summarized in Table 3. Of the fourteen compounds across three libraries displaying a destabilizing effect on β-catenin in the Ff-luc assay, only three were reproduced in the duplicate manual Ff-luc (Table 4; Fig. 12), indicating a false positive rate for the Ff-luc of approximately 80%. The apparent high false positive rate is unexpected given the significant reproducibility seen in the duplicate runs of the first 4000 Maybridge compounds. However, as only a small number of samples

62 54 Table 3: Summary of results from all compounds screened. Compounds from the three libraries screened were identified as inhibitors or activators based on the destabilizing or stabilizing effect in the Ff-luc assay. Hits are determined as points three standard deviations above or below the mean using B-score analysis. Data is presented as both totals and percent hit rate for Wnt-treated (Wnt3A), and untreated (Wo) high-throughput screening runs.

63 55 Table 4: Summary of secondary assay results for twenty-two selected inhibitors and random controls. Nine randomly selected controls (compound #10-18) and thirteen inhibitory compounds were selected from the three libraries screened. Compounds were tested for their effect in manual repeats of Ff-luc to assess reproducibility. And for effects upon specific signaling pathway transcriptional reporter assays to ascertain Wnt pathway specific effects. In signaling assays, HEK293T cells were transfected with the indicated pathway specific reporter plasmids, and treated overnight with compound and the appropriate ligand. Luciferase activity was measured and normalized with a β-gal control. Effects in the manual Ff-luc assay are reported as modulation of Ff-luc-β-catenin levels. Inhibitory or activating effects on pathway specific transcriptional regulation are indicated with directional arrows. Points in which no effect was seen are reported with a dashed line.

64 56 Figure 12: Re-testing of select compounds using the Ff-luc assay. Compound 21H7 was shown to destabilize the Ff-Luc-b-catenin construct. Other compounds displayed no effect or stabilized (compound 25A2) Ff-Luc-β-catenin. Manual repeats of the Ff-luc assay were preformed in 48 well plates at 0.5, 1, 2.5, 5 and 12.5 µm concentrations. Media was removed and luciferase values were measured from whole cell lysates in a luminometer.

65 57 (12% of total hits) were tested, this may not be a completely accurate estimate of false-positive rates. Of the randomly selected compounds not detected as hits during screening, none were seen to modulate β-catenin stability in the manual Ff-luc repeat, suggesting a low false negative rate Effects of hits on Wnt transcriptional activity and assessing pathway specificity Next, those compounds scoring positive in the screen were tested for effects on Wnt induced transcriptional activity in HEK 293T cells. Compound effects were monitored by transfecting the Wnt pathway specific luciferase reporter plasmid ptopflash. A construct containing a mutated TCF/LEF binding site, pfopflash, was transfected in parallel as control. Transfection efficiencies were corrected by cotransfection with a β-galactosidase-encoding reporter plasmid. Assays were performed using varying compound doses to facilitate determination of the effective dose range for each compound. β-catenin activated transcriptional activity from the Topflash reporter assay was abrogated by only five of the fourteen inhibitory compounds tested (Table 4). Moreover, when comparing to the manual Ff-luc assay, all three of the compounds shown to reproduce a β-catenin destabilizing effect also demonstrated an inhibitory effect upon Wnt mediated transcriptional activity (Table 4). Additionally, none of the randomly selected compounds not identified as inhibitors in the screen modulated the Wnt pathway. The compounds identified as inhibitors in the screen, manual repeats, or in Topflash assays, were next assessed for specific or preferential effect upon the Wnt signaling pathway. For this, we employed multiple pathway specific reporter assays, namely 3TP-lux, IBRE-luc and NFκB reporter plasmids that are responsive to the TGFβ (Transforming Growth Factor β), BMP (Bone Morphogenetic Protein) and IL1 (Interleukin 1) ligands, respectively. Only two compounds, 21H7 and 46D10, inhibited Wnt-dependent transcriptional activity, yet had minimal or no effect on the other pathways tested (Table 4). Therefore, in using these assays, we are able to assess properties of hit compounds and rule out general cell inhibitors (compounds 23H2, 25A2, and chelerythrine chloride) which display transcriptional effects on all pathways tested (Table 4). Compound 21H7 reduced Topflash reporter activity by 60% at 1μΜ concentration (Fig 13).

66 58 Figure 13: Compound 21H7 preferentially inhibits the Wnt pathway. HEK293T cells were transfected with the indicated reporter plasmids, and treated overnight with compound and pathway specific ligand. Luciferase values indicate pathway specific reporter gene expression. The inhibitory compound (21H7) displays a preferential inhibitory effect upon the Wnt pathway specific reporter Topflash assay in comparison to TGFβ, IL1 and BMP responsive transcriptional assays.

67 59 While only a 20% reduction of NFκB and BMP pathways was observed at this concentration. Therefore, these results indicate a preferential effect on the Wnt pathway. Although compound 46D10 (Table 4) displayed inhibitory properties, this activity was lost upon subsequent compound restocking and was not pursued further (data not shown). As the remaining inhibitory compounds affected other pathways tested, either promoting or inhibiting transcriptional activity indicative of non-specific effects (Summarized in Table 4), 21H7 was the only compound selected for further analysis. 3.5 Analysis of 21H7, a selective Wnt pathway inhibitor Compound 21H7 preferentially inhibits Wnt responsive Topflash activity over the non-responsive Fopflash We next assessed the effects of a wider range of 21H7 doses in a Topflash luciferase assay using Fopflash as control. Fopflash as previously reported is a construct identical to Topflash, except that the LEF/TCF binding sites are mutated and therefore the reporter is non-responsive to Wnt treatment. Topflash results indicate 21H7 displays a dose-dependent effect, having no activity at 0.1 μm, while at 0.5μM a 30% inhibition of transcriptional activity is observed. The effect levels out at approximately 80% inhibition at concentrations between 2.5 and 12.5 μm, and the IC 50 value is approximately 1 μm (Fig. 14). At inhibitory concentrations ( μm) in the Topflash assay, a preferential inhibitory effect in the Wnt treated samples (80%) is observed compared to the control (45%) treatment. Furthermore, analogous Fopflash experiments (Fig. 14) do not display the same differential effect between the Wnt treated and untreated data sets. In fact, the Fopflash data appears almost identical in both inhibitory and dose dependent effect to that of the untreated Topflash data. Analysis of the Fopflash and Topflash data combined strongly indicates that 21H7 is acting preferentially to block Wnt-dependent transcriptional activity.

68 60 Figure 14: 21H7 preferentially inhibits the transcriptional activity of Wnt3A treated cells. HEK293T cells were transfected with the Wnt responsive Topflash or non-responsive Fopflash reporter plasmids. One day after transfection, cells were treated with 21H7 or DMSO control and stimulated overnight with Wnt3A or control ligand. Data is presented as a percent of the control (DMSO) value. Preferential inhibition is seen in Wnt-treated Topflash data, but not with the non-wnt responsive Fopflash reporter.

69 H7 blocks Wnt-induced β-catenin stabilization Our screen was designed to identify molecules that effect Wnt-dependent β-catenin stabilization, thus we next examined the effects of 21H7 on endogenous β-catenin. For this, we utilized mouse fibroblastic L-cells, a line commonly used to measure Wnt induced β-catenin stabilization due to their low steady-state β-catenin levels and a pronounced Wnt3A induced stabilization of β-catenin. In multiple experiments, we found that Wnt3A treatment induces a steady rise in β- catenin levels for up to four hours at which time levels remained constant up to 16 hours. Therefore, we examined the effects of the compound at the four hour time point. As seen in Figure 15, cells stimulated with Wnt3A displayed a robust and dose-dependent increase in total β-catenin levels. The addition of 21H7 at low concentrations did not affect total β-catenin levels, but in the range of μm, dramatic destabilization of β-catenin and no effect on α-actin levels was observed. Similar results were observed with overnight treatment of both compound and ligand (data not shown), indicating the compound is effective in both short term and longer time frames. We noted that even at the highest concentrations, the compound did not restore β-catenin levels to that observed in controls. Nevertheless, these experiments indicate that 21H7 can significantly decrease endogenous β-catenin levels induced by the addition of Wnt ligand H7 inhibits Wnt dependent gene expression in colon cancer cell lines For a lead compound to have anticancer application, the compound should also demonstrate the ability to suppress the constitutive Wnt signaling typically observed in cancers. Colorectal cancer (CRC) cell lines contain a variety of activating mutations in key Wnt pathway components, typically APC or β-catenin, and in some cases both, that result in constitutively nuclear β-catenin and activation of Wnt target genes (Ilyas et al., 1997). Thus, we next utilized the Topflash/Fopflash reporter system to ascertain the ability of compound 21H7 to turn off constitutive Wnt signaling in CRC cell lines.

70 62 Figure 15: Compound 21H7 inhibits the Wnt3A induced stabilization of endogenous β- catenin levels in the presence of Wnt3A and increasing concentrations of compound 21H7. Cell lysates from mouse L-cells pre-treated with compound for 1 hour then stimulated with Wnt ligand for a four hour period were immunoblotted with anti-β-catenin and anti-β-actin to determine protein levels. The Wnt-induced stabilization of β-catenin is inhibited in a dose dependent manner by 21H7. The final compound concentration ranges from 0.1µM to 12.5 µm, while control lanes were treated with DMSO only.

71 63 Common amongst all CRC lines tested is the loss of Wnt induced transcriptional activation of Topflash signal (Fig.16 B), as a result of mutations that hyperactivate the Wnt pathway. SW620 cells harbor an APC truncation (Zhao et al., 2007), and upon compound treatment we detected a significant inhibition of Topflash transcriptional activity yet very little inhibition of the Fopflash construct (Fig. 16A), a pattern of preferential inhibition also seen with HEK293 cells (Fig 14). Although the pattern of preferential Topflash inhibition in β-catenin mutated Colo205 cells (Gayet et al., 2001) is seen, the effect was not as dramatic. However, in two additional cancer cell lines containing mutations in β-catenin (HepG2 cells) and APC (SW480 cells), the inhibitory effects on neither Topflash nor Fopflash reporters were detected. Together, this set of results indicates that 21H7 can inhibit Topflash activity in colorectal cancer cell lines harbouring diverse pathway mutations, however, penetrance of this effect appears to depend on cell background Expression of endogenous Wnt target genes is inhibited by 21H7 in CRC cell lines We next assessed the inhibitory effects of the compound on the Wnt3A induced expression of endogenous Wnt target genes in a range of colorectal cancer cell lines. Previous work in the lab (Labbe et al 2007) and other studies (Shtutman et al., 1999) have demonstrated Wnt-dependent induction of Axin2 and Cyclin D1, thus, we focused on those genes endogenous targets. Cells were treated with compounds or with DMSO as control for 16 hours, RNA was isolated and reverse transcribed, and relative gene expression levels normalized to GAPDH levels were quantified using Real-Time PCR. 21H7 induced a significant dose-dependent decrease in Axin2 levels in all CRC cell lines tested, though, the extent of inhibition varied between the lines. In the related cell lines SW480 and SW620 cells, which are heterozygotic for a truncated APC (Zhao et al., 2007), an 80-90% reduction in total Axin2 levels was seen (Fig. 17). Analysis of four other cell lines, each harboring different mutations in Wnt pathway components also revealed significant reductions in Axin2 mrna levels. Similar to SW480 cells, LS1034 cells have undergone APC loss of heterozygosity and possess a truncated form of the remaining allele (Sparks et al., 1998).

72 64 Figure 16: Differential suppression of Wnt-dependent gene expression in colon cancer cell lines harboring activating mutations in the Wnt pathway by 21H7. Cells transfected with individual reporter constructs were treated with the 21H7 and stimulated with Wnt3A or ligand control overnight. A) In a dose-dependent manner, 21H7 inhibits both Topflash and Fopflash activity. Data is presented as a percent of the control (DMSO) value. B) All colon cancer cell lines tested lack ligand responsiveness due to known hyperactivating mutations of the Wnt pathway.

