Skip to main content
Erschienen in: Molecular Cancer 1/2010

Open Access 01.12.2010 | Review

MicroRNA roles in beta-catenin pathway

verfasst von: Kai Huang, Jun-Xia Zhang, Lei Han, Yong-Ping You, Tao Jiang, Pei-Yu Pu, Chun-Sheng Kang

Erschienen in: Molecular Cancer | Ausgabe 1/2010

Abstract

β-catenin, a key factor in the Wnt signaling pathway, has essential functions in the regulation of cell growth and differentiation. Aberrant β-catenin signaling has been linked to various disease pathologies, including an important role in tumorigenesis. Here, we review the regulation of the Wnt signaling pathway as it relates to β-catenin signaling in tumorigenesis, with particular focus on the role of microRNAs. Finally, we discuss the potential of β-catenin targeted therapeutics for cancer treatment.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-9-252) contains supplementary material, which is available to authorized users.
Kai Huang, Jun-Xia Zhang contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

KH, JXZ, LH and YPY drafted and wrote the manuscript. TJ and PYP revised the manuscript critically for important and intellectual content. CSK contributed to the writing of the manuscript and supervised the project. All authors read and approved the final manuscript.

Introduction

Altered function of components of the canonical Wnt/β-catenin signaling pathway is associated with cancer, as multiple Wnt/β-catenin target genes are regulators of cell proliferation, metastatic potential and tumorigenesis [1, 2]. In the absence of Wnt, cytoplasmic β-catenin dissociated from the E-cadherin/β-catenin/α-catenin complex [3] is rapidly phosphorylated by activated glycogen synthase kinase 3β (GSK3β) at Ser33, Ser37, and Thr41[4] and phosphorylated by casein kinase Iα (CK Iα) at Ser45 [5]. These phophorylations prevent the nuclear accumulation of β-catenin, leading to its ubiquitination and subsequent degradation by the ubiquitin/proteasome system [6, 7]. Upon binding of Wnt to the transmembrane receptor Frizzled (FZD), in complex with co-receptors Low-density-lipoprotein receptor-related proteins 5 and 6 (LRP5/6) [8], the Wnt-FZD-LRP-5/6 complex phosphorylates and activates Disheveled (Dsh) [9]. Dsh activation inhibits GSK3β, subsequently decreases β-catenin degradation by the ubiquitination and proteasomal pathways. In turn, β-catenin accumulates in the cytoplasm and nucleus, where it interacts with coregulators of transcription including T cell factor/lymphocyte enhancer factor (Tcf/Lef) to form a β-catenin/Lef/Tcf complex [10]. This complex regulates transcription of multiple genes involved in cellular proliferation, differentiation, survival and apoptosis, including c-myc and cyclin D [11, 12]. Recent reports suggest that nuclear GSK3β can additionally inhibit β-catenin transcription indirectly, via binding and phosphorylation of Axin and then reducing the transcriptional activity of the β-catenin/Tcf/Lef complex [13]. Hyperactivation of β-catenin caused by the overexpression of Wnt or mutation of CTNNB1 (the gene which encodes β-catenin), GSK3β, Axin or APC is a common cause of carcinoma [14, 15]. Specifically, the mutation of APC is a leading cause of colorectal carcinomas [16], and the relative mRNA and protein expression of β-catenin positively correlates with histological malignancy in astrocytoma [17, 18].
Up to now, there have been more than ten thousands of β-catenin related publications in MEDLINE (Pubmed with: beta catenin). Over the last 10 years, the number of new entries about β-catenin in MEDLINE has grown at a 9.3% compounded annual growth rate, and the number of new entries in MEDLINE each year has grown at a compounded annual growth rate of 3.1% [19]. Furthermore, we queried Pubmed with: (catenin or CTNNB or CTNNB1) and ("1980/01/01"[PDAT]: "2009/05/24"[PDAT]), and identified 10018 articles describing putative interactions between β-catenin and other genes (543 genes) by text mining. 213 genes (including Tcf4 and Lef, ect.) interact with β-catenin (interaction relations is associate, bind, etc.) and a β-catenin interaction network was constructed (Fig 1). Therefore, great progress in biological function and molecular mechanism of β-catenin has been made, and new highlights for β-catenin pathway are deserved to summary. In this review, we focus on modulators of the Wnt/β-catenin signaling pathway, describing new findings of upstream regulators (Fig. 2), coregulators (Fig. 3) and downstream targets, with special focus on the function of microRNAs (Table 1). Finally, we aim to emphasize the importance of the Wnt/β-catenin signaling pathway in cancer, describing β-catenin-targeted reagents that hold promise as chemotherapeutics.
Table 1
Novel modulators of Wnt/β-catenin signaling pathway
upstream regulators
function
coregulators
function
downstream targets
transcription
EGFR [20, 21]
CtBP [53]
AKT1 [77]
AKT1 [26, 28]
Groucho [52]
STAT3 [86, 87]
miR-135a/b [50]
HDAC1/2 [61, 62]
Gbx2 [82]
miR-315 [51]
KLF4 [72]
MMP1 [20]
JNK [34, 35]
↑ or ↓
MAD2B [73]
Foxc1 [84]
miR-200a [36, 42]
VentX [74]
StarD7 [85]
miR-21 [43]
HDAC3/6 [64, 65]
E2F1 [87]
miR-203 [48]
c-Jun [66]
p16INK4a [83]
miR-8 [49]
TNIK [70]
miR-15/16 [93]
  
