Abstract
The canonical WNT/β-catenin signaling pathway governs a myriad of biological processes underlying development and maintenance of adult tissue homeostasis, including regulation of stem cell self-renewal, cell proliferation, differentiation, and apoptosis. WNTs are secreted lipid-modified glycoproteins that act as short-range ligands to activate receptor-mediated signaling pathways. The hallmark of the canonical pathway is the activation of β-catenin mediated transcriptional activity. Canonical WNTs control the β-catenin dynamics as the cytoplasmic level of β-catenin is tightly regulated via phosphorylation by the ‘destruction complex’, consisting of glycogen synthase kinase 3β (GSK3β), casein kinase 1α (CK1α), the scaffold protein AXIN, and the tumor suppressor adenomatous polyposis coli (APC). Aberrant regulation of this signaling cascade is associated with varieties of human diseases, especially cancers. Over the past decade, significant progress has been made in understanding the mechanisms of canonical WNT signaling. In this review, we focus on the current understanding of WNT signaling at the extracellular, cytoplasmic membrane, and intracellular/nuclear levels, including the emerging knowledge of crosstalk with other pathways. Recent progresses in developing novel WNT pathway-targeted therapies will also be reviewed. Thus, this review is intended to serve as a refresher of the current understanding about the physiologic and pathogenic roles of WNT/β-catenin signaling pathway, and to outline potential therapeutic opportunities by targeting the canonical WNT pathway.
INTRODUCTION
Originally identified as Int-1, the Wnt1 gene was discovered over 30 years ago as a gene activated by integration of mouse mammary tumor virus (MMTV) proviral DNA in virally induced breast tumors 1, 2. An early identified fly Wingless (Wg) gene, which regulates segment polarity during larval development 3, was found to be a WNT1 homolog 4. In the following years, studies of Drosophila genetics delineating the relationships among segment polarity mutations mapped out the core of the WNT/Wg signal transduction cascade by identifying Porcupine (PORC), disheveled (DVL), armadillo (β-catenin), and zeste-white 3/glycogen synthase kinase 3 (GSK3) genes 5-8. A fuller image of the WNT signaling pathway emerged when T-cell factor/lymphocyte enhancer factor (TCF/LEF) transcription factors were identified as WNT nuclear effectors 9, 10 and Frizzleds (FZDs) were identified as WNT obligate receptors 11, functioning together with co-receptors, such as low-density lipoprotein-receptor-related proteins (LRPs)/Arrow 12. The first case for the involvement of WNT signaling in human cancers was made when the hereditary cancer syndrome termed familial adenomatous polyposis (FAP) gene product, adenomatous polyposis coli (APC) 13, 14, was found to interact with β-catenin 15, 16, and was later shown to play a critical role in controlling β-catenin protein stability. For the past two decades, numerous components of this pathway and more disease connections have been uncovered 17-27.
In most mammalian genomes, the WNT family is comprised of 19 members that are characterized by a highly conserved cysteine-rich secreted glycoproteins, which present the technical challenges in efficient production, biochemical characterization and structural analysis of WNT proteins 28, although the structure of the Xenopus WNT8 protein as bound to Frizzled (FZD) was recently solved 29. The lipid components of WNTs are required for efficient signaling, including WNT protein secretion 30, 31. WNT palmitoylation is essential for WNT signaling and is carried out by PORC, a dedicated ER-localized O-acyltransferase and highly conserved component of the WNT pathway 32, 33. Loss of PORC leads to retention of WNT3A in the ER 34. In most cell/tissue contexts, WNTs act as short-range signaling 23.
The emerging evidence indicates that WNT signaling plays an essential role in regulating many biological processes, including embryonic development, tissue homoeostasis and maintenance of stem cells. Dysregulation of WNT signaling pathway is associated with various human diseases 17-27. Traditionally, WNT signaling is classified into two large categories: the canonical WNT (or β-catenin-dependent) and non-canonical WNT (or β-catenin-independent) pathways. Biologically, the canonical WNT/β-catenin signaling pathway usually plays crucial roles in regulating cell fate, proliferation and survival, while the non-canonical WNT signaling is more associated with differentiation, cell polarity and migration 25-27. Non-canonical WNT signaling can be initiated by WNT interaction with Frizzled receptors, or RYK and ROR receptor tyrosine kinases, and regulates small GTPases (such as RhoA, Rac and Cdc42) in DVL-dependent manner. Non-canonical WNT signaling can also activate calcium flux and kinase cascades, including protein kinase C (PKC), calcium/calmodulin-dependent protein kinase II (CaMKII) and JUN N-terminal kinase (JNK), leading to the activation of AP1- and NFAT-regulated gene expression 25-27. Increasing evidence indicates that the canonical and non-canonical pathways are intersecting signaling networks that coordinately regulate complex processes, such as embryonic development, stem cell maintenance, tissue homeostasis and wound healing 27. In this review, we mainly focus on the canonical WNT/β-catenin pathway in regulating stem cells and tumorigenesis, as well as potential anticancer therapeutic opportunities by targeting key steps of this signaling pathway.
THE CANONICAL WNT/β-CATENIN SIGNALING PATHWAY
A Simplified Overview
When specific WNT ligands are absent, cytoplasmic β-catenin is phosphorylated by the destruction complex formed by the three proteins APC, AXIN and GSK3β (Figure 1). Initial Casein Kinase 1 (CKI) phosphorylation occurs at Ser45 which primes the molecule for subsequent phosphorylation by GSK3β on Thr41, Ser37 and Ser33 20, 25. Phosphorylated β-catenin is recognized by E3 ubiquitin ligase β-Trcp, and degraded by ubiquitin proteasome pathway. Consequently, β-catenin in cytoplasm is kept at a low level. The nuclear transcription factor lymphoid enhancer-binding factor/T cell-specific factor (LEF/TCF) is associated with Groucho and histone deacetylases (HDACs), and represses the expression of WNT/β-catenin target genes 35-37.
Figure 1. Schematic depiction of the canonical WNT signaling pathway.
WNT ligands are post-translationally modified in ER with the participation of porcupine (PORC) and secreted into the extraexcellular space, where WNTs interact with receptors FZDs and co-receptors LRP5/6. Antagonists SFRPs and WIF can bind to WNT ligands while SFRPs can also interact with FZDs. Antagonists SOST and DKKs bind to LRP5/6 to compete with WNT ligands. WNT binding to the receptors initiates the disassembly of the destruction complex consisting of DVL/AXIN/APC/GSK3β/CK1, resulting in the release of the stabilized β-catenin to cytoplasm, which is subsequently transloated into cell nucleus and regulates WNT target genes in concert with co-activators BCL9, Pygo and CBP/p300. TANKs interact with AXIN and promote its degradation. RSPOs interact with LRG5 and membrane-bound E3 ubiquitin ligases ZNRF3/RNF43 and inhibit the ubiquitination-mediated degradation of FZDs. In WNT-OFF cells (the inserted box), the destruction complex is assembled, and the β-catenin protein is phosphorylated by CK1 and GSK3β, and doomed for proteasome-mediated protein degradation.
WNT proteins interact with the seven transmembrane receptors of FZD family and single pass transmembrane co-receptors, such as low-density lipoprotein receptor-related protein 5/6 (LRP5/6) or receptor tyrosine kinase-like orphan receptor 2 (Ror2), to induce intracellular signaling pathway. WNT ligands bind to the cysteine-rich domain of FZD, that trigger LRP5/6 phosphorylation and the formation of FZD-LRP5/6 heterotrimeric complex 38. The activation of DVL protein is phosphorylated and translocate to the FZD receptor 39, 40. In this context, the β-catenin destruction complex is disrupted which prevents β-catenin proteasomal degradation. Stabilized β-catenin accumulates in the cytoplasm and then translocated into the nucleus (Figure 1). Nuclear β-catenin displaces Groucho and forms a complex with the B-cell lymphoma 9 protein (BCL9), Pygopus, histone modifier CBP, as well as tissue-specific transcriptional activators 41, 42, and binds to LEF/TCF proteins to regulate the expression of WNT target genes in a cell type specific manner 43-50.
DVL Plays an Essential Switchboard Role in Channeling WNT Signaling
The scaffold protein DVL is the key cytoplasmic partner of WNT signaling. DVL inhibits AXIN function through a direct interaction with the DIX domain of DVL, which is an important step in the activation of canonical WNT signal pathway 51. DVL is involved in the formation of the FZD and LRP6 complex. FZD recruits DVL by binding to the PDZ domain of DVL. WNT promotes DVL-dependent LRP6 phosphorylation to regulate downstream gene expression 38. Furthermore, DVL shuttles between the cytoplasm and the nucleus to transduce canonical WNT signaling to GSK3β-destruction complex of β-catenin, resulting in the stabilization of β-catenin 52 The mutation in DVL nuclear localization signal domain leads to inhibition of the WNT/β-catenin signaling. Nuclear DVL, c-Jun, β-catenin form a complex leading to stabilization of β-catenin/TCF interaction. Interestingly, DVL was also shown to interact with transcription factor Hipk1 to regulate the transcription of WNT/β-catenin target genes 53 Forkhead box (FOX) transcription factors, FOXK1 and FOXK2, have been recently shown to positively regulate WNT/β-catenin signaling by translocating DVL into the nucleus 54.
Stabilization and Nuclear Translocation of the β-Catenin Protein Is the Essence of Canonical WNT Signaling
As a dual function adhesion and transcription coactivator protein, β-catenin is a key mediator of the canonical WNT pathway. A key step in transcriptional activation is the formation of a complex between β-catenin and TCF/LEF transcription factors. In the absence of β-catenin, TCF/LEF factors have no transcriptional activity and are bound by transcription repressors, such as Groucho (Figure 1). To generate a transcriptionally active complex, TCF/LEF and β-catenin recruit CBP or its homolog p300, as well as other components of the basal transcription machinery, to initiate transcription 55. CBP and p300 proteins promote histone acetylation 56. BCL9 is involved in the majority of β-catenin-regulated gene transcription. β-Catenin can bind to the HD2 domain of BCL9, which served as a specific cofactor for β-catenin regulated transcription.57 The Drosophila Pygopus (Pygo) protein is shown to promote the binding of β-catenin to WNT responding element (WRE) and transcriptional active sites by the combination with BCL9-β-catenin 58. It has recently been shown that β-catenin can interact with other transcription factors (e.g. FOXOs, nuclear receptors, Sox, Smad, Oct4) and plays an important role in various cellular processes 52, 59, 60. In fact, it was shown that FoxM1can physically interact with β-catenin and promote β-catenin nuclear localization and WNT target-gene expression 61. Therefore, the cell context-specific activities of canonical WNT signaling may be accomplished at least in part by differential recruitments of tissue-specific cofactors to the β-catenin/TCF/LEF complex to regulate the expression of different genes.
R-Spondins Emerge as WNT Signaling Activators
R-Spondin (RSPO) family consists of four members (RSPO1-4), emerging as a group of canonical WNT signaling activators 62, 63. RSPO proteins contain four major functional domains: two cysteine-rich furin-like (CR) domains, a thrombospondin type 1 (TSP1) domain and a C-terminal region containing basic amino acids. The CR domains (92–94 aa in length) of RSPO proteins are primarily responsible for activation of WNT/β-catenin signaling 64, 65. Deletion of furin-like motifs within the CR domain of RSPO abolishes its ability to activate canonical WNT signaling. RSPO activation of canonical WNT signaling pathway may depend on the phosphorylation of LRP5/6 receptors 66. It has been recently shown that Lgr proteins (or Leu-rich repeat-containing G protein-coupled receptor) can bind the furin domains of R-Spondins with high affinity to promote β-catenin signaling 67, 68. R-spondins, acting through Lgr receptors, inhibit the transmembrane E3 ubiquitin ligases RNF43/ZNRF3 that ubiquitinate and thus degrade FZD receptors 69, 70, leading to the stabilization of FZD receptors and subsequent enhancement of WNT signal. It should be pointed out that the R-Spondin (RSPO)/LGR axis is only found in vertebrate systems 71.
WNT Signaling Is Tightly Controlled by Its Naturally Occurring Secreted Antagonists
All essential pathways in mammalian cells are heavily negatively regulated. The canonical WNT signaling pathway is no exception. Numerous inhibitors of WNT signaling present outside of the cell and affect ligand–receptor interactions. The prototype antagonists, called secreted Frizzled-related proteins (SFRPs), possess FZD-like cysteine-rich domain (CRD) that competitively binds to WNT ligands and prevents the interaction between WNTs and FZD receptors 72-74. Another structurally unrelated secreted inhibitor called WNT inhibitory protein (WIF) can bind WNTs, thereby blocking interactions between WNT and WNT receptors 75.
Secreted inhibitors that target co-receptors LRP5/6 also exist. The Dickkopf (DKK) family of proteins can inhibit WNT/β-catenin signaling by competitively binding to the WNT co-receptors LRP5/6 76. It was suggested that DKK1 may inhibit WNT signaling via inducing LRP6 internalization or degradation through transmembrane Kremen (Krm) proteins 77, which was not however supported by recent biochemical and genetic studies 78-80. WISE and SOST constitute another family of LRP5/6 ligands/antagonists 81-83. SOST can disrupt WNT-induced Fz-LRP6 complex in vitro 83. Both DKK1 and SOST are strongly implicated in human diseases 22, 84, 85. It is noteworthy that none of the above naturally occurring secreted inhibitors has been identified in Drosophila.
CROSSTALK BETWEEN WNT AND OTHER MAJOR SIGNALING PATHWAYS
Given the wide spectrum of WNT effects on target cells, crosstalks with other signaling pathways play an important role in fine-tuning modulation of WNT signaling during development and in maintaining tissue homeostasis. Here, we discuss several well-established cross-talks between WNT signaling and other signaling pathways.
Some Growth Factor Signaling Pathways Can “Hijack” β-Catenin Signaling Activity
It is conceivable that any cellular signaling that can stabilize the β-catenin protein should be able to activate the downstream events regulated by the β-catenin-TCF/LEF transcriptional complex. As the essential mediator of the canonical WNT pathway, β-catenin is aberrantly activated in a multitude of human cancers without any known mutations in the upstream components of the pathway or any increase in WNT expression, suggesting that non-WNT factors may be also capable of activating β-catenin. In fact, several growth factor and developmental signaling pathways, such as hepatocyte growth factor (HGF) 86-89, epidermal growth factor (EGF) 90-92, insulin-like growth factors (IGFs) 93-98, vascular endothelial growth factor (VEGF) 99, 100, and fibroblast growth factors (FGFs) 101-106, have been found to cause the accumulation/stabilization of the β-catenin protein and/or to activate β-catenin activity.
For example, it was shown that EGFR activation involves a kinase signaling cascade that leads to the dissociation of β-catenin from α-catenin at the adherens junction and the eventual nuclear translocation of β-catenin 90, although EGF-induced activation of β-catenin may also involve histone deacetylase 6 107 or embryonic pyruvate kinase M2 (PKM2) 92. The HGF receptor Met is involved in phosphorylation of β-catenin at tyrosine residues 654 and 670 with subsequent nuclear translocation of β-catenin 87. A significant correlation exists between expression of c-Met and abnormal β-catenin expression in invasive breast carcinoma, implicating crosstalk between the two in breast cancer 88. IGF1 or insulin treatment increased β-catenin/TCF-mediated transcription and cytoplasmic stabilization of β-catenin 94, while IGF-1 also induced stabilization of β-catenin in prostate cancer and early melanoma cells 97, 98. FGF19 was shown to increase β-catenin transcriptional activity in colon cancer cells and in transgenic mice104, 105, whereas co-activation of the WNT and FGF pathways in colorectal carcinogenesis leads to a more malignant phenotype 102. Interestingly, FGFs also cooperate with Wnt1 in MMTV-induced mammary tumorigenesis 101, 103, 106. Nonetheless, molecular mechanisms for these cross-talks have yet to be fully elucidated.
Crosstalk between the WNT and TGFβ/BMP Signaling Pathways
The TGFβ family includes TGFβ and the bone morphogenetic proteins (BMPs), which share a canonical signaling cascade involving two types of receptors (type I and II) and a common set of signal transducers known as Smads 108-113. Upon TGFβ or BMP binding to the type II receptor, the type I receptor is recruited to form a heterodimer complex, leading to the phosphorylation of the C-terminus of receptor-regulated Smads (R-Smads) (Smad 1, 2, 3, 5, and 8), which are then able to complex with the common Smad4. The Smads complex then translocates to the nucleus, binds to DNA, and regulates target gene expression 108-113.
Crosstalk between the WNT and TGFβ/BMP pathways is quite complex and occurs at multiple levels. The ligand production for the two pathways is under regulation by each other. For example, BMP4 expression in human colon cancer cells is dependent on the expression of oncogenic β-catenin 114. Conversely, BMP2/4 is capable of regulating production of WNT8 115. Smad and β-catenin/TCF can form a complex that binds to DNA and regulates shared target gene expression during development 116, 117. WNT3a and BMP4 synergistically induce expression of common target genes, such as Id2, Msx1, and Msx2 50. Many genes have been identified that harbor both Smad and TCF/LEF binding sites within their regulatory sequences, including Tbx6, Msx2, Xtwin, Emx2, Slug, c-Myc 118-123. TGFβRII knockout specifically in the stromal cells leads to increased expression of WNT3a and development of prostatic cancer 124, 125. Furthermore, it has been shown that Smad7 can directly bind to β-catenin and induce its degradation by the recruitment of Smurf2 126. Smad7 was found to physically associate with β-catenin, leading to an accumulation of β-catenin in prostate cancer cells 127. Smad7 can also directly bind to AXIN, which induces disassembly of the degradation complex and subsequent stabilization of β-catenin at the adherens junction 128. It has been recently reported that antagonism between WNT and BMP at the cytoplasmic level can be mediated by a direct interaction of DVL and phosphorylated Smad1 129. Therefore, the crosstalk between TGFβ/BMP and WNT signaling pathways can be either synergistic or antagonistic depending on the cellular context 130.