73 65 Figure 16

74 66 Figure 17: Wnt target gene expression is inhibited by 21H7 in colon cancer (CRC) cell lines irrespective of the differing Wnt pathway mutations. Total cellular RNA was isolated from prostate cancer cell lines treated with 21H7 or DMSO control for a 16 hour period. The RNA was reverse transcribed with random hexamers and cdna s of established Wnt target genes were quantified by QPCR (using SYBRGreen in an ABI Prism 7000 sequence detection system). Levels of GAPDH cdna were also quantified and used to normalize results by comparative standard curve analysis. All six CRC cell lines possess activating mutations of key Wnt pathway components, as shown. Irrespective of their mutational status, all lines tested display a 21H7 induced decrease in mrna levels of established Wnt target genes, with differing magnitudes of effect.

75 67 Figure 17

76 68 Despite the similar mutational status, the reduction in Axin2 levels (roughly 50%) was not as robust as seen in SW480 and SW620 cells. The HCT116 and LS174T cell lines contain wild type APC, but possess β-catenin mutations at serine 45, a residue phosphorylated in a Wnt dependent manner (Gayet et al., 2001; Sparks et al., 1998). In HCT116 cells, this mutation consists of a heterozygous deletion of serine 45, while LS174T cells possess a homozygous serine to phenylalanine mutation. In addition to possessing similar mutations, HCT116 and LS174T cells displayed very similar dose-dependency with roughly 70% maximal reduction in Axin2 mrna levels (Fig.17). Colo205 cells displayed the lowest reduction of Axin2 mrna levels (45%) in the presence of 21H7. The Colo 205 cells possess compound mutations of the Wnt pathway, whereby neither APC nor β-catenin are wild-type. However, the mutation in β- catenin is an alanine to serine substitution at amino acid 287, which has not been examined for effects upon Wnt signaling. From the above data, it is clear that Axin2 mrna levels are affected by 21H7 in all cell lines regardless of their Wnt pathway mutational status. Moreover, the varied effect of inhibition between cell lines displays no clear pattern based the mutational status of the cells. CyclinD1 is another established Wnt target gene, though in general, Wnt dependent induction is not as robust as observed for Axin2 (Jho et al., 2002; Labbe et al., 2007; Tetsu and McCormick, 1999). Levels of CyclinD1 displayed a modest 21H7 induced reduction in most cell lines, for some lines, no change at was identified (Fig. 17). In addition, the effects on CyclinD1 levels do not appear to correlate with the mutational status of cells. LS 1034 and Colo 205 cell lines, which displayed the least reduction in Axin2 mrna levels, exhibit no significant decrease in total CyclinD1 mrna levels. The only cell line in which CyclinD1 and Axin2 levels were comparably affected, resulting in a maximal reduction of 70% in each case, was the LS174T line. In all other lines, 21H7 had a significantly more pronounced effect upon Axin2 levels than CyclinD1. Levels of CyclinD1 are regulated by a variety of other factors including NF- κb (Guttridge et al., 1999), STAT (Leslie et al., 2006) and Ras-dependent signaling (Gladden and Diehl, 2005), indicating the balance of CyclinD1 levels in cells is important to or affected by other factors. From the combined Axin2 and CyclinD1 data, it is clear that Wnt target gene mrna levels are affected by 21H7 in colorectal cancer cell lines regardless of their Wnt pathway mutational

77 69 status. The inhibitory effects did vary between cell lines and were significantly more pronounced for Axin2, however, the experiments indicate no clear pattern of inhibition based upon the mutational status of the cells H7 inhibits expression of endogenous Wnt target genes in prostate cancer cell lines. Excessive Wnt signaling has been implicated in a variety of cancers. Therefore, we next assessed the effects of 21H7 on the established prostate cancer cell lines, PC3 and Du145 cells, in which the mutational status of Wnt pathway components has not been examined by us or others. In both prostate lines tested, we identified a marked inhibition of the expression of both the Axin2 and CyclinD1 Wnt target genes (Fig. 18). This demonstrates that 21H7 can suppress Wnt transcriptional activity in both colon cancer and prostate cell lines. As Wnt pathway misregulation is prominent in a wide range of cancers including prostate (Yardy and Brewster, 2005), these indicate that 21H7 may have efficacy in a broad range or cancers Wnt signaling is inhibited by 21H7 at the level of β-catenin Having demonstrated the wide ranging and significant inhibitory effect of 21H7 on Wntdependent transcription, we next focused on examining where in the pathway 21H7 acts. While the Wnt pathway is becoming increasingly complicated, key steps can be viewed as a linear system, involving ligand-receptor binding, dishevelled activation, destruction complex dissociation, β-catenin stabilization, nuclear translocation and transcriptional activation (Fig. 19A). Therefore, to determine where 21H7 acts, we performed Topflash assays in the presence of individual Wnt-activating pathway components, with the goal of identifying whether the repressive activity of 21H7 occurs in their presence. Wnt signaling requires a Wnt induced Frizzled-LRP interaction (Holmen et al., 2005; Liu G, 2003). Interestingly, truncated forms of LRP omitting the N-terminal extracellular domain confers pathway activation by a yet undetermined mechanism (Mi and Johnson, 2005). Our data indicates that the inhibitory effect of 21H7 is not lost in the presence of LRP6ΔΝ, ruling out any

78 70 Figure 18: 21H7 inhibits Wnt target gene expression in prostate cancer cells. Total cellular RNA was isolated from prostate cancer cell lines treated with 21H7 or DMSO control for a 16 hour period. The RNA was reverse transcribed with random hexamers and cdna s of established Wnt target genes were quantified by QPCR (using SYBRGreen in an ABI Prism 7000 sequence detection system). Levels of GAPDH cdna was also quantified and used to normalize results by comparative standard curve analysis. 21H7 is able to decrease endogenous mrna levels of both Wnt target genes in the both prostate lines tested. The mutational status of APC and β-catenin has not been characterized in either cell line.

79 71 Figure 19: Epistatic analysis indicates 21H7 effects Wnt-dependent signaling at the level of β-catenin. HEK293T cells were transiently transfected with the Topflash reporter along with Wnt pathway activators LRP6, Dvl2 and β-catenin. A) Linear schematic of the canonical Wnt pathway noting the location of overexpressed components. B) 21H7 retains the ability to inhibit Wnt dependent transcriptional expressions in the presence of upstream activating components (LRP6 & Dvl2). C) In the presence of β-catenind91, a constitutively active mutation, 21H7 retains its inhibitory effect on Topflash activity and is therefore not inhibiting the Wnt signaling cascade.

80 72 effects on ligand binding or receptor interaction (Fig 19B). Likewise, the inhibitory effect appears to occur downstream of Dishevelled2 (Dvl2) as 21H7 still inhibited Topflash activity in a Dvl2 overexpression background. Similar results were observed on Topflash when overexpressing β-catenin (Fig 19B and C). Considering our previous data, that indicated an effect of 21H7 on β-catenin levels, we next assessed whether this effect was specifically on the Wnt induced β-catenin stabilization, or on basal levels. The β-cateninδ91 construct, lacking its amino terminus and hence the three residues phosphorylated during a Wnt response is a dominant positive mutant. The ability of our inhibitor to reduce Topflash transcriptional activity in the presence of overexpressed β-catenin and the mutant construct indicates a mode of action independent of upstream Wnt pathway activity, but likely promoting β-catenin destruction Structurally related compounds mimic the effects of 21H7 To obtain preliminary insights into structure-activity relationships we searched the entire Maybridge library for compounds displaying substructures, superstructures and compounds with general similarity to 21H7 using StructureBase software (IDBS). We focused the search to solely include compounds that had been screened, which resulted in three closely related compounds; 21A8, 12B11 and 21B8 (Fig. 20). Notably, none of the three compounds displayed any effect in the Ff-luc assay. We tested the most closely related compound, 21A8 for effects on Wnt-dependent induction of Axin2 and CyclinD1 expression by Real-time PCR. 21A8, which has an additional ether moiety, was seen to block Wnt-induced Axin2 and/or CyclinD1 expression similar to 21H7 (Fig. 21) and therefore appears to be a false negative identified through secondary analysis. Neither of the other two compounds, 21B8 or 12B11, were available to us at the time of testing and were not assessed for effect.

81 73 Figure 20: Screened Maybridge compounds with significant structural similarity to 21H7. Compound 21H7 and molecules of structural similarity which were screened are depicted. None of the additional compounds displayed any effect upon b-catenin levels in the Ff-luc highthroughput screen.

82 74 Figure 21: Compound 21A8, the molecule of greatest structural similarity to 21H7 inhibits the expression of Wnt target genes in colon cancer cell lines. RNA isolated from the CRC cell lines treated with 21A8 or DMSO control was reverse transcribed, and cdna of established Wnt target genes was quantified by QPCR. Levels of GAPDH cdna was also quantified and used to normalize results. Compound 21A8 displays an effect similar to 21H7 in reducing endogenous mrna levels of Wnt target genes in all colon cancer cell lines tested.

83 75 1 Discussion A large number of cancer therapeutic approaches rely on cytotoxic agents to induce general cell death. Albeit successful, these methods take an unfocused approach to offset the hyperproliferative effects of cancers in a nonspecific manner. Future generations of cancer drugs would ideally be targeted to specific molecular pathways. Targeting known tumorigenic pathways, or pathways highly correlated to certain cancers presents an ideal opportunity for less toxic chemopreventive measures. One cancer well suited to a directed approach is colorectal cancer, for which mutations of key Wnt pathway components appear very early in adenoma development in approximately 90% of human CRC s (Giles RH, 2003). Moreover, all such mutations confer constitutive Wnt pathway activation and the subsequent expression of a variety of anti-apoptotic and proliferative genes (Polakis, 2000). In considering how to inhibit the proliferative effect associated with hyperactive Wnt signaling, several approaches can be envisaged. Studies using antisense (Green et al., 2001) or gene targeting strategies (Kwong et al., 2002) have been met with considerable obstacles. A more feasible approach would be to identify small molecules directly targeted to a component of the signaling pathway. Such studies have already produced molecules which inhibit β-catenin-tcf interaction (Lepourcelet et al., 2004). Other inhibitors of the pathway that currently exist are downstream effectors of transcriptional activation (Fig. 2). We therefore sought to identify novel upstream small molecule inhibitors of Wnt signaling not only for the potential as a therapeutic agent, but as a research tool to elucidate the mechanisms involved in regulating β-catenin stability. To this end we developed and optimized an assay to identify small molecule effectors of β- catenin stabilization utilizing a high-throughput screen. The cell based screening approach offers several advantages; any active compound will be cell permeable, and compounds displaying nonspecific cytotoxicity can be recognized through secondary assays. Moreover, by choosing a midstream pathway component as readout, the strategy allowed us to identify molecules active at or upstream of β-catenin in the pathway, independent of any predetermined mechanism of