Snail [71]
miR-122a [94]
    
miR-375 [95]
↑: Regulator upregulates β-catenin transcriptional activity
↓: Regulator downregulates β-catenin transcriptional activity
→: Target transcription is upregulated by activated β-catenin signaling
←: Target transcription is downregulated by activated β-catenin signaling

Upstream Regulators of β-catenin transcriptional activity

EGFR activation phosphorylates β-catenin at Tyr654

Activation of EGFR induces phosphorylation and activation of CK2α via ERK. CK2α activation signals phosphorylation of α-catenin at S641, and triggers loss of α-catenin binding to β-catenin and subsequent activates β-catenin/Lef/Tcf transcriptional activity [20]. Additionally, EGFR activation leads to phosphorylation of β-catenin on Tyr654 residue. EGFR binds to β-catenin and induces its tyrosine phosphorylation [21], definitive evidence that EGFR directly phosphorylates β-catenin at Tyr654 remains elusive. However, Tyr654 phosphorylation results in dissociation of the E-cadherin/α-catenin/β-catenin complex [22]. Further, EGFR regulates β-catenin localization and stability, transcriptional activity, and tumor progression in oral cancer [23]. In addition, phosphorylation of β-catenin at Tyr654 and 670 mediated by interaction of the hepatocyte growth factor (HGF) with its cognate receptor (Met), regulates its nuclear translocation and activation [24]. Whether the Tyr654 residue of β-catenin is a direct target of EGFR requires further determination.

β-catenin is directly phosphorylated at Ser552 by AKT1

AKT1, a serine/threonin kinase also known as PKB, functions as a key mediator of the PI3K/AKT pathway. Activated AKT1 is phosphorylated at Thr308 and Ser473, thereby regulating β-catenin by inducing phosphorylation of effector molecules GSK3β, mTOR and BAD [2527]. Specifically, GSK3β phosphorylation at Ser9 by AKT1 inhibits the N-terminal phosphorylation of β-catenin [26]. A resent study, however, suggests that AKT1 can regulate β-catenin directly by inducing phosphorylation at Ser552 in vitro and in vivo [28]. This Ser552 residue was confirmed as a phosphorylation site by liquid chromatography-coupled ion trap mass spectrometry (LC-MS/MS), and validated by site directed mutagenesis. Ser552 phosphorylation results in β-catenin translocation from the cytosol into the nucleus, increasing Tcf4/Lef1 transcriptional activity and promoting tumor cell invasion. β-catenin phosphorylation of Ser552 was additionally identified in intestinal crypts [29]. Cells exhibiting nuclear β-catenin phosphorylation at Ser552 were frequently clustered together and found at sites of crypt fission and invagination during crypt budding, revealing an interesting role for AKT1 in stem cell biology. In sum, the phosphorylation of β-catenin at Ser552 by AKT1 increases nuclear translocation and retention, enhancing Tcf4/Lef1 complex transcriptional activity to promote tumor cell invasion and stem cell migration. The exact mechanism how the phosphorylation of β-catenin at Ser552 positively regulates the expression of its target genes remains unclear. One possibility exists that phosphorylation of Ser552 may modulate the translocation of β-catenin through nuclear pores, while a second possibility remains that phosphorylation by AKT1 occurs within the nucleus and affects function rather than localization [29]. Further investigation is needed to determine whether the phosphorylation of β-catenin at Ser552 exerts the same effect as stabilized β-catenin on binding to the Tcf4/Lef1 complex and activating overexpression of Wnt target genes.