Crosstalk between the WNT and Notch Signaling Pathways
Notch signaling pathway is a highly conserved mechanism of intercellular communication and essential for development, patterning, and tissue homeostasis 131-133. Notch signaling is transduced through direct cell-to-cell contact and requires activation at the cell surface by ligands of the DSL (Delta and Serrate/Jagged) family. The ligand-receptor interaction causes cleavage of the Notch receptor through intramembrane proteolysis and yields the Notch intracellular domain (NICD), which then translocates to the nucleus and activates downstream target genes through transcription factor RBPj and co-factors such as Mastermind 132, 133.
Crosstalk between the Notch and WNT pathways has been observed in many developmental and cellular processes, such as somitogenesis, intestinal epithelial cell fate, and hematopoietic stem cell maintenance 134-138. Notch can repress WNT signaling during development and homeostasis by associating with β-catenin 139, while WNT activation can antagonize Notch signaling through DVL 140. Numerous reports have detailed an opposing role for the WNT and Notch pathways in tumorigenesis as the deletion or inhibition of Notch results in basal cell carcinoma 141. Activation of the Delta-Notch pathway inhibits the WNT pathway in neuroblastoma cells 142. Notch activation in a human tongue cancer cell line suppressed WNT signaling and led to cell cycle arrest and apoptosis 143. The physical interaction between β-catenin and the cytoplasmic tail of membrane bound Notch resulted in degradation of β-catenin protein 39.
Nonetheless, WNT and Notch pathways seem to work synergistically in intestinal tumorigenesis adenomas as activation of Notch in APC mutant mice hyperactive for WNT signaling accelerates adenoma development 144. WNT and Notch appear to operate synergistically in other cancer types, such as liver cancer 145, prostate cancer 146, breast cancer 147, 148, and leukemia 134. Furthermore, Notch ligand Jagged1 can be transcriptionally activated by β-catenin 149. Nonetheless, detailed mechanisms underlying the crosstalk between WNT and Notch pathways remain to be fully elucidated.
Crosstalk between the WNT and Hedgehog Signaling Pathways
The Hedgehog pathway is essential for tissue growth, patterning, and morphogenesis 150. The three mammalian Hedgehogs (Hh), Sonic, Indian, and Desert hedgehog (Shh, Ihh, and Dhh) are secreted proteins that undergo cleavage and lipid modification to become active signaling molecules 150, 151. Binding of Hh ligand to its receptor, Patched, de-represses the transmembrane protein Smoothened (Smo), therebyinducing a signaling cascade which results in stabilization of Gli transcription factors 152, 153, which in turn regulate the expression of target genes 150, 151, 153. In basal cell carcinomas (BCC)s, elevated levels of WNT pathway components were detected in response to Hh signaling abnormalities and Gli1 expression, suggesting a requirement for ligand-driven, canonical WNT/β-catenin signaling for Hh pathway-driven tumorigenesis 154, 155. Elevated Gli1 expression was shown to lead to accumulation of nuclear β-catenin in endometirial cancers 156. WNT/β-catenin signaling induces expression of an RNA-binding protein, CRD-BP, leading to stabilization of Gli1 mRNA and increased Hedgehog signaling and survival of colorectal cancer cells 157. Inhibition of Smo rescues the lethality caused by the loss of APC in mice, suggesting Hh may be activated in parallel with or downstream of WNT signaling 158. Conversely, Hh signaling was shown to positively regulate the WNT pathway 159. Reduced expression of Smo in APC mice suppressed β-catenin-dependent transcription in intestinal adenoma cells independently of the canonical Hh pathway 160. Activation of the Hh pathway through Smo or Gli2 increases WNT activity in pancreatic adenocarcinoma 161. However, it was also reported that Ihh acts as an antagonist of WNT signaling in colonic epithelial cell differentiation 162. WNT and Hh signaling was also found to be inversely correlated in gastric cancer specimens as overexpression of Gli1 suppressed WNT signaling in a gastric cancer cell line 163. Therefore, the disparate regulations of WNT and Hh signaling pathways may be cell or tissue-specific or context-dependent and remain to be full understood.
Crosstalk between the WNT and Hippo/YAP/TAZ Signaling Pathways
The Hippo pathway is a potent regulator of cellular proliferation and differentiation, and has emerged as a crucial regulator of tissue development and homeostasis 164-168. The Yes-associated protein (YAP)/transcriptional coactivator with a PDZ-binding domain (TAZ) are the prime mediators of the Hippo pathway 166. Activation of the Hippo pathway leads to the phosphorylation and cytoplasmic retention of YAP/TAZ. While the nuclear localization of YAP/TAZ is essential for the transcriptional activities of the Hippo pathway, nonnuclear TAZ appears to be crucial for the regulation of canonical WNT signaling 169. The Hippo pathway seems to restrict WNT/β-catenin signaling by promoting an interaction between TAZ and DVL. Cytoplasmic YAP may also counterbalance the effect of WNT signaling by limiting DVL activity 170. The Hippo and WNT pathways also cooperate in the nucleus, where YAP interacts with β-catenin and induces the expression of canonical WNT target genes 171. It has been reported that YAP and TAZ are integral components of the β-catenin destruction complex that serves as cytoplasmic sink for YAP/TAZ 172. In WNT-ON cells, YAP/TAZ are physically dislodged from the destruction complex, allowing their nuclear accumulation and activation of WNT/YAP/TAZ-dependent biological effects 172. In WNT-OFF cells, YAP/TAZ are essential for β-TrCP recruitment to the complex and β-catenin inactivation172. However, a recent study indicates that APC can regulate Hippo-YAP signaling in a β-catenin destruction complex-independent fashion during intestinal tumorigenesis 173. In fact, activation of YAP is a general hallmark of tubular adenomas from FAP patients; and APC was shown to function as a scaffold protein that facilitates the Hippo kinase cascade by interacting with Sav1 and Lats1 173. More surprisingly, a recent study suggest that YAP/TAZ may function as bona fide downstream effectors of the alternative WNT-YAP/TAZ signaling pathway as WNT5a/b and WNT3a were shown to induce YAP/TAZ activation independent of canonical WNT/β-catenin signaling 174. The so-called “alternative WNT-YAP/TAZ signaling axis” consists of WNT-FZD/ROR-Gα12/13-Rho GTPases-Lats1/2 and outcomes include YAP/TAZ activation and TEAD-mediated transcription, leading to the fulfillment of YAP/TAZ-mediated biological functions of alternative WNT signaling, such as gene expression, osteogenic differentiation, cell migration, and antagonism of WNT/β-catenin signaling 174. While these findings define a G protein–mediated pathway for WNT signaling to YAP/TAZ, it remains to be fully understood to what extent such alternative pathway transduces either canonical and/or non-canonical WNT signaling.
WNT/β-CATENIN SIGNALING AND STEM CELLS SELF-RENEWAL
Emerging evidence has established the important and wide-range roles of canonical WNT signing in stem cell self-renewal and/or lineage-specific differentiation in diverse tissues and cell types in vivo 27. Given the short-range feature of the signaling gradient, WNT signals can function as ideal stem cell niche factors, which may control immediately adjacent stem cell, leading to the parsimonious control of progenitor cell fate 27, 175. Here, we discuss the roles of canonical WNT signaling in regulating several well-characterized stem cell systems.
WNT/β-Catenin Signaling in Embryonic and Pluripotent Stem Cells
Embryonic stem cells (ESCs) are generated from the inner cell mass (ICM) of the blastocyst, and possess the pluripotent capacity to retain their ability to make all cell types within the organism 176. WNT/β-catenin pathway is required for the establishment and self-renewal of ESCs cells 27, 175, 177. WNT3a or inhibitor of GSK3β was shown to promote the formation of ESC-like colonies 178. WNT/β-catenin signaling stimulates self-renewal by inhibiting the repressor activity of endogenously expressed TCF3, while WNT/β-catenin activation may also result in differentiation 27, 175. The effects of APC mutation or endogenous GSK3β preventing activation of WNT/β-catenin signaling result in unable to differentiate normally in either embryoid body (EB) or teratoma differentiation assays 179. WNT/β-catenin activation and GSK3β inhibitors can enhance somatic cell reprogramming and iPSC formation 180. The efficiency of WNT/β-catenin-stimulated reprogramming appears to be stage-dependent; and TCF3/4 and LEF1/TCF1 act temporally in this process. Pluripotency can be maintained as long as conditions favor expression of the core transcription factors Oct4, Sox2, and Nanog. β-Catenin was shown to interact with reprogramming factors Klf4, Oct4, and Sox2, further enhancing the expression of pluripotency related genes 27, 175, 181.
WNT/β-Catenin Signaling in Mesenchymal Stem Cells
Mesenchymal stromal cells (MSCs) derived from stroma of bone marrow (BM), adipose tissue, or placental tissue, have the potential to differentiate into multiple cell types 22, 111, 112, 182. Canonical WNT signaling plays a critical role in regulating cell fate decisions of MSCs. The activation of canonical WNT pathway can promote the osteogenic differentiation of MSCs by upregulating the expression of Cbfa1/Runx2 and alkaline phosphatase (ALP) 85, 183. Canonical WNT/β-catenin signaling was shown to induce overlapping target genes and to act synergistically with osteogenic BMPs in inducing osteogenic differentiation of MSCs 184-188. The adipogenic differentiation is enhanced in the absence of WNT signaling as activation of β-catenin via ectopic expression of WNT1 was shown to directly suppress PPARγ expression and prevent 3T3-L1 adipogenic differentiation 189. GSK3β mediates WNT inhibition of adipogenesis interfering with PPARγ transcriptional activation 190. In the cardiac differentiation process of MSCs, the WNT/β-catenin signaling pathway had been inhibited 191. Blocking the WNT/β-catenin signaling can enhance MSC-based granulation tissue formation and myocardial repair 192. Canonical WNT signals distinctively regulate MSCs in a biphasic manner depending on signal intensity. The proliferation and self-renewal of MSCs were promoted only under low levels of WNT/β-catenin, whereas osteogenic differentiation was promoted under high levels of WNT signaling 193.
WNT/β-Catenin Signaling in Intestinal Stem Cells
Intestinal stem cells have been demonstrated to be divided into two populations: thoase located at the base of the crypt and those at the position 4 from the base of the crypt 27, 194. The position 4 cells characterized by the stem cell marker Bmi1 are thought to be quiescent, slow cycling and apparently activated only during injury 194-196. The crypt base columnar (CBC) cells residing at the base of the crypt are rapid cycling and responsible for sustained tissue homeostasis, which can be identified by the expression of Lgr5 196, 197. WNT/β-catenin signaling is required for proper stem cell maintenance and differentiation in the intestine 197. The expressions of several WNT ligands and receptors (WNT3, 6, 9b, FZD 4, 6, 7, LRP5, SFRP5) are detected in epithelial cells of the intestinal crypt 198. WNT antagonist SFRP5 highly expressing in +4 cell surrounding area has been associated with the regulation of the stem cell niche in the intestine 199. Numerous target genes of the WNT/β-catenin pathway have been identified in the intestine. Sox9 as both a transcriptional target and a regulator of the WNT pathway has been shown to be required for paneth cell differentiation 200. The transcription factor achaete scute-like 2 (Ascl2) acts as an R-spondin/WNT-responsive gene and regulates the expression of the genes essential to the stem cell state together with β-catenin /TCF 201. EphB2/3 is required for the correct positioning of cells in the intestinal epithelium controlled by β-catenin and TCF 202. Notch and WNT signaling are required both for stem cell maintenance and for a proper balance of differentiation between secretory and absorptive cell lineages 194. In the absence of Notch signaling, stem cells preferentially generate secretory cells at the expense of absorptive cells as blocking Notch signaling disturbs the normal function of the intestine stem cells and lead to the mis-expression of prosecretory genes by inhibiting the WNT signaling pathway 194, 203.
WNT/β-Catenin Signaling in Hematopoietic Stem Cells
WNT signaling pathway plays a key role in the early stage of hematopoiesis 23, 175. WNT/β-catenin signaling is indispensable in the formation of vascular endothelial cells to hematopoietic stem cell (HSC) transformation process on early stage of hematopoiesis 21, 27. The WNT signaling is activated in the early stage of embryonic formation of red blood cells. Activation of the WNT/β-catenin pathway can increase the specific markers of pronormoblast and induce the formation of hematopoietic progenitors (MPP) 204. Knockout of WNT3a in mouse decreased the hematopoietic stem/progenitor cells in the fetal liver 205. Overexpression of activated N-terminal truncation β-catenin promoted the expansion of HSC 206. WNT/β-catenin inhibitors, DKK1 and Wif1, can disrupt the quiescent state of HSCs and result in the loss of HSC self-renewal and decrease hematopoietic reconstitution 207, 208. Survivin expression, which is regulated by WNT/CBP/β-catenin, is important during hematopoiesis and is prominently up-regulated in CD34+ hematopoietic stem/progenitor cells upon growth factor treatment, as survivin-deficient hematopoietic progenitors were shown to have defects in erythroid and megakaryocytic lineage formation 209, 210. WNT/β-catenin signaling, together with other pathways, such as Notch, PGE2, and BMPs, is important for maintaining the hematopoietic lineage balance. It was shown in zebrafish that the timed WNT to Notch relay signaling serves as an early upstream mechanism in HSC specification 211, 212. After the formation of mesoderm, the BMP signaling activates WNT signaling pathway and Cdx-Hox to promote hematopoiesis 211, 212. The BMP and WNT signaling pathways were shown to regulate hematopoiesis related genes and erythroid differentiation through the transcription factors Smad1 and TCF after acute injury of hematopoietic system 213.
WNT/β-Catenin Signaling in Hair Follicle Stem Cells
Hair follicle stem cells residing in the hair follicle (HF) are quiescent when the follicle is resting, but rapidly expand and differentiate response to hair periodical regeneration, maintenance of adult skin homeostasis and wound repair 214, 215. WNT/β-catenin signaling is required for embryonic HF morphogenesis 215. Forced activated β-catenin signaling converts embryonic ectoderm to hair follicle fate. The expression of nuclear β-catenin is described in hair germ progenitor cells at anagen onset, in hair follicle precursor cells during anagen, but undetectable in telogen HFs 216. Conditional loss of β-catenin in skin epithelia leads to hair follicle stem cells depletion, whereas hair follicle stem cells-specific ablation inhibits the proliferation of hair germ progenitor cells and fate specification of bulge stem cells 217. TCF3 and TCF4 are present in quiescent stem cells, where WNT/β-catenin activity is silent 218. Elevation of WNT/β-catenin depresses TCF3/TCF4/TLE-bound target genes, including chromatin-repressed genes, and then activates LEF1 to drive the progenitor cells along the hair differentiation lineage 219. Pygo2 was shown to function as an important regulator of WNT/β-catenin function in skin epithelia and β-catenin-induced activation of hair follicle stem/early progenitor cells 220. Furthermore, it is well established that canonical WNT signaling regulates the fate of hair follicle stem cells in concert with other signaling pathways in their niches. Notch ligand Jagged-1 was shown to be a WNT/β-catenin target gene in hair follicle formation of the adult epidermis 149. The antagonistic competition between BMP and WNT signaling balances hair follicle stem cell activity, as reduced BMP signaling and increased WNT signaling activated hair follicle stem cell toward hair fate and hair follicle cycle 221. WNT7b as a putative target of canonical BMP signaling serves as a key component required for normal hair follicle stem cells activation during the telogen-anagen transition 222.
WNT/β-CATENIN SIGNALING AND TUMORIGENESIS
Given the important roles and pleiotropic effects of canonical WNT signaling in virtually every organ system in normal tissue homeostasis and tissue injury repair, it is expected that dysregulation of this signaling pathway would be associated with a large array of human diseases, including neurological diseases, inflammatory and fibrotic disease, and disorders of endocrine function and bone metabolism in adults 22, 23, 27, 85. Here, we focus on the consequences of aberrant regulations of WNT/β-catenin signaling in the development of human cancers.
Aberrant Activation of WNT/β-Catenin and Tumorigenesis
WNT1 was initially discovered as a potential oncogene in mouse mammary glands, which was further substantiated by the fact that WNT1 transgenic mice developed mammary tumors 1, 223. These early studies strongly suggest a causative role for WNT1 in mammary tumorigenesis. Later studies demonstrated a pivotal relation between hyperactivated WNT/β-catenin signaling pathway and the initiation of colorectal cancer 224, 225. The high frequency of mutations in various components of WNT pathway in many types of human cancers further highlights the importance of activation of WNT/β-catenin signaling in tumorigenesis 226, 227. Germline inactivating mutations in APC, resulting in nuclear accumulation of β-catenin stability, are found in patients with familial adenomatous polyposis (FAP) 13, 228, while a nonsense mutation in the coding region of the APC gene causes multiple intestinal neoplasia (Min) phenotype in mice 229.
Dysregulation of the WNT/β-catenin pathway has also been widely found in non-colorectal cancers 18. For example, it was reported that up to 44% and 25% of hepatocellular carcinoma (HCC) tumors contain mutations of β-catenin in exon 3 or mutations in AXIN1, respectively 230, 231. Oncogenic mutations of β-catenin are commonly found in human skin cancers, including melanoma 232. Increasing evidence indicates that WNT/β-catenin signaling is involved in pancreatic ductal adenocarcinoma (PDAC) tumorigenesis 161, 233, 234. The results of pancreatic circulating tumor cell RNA studies implicated that WNT2 expression was up-regulated, suggesting WNT2 may be associated with PDAC metastasis 235. It is noteworthy that many types of human cancers exhibit nuclear and/or cytoplasmic β-catenin accumulation, indicating the activation of the canonical WNT pathway without any identifiable mutations in APC, AXINs, β-catenin, or other components of the canonical WNT pathway 232, 236. Furthermore, as one of the hallmarks of tumorigenesis telomerase is regulated by β-catenin 237. Conversely, WIF1, a component of the WNT pathway and a competitive inhibitor of WNT pathway, was down-regulated in prostate, breast, lung, bladder cancer and osteosarcoma 238, 239.
As discussed earlier, Yes-associated protein 1(YAP1) was show to be essential to the survival and transformation of β-catenin-active cancer cell lines, and YAP is induced by β-catenin in colorectal cancer cells and is up-regulated in APC mutant colorectal cancer cells 240, 241. The role of YAP and TAZ as mediators of WNT signaling is further supported by the findings from an animal model in which showed that both YAP and TAZ were required for loss of APC-induced crypt hyperplasia 172. Nonetheless, a recent study revealed that YAP is required for the development of APC-deficient adenomas, but APC functions as a scaffold protein to facilitate the Hippo kinase cascade by interacting with Sav1 and Lats1, which is independent from its involvement in the β-catenin destruction complex 173. These findings indicate that while the causative role of aberrantly activated WNT/β-catenin signaling in human cancer development is well established, the detailed molecular mechanisms underlying WNT/β-catenin signaling in tumorigenesis are far from being clearly understood.