84 76 action. Potentially, inhibitors or activators identified may even affect the signaling pathway through unknown mechanisms or levels of regulation. The final screening data was analyzed and is presented as B-score values which we believed best compensated for systemic assay variability. Percent activity measurements, like all control based measures do not account for systemic sources of error or positional variability (Brideau et al., 2003). Moreover, B-scores contain adjustments for both row and column positional effects not present in a simple fold over median measurement. As a result, the B-score proved to perform the best in our data analysis identifying outliers and minimizing noise (Fig. 8). Through the screen, we identified a number of effectors of β-catenin stability, hence Wnt pathway modulators. Hit rates for cell based assays are typically higher than in vitro and in silico approaches (Zaman, 2004), and our inhibitory hit rate from the Maybridge library (0.6%) is higher than other Wnt pathway in vitro (Lepourcelet et al., 2004) and in silico (Shan et al., 2005) screens. However, differences in hit rates are also influenced by the nature of the library being screened, as noted in the variation between libraries we used for screening. In both the Lopac and Prestwick libraries, destabilizing compounds were far more frequently identified than stabilizing molecules. An inverse of the ratio is seen in the Maybridge screening data, and can be attributed to the nature of the collection. Prestwick and Lopac libraries, which consist of biologically active small molecules are more likely to possess compounds effecting general cell viability. The Ff-luc assay is not ideally designed to identify general effectors such as these, and therefore hit rates are skewed. Weeding out compounds with general effects was accomplished through the use of manual assay repeats along with pathway specific reporter assays assessing the effects of these compounds on the activity of the Wnt pathway as a whole compared to its effects on other developmentally important signaling pathways (Fig 12, 13, Table 4). Interestingly, the only compound displaying a repeatable and preferential inhibitory effect upon the Wnt signaling pathway was also the only molecule (21H7) which preferentially destabilized β-catenin in the Wnt ligand treated run compared to the control run. We then used immunoblotting to detect changes in the levels of cytosolic β-catenin and found a concentration dependent decrease in total levels at micromolar concentrations. Notably, this effect has been seen with other small molecules in cell lines before

85 77 (Boon et al., 2004; Fujii et al., 2007), but never at low micromolar concentrations as displayed by compound 21H7. Because the effects of 21H7 could be occurring at or anywhere upstream of β-catenin, we utilized a variety of assays to ascertain the molecular target of 21H7. Topflash assays assessing the epistatic effects of the compound were inconclusive, indicating the compound definitely acts downstream of the receptor level and dishevelled activation. Moreover, the ability of the compound to retain its inhibitory effects on Topflash activity in the presence of overexpressed β- catenin and a β-catenin construct lacking residues phosphorylated during a Wnt response (Fig. 19) points to a mode of action in which β-catenin levels are regulated by a mechanism outside of the known Wnt pathway effectors. These results, taken together with the immunoblotting data strongly suggest the inhibitory mechanism is related to β-catenin itself. Due to the inherent nature of the Ff-luc assay which measures the levels of β-catenin, downstream components can be ruled out. While β-catenin levels are most profoundly kept in check through Wnt signaling via the E3 ligase βtrcp (Lustig and Behrens, 2003), the possibility exists of other ligases promoting β-catenin degradation by a means entirely unrelated to the Wnt pathway. In fact, the E3 ligase, Siah1 has been shown to work in this manner (Liu et al., 2001). In the future, it will be important to determine whether 21H7 mediates its effect in a Siah1 or βtrcp dependent manner through directed knock-down experiments using Topflash as readout. Experiments such as these could point towards a significantly more complex mechanism regulating β-catenin stability then what is currently known. More immediate experiments should be directed towards determining whether the experimental evidence pointing towards 21H7 in promoting the destruction of β-catenin is in fact correct. Multiple possibilities can be envisioned whereby a specific pool or subpopulation of β-catenin is solely affected, or perhaps that the effect of 21H7 is more general than we believe. To this end, Topflash assays using a combination of 21H7 and general proteosome inhibitors such as LLnL or MG132 (Xu and Attisano, 2000) would be resistant to the inhibitory effects. Likewise, in conjunction with β-catenin knock-down Topflash experiments should determine whether the effect is being mediated solely through β-catenin. The possibility could exist where 21H7 is acting upon one or more particular forms of β-catenin. Subpopulations of differentially activated

86 78 forms of β-catenin exist, whereby different phosphorylation states govern the activity of the molecule (van Noort et al., 2007). Identifying the cellular components directly or indirectly affected by our inhibitor will undoubtedly be difficult. Nevertheless, one approach we anticipate using will combine basic biomolecular techniques with high throughput technologies. We have successfully developed protocols for screening large sirna and cdna libraries for effects in Topflash assays (Manuscript in preparation). Eventually these screens can be used to identify whether 21H7 retain its inhibitory effect with overexpressed cdna s, or whether its effects can be either mimicked or enhanced by a particular sirna construct. For these experiments we will initially utilize a sirna (Dharmacon, sigenome Smartpool) and cdna library of known Wnt pathway components followed by the possibility of larger genome wide libraries. Constitutive activation of the Wnt pathway is found in nearly every case of colon cancer (Polakis, 2000). Mutations of key Wnt pathway components lead to a disruption of normal β- catenin breakdown resulting in increased Wnt/β-catenin target gene expression (Logan and Nusse, 2004). We therefore set out to explore the effect of 21H7 on a variety of colorectal cancer (CRC) cell lines possessing hyperactive Wnt signaling due to various mutations by examining transcript levels of established Wnt target genes (Axin2 and CyclinD1) via real-time quantitative PCR analysis. Axin2 levels were clearly affected by 21H7 in all colorectal cancer cell lines regardless of their Wnt pathway mutational status (Fig 17). While the effects did vary between lines, there was no correlation between degree of effect and mutational status, indicating 21H7 does exert its effect downstream of these mutations, supporting our model of inhibition by promoting β-catenin degradation. CyclinD1 mrna levels were not effected to the same extent as Axin2. In fact, transcript levels in both LS1034 or Colo205 cells displayed no significant reduction upon treatment with the inhibitor. This is most likely due to the fact that Cyclin D1 is not as robust a Wnt target, coupled with the fact that its levels are regulated by a variety of other pathways including STAT (Leslie et al., 2006), NFκB (Guttridge et al., 1999) and Ras (Gladden and Diehl, 2005). Interestingly, CyclinD1 it is not the sole Wnt target gene which has its levels influenced by other pathways. Work done in our lab (Labbe et al., 2007), point towards cross-talk between the Wnt and TGF-β pathways regulating Axin2 levels and playing a role during intestinal tumorogenesis.

87 79 It is therefore possible that differences in inhibitory effects displayed between CRC cell lines may be attributed to synergistic pathway activity. Therefore knowledge of TGF-β activity in all CRC cell lines assayed would be informative, made more noteworthy because 21H7 displays a slight inhibitory effect on TGF-β dependent transcriptional activity (Fig 13). As a lead compound with potential anticancer properties, 21H7 demonstrated the ability to not only inhibit Wnt transcriptional activity, but also block the effects associated with misregulated Wnt signaling in CRC cell lines. Thus, we next assessed the breadth of that effect. First, we determined whether 21H7 can suppress Wnt target gene expression in cell lines derived from other cancers, by using prostate derived cell lines where misregulated Wnt signaling has been implicated in prostate cancer (Yardy and Brewster, 2005). We identified a similar inhibitory effect by 21H7 in two different prostate cancer cell lines via QPCR analysis (Fig. 18). Our second approach focused on the effects of a structurally related compound. With the exception of an additional ether moiety, compound 21A8 is structurally identical 21H7 (Fig 20). The fact that 21A8 displayed matching patterns of inhibition through QPCR analysis in CRC cell lines signifies a structure-activity relationship. We have yet to acquire additional structurally related compounds, however, 21H7 will be treated as molecular starting point from which lead optimization will be pursued. For this purpose we will use StructureBase software (IDBS) to screen other chemical libraries for substructures, superstructures and compounds with general similarity. This will allow us to test additional compounds for increased selectivity, potency or ascertain which chemical moieties are key to the Wnt inhibitory effect. In considering the soundness of future therapeutic applications, it would be necessary to identify a positive phenotypic chemopreventive effect managed by 21H7 treatment. Our lead compound should demonstrate growth inhibitory effects preferential to CRC cell lines as compared to normal colonic epithelial cells (Emami KH, 2004). Therefore, via cell proliferation (MTT, ATCC) or soft agar growth assays (Lin et al., 2007), we could ascertain whether the mitogenic activity of Wnts can be suppressed in CRC cell lines habituated to a constitutively active pathway. More pertinent experiments should be directed towards animal studies. For this we could use Min (APC +/- ) mice, which harbor a mutation in APC leading to the development of numerous colonic and small-bowel adenomas (Moser et al., 1995). Quantifying the colonic polyps, adenomas, or rate of apoptosis in crypts with 21H7 treatment or vehicle alone would allow us to determine the effectiveness of our inhibitor as a CRC chemopreventive agent.

88 80 Moreover, future work towards potential therapeutic applications would require ADME/Tox studies completed in laboratory mice to determine whether patient applications are feasible. As equally important as an inhibitor would be for its cancer applications, identifying a novel midstream small molecule inhibitor of Wnt signaling could be of tremendous importance as a research tool in elucidating the mechanisms involved in regulating Wnt signaling. There are many unanswered questions within the Wnt signaling field, and if our inhibitor proves to be suitably selective, it could be utilized in assessing unanswered pathway questions. For instance, unlike canonical Wnt signaling, other well defined signaling pathways such as TGF-β, ΒΜP and MAPK exhibit significant amounts of cross-talk (von Bubnoff and Cho, 2001). Therefore, contingent on the specificity of 21H7 or its derivatives, we could address questions of Wnt pathway cross-talk in addition to that previously described by our lab (Labbe E, 2000). In addition to the importance of inhibitors, compounds identified as activators in the screen may inhibit β-trcp or GSK3β, two attractive targets for drug intervention. The E3 ligase recognizing β-catenin also recognizes IκB, the inhibitory subunit of NFκB. Preventing the degradation of the inhibitory subunit (IκB) will curb the nuclear activities of NFκB associated with a variety of cancers (Fuchs SY, 2004). Also, GSK3β activation has been shown to increase β-amyloid (Aβ) production, while its inhibition decrease Aβ levels (Phiel CJ, 2003). Heritable defects of earlyonset Alzheimers are linked to this increase in Aβ production, indicating the potential for GSK3β inhibitors to reduce Aβ levels (Fuchs SY, 2004). Therefore, while our secondary analyses have focused entirely on Wnt pathway inhibitors, the activitory hits which are greater in number may have significant medical and biochemical implications and merit future consideration. In summary, we have developed and optimized a high-throughput assay to investigate β-catenin stability, hence Wnt pathway activity in a human derived cell line using chemiluminescent readout. This format of Luciferase-tagging a molecule of interest to investigate its stability has never been reported as a high-throughput technique. Our study demonstrates the viability of this technique to identify novel pathway small molecule effectors. We provide evidence of an identified inhibitory compound (21H7) promoting the destruction of β-catenin and repressing Wnt-dependent gene expression in colorectal cancer cell lines. Furthermore, considering the

89 81 nearly identical effects of structurally similar compounds, these findings support future research designed to ascertain mechanism of action and to further mine the screening data for additional Wnt pathway effectors.