β-catenin is phosphorylated by JNK

The c-Jun N-terminal kinase (JNK) is a stress-activated protein kinase that is a member of an evolutionarily conserved sub-family of the mitogen-activated protein kinase (MAPK) family of serine/threonine protein kinases. In early Xenopus embryos, high level of nuclear JNK negatively regulates the canonical Wnt/β-catenin signaling pathway by expelling β-catenin from the nucleus [30]. A recent study demonstrates that JNK directly binds to and phosphorylates β-catenin at Ser37 and Thr41, regulating the formation of adherens junctions in human epithelial cell lines [31]. Pharmacologic inhibition of JNK with SP600125 or by expression of dominant negative JNK (JNKDN) decreased the phosphorylation of β-catenin and induced translocation of both β-catenin and E-cadherin to the cell surface. This event disrupts cell-cell adhesion stimulated by okadaic acid (OA) treatment, suggesting that cell adhesion is dynamically regulated by JNK. Further, data suggests that JNK regulation of cell adhesion may contribute to processes including wound healing, tumor proliferation and metastasis.
Loss of JNK 1/2 results in increased expression of β-catenin, and β-catenin/Tcf complex target genes, including c-myc [31, 32]. Phosphorylation and degradation of β-catenin by JNK 1/2 in these studies was blocked by pharmacologic inhibition or RNAi knockdown of GSK3β. Similarly, immunoblotting revealed that JNK1/2 activated GSK3β activation, promoting β-catenin degradation. The interaction between JNK1/2, β-catenin and GSK3β, confirmed by coimmunoprecipitation and confocal microscopy, suggests that β-catenin is phophorylated and degraded by JNK/β-catenin/GSK3β complex. Studies by Ximei Wu and colleagues demonstrated that JNK2 is more potent than JNK1 in phosphorylating β-catenin at Ser191 and Ser605 in ST2 cells [33]. In contrast to the phosphorylations at Ser33, Ser37 and Thr41 by GSK3β in canonical Wnt signaling pathway, the phosphorylations at Ser191 and Ser605 of β-catenin results in stabilization of the protein, translocation to the nucleus, and increased bind of and transcription by the Tcf/Lef complex. Further, JNK enhances the transcriptional activity of β-catenin by phophorylating and activating c-Jun/AP-1, a well characterized coactivator of the β-catenin/Tcf/Lef complex [34]. However, a recent report has revealed that phosphorylation of JNK1 induced by selenium suppresses β-catenin in vivo, resulting in cell growth inhibition [35]. Thus, the discrepancy in the regulation of β-catenin/Tcf complex transcriptional activity by JNK may vary depending on cell type and microenvironment.

Targeting β-catenin-mediated transcription by miR-200a

MicroRNAs are single-stranded noncoding RNAs of 21 to 23 nucleotides in length that repress translation or induce cleavage of target mRNAs that they are partially complementary to at the 3'or 5' untranslated region (UTR). MiR-200a was recently reported to downregulate β-catenin-mediated transcription via two different mechanisms. MiR-200a targets the mRNA of the E-cadherin repressor proteins ZEB1(also known as Tcf8) and ZEB2 (also known as SIP1), subsequently increases the total E-cadherin available for binding to β-catenin and induces formation of the cell-cell adhesion complex [36]. β-catenin is subsequently phosphorylated at Ser33, Ser 37, Thr 41 and Ser45 by the tumor destruction complex and degraded by the ubiquitin/proteasome system. Reduction of miRNA-200a upregulates free cytoplasmic and nuclear β-catenin levels and then induces epithelial to mesenchymal transition (EMT), revealing a role for β-catenin in this process [3741]. A second, novel mechanism for downregulation of β-catenin activity by miR-200a was recently proposed in meningiomas [42]. MiR-200a, down-regulated in most meningiomas, regulates expression of β-catenin and activation of Wnt/β-catenin signaling via directly targeting the 3'UTR of β-catenin mRNA. While miR-200b and miR-200c, two additional members of miR-200 family, show no impact on β-catenin expression.