WNT/β-Catenin Signaling and Cancer Metastasis
Cancer metastasis is a complex multistep process involving breaking through the extracellular matrix and basement membrane at the primary tumor sites 242, 243. WNT /β-catenin pathway related gene and target gene is associated with tumor invasion and metastasis, such as matrix metalloproteinase (MMP) 7, CD44, vascular endothelial growth factor, E-cadherin 244, 245. E-cadherin/β-catenin complex mediated cell adhesion is to establish and maintain normal polarity and cell tight junction of epithelial cells 244. Epithelial to mesenchymal transition (EMT) are known about the epithelial plasticity that are important in cancer metastasis 246-248. Activation of WNT/β-catenin signaling leads to the nuclear translocation of β-catenin to disturb the E-cadherin/β-catenin complex, contributing to the EMT process and cancer metastasis 246-248. WNT/β-catenin activity usually up-regulates the expression of EMT-promoting genes, including SNAI1/Snail 1, SNAI2/Snail 2 (also known as Slug), ZEB1, ZEB2, E47 and KLF8 247, 248. Activation of the canonical WNT/TCF pathway through LEF1 and HOXB9 was also identified as a determinant of metastasis to brain and bone during lung adenocarcinoma progression 249. Furthermore, the WNT /β-catenin signaling can up-regulate the expression of cyclooxygenase-2 (Cox2) to promote tumor angiogenesis, which subsequently promotes tumor metastasis. It was also reported that the metastasis-stroma interaction in human breast cancer metastasis was regulated by the HGF/nuclear Met/phospohho-c-Src/β-catenin-TCF/WNT pathway 250.
WNT/β-Catenin Signaling and Cancer Stem Cells
The WNT/β-catenin pathway is also involved in the regulation of cancer stem cells (CSCs) from many tissue types 23, 175, 194, 237. Many of the cancer stem cell surface markers, such as LGR5/GPR49, CD44, CD24, and Epcam, which are used to identify and isolate putative cancer stem cell populations in a variety of tissues, are WNT target genes 23, 175, 194. In breast cancer LGR5-expressing cells exhibit cancer stem cell-like properties, including the formation of self-renewing spheres and high tumorigenicity by activating WNT/β-catenin signaling 251. WNT3a can promote the self-renewal of cancer stem/progenitor cells in acute lymphocytic leukemia (ALL) and prostate cancer 252, 253. CD44 is closely associated with tumor growth, invasion, metastasis as an important tumor stem cell marker 254. In human colon cancer cell line LT97, CD44 positive cells were detected with the expression of nuclear β-catenin, while CD44 negative cells exhibited no nuclear β-catenin 255. However, it remains to be fully elucidated if WNT/β-catenin signaling regulates normal stem cells/progenitor cells versus cancer stem cells.
TARGETING WNT/β-CATENIN SIGNALING FOR CANCER TREATMENT
The broad involvement and pleiotropism of WNT signaling in stem cells and human diseases has attracted extraordinary amounts of interests in the development of novel strategies targeting this signaling pathway 21, 26, 256-260. One of the earliest such efforts involved the reintroduction of wild type APC into human colorectal cancer cell lines, which induced growth inhibition and apoptosis of the cancer cells 261. Similarly, expression of AXIN I also promoted apoptosis in cancer cell lines containing mutations in either β-catenin, APC or AXIN I 231. These experiments strongly suggest that therapeutic intervention targeting WNT signaling can be developed for anticancer therapies.
For the past decade, significant progresses have been made for the past decade in identifying the druggable targets of the WNT pathway and/or in developing novel small molecules that specifically target WNT/β-catenin signaling 26, 257-260. Although most of these drugs have not yet progressed to evaluation in clinical trials (Table 1), current genomics and proteomics studies enable more targeted approaches for high-throughput screening of the WNT/β-catenin pathway, which is expected to deliver clinical drugs in the coming decade. Here, we primarily focus on the recent development of potential anticancer therapies by targeting the WNT signaling pathway. As the WNT pathway lends itself ample targeting nodal points for drug development, numerous efforts have been devoted to targeting WNT signaling at different regulatory levels of the signaling cascade.
Table 1.
Currently Known Inhibitors of the Canonical WNT Signaling Pathway
Molecular Targets | Inhibitors | Anti-WNT and Anticancer Activities | Stage of Development | References |
---|---|---|---|---|
WNTs | Antibodies | WNT-blocking antibodies were developed and shown to inhibit proliferation and induce apoptosis in different cancers | preclinical | 21, 262-265 |
SFRPs/WIF | overexpress naturally occurring antagonists of WNT ligands | 266-268 | ||
SFRP peptides | SFRP1 and SFRP1-derived peptides can delay HCT116 xenograft tumor formation | 266 | ||
DNA demethylation agents | use DNA demethylation agents to reverse hypermethylation of SFRP promoters | 273 | ||
FZDs | OMP18R5 | humanized monoclonal anti-body that binds to FZD1, FZD2, FZD5, FZD7 and FZD8 | Phase Ia | 28 |
FZD8CRD | fusion protein consisting of the Fc region of IgG fused to the extracellular domain of FZD8 | preclinical | 270-271 | |
DVL | NSC668036 | inhibits the DVL PDZ domain, not reported in cancer | Preclinical | 274-276 |
3289-8625 | inhibits the growth of prostate cancer PC-3 cells | |||
FJ9 | disrupts the interaction between FZD7 and the PDZ domain of DVL, induces apoptosis and inhibits H460 lung cancer growth | |||
sulindac | inhibits proliferation of lung cancer A549 cells | FDA Approved | 300, 301 | |
TANKs | XAV-939 | inhibits colony formation of β-catenin-dependent DLD-1 cells | Preclinical | 277 |
JW55 | decreases canonical Wnt signaling in SW480 and HCT-15 colon carcinoma cell lines; reduces cell cycle progression and proliferation in SW480 cells in vitro | 279 | ||
G007-LK | suppresses APC mutation-driven colorectal tumor growth | 280 | ||
IWR-1 | inhibits L-cells expressing Wnt3A | 278 | ||
PORC | IWP | inhibits colorectal cancer cells invasion by WISP2 | Preclinical | 278 |
LGK-974 | inhibits growth of Mouse MMTV-WNT1 tumor model and human head and neck squamous cell carcinoma model | Phase I clinical trial | 259, 282 | |
WNT C59 | PORC inhibitor with 10-fold therapeutic dose over toxic dose | Preclinical | 283 | |
Activation of CK1α to promote β-catenin degradation | Pyrvinium | Pyrvinium synergizes with 5-fluorouracil in mediating the apoptosis of SW620 colorectal cancer cells and inhibits the proliferation of SW480 and HCT116 cells. | Preclinical | 317 |
β-catenin/TCF interaction | iCRT3, iCRT5, and iCRT14 | reduced the growth of colorectal cancer cells | Preclinical | 288 |
PKF115-584, CGP049090 and PKF118-310 | inhibit the growth of HCC cells in xenografts | 284-286 | ||
2,4 diamino-quinazoline | inhibitor lead of the β-catenin-TCF4 pathway | 287 | ||
PNU-74654 | a druglike β-catenin-TCF antagonist | 289 | ||
BC21 | an organo-copper complex as the top-ranked compound that can bind to the armadillo repeat | 290 | ||
AV-65 | inhibits progression of multiple myeloma in a mouse model | 291 | ||
stapled peptides | potent inhibitors to target the β-catenin-BCL9 interface and and the β-catenin-TCF4 interface | 292, 293 | ||
β-catenin/CBP interaction | ICG-001 | decreases xenograft growth of SW620 colon carcinoma cells | Preclinical | 294, 295 |
PRI-724 | downregulates survivin (BIRC5) expression in circulating tumour cells, supresses growth of refractory pancreatic cancer | Phase Ia | 259, 302 | |
β-catenin/TCF-regulated transcription; signal crosstalk; non-specific or overlapping targeting | CCT036477, CCT070535, and CCT031374 | Inhibits the growth of SW480 and HCT116 colorectal cell lines | Preclinical | 26 |
OSU03012 | PDK1 inhibitor OSU03012 inhibits the growth of various medulloblastoma cell lines | 307 | ||
Celecoxib | induces apoptosis in cervical cancer cells | FDA Approved | 303,304 | |
Imatinib | tyrosine kinase inhibitor that inihibits TCF/β-catenin activity | FDA Approved | 305 | |
PHA665752 | c-MET inhibitor shown to inhibit WNT/β-catenin signaling | Preclinical | 306 | |
OSU03012 | PDK1 inhibitor to suppress medulloblastoma xenograft tumors | 307 | ||
IQ-1 | protein phosphatase 2A (PP2A) regulatory subunits PR72 and PR130 shown to maintain pluripotency in murine ES cells in a WNT-dependent manner | 296 | ||
ID-8 | dual specificity YAK1 related kinases (DYRKs) shown to allow for long-term WNT-mediated maintenance of human ES cells | 297 | ||
Retinoic acids | induces Disabled homolog 2 (DAB2) | 308 | ||
Vitamin D | induces DKK1 and DKK4 | 308 | ||
Natural products | quercetin, epigallocatechin-3-gallate (EGCG), curcumin, resveratrol, ginsenoside Rg3, and tetrandrine | 308-314 | ||
Silibinin | suppressing LRP6 expression in human prostate and breast cancer cells | 315 | ||
Rottlerin | shown to induce LRP6 degradation and suppress both WNT/β-catenin and mTORC1 signaling pathways in prostate and breast cancer cells | 316 | ||
Niclosamide | promoting FZD1 endocytosis, down-regulating DVL2 protein, and inhibiting WNT3A-stimulated β-catenin stabilization and LEF/TCF activity; anticancer activity in WNT-independent manner as well | FDA Approved | 319-325 | |
Salinomycin | blocking the phosphorylation of LRP6 and induce its degradation | Preclinical | 326 | |
Monensin | a potent blocker of WNT-induced transcription and to inhibit the progression of intestinal tumors without any sign of toxicity on normal mucosa | 327 |
Targeting WNT Signaling at Extracellular Level
Directly targeting WNT ligands may prove to be an attractive strategy for targeting WNT signaling preferentially in cancer cells that exhibit aberrantly overexpressed WNTs. Several WNT-blocking antibodies were developed and shown to inhibit proliferation and induce apoptosis in different cancers 21, 262-265. Intraperitoneal injections of WNT3A-neutralizing antibodies decrease proliferation and induce apoptosis in a mouse model of prostate cancer 125.
FZD receptors are another class of logic targets for developing WNT-targeting biologics. One such agent OMP18R5 was developed by OncoMed Pharmaceuticals and is a humanized monoclonal antibody that binds to FZD1, FZD2, FZD5, FZD7 and FZD8 26. OMP18R5 recently completed the Phase Ia clinical trial in patients with advanced solid tumors 26. A total of 18 patients were treated and, the most common drug-related adverse events included fatigue, vomiting, abdominal pain, constipation, diarrhea and nausea. There were three cases of prolonged stable disease in patients with neuroendocrine tumors 26.
The naturally occurring FZD receptor antagonists SFRPs are logic agents to target WNT signaling. These factors are extracellular inhibitors that bind directly to WNT ligands or to Frizzled receptors. The SFRP1 or SFRP1-derived peptides were shown to delay HCT116 xenograft tumor formation in nude mice and reduced the proportion of mitotic 266. WNT inhibitory factors (WIFs) are also secreted proteins that competitively displace certain WNT ligands from their receptors. Overexpression of WIF1 was shown to inhibit osteosarcoma cell growth in soft agar assays and in xenograft assays 267. It's noteworthy that SFRPs may regulate cell proliferation of some cancer cells, such as prostate cancer cells, in a context-dependent manner, as the overexpression of SFRP4 or SFRP3 decreases the proliferation of human PC3 cells 268, whereas the overexpression of SFRP1 promotes the growth of BPH1 prostate cancer cells 269. As SFRPs and WIFs are associated with multiple WNTs, it's conceivable that altering SFRP and WIF levels may have pleiotropic effects on cancer cell proliferation.
Alternatively, a competitive inhibition of WNT signaling can be achieved by overexpression of the secreted forms of FZD receptors. In fact, it was reported that administration of a fusion protein consisting of the Fc region of IgG fused to the extracellular domain of FZD8 (FZD8CRD) inhibited the formation of tumor xenografts by two non-engineered cancer cell lines, the N-TERA2 human testicular cancer line and the PA1 human ovarian cancer cell line 270. It was also shown that soluble FZD7 can inhibit WNT signaling and sensitize hepatocellular carcinoma (HCC) cells towards doxorubicin 271. Interestingly, the FZD7 peptides derived from the domains that interact with DVL effectively inhibited the growth of HCC cells 272.
Furthermore, since the SFRP genes are usually silenced by hypermethylation, it is conceivable drugs that affect the DNA methylation can be used to alter the methylation status of SFRP gene promoters and hence re-activate the expression of SFRPs. It was reported that aberrant epigenetic modification of SFRP gene was one of the major mechanisms by which WNT signaling is activated in human gastric cancer cells, and sodium butyrate can modulate the SFRP1/2 expression through histone modification and promoter demethylation, causing anti-tumor effects 273.
Targeting WNT Signaling at Cytoplasmic Membrane Level
DVL is an essential mediator in the WNT signaling pathway and transduces extracellular WNT signals to downstream components. DVL utilizes its PDZ domain to bind to the carboxyl-terminal region of the FZD receptors. Thus, binders to the PDZ domain of DVL proteins may disrupt the WNT signaling cascade. Three compounds, namely NSC 668036, FJ9 and Compound 3289–8625, were identified through in silico screening and nuclear magnetic resonance (NMR) spectroscopy approaches, and were shown to block WNT signaling in vivo 274-276. The inhibitor NSC668036 provided a basis for rational design of high-affinity inhibitors of the PDZ domain and can block WNT signaling by interrupting the FZD-DVL interaction 274. The inhibitor FJ9 can disrupt the interaction between FZD7 and PDZ domain of DVL 275. The Compounds 3289-8625 was identified as a small molecule inhibitor of PDZ domain of DVL, and was shown to suppress the growth of prostate cancer PC-3 cells 276. These results strongly suggest that blocking the PDZ domain of DVL may offer ample opportunities for developing effective and specific inhibitors of the WNT signaling pathway.
Targeting WNT Signaling Intracellular and Nuclear Levels
Targeting PORC and TANKS
Recent studies have demonstrated that porcupine (PORC) and tankyrases (TANKs) may serve promising drug targets of the WNT signaling pathway. PORC is a member of the membrane-bound O-acyltransferase family and adds a palmitoyl group to WNT proteins, which is essential to their signaling ability and is required for WNT secretion 34. Tankyrase 1 (TANK1) and tankyrase 2 (TANK2) are members of the larger family of poly(ADP-ribose) polymerase (PARP) enzymes. TANKs interact with a highly conserved domain of AXIN and promote its ubiquitylation and degradation 277. Chen et al identified and characterized two classes of several small molecules called IWRs (inhibitors of WNT response, such as IWR-1) that stabilize the protein AXIN and IWPs (inhibitors of WNT production, such as IWP-2) that inhibit the PORC acyltransferase activity 278. The IWP inhibitors can efficiently inhibit WNT pathway by disrupting the WNT ligand in colon cancer cell line, while IWR-1 was confirmed as a TANK inhibitor 278. Huang et al also identified another class of TANK inhibitors XAV939, which was shown to induce the stabilization of AXIN 277. Another tankyrase inhibitor JW55 was shown to decrease canonical Wnt signaling in colon carcinoma cells and to reduce tumor growth in conditional APC mutant mice 279. Similarly, the compound G007-LK displayed favorable pharmacokinetic properties and inhibited in vivo tumor growth in a subset of APC-mutant colorectal cancer xenograft models 280. Recent efforts have been devoted to the development of more potent and selective second-generation of TANK inhibitors 281. Meanwhile, a new PORC inhibitor LGK974 was shown to potently inhibit WNT signaling and exhibit strong efficacy in rodent tumor models, yet well-tolerated 282, which has entered a Phase I trial by Novartis 259. Another PORC inhibitor WNT C59 was shown to have 10-fold higher than the therapeutic dose to cause extensive loss of intestinal proliferation 283.
Targeting β-Catenin/TCF Transcription Complex
Effectively disrupting the protein-protein interaction between TCF/LEF and β-catenin via small molecules is attractive but technically challenging. Nonetheless, an early high-throughput ELISA-based screening assay of approximately 7,000 natural products and 45,000 synthetic compounds, which was confirmed by the bioassay for axis duplication in Xenopus laevis embryos, identified two structurally related compounds, PKF115-584 and CGP049090 284. Interestingly, both PKF115-584 and CGP049090 were shown to disrupt β-catenin-APC interaction as well 284. While these compounds have not advanced to clinical trials, they indeed show anti-WNT efficacy in preclinical models of hepatocellular cancers 285 and hematologic cancers 286. Another high-throughput screen of a large compound library, 2,4-diamino-quinazoline was identified as an inhibitor lead of the β-catenin-TCF4 pathway 287. A cell-based high-throughput screening in D. melanogaster cells with a WNT-responsive luciferase reporter was carried out to screen 14,977 compounds and identified three candidates, namely iCRT3, iCRT5 and iCRT14, which were shown to disrupt the β-catenin-TCF interaction in vitro and to inhibit the expression of WNT target genes with cytotoxicity in colorectal cancer cells 288. A combination of virtual and biophysical screening identified the synthetic compound PNU-74654 as a druglike β-catenin-TCF antagonist 289. A virtual screen of the 1,990 small-molecule diversity set of the US National Cancer Institute identified the organo-copper complex BC21 as the top-ranked compound that can bind to the armadillo repeat 290. AV-65 was identified by screening from a library of more than 100,000 small-molecule chemical compounds for novel Wnt/β-catenin signaling inhibitors and was shown to diminish β-catenin protein levels and TCF transcriptional activity, as well as to prolong the survival of multiple myeloma-bearing mice 291. More recently a stapled peptide approach was used to identify potent inhibitors to target the β-catenin-BCL9 interface 292, and the β-catenin-TCF4 interface 293. While many of the above inhibitors possess high translational potential, their biological activity profiles and/or mechanisms of action remain to be fully defined.