90 82 References Adler, P.N. (2002) Planar signaling and morphogenesis in Drosophila. Dev Cell, 2, Aguilera, O., Fraga, M.F., Ballestar, E., Paz, M.F., Herranz, M., Espada, J., Garcia, J.M., Munoz, A., Esteller, M. and Gonzalez-Sancho, J.M. (2006) Epigenetic inactivation of the Wnt antagonist DICKKOPF-1 (DKK-1) gene in human colorectal cancer. Oncogene, 25, Amit, S., Hatzubai, A., Birman, Y., Andersen, J.S., Ben-Shushan, E., Mann, M., Ben-Neriah, Y. and Alkalay, I. (2002) Axin-mediated CKI phosphorylation of beta-catenin at Ser 45: a molecular switch for the Wnt pathway. Genes Dev, 16, Austin, C.P. (2003) The completed human genome: implications for chemical biology. Curr Opin Chem Biol, 7, Barker, N. and Clevers, H. (2006) Mining the Wnt pathway for cancer therapeutics. Nat Rev Drug Discov, 5, Bellows, D.S. and Tyers, M. (2004) Cell biology. Chemical genetics hits. Science, 306, Benchabane, H. and Wrana, J.L. (2003) GATA- and Smad1-dependent enhancers in the Smad7 gene differentially interpret bone morphogenetic protein concentrations. Mol Cell Biol, 23, Bennett, C.N., Longo, K.A., Wright, W.S., Suva, L.J., Lane, T.F., Hankenson, K.D. and MacDougald, O.A. (2005) Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc Natl Acad Sci U S A, 102, Biben, C., Stanley, E., Fabri, L., Kotecha, S., Rhinn, M., Drinkwater, C., Lah, M., Wang, C.C., Nash, A., Hilton, D., Ang, S.L., Mohun, T. and Harvey, R.P. (1998) Murine cerberus homologue mcer-1: a candidate anterior patterning molecule. Dev Biol, 194, Boon, E.M., Keller, J.J., Wormhoudt, T.A., Giardiello, F.M., Offerhaus, G.J., van der Neut, R. and Pals, S.T. (2004) Sulindac targets nuclear beta-catenin accumulation and Wnt signalling in adenomas of patients with familial adenomatous polyposis and in human colorectal cancer cell lines. Br J Cancer, 90, Bos, C.L., Kodach, L.L., van den Brink, G.R., Diks, S.H., van Santen, M.M., Richel, D.J., Peppelenbosch, M.P. and Hardwick, J.C. (2006) Effect of aspirin on the Wnt/beta-catenin pathway is mediated via protein phosphatase 2A. Oncogene, 25, Boyden LM, M.J., Belsky J, Mitzner L, Farhi A, Mitnick MA, Wu D, Insogna K, Lifton RP. (2002) High bone density due to a mutation in LDL-receptor-related protein 5. N Engl J Med., 346, Brideau, C., Gunter, B., Pikounis, B. and Liaw, A. (2003) Improved statistical methods for hit selection in high-throughput screening. J Biomol Screen, 8,

91 83 Caldwell, J.S. (2007) Cancer cell-based genomic and small molecule screens. Adv Cancer Res, 96, Clevers, H. (2006) Wnt/beta-catenin signaling in development and disease. Cell, 127, Coudreuse, D. and Korswagen, H.C. (2007) The making of Wnt: new insights into Wnt maturation, sorting and secretion. Development, 134, Dajani, R., Fraser, E., Roe, S.M., Yeo, M., Good, V.M., Thompson, V., Dale, T.C. and Pearl, L.H. (2003) Structural basis for recruitment of glycogen synthase kinase 3beta to the axin-apc scaffold complex. Embo J, 22, Day, T.F., Guo, X., Garrett-Beal, L. and Yang, Y. (2005) Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell, 8, Drews, J. (2000) Drug discovery: a historical perspective. Science, 287, Emami KH, N.C., Ma H, Kim DH, Jeong KW, Eguchi M, Moon RT, Teo JL, Kim HY, Moon SH, Ha JR, Kahn M. (2004) A small molecule inhibitor of beta-catenin/creb-binding protein transcription. Proc Natl Acad Sci U S A., 101, Fuchs SY, S.V., Kumar KG. (2004) The many faces of beta-trcp E3 ubiquitin ligases: reflections in the magic mirror of cancer. Oncogene, 15, Fujii, N., You, L., Xu, Z., Uematsu, K., Shan, J., He, B., Mikami, I., Edmondson, L.R., Neale, G., Zheng, J., Guy, R.K. and Jablons, D.M. (2007) An antagonist of dishevelled proteinprotein interaction suppresses beta-catenin-dependent tumor cell growth. Cancer Res, 67, Galiatsatos, P. and Foulkes, W.D. (2006) Familial adenomatous polyposis. Am J Gastroenterol, 101, Garcia-Fernandez, J., D'Aniello, S. and Escriva, H. (2007) Organizing chordates with an organizer. Bioessays, 29, Gayet, J., Zhou, X.P., Duval, A., Rolland, S., Hoang, J.M., Cottu, P. and Hamelin, R. (2001) Extensive characterization of genetic alterations in a series of human colorectal cancer cell lines. Oncogene, 20, Giles RH, v.e.j., Clevers H. (2003) Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta., 1653, Gladden, A.B. and Diehl, J.A. (2005) Location, location, location: the role of cyclin D1 nuclear localization in cancer. J Cell Biochem, 96, Glinka, A., Wu, W., Delius, H., Monaghan, A.P., Blumenstock, C. and Niehrs, C. (1998) Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature, 391,

92 84 Goodnow, R.A., Jr. (2001) Current practices in generation of small molecule new leads. J Cell Biochem Suppl, Suppl 37, Green, D.W., Roh, H., Pippin, J.A. and Drebin, J.A. (2001) Beta-catenin antisense treatment decreases beta-catenin expression and tumor growth rate in colon carcinoma xenografts. J Surg Res, 101, Gregorieff, A. and Clevers, H. (2005) Wnt signaling in the intestinal epithelium: from endoderm to cancer. Genes Dev, 19, Guttridge, D.C., Albanese, C., Reuther, J.Y., Pestell, R.G. and Baldwin, A.S., Jr. (1999) NFkappaB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Mol Cell Biol, 19, Haegel, H., Larue, L., Ohsugi, M., Fedorov, L., Herrenknecht, K. and Kemler, R. (1995) Lack of beta-catenin affects mouse development at gastrulation. Development, 121, Hagen, T. and Vidal-Puig, A. (2002) Characterisation of the phosphorylation of beta-catenin at the GSK-3 priming site Ser45. Biochem Biophys Res Commun, 294, Hamatani, T., Carter, M.G., Sharov, A.A. and Ko, M.S. (2004) Dynamics of global gene expression changes during mouse preimplantation development. Dev Cell, 6, He, T.C., Sparks, A.B., Rago, C., Hermeking, H., Zawel, L., da Costa, L.T., Morin, P.J., Vogelstein, B. and Kinzler, K.W. (1998) Identification of c-myc as a target of the APC pathway. Science, 281, Herzlinger, D., Qiao, J., Cohen, D., Ramakrishna, N. and Brown, A.M. (1994) Induction of kidney epithelial morphogenesis by cells expressing Wnt-1. Dev Biol, 166, Holmen, S.L., Giambernardi, T.A., Zylstra, C.R., Buckner-Berghuis, B.D., Resau, J.H., Hess, J.F., Glatt, V., Bouxsein, M.L., Ai, M., Warman, M.L. and Williams, B.O. (2004) Decreased BMD and limb deformities in mice carrying mutations in both Lrp5 and Lrp6. J Bone Miner Res, 19, Holmen, S.L., Zylstra, C.R., Mukherjee, A., Sigler, R.E., Faugere, M.C., Bouxsein, M.L., Deng, L., Clemens, T.L. and Williams, B.O. (2005) Essential Role of {beta}-catenin in Postnatal Bone Acquisition. J Biol Chem, 280, Hoppler, S. and Kavanagh, C.L. (2007) Wnt signalling: variety at the core. J Cell Sci, 120, Huelsken, J., Vogel, R., Brinkmann, V., Erdmann, B., Birchmeier, C. and Birchmeier, W. (2000) Requirement for beta-catenin in anterior-posterior axis formation in mice. J Cell Biol, 148, Ilyas, M., Tomlinson, I.P., Rowan, A., Pignatelli, M. and Bodmer, W.F. (1997) Beta-catenin mutations in cell lines established from human colorectal cancers. Proc Natl Acad Sci U S A, 94,

93 85 Jamieson, C.H., Ailles, L.E., Dylla, S.J., Muijtjens, M., Jones, C., Zehnder, J.L., Gotlib, J., Li, K., Manz, M.G., Keating, A., Sawyers, C.L. and Weissman, I.L. (2004) Granulocytemacrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med, 351, Jho, E.H., Zhang, T., Domon, C., Joo, C.K., Freund, J.N. and Costantini, F. (2002) Wnt/betacatenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol, 22, Johnson, M.L. and Rajamannan, N. (2006) Diseases of Wnt signaling. Rev Endocr Metab Disord, 7, Kamb, A., Wee, S. and Lengauer, C. (2007) Why is cancer drug discovery so difficult? Nat Rev Drug Discov, 6, Kemler, R., Hierholzer, A., Kanzler, B., Kuppig, S., Hansen, K., Taketo, M.M., de Vries, W.N., Knowles, B.B. and Solter, D. (2004) Stabilization of beta-catenin in the mouse zygote leads to premature epithelial-mesenchymal transition in the epiblast. Development, 131, Kikuchi, A., Yamamoto, H. and Kishida, S. (2007) Multiplicity of the interactions of Wnt proteins and their receptors. Cell Signal, 19, Kim, J., Zhang, X., Rieger-Christ, K.M., Summerhayes, I.C., Wazer, D.E., Paulson, K.E. and Yee, A.S. (2006) Suppression of Wnt signaling by the green tea compound (-)- epigallocatechin 3-gallate (EGCG) in invasive breast cancer cells. Requirement of the transcriptional repressor HBP1. J Biol Chem, 281, Kim, K.A., Kakitani, M., Zhao, J., Oshima, T., Tang, T., Binnerts, M., Liu, Y., Boyle, B., Park, E., Emtage, P., Funk, W.D. and Tomizuka, K. (2005) Mitogenic influence of human R- spondin1 on the intestinal epithelium. Science, 309, Kimelman, D. and Xu, W. (2006) beta-catenin destruction complex: insights and questions from a structural perspective. Oncogene, 25, Koornstra, J.J., Rijcken, F.E., Oldenhuis, C.N., Zwart, N., van der Sluis, T., Hollema, H., devries, E.G., Keller, J.J., Offerhaus, J.A., Giardiello, F.M. and Kleibeuker, J.H. (2005) Sulindac inhibits beta-catenin expression in normal-appearing colon of hereditary nonpolyposis colorectal cancer and familial adenomatous polyposis patients. Cancer Epidemiol Biomarkers Prev, 14, Korinek, V., Barker, N., Morin, P.J., van Wichen, D., de Weger, R., Kinzler, K.W., Vogelstein, B. and Clevers, H. (1997) Constitutive transcriptional activation by a beta-catenin-tcf complex in APC-/- colon carcinoma. Science, 275, Krishnan, V., Bryant, H.U. and Macdougald, O.A. (2006) Regulation of bone mass by Wnt signaling. J Clin Invest, 116,

94 86 Kwong, K.Y., Zou, Y., Day, C.P. and Hung, M.C. (2002) The suppression of colon cancer cell growth in nude mice by targeting beta-catenin/tcf pathway. Oncogene, 21, Labbe E, L.A., Attisano L. (2000) Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factorbeta and wnt pathways. Proc Natl Acad Sci U S A., 97, Labbe, E., Lock, L., Letamendia, A., Gorska, A.E., Gryfe, R., Gallinger, S., Moses, H.L. and Attisano, L. (2007) Transcriptional cooperation between the transforming growth factorbeta and Wnt pathways in mammary and intestinal tumorigenesis. Cancer Res, 67, Labbe, E., Silvestri, C., Hoodless, P.A., Wrana, J.L. and Attisano, L. (1998) Smad2 and Smad3 positively and negatively regulate TGF beta-dependent transcription through the forkhead DNA-binding protein FAST2. Mol Cell, 2, LaBonne, C. (2002) Vertebrate development: wnt signals at the crest. Curr Biol, 12, R Lepourcelet, M., Chen, Y.N., France, D.S., Wang, H., Crews, P., Petersen, F., Bruseo, C., Wood, A.W. and Shivdasani, R.A. (2004) Small-molecule antagonists of the oncogenic Tcf/betacatenin protein complex. Cancer Cell, 5, Leslie, K., Lang, C., Devgan, G., Azare, J., Berishaj, M., Gerald, W., Kim, Y.B., Paz, K., Darnell, J.E., Albanese, C., Sakamaki, T., Pestell, R. and Bromberg, J. (2006) Cyclin D1 is transcriptionally regulated by and required for transformation by activated signal transducer and activator of transcription 3. Cancer Res, 66, Lin, M.S., Chen, W.C., Bai, X. and Wang, Y.D. (2007) Activation of peroxisome proliferatoractivated receptor gamma inhibits cell growth via apoptosis and arrest of the cell cycle in human colorectal cancer. J Dig Dis, 8, Lin, R., Thompson, S. and Priess, J.R. (1995) pop-1 encodes an HMG box protein required for the specification of a mesoderm precursor in early C. elegans embryos. Cell, 83, Lipinski, C.A., Lombardo, F., Dominy, B. W. & Feeney, P. J. (1997) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev., 23, Liu, C., Kato, Y., Zhang, Z., Do, V.M., Yankner, B.A. and He, X. (1999) beta-trcp couples betacatenin phosphorylation-degradation and regulates Xenopus axis formation. Proc Natl Acad Sci U S A, 96, Liu, C., Li, Y., Semenov, M., Han, C., Baeg, G.H., Tan, Y., Zhang, Z., Lin, X. and He, X. (2002) Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell, 108, Liu G, B.A., Harris VK, Aaronson SA. (2003) A novel mechanism for Wnt activation of canonical signaling through the LRP6 receptor. Mol Cell Biol., 23,