MiR-21 targets WNT1 gene expression

Protein and mRNA analyses identified that WNT1 is translationally repressed by miR-21. Antagonism of the effects of miR-21, either by transfection with miRNA inhibitors or by exogenous addition of Wnt-1, inhibits human monocyte-derived dendritic cell (MDDC) differentiation, suggesting that miR-21 has a key regulatory role in MDDC differentiation [43]. Interestingly, miR-21 was previously shown to additionally target the tumor suppressor PTEN, RECK and PDCD4 and induce tumorgenesis [4447], suggesting a general role for miR-21 in tumor progression.

MiR-203 directly targets Lef1

MiR-203 directly targets the Wnt signaling transcription factor Lef1 in zebrafish [48]. The 3'UTR of Lef1 contains two potential miRNA recognition elements (MREs) for miR-203. Expression of Lef1 from mRNAs lacking 3'UTR recognition elements can rescue the effects of excess miR-203, demonstrating that these effects are due to specific regulation of Lef1 by miR-203. MiR-203 was found to be significantly downregulated during fin regeneration. Further, repression of Lef1 by miR-203 blocks fin regeneration, whereas loss of miR-203 results in excess Lef1 levels and fin overgrowth.

MiR-8 impacts Wnt/ β-catenin signaling via the Wg pathway

While it is known that mammalian miR-8 family members promote adipogenesis, possibly by inhibiting Wg signaling, the mechanism of this event remains elusive. Three potential mechanisms have been reported. Expression of miR-8 may potently antagonize Wg signaling by directly binding the 3'UTR of wntless (wls) and inhibiting Wg signaling in part by preventing Wg secretion [49]. MiR-8 additionally may impact downstream of the Wg signal, by repressing Tcf protein levels. While miR-8 does not directly target the two putative 3' UTRs of Tcf mRNA, it may directly target the Tcf mRNA independently of its 3'UTR or through an indirect mechanism. Finally, miR-8 may exert its impact by directly targeting CG32767, a positive regulator of the Wg pathway.

MiRNAs target APC/Axin

MiR-135a and miR-135b target the 3'UTR of APC and suppress its expression, subsequently increasing Wnt signaling by stabilizing β-catenin [50]. Similar to the correlation of loss-of-function APC to colorectal tumorigenesis, increasing expression of miR-135a and miR-135b promoted progression of colorectal adenomas to adenocarcinomas. This association is independent of the status of APC mutation or promoter hypermethylation in the tumors. Further, MiR-315 directly targets Axin and Notum, two negative regulators of Wg signaling, in Drosophila cells mice, resulting in Wnt pathway activation [51].

Coregulators of β-catenin transcriptional ativity

Coregulation of the β-catenin/Tcf4/Lef1 complex

The Tcf family consists of four members in vertebrates, including Tcf1, Tcf3, Tcf4 and Lef1. Each member contains a DNA-binding high mobility group (HMG) box, and a highly conserved β-catenin interacting region. In the absence of β-catenin, Tcf4 recruits co-repressors HDAC1, CtBP, and Groucho/transducin-like enhancer of Split (TLE) to silence expression of target genes [5254]. Groucho/TLE proteins repress the basal transcriptional machinery [55] as well as recruit HDACs, which contribute to corepression by direct action on chromatin [56]. A direct interaction between Groucho and Lef1 occurs via a small region in the context regulatory domain (aa237-256) and a region in the highly conserved HMG DNA binding domain (aa296-396) [52]. However, as β-catenin accumulates in the nucleus, Groucho/TLE is displaced from Tcf/Lef by β-catenin binding to C-terminal of Lef1 (residure252-397). This C-terminal DNA-binding domain overlaps with the Groucho/TLE binding site [57]. β-catenin then recruits co-activators through its N-terminal and C-terminal transactivation domains, including BCL9/Legless [58, 59] and p300/CBP [60].