Targeting β-Catenin/TCF Co-Activators
β-Catenin/TCF complex needs to recruit the transcriptional co-activator CBP or p300 to regulate the expression of downstream target genes. A small molecule ICG-001 was identified to specifically bind to the co-activator CBP, but not p300, with high affinity 294, 295. Subsequently, several small molecules (IQ-1 and ID-8), which selectively block the p300-β-catenin interaction, were also identified 296-298. The therapeutic potential of ICG-001 was examined in several preclinical tumor models and was shown to safely eliminate drug-resistant tumor-initiating cells 299-301. Another specific CBP/β-catenin interaction inhibitor PRI-724 was developed by Prism Pharma and partnered with Eisai Pharmaceuticals and entered an open-label Phase Ia safety study in individuals with solid tumors 259, 302.
Targeting WNT Signaling by Nonspecific Inhibitors and Repurposed Drugs
Given the pleiotropic effects of the canonical WNT signaling pathway, it is conceivable many anticancer drugs and/or small molecule inhibitors may target WNT signaling as a part of their mode of action. For example, non-steroidal anti-inflammatory drugs (NSAIDs) and the selective COX2 inhibitor celecoxib were shown to inhibit β-catenin dependent transcription in colorectal cancer cells 303, 304. Other molecules, including CCT036477, CCT070535 and CCT031374, also showed their inhibitory abilities in the SW480 and HCT116 colorectal cell lines 26. The tyrosine kinase inhibitor imatinib (Gleevec; Novartis) and c-MET inhibitor PHA665752 were shown to inhibit WNT/β-catenin signaling 305, 306. It was shown that inhibitors of phosphatidylinositol 3-kinase (PI3K)/AKT signaling can inhibit WNT/β-catenin signaling cross-talk as PDK1 inhibitor OSU03012 suppressed the growth of established medulloblastoma xenograft tumors in a dose-dependent manner and augmented the antitumor effects of mammalian target of rapamycin (mTOR) inhibitor CCI-779 307. Inhibitor IQ-1 of the protein phosphatase 2A (PP2A) regulatory subunits PR72 and PR130 was shown to maintain pluripotency in murine embryonic stem cells in a WNT-dependent manner 296, whereas the inhibitor ID-8 of the dual specificity YAK1-related kinases (DYRKs) was shown to allow for long-term WNT-mediated maintenance of human embryonic stem cells 297. It has been reported that nuclear receptor ligands retinoic acids may induce Disabled homolog 2 (DAB2), whereas vitamin D may induce DKK1 and DKK4 308.
Derivatives for some natural products, such as quercetin, epigallocatechin-3-gallate (EGCG), curcumin, resveratrol, ginsenoside Rg3, and tetrandrine, have been reported as potential WNT signaling inhibitors 308-314. Silibinin, a natural compound isolated from milk thistle seed extracts, was shown to inhibit WNT/β-catenin signaling by suppressing LRP6 expression in human prostate and breast cancer cells 315. Another natural plant polyphenol, Rottlerin, was shown to induce LRP6 degradation and suppress both WNT/β-catenin and mTORC1 signaling pathways in prostate and breast cancer cells 316.
Several inhibitors of WNT signaling have been identified by drug-repurposing screening of the libraries of FDA-approved drugs. For example, the anti-helminthic drug pyrvinium was identified as an agent that potentiates CK1α activity and thus promotes degradation of β-catenin and the co-activator Pygopus, leading to a reduction in WNT/β-catenin signaling 317. Another anti-helminthic niclosamide was shown to promote FZD1 endocytosis, down-regulate DVL2 protein, and inhibit WNT3A-stimulated β-catenin stabilization and LEF/TCF activity 318. Niclosamide was also shown to suppress cancer cell growth by inducing LRP6 degradation and inhibiting the Wnt/β-catenin pathway 319, and niclosamide can inhibit tumor growth in human colon cancer xenograft model 320. More recently, niclosamide was shown to inhibit cell proliferation and/or tumor growth in ovarian cancers, breast cancer, prostate cancer, and osteosarcoma cells 321-325, although niclosamide's anticancer activity may be also mediated by inhibiting other signaling pathways 322-325. The antibiotic potassium ionophores salinomycin and nigericin were shown to block the phosphorylation of LRP6 and induce its degradation, thereby down-regulating WNT signaling 326. Interestingly, another polyether ionophore antibiotic, monensin, was shown to be a potent blocker of WNT-induced transcription in the cells stimulated with WNTs or GSK3 inhibitors and to inhibit the progression of intestinal tumors without any sign of toxicity on normal mucosa 327. These findings suggest that many small molecule inhibitors may function as WNT signaling modulators.
CONCLUSIONS AND FUTURE DIRECTIONS
It has been three decades since the ground-breaking discovery of WNT signaling as a fundamental and evolutionarily conserved pathway. For the past decade there has been a rapid expansion in our understanding about the regulatory circuitry and complexity of this pathway although many details involved in the essential aspects of WNT signaling mechanisms remain to be fully elucidated. Numerous new components of Wnt/β-catenin signaling, such as RSPOs, LGRs, NZRF3/RNF43, PORC, and TANKS, have been identified and linked to signaling regulation, stem cell functions and tissue homeostasis. It has been well-established that WNT signaling plays important roles in regulating cell self-renewal and differentiation in many types of stem cells and cancer stem cells. The lipid-modified WNT signals act primarily over short ranges to control stem cell behavior within the spatial confines of the niche, which implies that in particular tissues WNT-dependent stem cells are spatially restricted to the vicinity of the WNT-producing niche, physically delimiting the stem cell compartment and preventing uncontrolled stem cell expansion. The short-range action feature of WNT signaling may also account for the context-dependent nature of this pathway as emerging evidence suggests that WNT/β-catenin signaling, as well as β-catenin-independent WNT signaling pathway, can either promote or inhibit cancer progression in a context-dependent manner. Our better understanding of WNT signaling has opened numerous avenues and drawn significant interests for developing novel and effective drugs that may specifically target distinct steps of the WNT signaling pathway although their efficacy and toxicity remain to be fully evaluated.
Nonetheless, the detailed mechanisms underlying WNT signaling under physiological and pathological conditions are far from clearly understood. Future directions should be directed to address the following questions: How is the specificity of individual WNT ligand's interaction with FZDs and co-receptors determined? Through what mechanisms do WNTs interact with co-receptors, such as RORs and RYK? What are the upstream regulatory signals of WNT signaling? How extensively does WNT signaling cross-talk with other major signaling pathways and/or act through what detailed mechanisms? How many downstream target genes are regulated by individual WNTs and/or how these target genes are different in different cell/tissue types? How is canonical or non-canonical WNT signaling determined and at what level(s) of the pathway? How differently do the secreted antagonists interact with individual WNT ligands in determining to transduce canonical or non-canonical signaling? Can components, such as GSK3β, of WNT signaling be used or hijacked by other signaling pathways? How is β-catenin transported into the nucleus? Can we identify any bona fide, safe and effective WNT inhibitors and eventually move them to treat human diseases? With the rapid technological advances in genomics and systems biology, we expect to get satisfactory answers for many of the above questions in next 5-10 years.
Acknowledgments
We apologize to the investigators whose original work was not cited due to space constraints. The authors’ research efforts were supported in part by research grants from the NIH (AT004418, AR50142, and AR054381 to TCH, RCH and HHL), the 973 Program of Ministry of Science and Technology (MOST) of China (#2011CB707900 to TCH), and the National Natural Science Foundation of China (#81371718, #81272172, and #81100309 to KY, GN, and YB). MKM was a recipient of Howard Hughes Medical Institute Medical Research Fellowship.
Footnotes
Conflict of Interest
The authors declare no conflict of interest.
References
- 1.Nusse R, Varmus HE. Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell. 1982;31(1):99–109. doi: 10.1016/0092-8674(82)90409-3. [DOI] [PubMed] [Google Scholar]
- 2.Nusse R, Varmus H. Three decades of Wnts: a personal perspective on how a scientific field developed. The EMBO journal. 2012;31(12):2670–2684. doi: 10.1038/emboj.2012.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Nusslein-Volhard C, Wieschaus E. Mutations affecting segment number and polarity in Drosophila. Nature. 1980;287(5785):795–801. doi: 10.1038/287795a0. [DOI] [PubMed] [Google Scholar]
- 4.Rijsewijk F, Schuermann M, Wagenaar E, et al. The Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless. Cell. 1987;50(4):649–657. doi: 10.1016/0092-8674(87)90038-9. [DOI] [PubMed] [Google Scholar]
- 5.Noordermeer J, Klingensmith J, Perrimon N, et al. dishevelled and armadillo act in the wingless signalling pathway in Drosophila. Nature. 1994;367(6458):80–83. doi: 10.1038/367080a0. [DOI] [PubMed] [Google Scholar]
- 6.Peifer M, McCrea PD, Green KJ, et al. The vertebrate adhesive junction proteins beta-catenin and plakoglobin and the Drosophila segment polarity gene armadillo form a multigene family with similar properties. The Journal of cell biology. 1992;118(3):681–691. doi: 10.1083/jcb.118.3.681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Siegfried E, Chou TB, Perrimon N. wingless signaling acts through zeste-white 3, the Drosophila homolog of glycogen synthase kinase-3, to regulate engrailed and establish cell fate. Cell. 1992;71(7):1167–1179. doi: 10.1016/s0092-8674(05)80065-0. [DOI] [PubMed] [Google Scholar]
- 8.Siegfried E, Wilder EL, Perrimon N. Components of wingless signalling in Drosophila. Nature. 1994;367(6458):76–80. doi: 10.1038/367076a0. [DOI] [PubMed] [Google Scholar]
- 9.Behrens J, von Kries JP, Kuhl M, et al. Functional interaction of beta-catenin with the transcription factor LEF-1. Nature. 1996;382(6592):638–642. doi: 10.1038/382638a0. [DOI] [PubMed] [Google Scholar]
- 10.Molenaar M, van de Wetering M, Oosterwegel M, et al. XTcf-3 transcription factor mediates beta-catenin-induced axis formation in Xenopus embryos. Cell. 1996;86(3):391–399. doi: 10.1016/s0092-8674(00)80112-9. [DOI] [PubMed] [Google Scholar]
- 11.Bhanot P, Brink M, Samos CH, et al. A new member of the frizzled family from Drosophila functions as a Wingless receptor. Nature. 1996;382(6588):225–230. doi: 10.1038/382225a0. [DOI] [PubMed] [Google Scholar]
- 12.Wehrli M, Dougan ST, Caldwell K, et al. arrow encodes an LDL-receptor-related protein essential for Wingless signalling. Nature. 2000;407(6803):527–530. doi: 10.1038/35035110. [DOI] [PubMed] [Google Scholar]
- 13.Kinzler KW, Nilbert MC, Su LK, et al. Identification of FAP locus genes from chromosome 5q21. Science (New York, NY. 1991;253(5020):661–665. doi: 10.1126/science.1651562. [DOI] [PubMed] [Google Scholar]
- 14.Nishisho I, Nakamura Y, Miyoshi Y, et al. Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science (New York, NY. 1991;253(5020):665–669. doi: 10.1126/science.1651563. [DOI] [PubMed] [Google Scholar]
- 15.Rubinfeld B, Souza B, Albert I, et al. Association of the APC gene product with beta-catenin. Science (New York, NY. 1993;262(5140):1731–1734. doi: 10.1126/science.8259518. [DOI] [PubMed] [Google Scholar]
- 16.Su LK, Vogelstein B, Kinzler KW. Association of the APC tumor suppressor protein with catenins. Science (New York, NY. 1993;262(5140):1734–1737. doi: 10.1126/science.8259519. [DOI] [PubMed] [Google Scholar]
- 17.Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781–810. doi: 10.1146/annurev.cellbio.20.010403.113126. [DOI] [PubMed] [Google Scholar]
- 18.Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature. 2005;434(7035):843–850. doi: 10.1038/nature03319. [DOI] [PubMed] [Google Scholar]
- 19.Klaus A, Birchmeier W. Wnt signalling and its impact on development and cancer. Nature reviews. 2008;8(5):387–398. doi: 10.1038/nrc2389. [DOI] [PubMed] [Google Scholar]
- 20.MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Developmental cell. 2009;17(1):9–26. doi: 10.1016/j.devcel.2009.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Luo J, Chen J, Deng ZL, et al. Wnt signaling and human diseases: what are the therapeutic implications? Laboratory investigation; a journal of technical methods and pathology. 2007;87(2):97–103. doi: 10.1038/labinvest.3700509. [DOI] [PubMed] [Google Scholar]
- 22.Wagner ER, Zhu G, Zhang BQ, et al. The therapeutic potential of the Wnt signaling pathway in bone disorders. Current molecular pharmacology. 4(1):14–25. [PubMed] [Google Scholar]
- 23.Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell. 2012;149(6):1192–1205. doi: 10.1016/j.cell.2012.05.012. [DOI] [PubMed] [Google Scholar]
- 24.Gough NR. Focus issue: Wnt and beta-catenin signaling in development and disease. Science signaling. 2012;5(206):eg2. doi: 10.1126/scisignal.2002806. [DOI] [PubMed] [Google Scholar]
- 25.Niehrs C. The complex world of WNT receptor signalling. Nature reviews. 2012;13(12):767–779. doi: 10.1038/nrm3470. [DOI] [PubMed] [Google Scholar]
- 26.Anastas JN, Moon RT. WNT signalling pathways as therapeutic targets in cancer. Nature reviews. 2013;13(1):11–26. doi: 10.1038/nrc3419. [DOI] [PubMed] [Google Scholar]
- 27.Clevers H, Loh KM, Nusse R. Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science (New York, NY. 2014;346(6205):1248012. doi: 10.1126/science.1248012. [DOI] [PubMed] [Google Scholar]
- 28.Willert K, Brown JD, Danenberg E, et al. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature. 2003;423(6938):448–452. doi: 10.1038/nature01611. [DOI] [PubMed] [Google Scholar]
- 29.Janda CY, Waghray D, Levin AM, et al. Structural basis of Wnt recognition by Frizzled. Science (New York, NY. 2012;337(6090):59–64. doi: 10.1126/science.1222879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Franch-Marro X, Wendler F, Griffith J, et al. In vivo role of lipid adducts on Wingless. Journal of cell science. 2008;121(Pt 10):1587–1592. doi: 10.1242/jcs.015958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kurayoshi M, Yamamoto H, Izumi S, et al. Post-translational palmitoylation and glycosylation of Wnt-5a are necessary for its signalling. The Biochemical journal. 2007;402(3):515–523. doi: 10.1042/BJ20061476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hofmann K. A superfamily of membrane-bound O-acyltransferases with implications for wnt signaling. Trends in biochemical sciences. 2000;25(3):111–112. doi: 10.1016/s0968-0004(99)01539-x. [DOI] [PubMed] [Google Scholar]
- 33.Kadowaki T, Wilder E, Klingensmith J, et al. The segment polarity gene porcupine encodes a putative multitransmembrane protein involved in Wingless processing. Genes & development. 1996;10(24):3116–3128. doi: 10.1101/gad.10.24.3116. [DOI] [PubMed] [Google Scholar]
- 34.Takada R, Satomi Y, Kurata T, et al. Monounsaturated fatty acid modification of Wnt protein: its role in Wnt secretion. Developmental cell. 2006;11(6):791–801. doi: 10.1016/j.devcel.2006.10.003. [DOI] [PubMed] [Google Scholar]
- 35.Cavallo RA, Cox RT, Moline MM, et al. Drosophila Tcf and Groucho interact to repress Wingless signalling activity. Nature. 1998;395(6702):604–608. doi: 10.1038/26982. [DOI] [PubMed] [Google Scholar]
- 36.Roose J, Molenaar M, Peterson J, et al. The Xenopus Wnt effector XTcf-3 interacts with Groucho-related transcriptional repressors. Nature. 1998;395(6702):608–612. doi: 10.1038/26989. [DOI] [PubMed] [Google Scholar]
- 37.Arce L, Pate KT, Waterman ML. Groucho binds two conserved regions of LEF-1 for HDAC-dependent repression. BMC Cancer. 2009;9:159. doi: 10.1186/1471-2407-9-159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bilic J, Huang YL, Davidson G, et al. Wnt induces LRP6 signalosomes and promotes dishevelled-dependent LRP6 phosphorylation. Science (New York, NY. 2007;316(5831):1619–1622. doi: 10.1126/science.1137065. [DOI] [PubMed] [Google Scholar]
- 39.Zeng X, Huang H, Tamai K, et al. Initiation of Wnt signaling: control of Wnt coreceptor Lrp6 phosphorylation/activation via frizzled, dishevelled and axin functions. Development (Cambridge, England) 2008;135(2):367–375. doi: 10.1242/dev.013540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Metcalfe C, Mendoza-Topaz C, Mieszczanek J, et al. Stability elements in the LRP6 cytoplasmic tail confer efficient signalling upon DIX-dependent polymerization. Journal of cell science. 2010;123(Pt 9):1588–1599. doi: 10.1242/jcs.067546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kramps T, Peter O, Brunner E, et al. Wnt/wingless signaling requires BCL9/legless-mediated recruitment of pygopus to the nuclear beta-catenin-TCF complex. Cell. 2002;109(1):47–60. doi: 10.1016/s0092-8674(02)00679-7. [DOI] [PubMed] [Google Scholar]