95 87 Liu, J., Stevens, J., Rote, C.A., Yost, H.J., Hu, Y., Neufeld, K.L., White, R.L. and Matsunami, N. (2001) Siah-1 mediates a novel beta-catenin degradation pathway linking p53 to the adenomatous polyposis coli protein. Mol Cell, 7, Lo Celso, C., Prowse, D.M. and Watt, F.M. (2004) Transient activation of beta-catenin signalling in adult mouse epidermis is sufficient to induce new hair follicles but continuous activation is required to maintain hair follicle tumours. Development, 131, Logan, C.Y. and Nusse, R. (2004) The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol, 20, Lokey, R.S. (2003) Forward chemical genetics: progress and obstacles on the path to a new pharmacopoeia. Curr Opin Chem Biol, 7, Luesch, H. (2006) Towards high-throughput characterization of small molecule mechanisms of action. Mol Biosyst, 2, Luo, J., Chen, J., Deng, Z.L., Luo, X., Song, W.X., Sharff, K.A., Tang, N., Haydon, R.C., Luu, H.H. and He, T.C. (2007) Wnt signaling and human diseases: what are the therapeutic implications? Lab Invest, 87, Lustig, B. and Behrens, J. (2003) The Wnt signaling pathway and its role in tumor development. J Cancer Res Clin Oncol, 129, Mao, B., Wu, W., Li, Y., Hoppe, D., Stannek, P., Glinka, A. and Niehrs, C. (2001) LDLreceptor-related protein 6 is a receptor for Dickkopf proteins. Nature, 411, Marikawa, Y. (2006) Wnt/beta-catenin signaling and body plan formation in mouse embryos. Semin Cell Dev Biol, 17, Mazieres, J., He, B., You, L., Xu, Z. and Jablons, D.M. (2005) Wnt signaling in lung cancer. Cancer Lett, 222, McMahon, A.P. and Moon, R.T. (1989) Ectopic expression of the proto-oncogene int-1 in Xenopus embryos leads to duplication of the embryonic axis. Cell, 58, McMillan, M. and Kahn, M. (2005) Investigating Wnt signaling: a chemogenomic safari. Drug Discov Today, 10, Mi, K. and Johnson, G.V. (2005) Role of the intracellular domains of LRP5 and LRP6 in activating the Wnt canonical pathway. J Cell Biochem, 95, Miller, J.R. (2002) The Wnts. Genome Biol, 3, REVIEWS3001. Mohamed, O.A., Clarke, H.J. and Dufort, D. (2004) Beta-catenin signaling marks the prospective site of primitive streak formation in the mouse embryo. Dev Dyn, 231,

96 88 Moser, A.R., Luongo, C., Gould, K.A., McNeley, M.K., Shoemaker, A.R. and Dove, W.F. (1995) ApcMin: a mouse model for intestinal and mammary tumorigenesis. Eur J Cancer, 31A, Nam, J.S., Turcotte, T.J., Smith, P.F., Choi, S. and Yoon, J.K. (2006) Mouse cristin/r-spondin family proteins are novel ligands for the Frizzled 8 and LRP6 receptors and activate betacatenin-dependent gene expression. J Biol Chem, 281, Nath, N., Kashfi, K., Chen, J. and Rigas, B. (2003) Nitric oxide-donating aspirin inhibits betacatenin/t cell factor (TCF) signaling in SW480 colon cancer cells by disrupting the nuclear beta-catenin-tcf association. Proc Natl Acad Sci U S A, 100, Nusse, R. and Varmus, H.E. (1982) Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell, 31, Park, C.H., Chang, J.Y., Hahm, E.R., Park, S., Kim, H.K. and Yang, C.H. (2005) Quercetin, a potent inhibitor against beta-catenin/tcf signaling in SW480 colon cancer cells. Biochem Biophys Res Commun, 328, Phiel CJ, W.C., Lee VM, Klein PS. (2003) GSK-3alpha regulates production of Alzheimer's disease amyloid-beta peptides. Nature, 423, Planutis, K., Planutiene, M., Moyer, M.P., Nguyen, A.V., Perez, C.A. and Holcombe, R.F. (2007) Regulation of norrin receptor frizzled-4 by Wnt2 in colon-derived cells. BMC Cell Biol, 8, 12. Polakis, P. (2000) Wnt signaling and cancer. Genes Dev, 14, Polychronopoulos, P., Magiatis, P., Skaltsounis, A.L., Myrianthopoulos, V., Mikros, E., Tarricone, A., Musacchio, A., Roe, S.M., Pearl, L., Leost, M., Greengard, P. and Meijer, L. (2004) Structural basis for the synthesis of indirubins as potent and selective inhibitors of glycogen synthase kinase-3 and cyclin-dependent kinases. J Med Chem, 47, Reifenberger, J., Knobbe, C.B., Wolter, M., Blaschke, B., Schulte, K.W., Pietsch, T., Ruzicka, T. and Reifenberger, G. (2002) Molecular genetic analysis of malignant melanomas for aberrations of the WNT signaling pathway genes CTNNB1, APC, ICAT and BTRC. Int J Cancer, 100, Rocheleau, C.E., Downs, W.D., Lin, R., Wittmann, C., Bei, Y., Cha, Y.H., Ali, M., Priess, J.R. and Mello, C.C. (1997) Wnt signaling and an APC-related gene specify endoderm in early C. elegans embryos. Cell, 90, Seeling, J.M., Miller, J.R., Gil, R., Moon, R.T., White, R. and Virshup, D.M. (1999) Regulation of beta-catenin signaling by the B56 subunit of protein phosphatase 2A. Science, 283, Shan, J., Shi, D.L., Wang, J. and Zheng, J. (2005) Identification of a specific inhibitor of the dishevelled PDZ domain. Biochemistry, 44,

97 89 Shibamoto, S., Higano, K., Takada, R., Ito, F., Takeichi, M. and Takada, S. (1998) Cytoskeletal reorganization by soluble Wnt-3a protein signalling. Genes Cells, 3, Shin, S.Y., Kim, C.G., Jho, E.H., Rho, M.S., Kim, Y.S., Kim, Y.H. and Lee, Y.H. (2004) Hydrogen peroxide negatively modulates Wnt signaling through downregulation of betacatenin. Cancer Lett, 212, Shtutman, M., Zhurinsky, J., Simcha, I., Albanese, C., D'Amico, M., Pestell, R. and Ben-Ze'ev, A. (1999) The cyclin D1 gene is a target of the beta-catenin/lef-1 pathway. Proc Natl Acad Sci U S A, 96, Slusarski, D.C., Corces, V.G. and Moon, R.T. (1997) Interaction of Wnt and a Frizzled homologue triggers G-protein-linked phosphatidylinositol signalling. Nature, 390, Sparks, A.B., Morin, P.J., Vogelstein, B. and Kinzler, K.W. (1998) Mutational analysis of the APC/beta-catenin/Tcf pathway in colorectal cancer. Cancer Res, 58, Spink, K.E., Polakis, P. and Weis, W.I. (2000) Structural basis of the Axin-adenomatous polyposis coli interaction. Embo J, 19, Strovel, E.T., Wu, D. and Sussman, D.J. (2000) Protein phosphatase 2Calpha dephosphorylates axin and activates LEF-1-dependent transcription. J Biol Chem, 275, Suzuki, H., Watkins, D.N., Jair, K.W., Schuebel, K.E., Markowitz, S.D., Chen, W.D., Pretlow, T.P., Yang, B., Akiyama, Y., Van Engeland, M., Toyota, M., Tokino, T., Hinoda, Y., Imai, K., Herman, J.G. and Baylin, S.B. (2004) Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet, 36, Takeda, H., Lyle, S., Lazar, A.J., Zouboulis, C.C., Smyth, I. and Watt, F.M. (2006) Nat Med, 12, Tamai, K., Zeng, X., Liu, C., Zhang, X., Harada, Y., Chang, Z. and He, X. (2004) A mechanism for Wnt coreceptor activation. Mol Cell, 13, Taniguchi, H., Yamamoto, H., Hirata, T., Miyamoto, N., Oki, M., Nosho, K., Adachi, Y., Endo, T., Imai, K. and Shinomura, Y. (2005) Frequent epigenetic inactivation of Wnt inhibitory factor-1 in human gastrointestinal cancers. Oncogene, 24, Tekmal, R.R. and Keshava, N. (1997) Role of MMTV integration locus cellular genes in breast cancer. Front Biosci, 2, d Tetsu, O. and McCormick, F. (1999) Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature, 398, Thomas, K.R. and Capecchi, M.R. (1990) Targeted disruption of the murine int-1 protooncogene resulting in severe abnormalities in midbrain and cerebellar development. Nature, 346,

98 90 van de Wetering, M., Sancho, E., Verweij, C., de Lau, W., Oving, I., Hurlstone, A., van der Horn, K., Batlle, E., Coudreuse, D., Haramis, A.P., Tjon-Pon-Fong, M., Moerer, P., van den Born, M., Soete, G., Pals, S., Eilers, M., Medema, R. and Clevers, H. (2002) The beta-catenin/tcf-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell, 111, van Noort, M., Weerkamp, F., Clevers, H.C. and Staal, F.J. (2007) Wnt signaling and phosphorylation status of beta-catenin: importance of the correct antibody tools. Blood, 110, von Bubnoff, A. and Cho, K.W. (2001) Intracellular BMP signaling regulation in vertebrates: pathway or network? Dev Biol, 239, Wang, Q.T., Piotrowska, K., Ciemerych, M.A., Milenkovic, L., Scott, M.P., Davis, R.W. and Zernicka-Goetz, M. (2004) A genome-wide study of gene activity reveals developmental signaling pathways in the preimplantation mouse embryo. Dev Cell, 6, Wharton, K.A., Jr. (2003) Runnin' with the Dvl: proteins that associate with Dsh/Dvl and their significance to Wnt signal transduction. Dev Biol, 253, Willert, K., Shibamoto, S. and Nusse, R. (1999) Wnt-induced dephosphorylation of axin releases beta-catenin from the axin complex. Genes Dev, 13, Wodarz, A. and Nusse, R. (1998) Mechanisms of Wnt signaling in development. Annu Rev Cell Dev Biol, 14, Wrana, J.L., Attisano, L., Carcamo, J., Zentella, A., Doody, J., Laiho, M., Wang, X.F. and Massague, J. (1992) TGF beta signals through a heteromeric protein kinase receptor complex. Cell, 71, Xu, J. and Attisano, L. (2000) Mutations in the tumor suppressors Smad2 and Smad4 inactivate transforming growth factor beta signaling by targeting Smads to the ubiquitin-proteasome pathway. Proc Natl Acad Sci U S A, 97, Xu, L., Corcoran, R.B., Welsh, J.W., Pennica, D. and Levine, A.J. (2000) WISP-1 is a Wnt-1- and beta-catenin-responsive oncogene. Genes Dev, 14, Yardy, G.W. and Brewster, S.F. (2005) Wnt signalling and prostate cancer. Prostate Cancer Prostatic Dis, 8, Yu, H. and Adedoyin, A. (2003) ADME-Tox in drug discovery: integration of experimental and computational technologies. Drug Discov Today, 8, Zaman, G.J. (2004) Cell-based screening. Drug Discov Today, 9, Zhang, T., Otevrel, T., Gao, Z., Ehrlich, S.M., Fields, J.Z. and Boman, B.M. (2001a) Evidence that APC regulates survivin expression: a possible mechanism contributing to the stem cell origin of colon cancer. Cancer Res, 61,

99 91 Zhang, X., Gaspard, J.P. and Chung, D.C. (2001b) Regulation of vascular endothelial growth factor by the Wnt and K-ras pathways in colonic neoplasia. Cancer Res, 61, Zhao, L., Liu, L., Wang, S., Zhang, Y.F., Yu, L. and Ding, Y.Q. (2007) Differential proteomic analysis of human colorectal carcinoma cell lines metastasis-associated proteins. J Cancer Res Clin Oncol, 133,

100 92 Appendices Appendix 1. Small molecule hits detected from screening the Prestwick chemical library. Positive and negative effectors of β-catenin stabilization are listed with their common chemical name, plate/well location and their associated B-score. Negative scores indicate compounds which destabilize β-catenin, positive values have the opposite effect.