β-catenin transcriptional activity is regulated by Histone deacetylases

Histone deacetylases (HDACs) are a class of enzymes that remove acetyl groups from ε-N-acetyl lysine amino acids on a histone. HDACs attenuate β-catenin transcriptional activity by binding to Lef, forming a HDAC-Lef complex that hypoacetylates the promoter region of β-catenin/Tcf/Lef complex target genes [52, 61]. Upon β-catenin recruitment and accumulation in the nucleus, however, HDACs dissociate from Lef and form a HDAC/β-catenin complex, attenuating the enzymatic activity of HDAC and allowing residual β-catenin to bind to Lef and recover the transcriptional activity of the β-catenin/Tcf/Lef complex [61]. Recently, HDAC1/2 were demonstrated to regulate oligodendrocyte differentiation, at least in part, via disruption of β-catenin/Tcf interactions [62]. HDAC1/2 competes with β-catenin for Tcf4 interaction, thus regulating β-catenin/Tcf4 complex transcriptional activity. Specifically, HDAC1/2 regulate expression of differentiation inhibitors2/4 (ID2/4), target genes of the β-catenin/Tcf4 complex that negatively regulate oligodendrocyte differentiate. The displacement of Tcf4 from β-catenin by HDAC1/2 or the formation of ternary complex HDAC/β-catenin/Tcf4 switches off transcription of ID2/4. Thus, transcriptional co-repressors HDAC1 and HDAC2 compete with β-catenin for Tcf4 interaction to promote oligodendrocyte differentiation in a manner, at least in part, dependent on the expression of β-catenin/Tcf4 complex target genes ID2/4. Similarly, in the zebrafish retina, HDAC1 antagonizes Wnt signaling to suppress both cell-cycle progression and subsequent inhibition of neurogenesis [63].
Recent evidence identifies that HDAC3 expression may enhance transcription activity of the Wnt/β-catenin pathway [64]. Stable HDAC3 knockdown attenuated activation of the Wnt pathway by increasing plasma membrane localization and reducing nuclear accumulation of β-catenin. Further, knockdown of HDAC3 induced increased expression of TLE1/4, which provided competition with β-catenin for interaction with Tcf4/Lef1, thereby antagonizing transcription. Moreover, HDAC6 may similarly enhance transcription activity of the Wnt/β-catenin pathway. HDAC6 deacetylates β-catenin at lysine 49 and inhibits its phosphorylation at Ser45, resulting in its nuclear translocation and accumulation [65].

c-Jun functions as a coactivator of the β-catenin/Tcf complex

c-Jun is well known as a downstream effector of the β-catenin/Tcf complex [66]. c-Jun heterodimerizes and forms a functional transcription factor complex termed AP-1 in combination with c-Fos. Recently, c-Jun and AP-1 have been demonstrated by genome wide ChIP-on-chip analysis to interact with β-catenin and form a c-Jun/AP-1/β-catenin/Tcf complex, which prevents β-catenin phosphorylation and degradation and increases its activity [67]. A recent study additionally proved that c-Jun directly binds to the β-catenin/Tcf complex [68], suggesting that c-Jun functions as an adaptor protein to mediate the association of Dsh with the β-catenin/Tcf complex on the promoter of Wnt target genes. The Dsh/c-Jun/β-catenin/Tcf complex similarly increases β-catenin stabilization and positively regulates the activity of Wnt signaling pathway. Each of these functions of c-Jun is dependent on its phosphorylation by JNK. In contrast, Wnt3a-induced Tcf reporter activity is not affected by knockdown of endogenous c-Fos, indicating that c-Fos does not impact canonical Wnt signaling as c-Jun [69]. Whether c-Fos binds to β-catenin or the c-Fos/β/catenin/Tcf complex is an avenue for further investigation.

Additional coregulators of the β-catenin/Tcf/Lef complex

Multiple novel upstream regulators of β-catenin/Tcf/Lef complex transcriptional activity have been identified. Positive modulators of β-catenin activity include Nck-interacting kinase (TNIK) and Snail. TNIK expression is enriched in the nuclei of Wnt-activated intestinal crypts, but not cells of the villus, where it is specifically recruited to the promoters of Wnt target genes Axin2 and c-Myc [70]. In vitro immunoprecipitation and kinase assays reveal that TNIK directly binds to both Tcf4 and β-catenin in these cells, phosphorylating and activating the transcription activity of Tcf4. Snail interacts with β-catenin at its N-terminus and increases its transcriptional activity independent of Tcf4. Snail, which is transcriptionally repressed by GSK3β, up-regulates TGF-3β gene expression through β-catenin/Tcf4 and promotes EMT [71]. In contrast, multiple negative regulators of β-catenin signaling exist as well. Krüppel-like factor 4 (KLF4), a transcription factor highly expressed in normal human intestine and critical for intestinal differentiation, inhibits Wnt signaling by direct interaction with the C-terminal transactivation domain of β-catenin, blocking recruitment of p300/CBP to this domain. KLF4 inhibition of p300/CBP recruitment results in inhibition of both β-catenin acetylation as well as histone acetylation of Wnt target genes. KLF4 additionally directly interacts with Tcf4 independent of β-catenin [72]. MAD2B, a novel Tcf4 coregulator identified by coimmunoprecipitation, downregulates β-catenin/Tcf/Lef complex activity by interrupting the DNA binding ability of Tcf4 [73]. Finally, VentX, a human Xom homologue, is a Lef/Tcf-associated inhibitor of canonical Wnt/β-catenin signaling and a negative regulator of cell proliferation[74].