- 42.Li J, Sutter C, Parker DS, et al. CBP/p300 are bimodal regulators of Wnt signaling. The EMBO journal. 2007;26(9):2284–2294. doi: 10.1038/sj.emboj.7601667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.He TC, Chan TA, Vogelstein B, et al. PPARdelta is an APC-regulated target of nonsteroidal anti-inflammatory drugs. Cell. 1999;99(3):335–345. doi: 10.1016/s0092-8674(00)81664-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.He TC, Sparks AB, Rago C, et al. Identification of c-MYC as a target of the APC pathway. Science (New York, NY. 1998;281(5382):1509–1512. doi: 10.1126/science.281.5382.1509. [DOI] [PubMed] [Google Scholar]
- 45.Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 1999;398(6726):422–426. doi: 10.1038/18884. [DOI] [PubMed] [Google Scholar]
- 46.Shtutman M, Zhurinsky J, Simcha I, et al. The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(10):5522–5527. doi: 10.1073/pnas.96.10.5522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yan D, Wiesmann M, Rohan M, et al. Elevated expression of axin2 and hnkd mRNA provides evidence that Wnt/beta -catenin signaling is activated in human colon tumors. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(26):14973–14978. doi: 10.1073/pnas.261574498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lustig B, Jerchow B, Sachs M, et al. Negative feedback loop of Wnt signaling through upregulation of conductin/axin2 in colorectal and liver tumors. Molecular and cellular biology. 2002;22(4):1184–1193. doi: 10.1128/MCB.22.4.1184-1193.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jho EH, Zhang T, Domon C, et al. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Molecular and cellular biology. 2002;22(4):1172–1183. doi: 10.1128/MCB.22.4.1172-1183.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Willert J, Epping M, Pollack JR, et al. A transcriptional response to Wnt protein in human embryonic carcinoma cells. BMC developmental biology. 2002;2:8. doi: 10.1186/1471-213x-2-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kikuchi A, Yamamoto H, Sato A, et al. New insights into the mechanism of Wnt signaling pathway activation. Int Rev Cell Mol Biol. 2011;291:21–71. doi: 10.1016/B978-0-12-386035-4.00002-1. [DOI] [PubMed] [Google Scholar]
- 52.Gan XQ, Wang JY, Xi Y, et al. Nuclear Dvl, c-Jun, beta-catenin, and TCF form a complex leading to stabilization of beta-catenin-TCF interaction. The Journal of cell biology. 2008;180(6):1087–1100. doi: 10.1083/jcb.200710050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Louie SH, Yang XY, Conrad WH, et al. Modulation of the beta-catenin signaling pathway by the dishevelled-associated protein Hipk1. PloS one. 2009;4(2):e4310. doi: 10.1371/journal.pone.0004310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang W, Li X, Lee M, et al. FOXKs promote Wnt/beta-catenin signaling by translocating DVL into the nucleus. Developmental cell. 2015;32(6):707–718. doi: 10.1016/j.devcel.2015.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Teo JL, Ma H, Nguyen C, et al. Specific inhibition of CBP/beta-catenin interaction rescues defects in neuronal differentiation caused by a presenilin-1 mutation. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(34):12171–12176. doi: 10.1073/pnas.0504600102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sasaki T, Hwang H, Nguyen C, et al. The small molecule Wnt signaling modulator ICG-001 improves contractile function in chronically infarcted rat myocardium. PloS one. 2013;8(9):e75010. doi: 10.1371/journal.pone.0075010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kessler R, Hausmann G, Basler K. The PHD domain is required to link Drosophila Pygopus to Legless/beta-catenin and not to histone H3. Mechanisms of development. 2009;126(8-9):752–759. doi: 10.1016/j.mod.2009.04.003. [DOI] [PubMed] [Google Scholar]
- 58.Miller TC, Rutherford TJ, Johnson CM, et al. Allosteric remodelling of the histone H3 binding pocket in the Pygo2 PHD finger triggered by its binding to the B9L/BCL9 co-factor. J Mol Biol. 2010;401(5):969–984. doi: 10.1016/j.jmb.2010.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.van den Bosch MH, Blom AB, van Lent PL, et al. Canonical Wnt signaling skews TGF-beta signaling in chondrocytes towards signaling via ALK1 and Smad 1/5/8. Cell Signal. 2014;26(5):951–958. doi: 10.1016/j.cellsig.2014.01.021. [DOI] [PubMed] [Google Scholar]
- 60.Funa NS, Schachter KA, Lerdrup M, et al. beta-Catenin Regulates Primitive Streak Induction through Collaborative Interactions with SMAD2/SMAD3 and OCT4. Cell Stem Cell. 2015;16(6):639–652. doi: 10.1016/j.stem.2015.03.008. [DOI] [PubMed] [Google Scholar]
- 61.Zhang N, Wei P, Gong A, et al. FoxM1 promotes beta-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis. Cancer cell. 2011;20(4):427–442. doi: 10.1016/j.ccr.2011.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kim KA, Zhao J, Andarmani S, et al. R-Spondin proteins: a novel link to beta-catenin activation. Cell Cycle. 2006;5(1):23–26. doi: 10.4161/cc.5.1.2305. [DOI] [PubMed] [Google Scholar]
- 63.Nam JS, Turcotte TJ, Smith PF, et al. Mouse cristin/R-spondin family proteins are novel ligands for the Frizzled 8 and LRP6 receptors and activate beta-catenin-dependent gene expression. The Journal of biological chemistry. 2006;281(19):13247–13257. doi: 10.1074/jbc.M508324200. [DOI] [PubMed] [Google Scholar]
- 64.Kazanskaya O, Glinka A, del Barco Barrantes I, et al. R-Spondin2 is a secreted activator of Wnt/beta-catenin signaling and is required for Xenopus myogenesis. Developmental cell. 2004;7(4):525–534. doi: 10.1016/j.devcel.2004.07.019. [DOI] [PubMed] [Google Scholar]
- 65.Kim KA, Wagle M, Tran K, et al. R-Spondin family members regulate the Wnt pathway by a common mechanism. Mol Biol Cell. 2008;19(6):2588–2596. doi: 10.1091/mbc.E08-02-0187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wei Q, Yokota C, Semenov MV, et al. R-spondin1 is a high affinity ligand for LRP6 and induces LRP6 phosphorylation and beta-catenin signaling. The Journal of biological chemistry. 2007;282(21):15903–15911. doi: 10.1074/jbc.M701927200. [DOI] [PubMed] [Google Scholar]
- 67.Carmon KS, Gong X, Lin Q, et al. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(28):11452–11457. doi: 10.1073/pnas.1106083108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.de Lau W, Barker N, Low TY, et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature. 2011;476(7360):293–297. doi: 10.1038/nature10337. [DOI] [PubMed] [Google Scholar]
- 69.Hao HX, Xie Y, Zhang Y, et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature. 2012;485(7397):195–200. doi: 10.1038/nature11019. [DOI] [PubMed] [Google Scholar]
- 70.Koo BK, Spit M, Jordens I, et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature. 2012;488(7413):665–669. doi: 10.1038/nature11308. [DOI] [PubMed] [Google Scholar]
- 71.de Lau WB, Snel B, Clevers HC. The R-spondin protein family. Genome biology. 2012;13(3):242. doi: 10.1186/gb-2012-13-3-242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Finch PW, He X, Kelley MJ, et al. Purification and molecular cloning of a secreted, Frizzled-related antagonist of Wnt action. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(13):6770–6775. doi: 10.1073/pnas.94.13.6770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kawano Y, Kypta R. Secreted antagonists of the Wnt signalling pathway. Journal of cell science. 2003;116(Pt 13):2627–2634. doi: 10.1242/jcs.00623. [DOI] [PubMed] [Google Scholar]
- 74.Ladher RK, Church VL, Allen S, et al. Cloning and expression of the Wnt antagonists Sfrp-2 and Frzb during chick development. Dev Biol. 2000;218(2):183–198. doi: 10.1006/dbio.1999.9586. [DOI] [PubMed] [Google Scholar]
- 75.Hsieh JC, Kodjabachian L, Rebbert ML, et al. A new secreted protein that binds to Wnt proteins and inhibits their activities. Nature. 1999;398(6726):431–436. doi: 10.1038/18899. [DOI] [PubMed] [Google Scholar]
- 76.Glinka A, Wu W, Delius H, et al. Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature. 1998;391(6665):357–362. doi: 10.1038/34848. [DOI] [PubMed] [Google Scholar]
- 77.Mao B, Wu W, Davidson G, et al. Kremen proteins are Dickkopf receptors that regulate Wnt/beta-catenin signalling. Nature. 2002;417(6889):664–667. doi: 10.1038/nature756. [DOI] [PubMed] [Google Scholar]
- 78.Ellwanger K, Saito H, Clement-Lacroix P, et al. Targeted disruption of the Wnt regulator Kremen induces limb defects and high bone density. Molecular and cellular biology. 2008;28(15):4875–4882. doi: 10.1128/MCB.00222-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Semenov MV, Zhang X, He X. DKK1 antagonizes Wnt signaling without promotion of LRP6 internalization and degradation. The Journal of biological chemistry. 2008;283(31):21427–21432. doi: 10.1074/jbc.M800014200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang K, Zhang Y, Li X, et al. Characterization of the Kremen-binding site on Dkk1 and elucidation of the role of Kremen in Dkk-mediated Wnt antagonism. The Journal of biological chemistry. 2008;283(34):23371–23375. doi: 10.1074/jbc.M802376200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Itasaki N, Jones CM, Mercurio S, et al. Wise, a context-dependent activator and inhibitor of Wnt signalling. Development (Cambridge, England) 2003;130(18):4295–4305. doi: 10.1242/dev.00674. [DOI] [PubMed] [Google Scholar]
- 82.Li X, Zhang Y, Kang H, et al. Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. The Journal of biological chemistry. 2005;280(20):19883–19887. doi: 10.1074/jbc.M413274200. [DOI] [PubMed] [Google Scholar]
- 83.Semenov M, Tamai K, He X. SOST is a ligand for LRP5/LRP6 and a Wnt signaling inhibitor. The Journal of biological chemistry. 2005;280(29):26770–26775. doi: 10.1074/jbc.M504308200. [DOI] [PubMed] [Google Scholar]
- 84.Johnson ML, Harnish K, Nusse R, et al. LRP5 and Wnt signaling: a union made for bone. J Bone Miner Res. 2004;19(11):1749–1757. doi: 10.1359/JBMR.040816. [DOI] [PubMed] [Google Scholar]
- 85.Kim JH, Liu X, Wang J, et al. Wnt signaling in bone formation and its therapeutic potential for bone diseases. Therapeutic advances in musculoskeletal disease. 2013;5(1):13–31. doi: 10.1177/1759720X12466608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Apte U, Zeng G, Muller P, et al. Activation of Wnt/beta-catenin pathway during hepatocyte growth factor-induced hepatomegaly in mice. Hepatology (Baltimore, Md. 2006;44(4):992–1002. doi: 10.1002/hep.21317. [DOI] [PubMed] [Google Scholar]
- 87.Monga SP, Mars WM, Pediaditakis P, et al. Hepatocyte growth factor induces Wnt-independent nuclear translocation of beta-catenin after Met-beta-catenin dissociation in hepatocytes. Cancer research. 2002;62(7):2064–2071. [PubMed] [Google Scholar]
- 88.Nakopoulou L, Gakiopoulou H, Keramopoulos A, et al. c-met tyrosine kinase receptor expression is associated with abnormal beta-catenin expression and favourable prognostic factors in invasive breast carcinoma. Histopathology. 2000;36(4):313–325. doi: 10.1046/j.1365-2559.2000.00847.x. [DOI] [PubMed] [Google Scholar]
- 89.Rasola A, Fassetta M, De Bacco F, et al. A positive feedback loop between hepatocyte growth factor receptor and beta-catenin sustains colorectal cancer cell invasive growth. Oncogene. 2007;26(7):1078–1087. doi: 10.1038/sj.onc.1209859. [DOI] [PubMed] [Google Scholar]
- 90.Ji H, Wang J, Nika H, et al. EGF-induced ERK activation promotes CK2-mediated disassociation of alpha-Catenin from beta-Catenin and transactivation of beta-Catenin. Molecular cell. 2009;36(4):547–559. doi: 10.1016/j.molcel.2009.09.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ochiai A, Akimoto S, Kanai Y, et al. c-erbB-2 gene product associates with catenins in human cancer cells. Biochemical and biophysical research communications. 1994;205(1):73–78. doi: 10.1006/bbrc.1994.2631. [DOI] [PubMed] [Google Scholar]
- 92.Yang W, Xia Y, Ji H, et al. Nuclear PKM2 regulates beta-catenin transactivation upon EGFR activation. Nature. 2011;480(7375):118–122. doi: 10.1038/nature10598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Bommer GT, Feng Y, Iura A, et al. IRS1 regulation by Wnt/beta-catenin signaling and varied contribution of IRS1 to the neoplastic phenotype. The Journal of biological chemistry. 2010;285(3):1928–1938. doi: 10.1074/jbc.M109.060319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Desbois-Mouthon C, Cadoret A, Blivet-Van Eggelpoel MJ, et al. Insulin and IGF-1 stimulate the beta-catenin pathway through two signalling cascades involving GSK-3beta inhibition and Ras activation. Oncogene. 2001;20(2):252–259. doi: 10.1038/sj.onc.1204064. [DOI] [PubMed] [Google Scholar]
- 95.Longato L, de la Monte S, Kuzushita N, et al. Overexpression of insulin receptor substrate-1 and hepatitis Bx genes causes premalignant alterations in the liver. Hepatology (Baltimore, Md. 2009;49(6):1935–1943. doi: 10.1002/hep.22856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Ramocki NM, Wilkins HR, Magness ST, et al. Insulin receptor substrate-1 deficiency promotes apoptosis in the putative intestinal crypt stem cell region, limits Apcmin/+ tumors, and regulates Sox9. Endocrinology. 2008;149(1):261–267. doi: 10.1210/en.2007-0869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Satyamoorthy K, Li G, Vaidya B, et al. Insulin-like growth factor-1 induces survival and growth of biologically early melanoma cells through both the mitogen-activated protein kinase and beta-catenin pathways. Cancer research. 2001;61(19):7318–7324. [PubMed] [Google Scholar]
- 98.Verras M, Sun Z. Beta-catenin is involved in insulin-like growth factor 1-mediated transactivation of the androgen receptor. Molecular endocrinology (Baltimore, Md. 2005;19(2):391–398. doi: 10.1210/me.2004-0208. [DOI] [PubMed] [Google Scholar]
- 99.Birdsey GM, Shah AV, Dufton N, et al. The endothelial transcription factor ERG promotes vascular stability and growth through Wnt/beta-catenin signaling. Developmental cell. 2015;32(1):82–96. doi: 10.1016/j.devcel.2014.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Naik S, Dothager RS, Marasa J, et al. Vascular Endothelial Growth Factor Receptor-1 Is Synthetic Lethal to Aberrant {beta}-Catenin Activation in Colon Cancer. Clin Cancer Res. 2009;15(24):7529–7537. doi: 10.1158/1078-0432.CCR-09-0336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Clausse N, Baines D, Moore R, et al. Activation of both Wnt-1 and Fgf-3 by insertion of mouse mammary tumor virus downstream in the reverse orientation: a reappraisal of the enhancer insertion model. Virology. 1993;194(1):157–165. doi: 10.1006/viro.1993.1245. [DOI] [PubMed] [Google Scholar]
- 102.Katoh M, Katoh M. Cross-talk of WNT and FGF signaling pathways at GSK3beta to regulate beta-catenin and SNAIL signaling cascades. Cancer biology & therapy. 2006;5(9):1059–1064. doi: 10.4161/cbt.5.9.3151. [DOI] [PubMed] [Google Scholar]
- 103.Lee FS, Lane TF, Kuo A, et al. Insertional mutagenesis identifies a member of the Wnt gene family as a candidate oncogene in the mammary epithelium of int-2/Fgf-3 transgenic mice. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(6):2268–2272. doi: 10.1073/pnas.92.6.2268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Nicholes K, Guillet S, Tomlinson E, et al. A mouse model of hepatocellular carcinoma: ectopic expression of fibroblast growth factor 19 in skeletal muscle of transgenic mice. The American journal of pathology. 2002;160(6):2295–2307. doi: 10.1016/S0002-9440(10)61177-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Pai R, Dunlap D, Qing J, et al. Inhibition of fibroblast growth factor 19 reduces tumor growth by modulating beta-catenin signaling. Cancer research. 2008;68(13):5086–5095. doi: 10.1158/0008-5472.CAN-07-2325. [DOI] [PubMed] [Google Scholar]
- 106.Shackleford GM, MacArthur CA, Kwan HC, et al. Mouse mammary tumor virus infection accelerates mammary carcinogenesis in Wnt-1 transgenic mice by insertional activation of int-2/Fgf-3 and hst/Fgf-4. Proceedings of the National Academy of Sciences of the United States of America. 1993;90(2):740–744. doi: 10.1073/pnas.90.2.740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Li Y, Zhang X, Polakiewicz RD, et al. HDAC6 is required for epidermal growth factor-induced beta-catenin nuclear localization. The Journal of biological chemistry. 2008;283(19):12686–12690. doi: 10.1074/jbc.C700185200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Chen D, Zhao M, Mundy GR. Bone morphogenetic proteins. Growth factors (Chur, Switzerland) 2004;22(4):233–241. doi: 10.1080/08977190412331279890. [DOI] [PubMed] [Google Scholar]
- 109.Dumont N, Arteaga CL. Targeting the TGF beta signaling network in human neoplasia. Cancer cell. 2003;3(6):531–536. doi: 10.1016/s1535-6108(03)00135-1. [DOI] [PubMed] [Google Scholar]
- 110.Yingling JM, Blanchard KL, Sawyer JS. Development of TGF-beta signalling inhibitors for cancer therapy. Nat Rev Drug Discov. 2004;3(12):1011–1022. doi: 10.1038/nrd1580. [DOI] [PubMed] [Google Scholar]
- 111.Wang RN, Green J, Wang Z, et al. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes & Diseases. 2014;1(1):87–105. doi: 10.1016/j.gendis.2014.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Luther G, Wagner ER, Zhu G, et al. BMP-9 induced osteogenic differentiation of mesenchymal stem cells: molecular mechanism and therapeutic potential. Current gene therapy. 11(3):229–240. doi: 10.2174/156652311795684777. [DOI] [PubMed] [Google Scholar]