101 93 Appendix 2. Small molecule hits detected from screening the Lopac chemical library. Positive and negative effectors of β-catenin stabilization are listed with their common chemical name, plate/well location and their associated B-score. Negative scores indicate compounds which destabilize β-catenin, positive values have the opposite effect. The known activity of each compound listed is data provided with the Lopac library (Sigma)

Wnt signaling. Ramray Bhat.

Wnt signaling. Ramray Bhat. Wnt signaling Ramray Bhat ramray@mrdg.iisc.ernet.in Starting with animal biology and viral infections The discovery of certain laboratory murine strains that were highly susceptible to mammary gland cancer.

More information

CHAPTER 6 SUMMARIZING DISCUSSION

CHAPTER 6 SUMMARIZING DISCUSSION CHAPTER 6 SUMMARIZING DISCUSSION More than 20 years ago the founding member of the Wnt gene family, Wnt-1/Int1, was discovered as a proto-oncogene activated in mammary gland tumors by the mouse mammary

More information

A Genetic Program for Embryonic Development

A Genetic Program for Embryonic Development Concept 18.4: A program of differential gene expression leads to the different cell types in a multicellular organism During embryonic development, a fertilized egg gives rise to many different cell types

More information

RAS Genes. The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes.

RAS Genes. The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes. ۱ RAS Genes The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes. Oncogenic ras genes in human cells include H ras, N ras,

More information

Neoplasia 18 lecture 6. Dr Heyam Awad MD, FRCPath

Neoplasia 18 lecture 6. Dr Heyam Awad MD, FRCPath Neoplasia 18 lecture 6 Dr Heyam Awad MD, FRCPath ILOS 1. understand the role of TGF beta, contact inhibition and APC in tumorigenesis. 2. implement the above knowledge in understanding histopathology reports.

More information

Protein Kinase C Modulates Wnt Signaling In Colon Tumoral Cell Lines

Protein Kinase C Modulates Wnt Signaling In Colon Tumoral Cell Lines Protein Kinase C Modulates Wnt Signaling In Colon Tumoral Cell Lines Dra Martha Robles Flores Department of Biochemistry Facultad de Medicina Universidad Nacional Autónoma de México Tissue anatomy of the

More information

Problem Set 5 KEY

Problem Set 5 KEY 2006 7.012 Problem Set 5 KEY ** Due before 5 PM on THURSDAY, November 9, 2006. ** Turn answers in to the box outside of 68-120. PLEASE WRITE YOUR ANSWERS ON THIS PRINTOUT. 1. You are studying the development

More information

Summary and Concluding Remarks

Summary and Concluding Remarks Summary and Concluding Remarks Chapter 6 The intestinal epithelium provides an excellent model system for investigating molecular mechanisms regulating cell lineage establishment, stem cell proliferation,

More information

oncogenes-and- tumour-suppressor-genes)

oncogenes-and- tumour-suppressor-genes) Special topics in tumor biochemistry oncogenes-and- tumour-suppressor-genes) Speaker: Prof. Jiunn-Jye Chuu E-Mail: jjchuu@mail.stust.edu.tw Genetic Basis of Cancer Cancer-causing mutations Disease of aging

More information

Axis Formation and Mesoderm Induction

Axis Formation and Mesoderm Induction Developmental Biology Biology 4361 Axis Formation and Mesoderm Induction October 27, 2005 Amphibian anteroposterior specification polarized eggs animal/vegetal pigment yolk v. clear cytoplasm mitochondrial

More information

C. elegans Embryonic Development

C. elegans Embryonic Development Autonomous Specification in Tunicate development Autonomous & Conditional Specification in C. elegans Embryonic Development Figure 8.36 Bilateral Symmetry in the Egg of the Tunicate Styela partita Fig.

More information

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation Cancer The fundamental defect is unregulated cell division. Properties of Cancerous Cells Altered growth and proliferation Loss of growth factor dependence Loss of contact inhibition Immortalization Alterated

More information

The functional investigation of the interaction between TATA-associated factor 3 (TAF3) and p53 protein

The functional investigation of the interaction between TATA-associated factor 3 (TAF3) and p53 protein THESIS BOOK The functional investigation of the interaction between TATA-associated factor 3 (TAF3) and p53 protein Orsolya Buzás-Bereczki Supervisors: Dr. Éva Bálint Dr. Imre Miklós Boros University of

More information

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation Cancer The fundamental defect is unregulated cell division. Properties of Cancerous Cells Altered growth and proliferation Loss of growth factor dependence Loss of contact inhibition Immortalization Alterated

More information

Regulation of Gene Expression in Eukaryotes

Regulation of Gene Expression in Eukaryotes Ch. 19 Regulation of Gene Expression in Eukaryotes BIOL 222 Differential Gene Expression in Eukaryotes Signal Cells in a multicellular eukaryotic organism genetically identical differential gene expression

More information

Introduction. Cancer Biology. Tumor-suppressor genes. Proto-oncogenes. DNA stability genes. Mechanisms of carcinogenesis.

Introduction. Cancer Biology. Tumor-suppressor genes. Proto-oncogenes. DNA stability genes. Mechanisms of carcinogenesis. Cancer Biology Chapter 18 Eric J. Hall., Amato Giaccia, Radiobiology for the Radiologist Introduction Tissue homeostasis depends on the regulated cell division and self-elimination (programmed cell death)

More information

Wnt/β-catenin s regulatory role in embryonic stem cell pluripotency and differentiation

Wnt/β-catenin s regulatory role in embryonic stem cell pluripotency and differentiation Wnt/β-catenin s regulatory role in embryonic stem cell pluripotency and differentiation Wim de Jonge 3361039 Supervisor: Prof. Dr. S.J.L. van den Heuvel Group: Developmental Biology, Faculty of Science,

More information

Biol403 MAP kinase signalling

Biol403 MAP kinase signalling Biol403 MAP kinase signalling The mitogen activated protein kinase (MAPK) pathway is a signalling cascade activated by a diverse range of effectors. The cascade regulates many cellular activities including

More information

Principles of Genetics and Molecular Biology

Principles of Genetics and Molecular Biology Cell signaling Dr. Diala Abu-Hassan, DDS, PhD School of Medicine Dr.abuhassand@gmail.com Principles of Genetics and Molecular Biology www.cs.montana.edu Modes of cell signaling Direct interaction of a

More information

Molecular biology :- Cancer genetics lecture 11

Molecular biology :- Cancer genetics lecture 11 Molecular biology :- Cancer genetics lecture 11 -We have talked about 2 group of genes that is involved in cellular transformation : proto-oncogenes and tumour suppressor genes, and it isn t enough to

More information

Cell Cell Communication

Cell Cell Communication IBS 8102 Cell, Molecular, and Developmental Biology Cell Cell Communication January 29, 2008 Communicate What? Why do cells communicate? To govern or modify each other for the benefit of the organism differentiate

More information

Lecture 15. Signal Transduction Pathways - Introduction

Lecture 15. Signal Transduction Pathways - Introduction Lecture 15 Signal Transduction Pathways - Introduction So far.. Regulation of mrna synthesis Regulation of rrna synthesis Regulation of trna & 5S rrna synthesis Regulation of gene expression by signals

More information

Cell Biology Lecture 9 Notes Basic Principles of cell signaling and GPCR system

Cell Biology Lecture 9 Notes Basic Principles of cell signaling and GPCR system Cell Biology Lecture 9 Notes Basic Principles of cell signaling and GPCR system Basic Elements of cell signaling: Signal or signaling molecule (ligand, first messenger) o Small molecules (epinephrine,

More information

MBios 401/501: Lecture 12.1 Signaling IV. Slide 1

MBios 401/501: Lecture 12.1 Signaling IV. Slide 1 MBios 401/501: Lecture 12.1 Signaling IV Slide 1 Pathways that require regulated proteolysis 1. Notch and Delta 2. Wnt/ b-catenin 3. Hedgehog 4. NFk-B Our last topic on cell signaling are pathways that

More information

Signal Transduction: G-Protein Coupled Receptors

Signal Transduction: G-Protein Coupled Receptors Signal Transduction: G-Protein Coupled Receptors Federle, M. (2017). Lectures 4-5: Signal Transduction parts 1&2: nuclear receptors and GPCRs. Lecture presented at PHAR 423 Lecture in UIC College of Pharmacy,

More information

Biochemistry of Cancer

Biochemistry of Cancer 4/6/15 Biochemistry of Cancer Tumor Suppressor Genes 1 Tumor Suppressor Genes Recessive phenotype Oncogenes (viral or cellular) dominate a phenotype i.e. acfvafon of cancer like behaviors. Ras, Myc, Erb

More information

CHAPTER 3. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 23: (2005)

CHAPTER 3. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology 23: (2005) CHAPTER 3 Expression pattern of Wnt signaling components in the adult intestine Gastroenterology 23:1825-1833 (2005) Chapter 3 Expression pattern of Wnt signaling components in the adult intestine Alex

More information

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells (b). TRIM33 was immunoprecipitated, and the amount of

More information

Cell Cell Communication

Cell Cell Communication IBS 8102 Cell, Molecular, and Developmental Biology Cell Cell Communication January 29, 2008 Communicate What? Why do cells communicate? To govern or modify each other for the benefit of the organism differentiate

More information

mirna Dr. S Hosseini-Asl

mirna Dr. S Hosseini-Asl mirna Dr. S Hosseini-Asl 1 2 MicroRNAs (mirnas) are small noncoding RNAs which enhance the cleavage or translational repression of specific mrna with recognition site(s) in the 3 - untranslated region

More information

G-Protein Signaling. Introduction to intracellular signaling. Dr. SARRAY Sameh, Ph.D

G-Protein Signaling. Introduction to intracellular signaling. Dr. SARRAY Sameh, Ph.D G-Protein Signaling Introduction to intracellular signaling Dr. SARRAY Sameh, Ph.D Cell signaling Cells communicate via extracellular signaling molecules (Hormones, growth factors and neurotransmitters

More information

VIII Curso Internacional del PIRRECV. Some molecular mechanisms of cancer

VIII Curso Internacional del PIRRECV. Some molecular mechanisms of cancer VIII Curso Internacional del PIRRECV Some molecular mechanisms of cancer Laboratorio de Comunicaciones Celulares, Centro FONDAP Estudios Moleculares de la Celula (CEMC), ICBM, Facultad de Medicina, Universidad

More information

Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment

Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment In multicellular eukaryotes, gene expression regulates development

More information

Wnt signals across the plasma membrane to activate the β-catenin pathway by forming oligomers containing its receptors, Frizzled and LRP

Wnt signals across the plasma membrane to activate the β-catenin pathway by forming oligomers containing its receptors, Frizzled and LRP Research article 5103 Wnt signals across the plasma membrane to activate the β-catenin pathway by forming oligomers containing its receptors, Frizzled and LRP Feng Cong*,, Liang Schweizer and Harold Varmus

More information

A class of genes that normally suppress cell proliferation. p53 and Rb..ect. suppressor gene products can release cells. hyperproliferation.