- 113.Lamplot JD, Qin J, Nan G, et al. BMP9 signaling in stem cell differentiation and osteogenesis. American journal of stem cells. 2(1):1–21. [PMC free article] [PubMed] [Google Scholar]
- 114.Kim JS, Crooks H, Dracheva T, et al. Oncogenic beta-catenin is required for bone morphogenetic protein 4 expression in human cancer cells. Cancer research. 2002;62(10):2744–2748. [PubMed] [Google Scholar]
- 115.Hoppler S, Moon RT. BMP-2/-4 and Wnt-8 cooperatively pattern the Xenopus mesoderm. Mechanisms of development. 1998;71(1-2):119–129. doi: 10.1016/s0925-4773(98)00004-5. [DOI] [PubMed] [Google Scholar]
- 116.Labbe E, Letamendia A, Attisano L. Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-beta and wnt pathways. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(15):8358–8363. doi: 10.1073/pnas.150152697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Nishita M, Hashimoto MK, Ogata S, et al. Interaction between Wnt and TGF-beta signalling pathways during formation of Spemann's organizer. Nature. 2000;403(6771):781–785. doi: 10.1038/35001602. [DOI] [PubMed] [Google Scholar]
- 118.Szeto DP, Kimelman D. Combinatorial gene regulation by Bmp and Wnt in zebrafish posterior mesoderm formation. Development (Cambridge, England) 2004;131(15):3751–3760. doi: 10.1242/dev.01236. [DOI] [PubMed] [Google Scholar]
- 119.Hussein SM, Duff EK, Sirard C. Smad4 and beta-catenin co-activators functionally interact with lymphoid-enhancing factor to regulate graded expression of Msx2. The Journal of biological chemistry. 2003;278(49):48805–48814. doi: 10.1074/jbc.M305472200. [DOI] [PubMed] [Google Scholar]
- 120.Letamendia A, Labbe E, Attisano L. Transcriptional regulation by Smads: crosstalk between the TGF-beta and Wnt pathways. The Journal of bone and joint surgery. 2001;83-A(Suppl 1)(Pt 1):S31–39. [PubMed] [Google Scholar]
- 121.Theil T, Aydin S, Koch S, et al. Wnt and Bmp signalling cooperatively regulate graded Emx2 expression in the dorsal telencephalon. Development (Cambridge, England) 2002;129(13):3045–3054. doi: 10.1242/dev.129.13.3045. [DOI] [PubMed] [Google Scholar]
- 122.Sakai D, Tanaka Y, Endo Y, et al. Regulation of Slug transcription in embryonic ectoderm by beta-catenin-Lef/Tcf and BMP-Smad signaling. Dev Growth Differ. 2005;47(7):471–482. doi: 10.1111/j.1440-169X.2005.00821.x. [DOI] [PubMed] [Google Scholar]
- 123.Hu MC, Rosenblum ND. Smad1, beta-catenin and Tcf4 associate in a molecular complex with the Myc promoter in dysplastic renal tissue and cooperate to control Myc transcription. Development (Cambridge, England) 2005;132(1):215–225. doi: 10.1242/dev.01573. [DOI] [PubMed] [Google Scholar]
- 124.Bhowmick NA, Chytil A, Plieth D, et al. TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science (New York, NY. 2004;303(5659):848–851. doi: 10.1126/science.1090922. [DOI] [PubMed] [Google Scholar]
- 125.Li X, Placencio V, Iturregui JM, et al. Prostate tumor progression is mediated by a paracrine TGF-beta/Wnt3a signaling axis. Oncogene. 2008;27(56):7118–7130. doi: 10.1038/onc.2008.293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Han G, Li AG, Liang YY, et al. Smad7-induced beta-catenin degradation alters epidermal appendage development. Developmental cell. 2006;11(3):301–312. doi: 10.1016/j.devcel.2006.06.014. [DOI] [PubMed] [Google Scholar]
- 127.Edlund S, Lee SY, Grimsby S, et al. Interaction between Smad7 and beta-catenin: importance for transforming growth factor beta-induced apoptosis. Molecular and cellular biology. 2005;25(4):1475–1488. doi: 10.1128/MCB.25.4.1475-1488.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Tang Y, Liu Z, Zhao L, et al. Smad7 stabilizes beta-catenin binding to E-cadherin complex and promotes cell-cell adhesion. The Journal of biological chemistry. 2008;283(35):23956–23963. doi: 10.1074/jbc.M800351200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Song J, McColl J, Camp E, et al. Smad1 transcription factor integrates BMP2 and Wnt3a signals in migrating cardiac progenitor cells. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(20):7337–7342. doi: 10.1073/pnas.1321764111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Itasaki N, Hoppler S. Crosstalk between Wnt and bone morphogenic protein signaling: a turbulent relationship. Dev Dyn. 2010;239(1):16–33. doi: 10.1002/dvdy.22009. [DOI] [PubMed] [Google Scholar]
- 131.Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: cell fate control and signal integration in development. Science (New York, NY. 1999;284(5415):770–776. doi: 10.1126/science.284.5415.770. [DOI] [PubMed] [Google Scholar]
- 132.Fortini ME. Notch signaling: the core pathway and its posttranslational regulation. Developmental cell. 2009;16(5):633–647. doi: 10.1016/j.devcel.2009.03.010. [DOI] [PubMed] [Google Scholar]
- 133.Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137(2):216–233. doi: 10.1016/j.cell.2009.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Duncan AW, Rattis FM, DiMascio LN, et al. Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance. Nature immunology. 2005;6(3):314–322. doi: 10.1038/ni1164. [DOI] [PubMed] [Google Scholar]
- 135.Galceran J, Sustmann C, Hsu SC, et al. LEF1-mediated regulation of Delta-like1 links Wnt and Notch signaling in somitogenesis. Genes & development. 2004;18(22):2718–2723. doi: 10.1101/gad.1249504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Nakamura T, Tsuchiya K, Watanabe M. Crosstalk between Wnt and Notch signaling in intestinal epithelial cell fate decision. Journal of gastroenterology. 2007;42(9):705–710. doi: 10.1007/s00535-007-2087-z. [DOI] [PubMed] [Google Scholar]
- 137.Collu GM, Hidalgo-Sastre A, Brennan K. Wnt-Notch signalling crosstalk in development and disease. Cell Mol Life Sci. 2014;71(18):3553–3567. doi: 10.1007/s00018-014-1644-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Munoz-Descalzo S, de Navascues J, Arias AM. Wnt-Notch signalling: an integrated mechanism regulating transitions between cell states. Bioessays. 2012;34(2):110–118. doi: 10.1002/bies.201100102. [DOI] [PubMed] [Google Scholar]
- 139.Hayward P, Brennan K, Sanders P, et al. Notch modulates Wnt signalling by associating with Armadillo/beta-catenin and regulating its transcriptional activity. Development (Cambridge, England) 2005;132(8):1819–1830. doi: 10.1242/dev.01724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Axelrod JD, Matsuno K, Artavanis-Tsakonas S, et al. Interaction between Wingless and Notch signaling pathways mediated by dishevelled. Science (New York, NY. 1996;271(5257):1826–1832. doi: 10.1126/science.271.5257.1826. [DOI] [PubMed] [Google Scholar]
- 141.Nicolas M, Wolfer A, Raj K, et al. Notch1 functions as a tumor suppressor in mouse skin. Nature genetics. 2003;33(3):416–421. doi: 10.1038/ng1099. [DOI] [PubMed] [Google Scholar]
- 142.Revet I, Huizenga G, Koster J, et al. MSX1 induces the Wnt pathway antagonist genes DKK1, DKK2, DKK3, and SFRP1 in neuroblastoma cells, but does not block Wnt3 and Wnt5A signalling to DVL3. Cancer letters. 2010;289(2):195–207. doi: 10.1016/j.canlet.2009.08.019. [DOI] [PubMed] [Google Scholar]
- 143.Duan L, Yao J, Wu X, et al. Growth suppression induced by Notch1 activation involves Wnt-beta-catenin down-regulation in human tongue carcinoma cells. Biology of the cell / under the auspices of the European Cell Biology Organization. 2006;98(8):479–490. doi: 10.1042/BC20060020. [DOI] [PubMed] [Google Scholar]
- 144.Fre S, Pallavi SK, Huyghe M, et al. Notch and Wnt signals cooperatively control cell proliferation and tumorigenesis in the intestine. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(15):6309–6314. doi: 10.1073/pnas.0900427106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Lopez-Terrada D, Gunaratne PH, Adesina AM, et al. Histologic subtypes of hepatoblastoma are characterized by differential canonical Wnt and Notch pathway activation in DLK+ precursors. Human pathology. 2009;40(6):783–794. doi: 10.1016/j.humpath.2008.07.022. [DOI] [PubMed] [Google Scholar]
- 146.Moreno CS. The Sex-determining region Y-box 4 and homeobox C6 transcriptional networks in prostate cancer progression: crosstalk with the Wnt, Notch, and PI3K pathways. The American journal of pathology. 2010;176(2):518–527. doi: 10.2353/ajpath.2010.090657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Collu GM, Brennan K. Cooperation between Wnt and Notch signalling in human breast cancer. Breast Cancer Res. 2007;9(3):105. doi: 10.1186/bcr1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Ayyanan A, Civenni G, Ciarloni L, et al. Increased Wnt signaling triggers oncogenic conversion of human breast epithelial cells by a Notch-dependent mechanism. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(10):3799–3804. doi: 10.1073/pnas.0600065103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Estrach S, Ambler CA, Lo Celso C, et al. Jagged 1 is a beta-catenin target gene required for ectopic hair follicle formation in adult epidermis. Development (Cambridge, England) 2006;133(22):4427–4438. doi: 10.1242/dev.02644. [DOI] [PubMed] [Google Scholar]
- 150.Ingham PW, McMahon AP. Hedgehog signaling in animal development: paradigms and principles. Genes & development. 2001;15(23):3059–3087. doi: 10.1101/gad.938601. [DOI] [PubMed] [Google Scholar]
- 151.van Den Heuvel M. Fat hedgehogs, slower or richer? Sci STKE. 2001;2001(100):e31. doi: 10.1126/stke.2001.100.pe31. [DOI] [PubMed] [Google Scholar]
- 152.Taipale J, Beachy PA. The Hedgehog and Wnt signalling pathways in cancer. Nature. 2001;411(6835):349–354. doi: 10.1038/35077219. [DOI] [PubMed] [Google Scholar]
- 153.Hooper JE, Scott MP. Communicating with Hedgehogs. Nature reviews. 2005;6(4):306–317. doi: 10.1038/nrm1622. [DOI] [PubMed] [Google Scholar]
- 154.Bonifas JM, Pennypacker S, Chuang PT, et al. Activation of expression of hedgehog target genes in basal cell carcinomas. The Journal of investigative dermatology. 2001;116(5):739–742. doi: 10.1046/j.1523-1747.2001.01315.x. [DOI] [PubMed] [Google Scholar]
- 155.Yang SH, Andl T, Grachtchouk V, et al. Pathological responses to oncogenic Hedgehog signaling in skin are dependent on canonical Wnt/beta3-catenin signaling. Nature genetics. 2008;40(9):1130–1135. doi: 10.1038/ng.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Liao X, Siu MK, Au CW, et al. Aberrant activation of hedgehog signaling pathway contributes to endometrial carcinogenesis through beta-catenin. Mod Pathol. 2009;22(6):839–847. doi: 10.1038/modpathol.2009.45. [DOI] [PubMed] [Google Scholar]
- 157.Noubissi FK, Goswami S, Sanek NA, et al. Wnt signaling stimulates transcriptional outcome of the Hedgehog pathway by stabilizing GLI1 mRNA. Cancer research. 2009;69(22):8572–8578. doi: 10.1158/0008-5472.CAN-09-1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Varnat F, Zacchetti G, Ruiz i Altaba A. Hedgehog pathway activity is required for the lethality and intestinal phenotypes of mice with hyperactive Wnt signaling. Mechanisms of development. 2010;127(1-2):73–81. doi: 10.1016/j.mod.2009.10.005. [DOI] [PubMed] [Google Scholar]
- 159.Kim BM, Choi MY. New insights into the role of Hedgehog signaling in gastrointestinal development and cancer. Gastroenterology. 2009;137(2):422–424. doi: 10.1053/j.gastro.2009.06.021. [DOI] [PubMed] [Google Scholar]
- 160.Arimura S, Matsunaga A, Kitamura T, et al. Reduced level of smoothened suppresses intestinal tumorigenesis by down-regulation of Wnt signaling. Gastroenterology. 2009;137(2):629–638. doi: 10.1053/j.gastro.2009.04.059. [DOI] [PubMed] [Google Scholar]
- 161.Pasca di Magliano M, Biankin AV, Heiser PW, et al. Common activation of canonical Wnt signaling in pancreatic adenocarcinoma. PloS one. 2007;2(11):e1155. doi: 10.1371/journal.pone.0001155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.van den Brink GR, Bleuming SA, Hardwick JC, et al. Indian Hedgehog is an antagonist of Wnt signaling in colonic epithelial cell differentiation. Nature genetics. 2004;36(3):277–282. doi: 10.1038/ng1304. [DOI] [PubMed] [Google Scholar]
- 163.Yanai K, Nakamura M, Akiyoshi T, et al. Crosstalk of hedgehog and Wnt pathways in gastric cancer. Cancer letters. 2008;263(1):145–156. doi: 10.1016/j.canlet.2007.12.030. [DOI] [PubMed] [Google Scholar]
- 164.Dong J, Feldmann G, Huang J, et al. Elucidation of a universal size-control mechanism in Drosophila and mammals. Cell. 2007;130(6):1120–1133. doi: 10.1016/j.cell.2007.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Varelas X. The Hippo pathway effectors TAZ and YAP in development, homeostasis and disease. Development (Cambridge, England) 2014;141(8):1614–1626. doi: 10.1242/dev.102376. [DOI] [PubMed] [Google Scholar]
- 166.Hansen CG, Moroishi T, Guan KL. YAP and TAZ: a nexus for Hippo signaling and beyond. Trends in cell biology. 2015 doi: 10.1016/j.tcb.2015.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Piccolo S, Dupont S, Cordenonsi M. The biology of YAP/TAZ: hippo signaling and beyond. Physiological reviews. 2014;94(4):1287–1312. doi: 10.1152/physrev.00005.2014. [DOI] [PubMed] [Google Scholar]
- 168.Wang Z, Ye J, Deng Y, et al. Wnt Hippo: A balanced act or dynamic duo? Genes & Diseases. 2014;1(2):127–128. doi: 10.1016/j.gendis.2014.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Ramos A, Camargo FD. The Hippo signaling pathway and stem cell biology. Trends in cell biology. 2012;22(7):339–346. doi: 10.1016/j.tcb.2012.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Barry ER, Morikawa T, Butler BL, et al. Restriction of intestinal stem cell expansion and the regenerative response by YAP. Nature. 2013;493(7430):106–110. doi: 10.1038/nature11693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Heallen T, Zhang M, Wang J, et al. Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science (New York, NY. 2011;332(6028):458–461. doi: 10.1126/science.1199010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Azzolin L, Panciera T, Soligo S, et al. YAP/TAZ incorporation in the beta-catenin destruction complex orchestrates the Wnt response. Cell. 2014;158(1):157–170. doi: 10.1016/j.cell.2014.06.013. [DOI] [PubMed] [Google Scholar]
- 173.Cai J, Maitra A, Anders RA, et al. beta-Catenin destruction complex-independent regulation of Hippo-YAP signaling by APC in intestinal tumorigenesis. Genes & development. 2015;29(14):1493–1506. doi: 10.1101/gad.264515.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Park HW, Kim YC, Yu B, et al. Alternative Wnt Signaling Activates YAP/TAZ. Cell. 2015;162(4):780–794. doi: 10.1016/j.cell.2015.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Holland JD, Klaus A, Garratt AN, et al. Wnt signaling in stem and cancer stem cells. Curr Opin Cell Biol. 2013;25(2):254–264. doi: 10.1016/j.ceb.2013.01.004. [DOI] [PubMed] [Google Scholar]
- 176.Hanna J, Markoulaki S, Mitalipova M, et al. Metastable pluripotent states in NOD-mouse-derived ESCs. Cell Stem Cell. 2009;4(6):513–524. doi: 10.1016/j.stem.2009.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Merrill BJ. Wnt pathway regulation of embryonic stem cell self-renewal. Cold Spring Harb Perspect Biol. 2012;4(9):a007971. doi: 10.1101/cshperspect.a007971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Sato N, Meijer L, Skaltsounis L, et al. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat Med. 2004;10(1):55–63. doi: 10.1038/nm979. [DOI] [PubMed] [Google Scholar]
- 179.Abu-Remaileh M, Gerson A, Farago M, et al. Oct-3/4 regulates stem cell identity and cell fate decisions by modulating Wnt/beta-catenin signalling. The EMBO journal. 2010;29(19):3236–3248. doi: 10.1038/emboj.2010.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Lluis F, Pedone E, Pepe S, et al. Periodic activation of Wnt/beta-catenin signaling enhances somatic cell reprogramming mediated by cell fusion. Cell Stem Cell. 2008;3(5):493–507. doi: 10.1016/j.stem.2008.08.017. [DOI] [PubMed] [Google Scholar]
- 181.Zhang P, Chang WH, Fong B, et al. Regulation of induced pluripotent stem (iPS) cell induction by Wnt/beta-catenin signaling. The Journal of biological chemistry. 2014;289(13):9221–9232. doi: 10.1074/jbc.M113.542845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Rastegar F, Shenaq D, Huang J, et al. Mesenchymal stem cells: Molecular characteristics and clinical applications. World journal of stem cells. 2(4):67–80. doi: 10.4252/wjsc.v2.i4.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Day TF, Guo X, Garrett-Beal L, et al. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Developmental cell. 2005;8(5):739–750. doi: 10.1016/j.devcel.2005.03.016. [DOI] [PubMed] [Google Scholar]
- 184.Luo Q, Kang Q, Si W, et al. Connective tissue growth factor (CTGF) is regulated by Wnt and bone morphogenetic proteins signaling in osteoblast differentiation of mesenchymal stem cells. The Journal of biological chemistry. 2004;279(53):55958–55968. doi: 10.1074/jbc.M407810200. [DOI] [PubMed] [Google Scholar]
- 185.Si W, Kang Q, Luu HH, et al. CCN1/Cyr61 is regulated by the canonical Wnt signal and plays an important role in Wnt3A-induced osteoblast differentiation of mesenchymal stem cells. Molecular and cellular biology. 2006;26(8):2955–2964. doi: 10.1128/MCB.26.8.2955-2964.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Tang N, Song WX, Luo J, et al. BMP-9-induced osteogenic differentiation of mesenchymal progenitors requires functional canonical Wnt/beta-catenin signalling. Journal of cellular and molecular medicine. 2009;13(8B):2448–2464. doi: 10.1111/j.1582-4934.2008.00569.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Wang J, Zhang H, Zhang W, et al. Bone morphogenetic protein-9 effectively induces osteo/odontoblastic differentiation of the reversibly immortalized stem cells of dental apical papilla. Stem cells and development. 23(12):1405–1416. doi: 10.1089/scd.2013.0580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Zhang H, Wang J, Deng F, et al. Canonical Wnt signaling acts synergistically on BMP9-induced osteo/odontoblastic differentiation of stem cells of dental apical papilla (SCAPs). Biomaterials. 39:145–154. doi: 10.1016/j.biomaterials.2014.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Liu J, Farmer SR. Regulating the balance between peroxisome proliferator-activated receptor gamma and beta-catenin signaling during adipogenesis. A glycogen synthase kinase 3beta phosphorylation-defective mutant of beta-catenin inhibits expression of a subset of adipogenic genes. The Journal of biological chemistry. 2004;279(43):45020–45027. doi: 10.1074/jbc.M407050200. [DOI] [PubMed] [Google Scholar]
- 190.Taipaleenmaki H, Abdallah BM, AlDahmash A, et al. Wnt signalling mediates the cross-talk between bone marrow derived pre-adipocytic and pre-osteoblastic cell populations. Exp Cell Res. 2011;317(6):745–756. doi: 10.1016/j.yexcr.2010.12.015. [DOI] [PubMed] [Google Scholar]
- 191.Gao Q, Guo M, Jiang X, et al. A cocktail method for promoting cardiomyocyte differentiation from bone marrow-derived mesenchymal stem cells. Stem cells international. 2014;2014:162024. doi: 10.1155/2014/162024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Alfaro MP, Pagni M, Vincent A, et al. The Wnt modulator sFRP2 enhances mesenchymal stem cell engraftment, granulation tissue formation and myocardial repair. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(47):18366–18371. doi: 10.1073/pnas.0803437105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.De Boer J, Wang HJ, Van Blitterswijk C. Effects of Wnt signaling on proliferation and differentiation of human mesenchymal stem cells. Tissue Eng. 2004;10(3-4):393–401. doi: 10.1089/107632704323061753. [DOI] [PubMed] [Google Scholar]
- 194.Clevers H. The intestinal crypt, a prototype stem cell compartment. Cell. 2013;154(2):274–284. doi: 10.1016/j.cell.2013.07.004. [DOI] [PubMed] [Google Scholar]
- 195.Tian H, Biehs B, Warming S, et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature. 2011;478(7368):255–259. doi: 10.1038/nature10408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Yan KS, Chia LA, Li X, et al. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(2):466–471. doi: 10.1073/pnas.1118857109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Gehart H, Clevers H. Repairing organs: lessons from intestine and liver. Trends Genet. 2015;31(6):344–351. doi: 10.1016/j.tig.2015.04.005. [DOI] [PubMed] [Google Scholar]
- 198.Gregorieff A, Pinto D, Begthel H, et al. Expression pattern of Wnt signaling components in the adult intestine. Gastroenterology. 2005;129(2):626–638. doi: 10.1016/j.gastro.2005.06.007. [DOI] [PubMed] [Google Scholar]
- 199.Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science (New York, NY. 2010;327(5965):542–545. doi: 10.1126/science.1180794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Bastide P, Darido C, Pannequin J, et al. Sox9 regulates cell proliferation and is required for Paneth cell differentiation in the intestinal epithelium. The Journal of cell biology. 2007;178(4):635–648. doi: 10.1083/jcb.200704152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.van der Flier LG, van Gijn ME, Hatzis P, et al. Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell. 2009;136(5):903–912. doi: 10.1016/j.cell.2009.01.031. [DOI] [PubMed] [Google Scholar]
- 202.Batlle E, Henderson JT, Beghtel H, et al. Beta-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell. 2002;111(2):251–263. doi: 10.1016/s0092-8674(02)01015-2. [DOI] [PubMed] [Google Scholar]
- 203.Tian H, Biehs B, Chiu C, et al. Opposing activities of Notch and Wnt signaling regulate intestinal stem cells and gut homeostasis. Cell Rep. 2015;11(1):33–42. doi: 10.1016/j.celrep.2015.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Nostro MC, Cheng X, Keller GM, et al. Wnt, activin, and BMP signaling regulate distinct stages in the developmental pathway from embryonic stem cells to blood. Cell Stem Cell. 2008;2(1):60–71. doi: 10.1016/j.stem.2007.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Luis TC, Weerkamp F, Naber BA, et al. Wnt3a deficiency irreversibly impairs hematopoietic stem cell self-renewal and leads to defects in progenitor cell differentiation. Blood. 2009;113(3):546–554. doi: 10.1182/blood-2008-06-163774. [DOI] [PubMed] [Google Scholar]
- 206.Zhao C, Blum J, Chen A, et al. Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer cell. 2007;12(6):528–541. doi: 10.1016/j.ccr.2007.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Schaniel C, Sirabella D, Qiu J, et al. Wnt-inhibitory factor 1 dysregulation of the bone marrow niche exhausts hematopoietic stem cells. Blood. 2011;118(9):2420–2429. doi: 10.1182/blood-2010-09-305664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Reya T, Duncan AW, Ailles L, et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature. 2003;423(6938):409–414. doi: 10.1038/nature01593. [DOI] [PubMed] [Google Scholar]
- 209.Fukuda S, Pelus LM. Regulation of the inhibitor-of-apoptosis family member survivin in normal cord blood and bone marrow CD34(+) cells by hematopoietic growth factors: implication of survivin expression in normal hematopoiesis. Blood. 2001;98(7):2091–2100. doi: 10.1182/blood.v98.7.2091. [DOI] [PubMed] [Google Scholar]
- 210.Gurbuxani S, Xu Y, Keerthivasan G, et al. Differential requirements for survivin in hematopoietic cell development. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(32):11480–11485. doi: 10.1073/pnas.0500303102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Clements WK, Kim AD, Ong KG, et al. A somitic Wnt16/Notch pathway specifies haematopoietic stem cells. Nature. 2011;474(7350):220–224. doi: 10.1038/nature10107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Loeffler D, Kokkaliaris KD, Schroeder T. Wnt to notch relay signaling induces definitive hematopoiesis. Cell Stem Cell. 2011;9(1):2–4. doi: 10.1016/j.stem.2011.06.011. [DOI] [PubMed] [Google Scholar]
- 213.Singbrant S, Karlsson G, Ehinger M, et al. Canonical BMP signaling is dispensable for hematopoietic stem cell function in both adult and fetal liver hematopoiesis, but essential to preserve colon architecture. Blood. 2010;115(23):4689–4698. doi: 10.1182/blood-2009-05-220988. [DOI] [PubMed] [Google Scholar]
- 214.Fuchs E. Scratching the surface of skin development. Nature. 2007;445(7130):834–842. doi: 10.1038/nature05659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Lim X, Nusse R. Wnt signaling in skin development, homeostasis, and disease. Cold Spring Harb Perspect Biol. 2013;5(2) doi: 10.1101/cshperspect.a008029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.DasGupta R, Fuchs E. Multiple roles for activated LEF/TCF transcription complexes during hair follicle development and differentiation. Development (Cambridge, England) 1999;126(20):4557–4568. doi: 10.1242/dev.126.20.4557. [DOI] [PubMed] [Google Scholar]
- 217.Huelsken J, Vogel R, Erdmann B, et al. beta-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell. 2001;105(4):533–545. doi: 10.1016/s0092-8674(01)00336-1. [DOI] [PubMed] [Google Scholar]
- 218.Nguyen H, Merrill BJ, Polak L, et al. Tcf3 and Tcf4 are essential for long-term homeostasis of skin epithelia. Nature genetics. 2009;41(10):1068–1075. doi: 10.1038/ng.431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Lien WH, Polak L, Lin M, et al. In vivo transcriptional governance of hair follicle stem cells by canonical Wnt regulators. Nat Cell Biol. 2014;16(2):179–190. doi: 10.1038/ncb2903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Sun P, Watanabe K, Fallahi M, et al. Pygo2 regulates beta-catenin-induced activation of hair follicle stem/progenitor cells and skin hyperplasia. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(28):10215–10220. doi: 10.1073/pnas.1311395111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Kandyba E, Leung Y, Chen YB, et al. Competitive balance of intrabulge BMP/Wnt signaling reveals a robust gene network ruling stem cell homeostasis and cyclic activation. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(4):1351–1356. doi: 10.1073/pnas.1121312110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Kandyba E, Kobielak K. Wnt7b is an important intrinsic regulator of hair follicle stem cell homeostasis and hair follicle cycling. Stem Cells. 2014;32(4):886–901. doi: 10.1002/stem.1599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Nusse R, van Ooyen A, Cox D, et al. Mode of proviral activation of a putative mammary oncogene (int-1) on mouse chromosome 15. Nature. 1984;307(5947):131–136. doi: 10.1038/307131a0. [DOI] [PubMed] [Google Scholar]
- 224.Korinek V, Barker N, Morin PJ, et al. Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC−/− colon carcinoma. Science (New York, NY. 1997;275(5307):1784–1787. doi: 10.1126/science.275.5307.1784. [DOI] [PubMed] [Google Scholar]
- 225.Morin PJ, Sparks AB, Korinek V, et al. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science (New York, NY. 1997;275(5307):1787–1790. doi: 10.1126/science.275.5307.1787. [DOI] [PubMed] [Google Scholar]
- 226.Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer. Cell. 1996;87(2):159–170. doi: 10.1016/s0092-8674(00)81333-1. [DOI] [PubMed] [Google Scholar]
- 227.Kinzler KW, Vogelstein B. Landscaping the cancer terrain. Science (New York, NY. 1998;280(5366):1036–1037. doi: 10.1126/science.280.5366.1036. [DOI] [PubMed] [Google Scholar]
- 228.Clements WM, Lowy AM, Groden J. Adenomatous polyposis coli/beta-catenin interaction and downstream targets: altered gene expression in gastrointestinal tumors. Clin Colorectal Cancer. 2003;3(2):113–120. doi: 10.3816/ccc.2003.n.018. [DOI] [PubMed] [Google Scholar]
- 229.Su LK, Kinzler KW, Vogelstein B, et al. Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science (New York, NY. 1992;256(5057):668–670. doi: 10.1126/science.1350108. [DOI] [PubMed] [Google Scholar]
- 230.Miyoshi Y, Iwao K, Nagasawa Y, et al. Activation of the beta-catenin gene in primary hepatocellular carcinomas by somatic alterations involving exon 3. Cancer research. 1998;58(12):2524–2527. [PubMed] [Google Scholar]
- 231.Satoh S, Daigo Y, Furukawa Y, et al. AXIN1 mutations in hepatocellular carcinomas, and growth suppression in cancer cells by virus-mediated transfer of AXIN1. Nature genetics. 2000;24(3):245–250. doi: 10.1038/73448. [DOI] [PubMed] [Google Scholar]
- 232.Polakis P. The many ways of Wnt in cancer. Curr Opin Genet Dev. 2007;17(1):45–51. doi: 10.1016/j.gde.2006.12.007. [DOI] [PubMed] [Google Scholar]
- 233.Zhang Y, Morris JPt, Yan W, et al. Canonical wnt signaling is required for pancreatic carcinogenesis. Cancer research. 2013;73(15):4909–4922. doi: 10.1158/0008-5472.CAN-12-4384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Xie D, Xie K. Pancreatic cancer stromal biology and therapy. Genes & Diseases. 2015;2(2):133–143. doi: 10.1016/j.gendis.2015.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Yu M, Ting DT, Stott SL, et al. RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis. Nature. 2012;487(7408):510–513. doi: 10.1038/nature11217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Haydon RC, Deyrup A, Ishikawa A, et al. Cytoplasmic and/or nuclear accumulation of the beta-catenin protein is a frequent event in human osteosarcoma. International journal of cancer. 2002;102(4):338–342. doi: 10.1002/ijc.10719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Hoffmeyer K, Raggioli A, Rudloff S, et al. Wnt/beta-catenin signaling regulates telomerase in stem cells and cancer cells. Science (New York, NY. 2012;336(6088):1549–1554. doi: 10.1126/science.1218370. [DOI] [PubMed] [Google Scholar]
- 238.Wissmann C, Wild PJ, Kaiser S, et al. WIF1, a component of the Wnt pathway, is down-regulated in prostate, breast, lung, and bladder cancer. J Pathol. 2003;201(2):204–212. doi: 10.1002/path.1449. [DOI] [PubMed] [Google Scholar]
- 239.Kansara M, Tsang M, Kodjabachian L, et al. Wnt inhibitory factor 1 is epigenetically silenced in human osteosarcoma, and targeted disruption accelerates osteosarcomagenesis in mice. J Clin Invest. 2009;119(4):837–851. doi: 10.1172/JCI37175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Mullighan CG, Zhang J, Kasper LH, et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature. 2011;471(7337):235–239. doi: 10.1038/nature09727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Rosenbluh J, Nijhawan D, Cox AG, et al. beta-Catenin-driven cancers require a YAP1 transcriptional complex for survival and tumorigenesis. Cell. 2012;151(7):1457–1473. doi: 10.1016/j.cell.2012.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Mehlen P, Puisieux A. Metastasis: a question of life or death. Nature reviews. 2006;6(6):449–458. doi: 10.1038/nrc1886. [DOI] [PubMed] [Google Scholar]
- 243.Coghlin C, Murray GI. Current and emerging concepts in tumour metastasis. J Pathol. 2010;222(1):1–15. doi: 10.1002/path.2727. [DOI] [PubMed] [Google Scholar]
- 244.Heuberger J, Birchmeier W. Interplay of cadherin-mediated cell adhesion and canonical Wnt signaling. Cold Spring Harb Perspect Biol. 2010;2(2):a002915. doi: 10.1101/cshperspect.a002915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Cathcart J, Pulkoski-Gross A, Cao J. Targeting Matrix Metalloproteinases in Cancer: Bringing New Life to Old Ideas. Genes & Diseases. 2015;2(1):26–34. doi: 10.1016/j.gendis.2014.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Vermeulen L, De Sousa EMF, van der Heijden M, et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010;12(5):468–476. doi: 10.1038/ncb2048. [DOI] [PubMed] [Google Scholar]
- 247.Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nature reviews. 2014;15(3):178–196. doi: 10.1038/nrm3758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Gonzalez DM, Medici D. Signaling mechanisms of the epithelial-mesenchymal transition. Science signaling. 2014;7(344):re8. doi: 10.1126/scisignal.2005189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Nguyen DX, Chiang AC, Zhang XH, et al. WNT/TCF signaling through LEF1 and HOXB9 mediates lung adenocarcinoma metastasis. Cell. 2009;138(1):51–62. doi: 10.1016/j.cell.2009.04.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Previdi S, Maroni P, Matteucci E, et al. Interaction between human-breast cancer metastasis and bone microenvironment through activated hepatocyte growth factor/Met and beta-catenin/Wnt pathways. Eur J Cancer. 2010;46(9):1679–1691. doi: 10.1016/j.ejca.2010.02.036. [DOI] [PubMed] [Google Scholar]
- 251.Yang L, Tang H, Kong Y, et al. LGR5 Promotes Breast Cancer Progression and Maintains Stem-Like Cells Through Activation of Wnt/beta-Catenin Signaling. Stem Cells. 2015 doi: 10.1002/stem.2083. [DOI] [PubMed] [Google Scholar]
- 252.Kawaguchi-Ihara N, Murohashi I, Nara N, et al. Promotion of the self-renewal capacity of human acute leukemia cells by Wnt3A. Anticancer Res. 2008;28(5A):2701–2704. [PubMed] [Google Scholar]
- 253.Bisson I, Prowse DM. WNT signaling regulates self-renewal and differentiation of prostate cancer cells with stem cell characteristics. Cell Res. 2009;19(6):683–697. doi: 10.1038/cr.2009.43. [DOI] [PubMed] [Google Scholar]
- 254.Molejon MI, Tellechea JI, Moutardier V, et al. Targeting CD44 as a novel therapeutic approach for treating pancreatic cancer recurrence. Oncoscience. 2015;2(6):572–575. doi: 10.18632/oncoscience.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Schulenburg A, Cech P, Herbacek I, et al. CD44-positive colorectal adenoma cells express the potential stem cell markers musashi antigen (msi1) and ephrin B2 receptor (EphB2). J Pathol. 2007;213(2):152–160. doi: 10.1002/path.2220. [DOI] [PubMed] [Google Scholar]
- 256.Verkaar F, Zaman GJ. New avenues to target Wnt/beta-catenin signaling. Drug Discov Today. 2011;16(1-2):35–41. doi: 10.1016/j.drudis.2010.11.007. [DOI] [PubMed] [Google Scholar]
- 257.Polakis P. Drugging Wnt signalling in cancer. The EMBO journal. 2012;31(12):2737–2746. doi: 10.1038/emboj.2012.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Jamieson C, Sharma M, Henderson BR. Targeting the beta-catenin nuclear transport pathway in cancer. Semin Cancer Biol. 2014;27:20–29. doi: 10.1016/j.semcancer.2014.04.012. [DOI] [PubMed] [Google Scholar]
- 259.Kahn M. Can we safely target the WNT pathway? Nat Rev Drug Discov. 2014;13(7):513–532. doi: 10.1038/nrd4233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Madan B, Virshup DM. Targeting Wnts at the source--new mechanisms, new biomarkers, new drugs. Mol Cancer Ther. 2015;14(5):1087–1094. doi: 10.1158/1535-7163.MCT-14-1038. [DOI] [PubMed] [Google Scholar]