A class of genes that normally suppress cell proliferation. p53 and Rb..ect. suppressor gene products can release cells. hyperproliferation. Tumor Suppressor Genes A class of genes that normally suppress cell proliferation. p53 and Rb..ect Mutations that inactivate the tumor suppressor gene products can release cells from growth suppression

More information

Biology Developmental Biology Spring Quarter Midterm 1 Version A

Biology Developmental Biology Spring Quarter Midterm 1 Version A Biology 411 - Developmental Biology Spring Quarter 2013 Midterm 1 Version A 75 Total Points Open Book Choose 15 out the 20 questions to answer (5 pts each). Only the first 15 questions that are answered

More information

number Done by Corrected by Doctor Maha Shomaf

number Done by Corrected by Doctor Maha Shomaf number 19 Done by Waseem Abo-Obeida Corrected by Abdullah Zreiqat Doctor Maha Shomaf Carcinogenesis: the molecular basis of cancer. Non-lethal genetic damage lies at the heart of carcinogenesis and leads

More information

WNT2 and WNT7B Cooperative Signaling in Lung Development

WNT2 and WNT7B Cooperative Signaling in Lung Development University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 1-1-2012 WNT2 and WNT7B Cooperative Signaling in Lung Development Mayumi Miller University of Pennsylvania, mayumimiller@gmail.com

More information

NFκB What is it and What s the deal with radicals?

NFκB What is it and What s the deal with radicals? The Virtual Free Radical School NFκB What is it and What s the deal with radicals? Emily Ho, Ph.D Linus Pauling Institute Scientist Department of Nutrition and Food Management Oregon State University 117

More information

From crypt stem cell to colorectal cancer

From crypt stem cell to colorectal cancer 19 3 2007 6 Chinese Bulletin of Life Sciences Vol. 19, No. 3 Jun., 2007 1004-0374(2007)03-0321-05 ( 510405) Wnt Notch BMP R735.35; R730.21 A From crypt stem cell to colorectal cancer WEN Bin*, CHEN Weiwen

More information

WNT signalling pathways as therapeutic targets in cancer

WNT signalling pathways as therapeutic targets in cancer WNT signalling pathways as therapeutic targets in cancer Jamie N. Anastas 1,2,3,4 and Randall T. Moon 1,2,4 Abstract Since the initial discovery of the oncogenic activity of WNT1 in mouse mammary glands,

More information

Negative Regulation of c-myc Oncogenic Activity Through the Tumor Suppressor PP2A-B56α

Negative Regulation of c-myc Oncogenic Activity Through the Tumor Suppressor PP2A-B56α Negative Regulation of c-myc Oncogenic Activity Through the Tumor Suppressor PP2A-B56α Mahnaz Janghorban, PhD Dr. Rosalie Sears lab 2/8/2015 Zanjan University Content 1. Background (keywords: c-myc, PP2A,

More information

Section D: The Molecular Biology of Cancer

Section D: The Molecular Biology of Cancer CHAPTER 19 THE ORGANIZATION AND CONTROL OF EUKARYOTIC GENOMES Section D: The Molecular Biology of Cancer 1. Cancer results from genetic changes that affect the cell cycle 2. Oncogene proteins and faulty

More information

Principles of cell signaling Lecture 4

Principles of cell signaling Lecture 4 Principles of cell signaling Lecture 4 Johan Lennartsson Molecular Cell Biology (1BG320), 2014 Johan.Lennartsson@licr.uu.se 1 Receptor tyrosine kinase-induced signal transduction Erk MAP kinase pathway

More information

Src-INACTIVE / Src-INACTIVE

Src-INACTIVE / Src-INACTIVE Biology 169 -- Exam 1 February 2003 Answer each question, noting carefully the instructions for each. Repeat- Read the instructions for each question before answering!!! Be as specific as possible in each

More information

Complexity DNA. Genome RNA. Transcriptome. Protein. Proteome. Metabolites. Metabolome

Complexity DNA. Genome RNA. Transcriptome. Protein. Proteome. Metabolites. Metabolome DNA Genome Complexity RNA Transcriptome Systems Biology Linking all the components of a cell in a quantitative and temporal manner Protein Proteome Metabolites Metabolome Where are the functional elements?

More information

The Biology and Genetics of Cells and Organisms The Biology of Cancer

The Biology and Genetics of Cells and Organisms The Biology of Cancer The Biology and Genetics of Cells and Organisms The Biology of Cancer Mendel and Genetics How many distinct genes are present in the genomes of mammals? - 21,000 for human. - Genetic information is carried

More information

S Award Number: DAMD TITLE: Potential Role of Chibby as a Tumor Suppressor in Breast Cancer PRINCIPAL INVESTIGATOR:

S Award Number: DAMD TITLE: Potential Role of Chibby as a Tumor Suppressor in Breast Cancer PRINCIPAL INVESTIGATOR: AD Award Number: DAMD17-03-1-0748 TITLE: Potential Role of Chibby as a Tumor Suppressor in Breast Cancer PRINCIPAL INVESTIGATOR: Richard W. Carthew, Ph.D. CONTRACTING ORGANIZATION: Northwestern University

More information

Chapter 7 Conclusions

Chapter 7 Conclusions VII-1 Chapter 7 Conclusions VII-2 The development of cell-based therapies ranging from well-established practices such as bone marrow transplant to next-generation strategies such as adoptive T-cell therapy

More information

HORMONES (Biomedical Importance)

HORMONES (Biomedical Importance) hormones HORMONES (Biomedical Importance) Hormones are the chemical messengers of the body. They are defined as organic substances secreted into blood stream to control the metabolic and biological activities.

More information

Cell Signaling part 2

Cell Signaling part 2 15 Cell Signaling part 2 Functions of Cell Surface Receptors Other cell surface receptors are directly linked to intracellular enzymes. The largest family of these is the receptor protein tyrosine kinases,

More information

CHAPTER VII CONCLUDING REMARKS AND FUTURE DIRECTION. Androgen deprivation therapy is the most used treatment of de novo or recurrent

CHAPTER VII CONCLUDING REMARKS AND FUTURE DIRECTION. Androgen deprivation therapy is the most used treatment of de novo or recurrent CHAPTER VII CONCLUDING REMARKS AND FUTURE DIRECTION Stathmin in Prostate Cancer Development and Progression Androgen deprivation therapy is the most used treatment of de novo or recurrent metastatic PCa.

More information

Signaling Vascular Morphogenesis and Maintenance

Signaling Vascular Morphogenesis and Maintenance Signaling Vascular Morphogenesis and Maintenance Douglas Hanahan Science 277: 48-50, in Perspectives (1997) Blood vessels are constructed by two processes: vasculogenesis, whereby a primitive vascular

More information

Biol220 Cell Signalling Cyclic AMP the classical secondary messenger

Biol220 Cell Signalling Cyclic AMP the classical secondary messenger Biol220 Cell Signalling Cyclic AMP the classical secondary messenger The classical secondary messenger model of intracellular signalling A cell surface receptor binds the signal molecule (the primary

More information

ulcer healing role 118 Bicarbonate, prostaglandins in duodenal cytoprotection 235, 236

ulcer healing role 118 Bicarbonate, prostaglandins in duodenal cytoprotection 235, 236 Subject Index Actin cellular forms 48, 49 epidermal growth factor, cytoskeletal change induction in mucosal repair 22, 23 wound repair 64, 65 polyamine effects on cytoskeleton 49 51 S-Adenosylmethionine

More information

Lecture IV. Mechanisms of Neural. Neural Development

Lecture IV. Mechanisms of Neural. Neural Development Lecture IV. Mechanisms of Neural Bio 3411 Monday 1 Readings NEUROSCIENCE: 5 th ed, pp 477-506 (sorta) 4 th ed, pp 545-575 (sorta) References : Fainsod, A., Steinbeisser, H., & De Robertis, E. M. (1994).

More information

Biochemistry of Carcinogenesis. Lecture # 35 Alexander N. Koval

Biochemistry of Carcinogenesis. Lecture # 35 Alexander N. Koval Biochemistry of Carcinogenesis Lecture # 35 Alexander N. Koval What is Cancer? The term "cancer" refers to a group of diseases in which cells grow and spread unrestrained throughout the body. It is difficult

More information

Propagation of the Signal

Propagation of the Signal OpenStax-CNX module: m44452 1 Propagation of the Signal OpenStax College This work is produced by OpenStax-CNX and licensed under the Creative Commons Attribution License 3.0 By the end of this section,

More information

Tumor suppressor genes D R. S H O S S E I N I - A S L

Tumor suppressor genes D R. S H O S S E I N I - A S L Tumor suppressor genes 1 D R. S H O S S E I N I - A S L What is a Tumor Suppressor Gene? 2 A tumor suppressor gene is a type of cancer gene that is created by loss-of function mutations. In contrast to

More information

KEY CONCEPT QUESTIONS IN SIGNAL TRANSDUCTION

KEY CONCEPT QUESTIONS IN SIGNAL TRANSDUCTION Signal Transduction - Part 2 Key Concepts - Receptor tyrosine kinases control cell metabolism and proliferation Growth factor signaling through Ras Mutated cell signaling genes in cancer cells are called

More information

Early Embryonic Development

Early Embryonic Development Early Embryonic Development Maternal effect gene products set the stage by controlling the expression of the first embryonic genes. 1. Transcription factors 2. Receptors 3. Regulatory proteins Maternal

More information

Introduction to Cancer Biology

Introduction to Cancer Biology Introduction to Cancer Biology Robin Hesketh Multiple choice questions (choose the one correct answer from the five choices) Which ONE of the following is a tumour suppressor? a. AKT b. APC c. BCL2 d.

More information

Frizzled Activation by Wnt-l Is Required for j3 -Catenin-T Cell Factor Dependent Transcription in Esophageal Cancer

Frizzled Activation by Wnt-l Is Required for j3 -Catenin-T Cell Factor Dependent Transcription in Esophageal Cancer 75 Wnt-l activate TCF transcription Frizzled Activation by Wnt-l Is Required for j3 -Catenin-T Cell Factor Dependent Transcription in Esophageal Cancer Takaaki Mizushima, Koji Ochi, Yasuyuki Emori, Hiroaki

More information

Regulation of cell function by intracellular signaling

Regulation of cell function by intracellular signaling Regulation of cell function by intracellular signaling Objectives: Regulation principle Allosteric and covalent mechanisms, Popular second messengers, Protein kinases, Kinase cascade and interaction. regulation

More information

Chapter 15: Signal transduction

Chapter 15: Signal transduction Chapter 15: Signal transduction Know the terminology: Enzyme-linked receptor, G-protein linked receptor, nuclear hormone receptor, G-protein, adaptor protein, scaffolding protein, SH2 domain, MAPK, Ras,

More information

C-Phycocyanin (C-PC) is a n«sjfc&c- waefc-jduble phycobiliprotein. pigment isolated from Spirulina platensis. This water- soluble protein pigment is

C-Phycocyanin (C-PC) is a n«sjfc&c- waefc-jduble phycobiliprotein. pigment isolated from Spirulina platensis. This water- soluble protein pigment is ' ^Summary C-Phycocyanin (C-PC) is a n«sjfc&c- waefc-jduble phycobiliprotein pigment isolated from Spirulina platensis. This water- soluble protein pigment is of greater importance because of its various

More information

The Hallmarks of Cancer

The Hallmarks of Cancer The Hallmarks of Cancer This illustration encompasses the six hallmark capabilities originally proposed in our 2000 perspective. The past decade has witnessed remarkable progress toward understanding the

More information

Wnt regulates developmental and oncogenic processes

Wnt regulates developmental and oncogenic processes Casein kinase I in the Wnt pathway: Regulation of -catenin function Chie Sakanaka*, Peng Leong*, Licen Xu*, Stephen D. Harrison*, and Lewis T. Williams* * Chiron Corporation, Emeryville, CA 94608; and

More information

INTERACTION DRUG BODY

INTERACTION DRUG BODY INTERACTION DRUG BODY What the drug does to the body What the body does to the drug Receptors - intracellular receptors - membrane receptors - Channel receptors - G protein-coupled receptors - Tyrosine-kinase

More information

Tetrapod Limb Development

Tetrapod Limb Development IBS 8102 Cell, Molecular and Developmental Biology Tetrapod Limb Development February 11, 2008 Tetrapod Limbs Merlin D. Tuttle Vicki Lockard and Paul Barry Father Alejandro Sanchez Anne Fischer Limb Patterning

More information

Chapter 9. Cellular Signaling

Chapter 9. Cellular Signaling Chapter 9 Cellular Signaling Cellular Messaging Page 215 Cells can signal to each other and interpret the signals they receive from other cells and the environment Signals are most often chemicals The

More information

Ch. 18 Regulation of Gene Expression

Ch. 18 Regulation of Gene Expression Ch. 18 Regulation of Gene Expression 1 Human genome has around 23,688 genes (Scientific American 2/2006) Essential Questions: How is transcription regulated? How are genes expressed? 2 Bacteria regulate

More information

Urogenital Development

Urogenital Development 2-5-03 Urogenital Development Greg Dressler Assoc. Professor Dept. of Pathology x46490 Dressler@umich.edu The Origin of the Kidney In the vertebrate embryo, the first stage of kidney development occurs

More information

Signaling. Dr. Sujata Persad Katz Group Centre for Pharmacy & Health research

Signaling. Dr. Sujata Persad Katz Group Centre for Pharmacy & Health research Signaling Dr. Sujata Persad 3-020 Katz Group Centre for Pharmacy & Health research E-mail:sujata.persad@ualberta.ca 1 Growth Factor Receptors and Other Signaling Pathways What we will cover today: How

More information

Insulin Resistance. Biol 405 Molecular Medicine

Insulin Resistance. Biol 405 Molecular Medicine Insulin Resistance Biol 405 Molecular Medicine Insulin resistance: a subnormal biological response to insulin. Defects of either insulin secretion or insulin action can cause diabetes mellitus. Insulin-dependent

More information

Cell Communication. Local and Long Distance Signaling

Cell Communication. Local and Long Distance Signaling Cell Communication Cell to cell communication is essential for multicellular organisms Some universal mechanisms of cellular regulation providing more evidence for the evolutionary relatedness of all life

More information

Cancer Genetics. What is Cancer? Cancer Classification. Medical Genetics. Uncontrolled growth of cells. Not all tumors are cancerous

Cancer Genetics. What is Cancer? Cancer Classification. Medical Genetics. Uncontrolled growth of cells. Not all tumors are cancerous Session8 Medical Genetics Cancer Genetics J avad Jamshidi F a s a U n i v e r s i t y o f M e d i c a l S c i e n c e s, N o v e m b e r 2 0 1 7 What is Cancer? Uncontrolled growth of cells Not all tumors

More information

Chapter 6 Communication, Integration, and Homeostasis

Chapter 6 Communication, Integration, and Homeostasis Chapter 6 Communication, Integration, and Homeostasis About This Chapter Cell-to-cell communication Signal pathways Novel signal molecules Modulation of signal pathways Homeostatic reflex pathways Cell-to-Cell

More information

Molecular Cell Biology - Problem Drill 19: Cell Signaling Pathways and Gene Expression

Molecular Cell Biology - Problem Drill 19: Cell Signaling Pathways and Gene Expression Molecular Cell Biology - Problem Drill 19: Cell Signaling Pathways and Gene Expression Question No. 1 of 10 1. Which statement about cell signaling is correct? Question #1 (A) Cell signaling involves receiving

More information

Multistep nature of cancer development. Cancer genes

Multistep nature of cancer development. Cancer genes Multistep nature of cancer development Phenotypic progression loss of control over cell growth/death (neoplasm) invasiveness (carcinoma) distal spread (metastatic tumor) Genetic progression multiple genetic

More information

Signal Transduction Cascades

Signal Transduction Cascades Signal Transduction Cascades Contents of this page: Kinases & phosphatases Protein Kinase A (camp-dependent protein kinase) G-protein signal cascade Structure of G-proteins Small GTP-binding proteins,

More information

BCHM3972 Human Molecular Cell Biology (Advanced) 2013 Course University of Sydney

BCHM3972 Human Molecular Cell Biology (Advanced) 2013 Course University of Sydney BCHM3972 Human Molecular Cell Biology (Advanced) 2013 Course University of Sydney Page 2: Immune Mechanisms & Molecular Biology of Host Defence (Prof Campbell) Page 45: Infection and Implications for Cell

More information

Drug Receptor Interactions and Pharmacodynamics

Drug Receptor Interactions and Pharmacodynamics Drug Receptor Interactions and Pharmacodynamics Dr. Raz Mohammed MSc Pharmacology School of Pharmacy 22.10.2017 Lec 6 Pharmacodynamics definition Pharmacodynamics describes the actions of a drug on the

More information

Determination Differentiation. determinated precursor specialized cell

Determination Differentiation. determinated precursor specialized cell Biology of Cancer -Developmental Biology: Determination and Differentiation -Cell Cycle Regulation -Tumor genes: Proto-Oncogenes, Tumor supressor genes -Tumor-Progression -Example for Tumor-Progression:

More information

Early cell death (FGF) B No RunX transcription factor produced Yes No differentiation

Early cell death (FGF) B No RunX transcription factor produced Yes No differentiation Solution Key - Practice Questions Question 1 a) A recent publication has shown that the fat stem cells (FSC) can act as bone stem cells to repair cavities in the skull, when transplanted into immuno-compromised

More information

BIOL212 Biochemistry of Disease. Metabolic Disorders - Obesity

BIOL212 Biochemistry of Disease. Metabolic Disorders - Obesity BIOL212 Biochemistry of Disease Metabolic Disorders - Obesity Obesity Approx. 23% of adults are obese in the U.K. The number of obese children has tripled in 20 years. 10% of six year olds are obese, rising

More information

Regulators of Cell Cycle Progression

Regulators of Cell Cycle Progression Regulators of Cell Cycle Progression Studies of Cdk s and cyclins in genetically modified mice reveal a high level of plasticity, allowing different cyclins and Cdk s to compensate for the loss of one

More information

Development of Carcinoma Pathways

Development of Carcinoma Pathways The Construction of Genetic Pathway to Colorectal Cancer Moriah Wright, MD Clinical Fellow in Colorectal Surgery Creighton University School of Medicine Management of Colon and Diseases February 23, 2019

More information

HOMEWORK RUBRICS MECHANOTRANSDUCTION UNIT: HOMEWORK #1 (20 pts towards your grade)

HOMEWORK RUBRICS MECHANOTRANSDUCTION UNIT: HOMEWORK #1 (20 pts towards your grade) HOMEWORK RUBRICS MECHANOTRANSDUCTION UNIT: HOMEWORK #1 (20 pts towards your grade) 1. Mesenchymal stem cells (MSC) cultured on extracellular matrices with different stiffness exhibit diverse lineage commitment

More information

Wnt signaling in disease and in development

Wnt signaling in disease and in development Blay JY, Schemann S, Favrot MC MINIREVIEW Roel NUSSE * Department of Developmental Biology, Howard Hughes Medical Institute Beckman Center,School of Medicine, Stanford University, USA. ABSCTACT The highly

More information

Vets 111/Biov 111 Cell Signalling-2. Secondary messengers the cyclic AMP intracellular signalling system

Vets 111/Biov 111 Cell Signalling-2. Secondary messengers the cyclic AMP intracellular signalling system Vets 111/Biov 111 Cell Signalling-2 Secondary messengers the cyclic AMP intracellular signalling system The classical secondary messenger model of intracellular signalling A cell surface receptor binds

More information

LESSON 3.2 WORKBOOK. How do normal cells become cancer cells? Workbook Lesson 3.2

LESSON 3.2 WORKBOOK. How do normal cells become cancer cells? Workbook Lesson 3.2 For a complete list of defined terms, see the Glossary. Transformation the process by which a cell acquires characteristics of a tumor cell. LESSON 3.2 WORKBOOK How do normal cells become cancer cells?

More information

Page 32 AP Biology: 2013 Exam Review CONCEPT 6 REGULATION

Page 32 AP Biology: 2013 Exam Review CONCEPT 6 REGULATION Page 32 AP Biology: 2013 Exam Review CONCEPT 6 REGULATION 1. Feedback a. Negative feedback mechanisms maintain dynamic homeostasis for a particular condition (variable) by regulating physiological processes,

More information

609G: Concepts of Cancer Genetics and Treatments (3 credits)

609G: Concepts of Cancer Genetics and Treatments (3 credits) Master of Chemical and Life Sciences Program College of Computer, Mathematical, and Natural Sciences 609G: Concepts of Cancer Genetics and Treatments (3 credits) Text books: Principles of Cancer Genetics,

More information

INTRODUCTION TO THE BIOCHEMISTRY OF HORMONES AND THEIR RECPTORS

INTRODUCTION TO THE BIOCHEMISTRY OF HORMONES AND THEIR RECPTORS INTRODUCTION TO THE BIOCHEMISTRY OF HORMONES AND THEIR RECPTORS 1 Introduction to the Biochemistry of Hormones and their Receptors Lectuctre1 Sunday 17/2/ Objectives: 1. To understand the biochemical nature

More information

A Theoretical Model of the Wnt Signaling Pathway in the Epithelial Mesenchymal Transition

A Theoretical Model of the Wnt Signaling Pathway in the Epithelial Mesenchymal Transition Gasior et al. Theoretical Biology and Medical Modelling (2017) 14:19 DOI 10.1186/s12976-017-0064-7 RESEARCH Open Access A Theoretical Model of the Wnt Signaling Pathway in the Epithelial Mesenchymal Transition

More information

Lecture 10. G1/S Regulation and Cell Cycle Checkpoints. G1/S regulation and growth control G2 repair checkpoint Spindle assembly or mitotic checkpoint

Lecture 10. G1/S Regulation and Cell Cycle Checkpoints. G1/S regulation and growth control G2 repair checkpoint Spindle assembly or mitotic checkpoint Lecture 10 G1/S Regulation and Cell Cycle Checkpoints Outline: G1/S regulation and growth control G2 repair checkpoint Spindle assembly or mitotic checkpoint Paper: The roles of Fzy/Cdc20 and Fzr/Cdh1

More information

Animal Tissue Culture SQG 3242 Biology of Cultured Cells. Dr. Siti Pauliena Mohd Bohari

Animal Tissue Culture SQG 3242 Biology of Cultured Cells. Dr. Siti Pauliena Mohd Bohari Animal Tissue Culture SQG 3242 Biology of Cultured Cells Dr. Siti Pauliena Mohd Bohari The Culture Environment Changes of Cell s microenvironment needed that favor the spreading, migration, and proliferation

More information

2013 W. H. Freeman and Company. 12 Signal Transduction

2013 W. H. Freeman and Company. 12 Signal Transduction 2013 W. H. Freeman and Company 12 Signal Transduction CHAPTER 12 Signal Transduction Key topics: General features of signal transduction Structure and function of G protein coupled receptors Structure

More information

Close to site of release (at synapse); binds to receptors in

Close to site of release (at synapse); binds to receptors in Chapter 18: The Endocrine System Chemical Messengers 1. Neural 2. Endocrine 3. Neuroendocrine 4. Paracrine 5. Autocrine Endocrine System --Endocrine and nervous systems work together --Endocrine vs. Nervous

More information