- 261.Morin PJ, Vogelstein B, Kinzler KW. Apoptosis and APC in colorectal tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(15):7950–7954. doi: 10.1073/pnas.93.15.7950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Chen S, Guttridge DC, You Z, et al. Wnt-1 signaling inhibits apoptosis by activating beta-catenin/T cell factor-mediated transcription. The Journal of cell biology. 2001;152(1):87–96. doi: 10.1083/jcb.152.1.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.He B, You L, Uematsu K, et al. A monoclonal antibody against Wnt-1 induces apoptosis in human cancer cells. Neoplasia. 2004;6(1):7–14. doi: 10.1016/s1476-5586(04)80048-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Mikami I, You L, He B, et al. Efficacy of Wnt-1 monoclonal antibody in sarcoma cells. BMC Cancer. 2005;5:53. doi: 10.1186/1471-2407-5-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Wei W, Chua MS, Grepper S, et al. Blockade of Wnt-1 signaling leads to anti-tumor effects in hepatocellular carcinoma cells. Mol Cancer. 2009;8:76. doi: 10.1186/1476-4598-8-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Lavergne E, Hendaoui I, Coulouarn C, et al. Blocking Wnt signaling by SFRP-like molecules inhibits in vivo cell proliferation and tumor growth in cells carrying active beta-catenin. Oncogene. 2011;30(4):423–433. doi: 10.1038/onc.2010.432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Rubin EM, Guo Y, Tu K, et al. Wnt inhibitory factor 1 decreases tumorigenesis and metastasis in osteosarcoma. Mol Cancer Ther. 2010;9(3):731–741. doi: 10.1158/1535-7163.MCT-09-0147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Horvath LG, Henshall SM, Kench JG, et al. Membranous expression of secreted frizzled-related protein 4 predicts for good prognosis in localized prostate cancer and inhibits PC3 cellular proliferation in vitro. Clin Cancer Res. 2004;10(2):615–625. doi: 10.1158/1078-0432.ccr-0707-03. [DOI] [PubMed] [Google Scholar]
- 269.Joesting MS, Perrin S, Elenbaas B, et al. Identification of SFRP1 as a candidate mediator of stromal-to-epithelial signaling in prostate cancer. Cancer research. 2005;65(22):10423–10430. doi: 10.1158/0008-5472.CAN-05-0824. [DOI] [PubMed] [Google Scholar]
- 270.DeAlmeida VI, Miao L, Ernst JA, et al. The soluble wnt receptor Frizzled8CRD-hFc inhibits the growth of teratocarcinomas in vivo. Cancer research. 2007;67(11):5371–5379. doi: 10.1158/0008-5472.CAN-07-0266. [DOI] [PubMed] [Google Scholar]
- 271.Wei W, Chua MS, Grepper S, et al. Soluble Frizzled-7 receptor inhibits Wnt signaling and sensitizes hepatocellular carcinoma cells towards doxorubicin. Mol Cancer. 2011;10:16. doi: 10.1186/1476-4598-10-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Nambotin SB, Lefrancois L, Sainsily X, et al. Pharmacological inhibition of Frizzled-7 displays anti-tumor properties in hepatocellular carcinoma. J Hepatol. 2011;54(2):288–299. doi: 10.1016/j.jhep.2010.06.033. [DOI] [PubMed] [Google Scholar]
- 273.Shin H, Kim JH, Lee YS, et al. Change in gene expression profiles of secreted frizzled-related proteins (SFRPs) by sodium butyrate in gastric cancers: induction of promoter demethylation and histone modification causing inhibition of Wnt signaling. International journal of oncology. 2012;40(5):1533–1542. doi: 10.3892/ijo.2012.1327. [DOI] [PubMed] [Google Scholar]
- 274.Shan J, Shi DL, Wang J, et al. Identification of a specific inhibitor of the dishevelled PDZ domain. Biochemistry. 2005;44(47):15495–15503. doi: 10.1021/bi0512602. [DOI] [PubMed] [Google Scholar]
- 275.Fujii N, You L, Xu Z, et al. An antagonist of dishevelled protein-protein interaction suppresses beta-catenin-dependent tumor cell growth. Cancer research. 2007;67(2):573–579. doi: 10.1158/0008-5472.CAN-06-2726. [DOI] [PubMed] [Google Scholar]
- 276.Grandy D, Shan J, Zhang X, et al. Discovery and characterization of a small molecule inhibitor of the PDZ domain of dishevelled. The Journal of biological chemistry. 2009;284(24):16256–16263. doi: 10.1074/jbc.M109.009647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Huang SM, Mishina YM, Liu S, et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature. 2009;461(7264):614–620. doi: 10.1038/nature08356. [DOI] [PubMed] [Google Scholar]
- 278.Chen B, Dodge ME, Tang W, et al. Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer. Nat Chem Biol. 2009;5(2):100–107. doi: 10.1038/nchembio.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Waaler J, Machon O, Tumova L, et al. A novel tankyrase inhibitor decreases canonical Wnt signaling in colon carcinoma cells and reduces tumor growth in conditional APC mutant mice. Cancer research. 2012;72(11):2822–2832. doi: 10.1158/0008-5472.CAN-11-3336. [DOI] [PubMed] [Google Scholar]
- 280.Lau T, Chan E, Callow M, et al. A novel tankyrase small-molecule inhibitor suppresses APC mutation-driven colorectal tumor growth. Cancer research. 2013;73(10):3132–3144. doi: 10.1158/0008-5472.CAN-12-4562. [DOI] [PubMed] [Google Scholar]
- 281.Lehtio L, Chi NW, Krauss S. Tankyrases as drug targets. Febs J. 2013;280(15):3576–3593. doi: 10.1111/febs.12320. [DOI] [PubMed] [Google Scholar]
- 282.Liu J, Pan S, Hsieh MH, et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(50):20224–20229. doi: 10.1073/pnas.1314239110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Kabiri Z, Greicius G, Madan B, et al. Stroma provides an intestinal stem cell niche in the absence of epithelial Wnts. Development (Cambridge, England) 2014;141(11):2206–2215. doi: 10.1242/dev.104976. [DOI] [PubMed] [Google Scholar]
- 284.Lepourcelet M, Chen YN, France DS, et al. Small-molecule antagonists of the oncogenic Tcf/beta-catenin protein complex. Cancer cell. 2004;5(1):91–102. doi: 10.1016/s1535-6108(03)00334-9. [DOI] [PubMed] [Google Scholar]
- 285.Wei W, Chua MS, Grepper S, et al. Small molecule antagonists of Tcf4/beta-catenin complex inhibit the growth of HCC cells in vitro and in vivo. International journal of cancer. 2010;126(10):2426–2436. doi: 10.1002/ijc.24810. [DOI] [PubMed] [Google Scholar]
- 286.Minke KS, Staib P, Puetter A, et al. Small molecule inhibitors of WNT signaling effectively induce apoptosis in acute myeloid leukemia cells. Eur J Haematol. 2009;82(3):165–175. doi: 10.1111/j.1600-0609.2008.01188.x. [DOI] [PubMed] [Google Scholar]
- 287.Chen Z, Venkatesan AM, Dehnhardt CM, et al. 2,4-Diamino-quinazolines as inhibitors of beta-catenin/Tcf-4 pathway: Potential treatment for colorectal cancer. Bioorg Med Chem Lett. 2009;19(17):4980–4983. doi: 10.1016/j.bmcl.2009.07.070. [DOI] [PubMed] [Google Scholar]
- 288.Gonsalves FC, Klein K, Carson BB, et al. An RNAi-based chemical genetic screen identifies three small-molecule inhibitors of the Wnt/wingless signaling pathway. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(15):5954–5963. doi: 10.1073/pnas.1017496108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Trosset JY, Dalvit C, Knapp S, et al. Inhibition of protein-protein interactions: the discovery of druglike beta-catenin inhibitors by combining virtual and biophysical screening. Proteins. 2006;64(1):60–67. doi: 10.1002/prot.20955. [DOI] [PubMed] [Google Scholar]
- 290.Tian W, Han X, Yan M, et al. Structure-based discovery of a novel inhibitor targeting the beta-catenin/Tcf4 interaction. Biochemistry. 2012;51(2):724–731. doi: 10.1021/bi201428h. [DOI] [PubMed] [Google Scholar]
- 291.Yao H, Ashihara E, Strovel JW, et al. AV-65, a novel Wnt/beta-catenin signal inhibitor, successfully suppresses progression of multiple myeloma in a mouse model. Blood Cancer J. 2011;1(11):e43. doi: 10.1038/bcj.2011.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Takada K, Zhu D, Bird GH, et al. Targeted disruption of the BCL9/beta-catenin complex inhibits oncogenic Wnt signaling. Sci Transl Med. 2012;4(148):148ra117. doi: 10.1126/scitranslmed.3003808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Grossmann TN, Yeh JT, Bowman BR, et al. Inhibition of oncogenic Wnt signaling through direct targeting of beta-catenin. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(44):17942–17947. doi: 10.1073/pnas.1208396109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Emami KH, Nguyen C, Ma H, et al. A small molecule inhibitor of beta-catenin/CREB-binding protein transcription [corrected]. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(34):12682–12687. doi: 10.1073/pnas.0404875101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Eguchi M, Nguyen C, Lee SC, et al. ICG-001, a novel small molecule regulator of TCF/beta-catenin transcription. Med Chem. 2005;1(5):467–472. doi: 10.2174/1573406054864098. [DOI] [PubMed] [Google Scholar]
- 296.Miyabayashi T, Teo JL, Yamamoto M, et al. Wnt/beta-catenin/CBP signaling maintains long-term murine embryonic stem cell pluripotency. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(13):5668–5673. doi: 10.1073/pnas.0701331104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Hasegawa K, Yasuda SY, Teo JL, et al. Wnt signaling orchestration with a small molecule DYRK inhibitor provides long-term xeno-free human pluripotent cell expansion. Stem Cells Transl Med. 2012;1(1):18–28. doi: 10.5966/sctm.2011-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Marson A, Foreman R, Chevalier B, et al. Wnt signaling promotes reprogramming of somatic cells to pluripotency. Cell Stem Cell. 2008;3(2):132–135. doi: 10.1016/j.stem.2008.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Wend P, Fang L, Zhu Q, et al. Wnt/beta-catenin signalling induces MLL to create epigenetic changes in salivary gland tumours. The EMBO journal. 2013;32(14):1977–1989. doi: 10.1038/emboj.2013.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Gang EJ, Hsieh YT, Pham J, et al. Small-molecule inhibition of CBP/catenin interactions eliminates drug-resistant clones in acute lymphoblastic leukemia. Oncogene. 2014;33(17):2169–2178. doi: 10.1038/onc.2013.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.He K, Xu T, Xu Y, et al. Cancer cells acquire a drug resistant, highly tumorigenic, cancer stem-like phenotype through modulation of the PI3K/Akt/beta-catenin/CBP pathway. International journal of cancer. 2014;134(1):43–54. doi: 10.1002/ijc.28341. [DOI] [PubMed] [Google Scholar]
- 302.Lenz HJ, Kahn M. Safely targeting cancer stem cells via selective catenin coactivator antagonism. Cancer Sci. 2014;105(9):1087–1092. doi: 10.1111/cas.12471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Smith ML, Hawcroft G, Hull MA. The effect of non-steroidal anti-inflammatory drugs on human colorectal cancer cells: evidence of different mechanisms of action. Eur J Cancer. 2000;36(5):664–674. doi: 10.1016/s0959-8049(99)00333-0. [DOI] [PubMed] [Google Scholar]
- 304.Dihlmann S, Siermann A, von Knebel Doeberitz M. The nonsteroidal anti-inflammatory drugs aspirin and indomethacin attenuate beta-catenin/TCF-4 signaling. Oncogene. 2001;20(5):645–653. doi: 10.1038/sj.onc.1204123. [DOI] [PubMed] [Google Scholar]
- 305.Zhou L, An N, Haydon RC, et al. Tyrosine kinase inhibitor STI-571/Gleevec down-regulates the beta-catenin signaling activity. Cancer letters. 2003;193(2):161–170. doi: 10.1016/s0304-3835(03)00013-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Christensen JG, Schreck R, Burrows J, et al. A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo. Cancer research. 2003;63(21):7345–7355. [PubMed] [Google Scholar]
- 307.Baryawno N, Sveinbjornsson B, Eksborg S, et al. Small-molecule inhibitors of phosphatidylinositol 3-kinase/Akt signaling inhibit Wnt/beta-catenin pathway cross-talk and suppress medulloblastoma growth. Cancer research. 2010;70(1):266–276. doi: 10.1158/0008-5472.CAN-09-0578. [DOI] [PubMed] [Google Scholar]
- 308.Pendas-Franco N, Aguilera O, Pereira F, et al. Vitamin D and Wnt/beta-catenin pathway in colon cancer: role and regulation of DICKKOPF genes. Anticancer Res. 2008;28(5A):2613–2623. [PubMed] [Google Scholar]
- 309.Jaiswal AS, Marlow BP, Gupta N, et al. Beta-catenin-mediated transactivation and cell-cell adhesion pathways are important in curcumin (diferuylmethane)-induced growth arrest and apoptosis in colon cancer cells. Oncogene. 2002;21(55):8414–8427. doi: 10.1038/sj.onc.1205947. [DOI] [PubMed] [Google Scholar]
- 310.Kim J, Zhang X, Rieger-Christ KM, et al. Suppression of Wnt signaling by the green tea compound (−)-epigallocatechin 3-gallate (EGCG) in invasive breast cancer cells. Requirement of the transcriptional repressor HBP1. The Journal of biological chemistry. 2006;281(16):10865–10875. doi: 10.1074/jbc.M513378200. [DOI] [PubMed] [Google Scholar]
- 311.Park CH, Chang JY, Hahm ER, et al. Quercetin, a potent inhibitor against beta-catenin/Tcf signaling in SW480 colon cancer cells. Biochemical and biophysical research communications. 2005;328(1):227–234. doi: 10.1016/j.bbrc.2004.12.151. [DOI] [PubMed] [Google Scholar]
- 312.Roccaro AM, Leleu X, Sacco A, et al. Resveratrol exerts antiproliferative activity and induces apoptosis in Waldenstrom's macroglobulinemia. Clin Cancer Res. 2008;14(6):1849–1858. doi: 10.1158/1078-0432.CCR-07-1750. [DOI] [PubMed] [Google Scholar]
- 313.He BC, Gao JL, Luo X, et al. Ginsenoside Rg3 inhibits colorectal tumor growth through the down-regulation of Wnt/ss-catenin signaling. International journal of oncology. 38(2):437–445. doi: 10.3892/ijo.2010.858. [DOI] [PubMed] [Google Scholar]
- 314.He BC, Gao JL, Zhang BQ, et al. Tetrandrine inhibits Wnt/beta-catenin signaling and suppresses tumor growth of human colorectal cancer. Molecular pharmacology. 79(2):211–219. doi: 10.1124/mol.110.068668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315.Lu W, Lin C, King TD, et al. Silibinin inhibits Wnt/beta-catenin signaling by suppressing Wnt co-receptor LRP6 expression in human prostate and breast cancer cells. Cell Signal. 2012;24(12):2291–2296. doi: 10.1016/j.cellsig.2012.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.Lu W, Lin C, Li Y. Rottlerin induces Wnt co-receptor LRP6 degradation and suppresses both Wnt/beta-catenin and mTORC1 signaling in prostate and breast cancer cells. Cell Signal. 2014;26(6):1303–1309. doi: 10.1016/j.cellsig.2014.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Thorne CA, Hanson AJ, Schneider J, et al. Small-molecule inhibition of Wnt signaling through activation of casein kinase 1alpha. Nat Chem Biol. 2010;6(11):829–836. doi: 10.1038/nchembio.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318.Chen M, Wang J, Lu J, et al. The anti-helminthic niclosamide inhibits Wnt/Frizzled1 signaling. Biochemistry. 2009;48(43):10267–10274. doi: 10.1021/bi9009677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Lu W, Lin C, Roberts MJ, et al. Niclosamide suppresses cancer cell growth by inducing Wnt co-receptor LRP6 degradation and inhibiting the Wnt/beta-catenin pathway. PloS one. 2011;6(12):e29290. doi: 10.1371/journal.pone.0029290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Osada T, Chen M, Yang XY, et al. Antihelminth compound niclosamide downregulates Wnt signaling and elicits antitumor responses in tumors with activating APC mutations. Cancer research. 2011;71(12):4172–4182. doi: 10.1158/0008-5472.CAN-10-3978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Arend RC, Londono-Joshi AI, Samant RS, et al. Inhibition of Wnt/beta-catenin pathway by niclosamide: a therapeutic target for ovarian cancer. Gynecol Oncol. 2014;134(1):112–120. doi: 10.1016/j.ygyno.2014.04.005. [DOI] [PubMed] [Google Scholar]
- 322.Liu C, Lou W, Armstrong C, et al. Niclosamide suppresses cell migration and invasion in enzalutamide resistant prostate cancer cells via Stat3-AR axis inhibition. Prostate. 2015;75(13):1341–1353. doi: 10.1002/pros.23015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323.Liu C, Lou W, Zhu Y, et al. Niclosamide inhibits androgen receptor variants expression and overcomes enzalutamide resistance in castration-resistant prostate cancer. Clin Cancer Res. 2014;20(12):3198–3210. doi: 10.1158/1078-0432.CCR-13-3296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.Londono-Joshi AI, Arend RC, Aristizabal L, et al. Effect of niclosamide on basal-like breast cancers. Mol Cancer Ther. 2014;13(4):800–811. doi: 10.1158/1535-7163.MCT-13-0555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Liao Z, Nan G, Yan Z, et al. The Anthelmintic Drug Niclosamide Inhibits the Proliferative Activity of Human Osteosarcoma Cells by Targeting Multiple Signal Pathways. Current cancer drug targets. 2015 doi: 10.2174/1568009615666150629132157. [DOI] [PubMed] [Google Scholar]
- 326.Lu D, Choi MY, Yu J, et al. Salinomycin inhibits Wnt signaling and selectively induces apoptosis in chronic lymphocytic leukemia cells. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(32):13253–13257. doi: 10.1073/pnas.1110431108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327.Tumova L, Pombinho AR, Vojtechova M, et al. Monensin inhibits canonical Wnt signaling in human colorectal cancer cells and suppresses tumor growth in multiple intestinal neoplasia mice. Mol Cancer Ther. 2014;13(4):812–822. doi: 10.1158/1535-7163.MCT-13-0625. [DOI] [PubMed] [Google Scholar]