Downstream targets of β-catenin

Upon translocation to the nucleus, β-catenin interacts with Tcf4 and Lef1 to regulate a wide range of gene expression at the transcriptional level [75]. The complex binds to A-C/G-A/T-T-C-A-A-A-G motifs, an evolutionarily conserved consensus motif on the promoter of target genes [76]. While c-myc, cyclin D and c-Jun expression are well-characterized as regulated by this complex [11, 12, 66], multiple new targets have recently been identified http://​www.​stanford.​edu/​~rnusse/​pathways/​targets.​html. Here, we summarize the novel downstream targets of β-catenin and the positive feedback loops between β-catenin and the downstream targets (Fig 4).

AKT1 is regulated by β-catenin at the transcriptional level

Several non-steroidal anti-inflammatory drugs (NSAIDs) suppress β-catenin expression in human cancer cell lines, including aspirin, indomethacin, sulindac and etodolac [7779]. Nitric oxide-donating aspirin (NO-ASA) inhibits the transcriptional activity of β-catenin/Tcf far more potently than aspirin, and inhibits the growth of colorectal cancer cells (CRC) more efficiently [80]. Aspirin and indomethacin downregulate β-catenin activity through increasing the stabilization of phosphorylated β-catenin in time- and concentration-dependent manners. These studies identified that aspirin induces a decrease in expression of AKT1, which is regulated by the β-catenin/Tcf complex as revealed by reporter assay [77]. Functionally, active AKT1 induced by β-catenin decreases Bax activation, oligomerization, and translocation to mitochondria, thus antagonizing mitochondrial injury and apoptosis [81]. As AKT2 and AKT3 are closely related and highly conserved homologs of AKT1, whether AKT2 and AKT3 are targets of Wnt/β-catenin signaling will be subject to additional studies.

Determination of novel β-catenin-regulated genes by ChIP assay

ChIP and transgenic analysis identified that the Gbx2 regulatory elements that drive expression in the neural crest (NC) respond directly to Wnt/β-catenin signaling. Loss-of-function experiments using antisense morpholinos against Gbx2 inhibit NC protein expression and expand the preplacodal domain, whereas Gbx2 overexpression leads to transformation of the preplacodal domain into NC cells. Previous studies identified a region of 500 bp upstream of Gbx2 that contains three (1-3) Lef/Tcf consensus sequences, termed the Gbx2 enhancer [82]. Similarly, the β-catenin/Tcf4 complex binds to a specific site on matrix metalloproteinase 1 (MMP1) promoter and governs MMP1 gene and protein expression, regulating cell migration in collagen and gelatin [20]. Further, β-catenin directly represses p16INK4a expression by binding to its promoter. Activated β-catenin directly represses the expression of p16INK4a through an evolutionarily conserved Lef/Tcf site in its promoter [83]. ChIP analysis additioanlly determined that β-catenin bound to conserved regions of mouse genomic DNA proximal to the Foxc1 transcriptional start site, revealing that Foxc1 is a direct target of β-catenin [84]. Despite β-catenin regulation of Foxc2 expression, no transcriptional start site has yet been identified. Finally, ChIP analysis revealed that β-catenin and Tcf4 activated the human StarD7 gene interacting with its promoter region [85].
Previously, WNT/β-catenin was suggested to regulate STAT3 at the mRNA and protein level, suggesting that STAT3 maybe a direct target of β-catenin [86]. Further investigation was performed by using EMSA and ChIP assay, confirming that the β-catenin/Tcf4 complex directly bound to the Tcf4 binding element site (TBE) of the STAT3 gene promoter [87]. These data confirm that STAT3 is regulated by β-catenin in the transcriptional level. Of note, STAT3 activation increases the nuclear accumulation of β-catenin, leading to a positive feedback loop between β-catenin and STAT3 [88]. Similarly, the E2F1 promoter was found to contain two putative Tcf-binding elements, and promoter activity is inhibited by overexpression of β-catenin/Tcf [89]. This event represents a positive feedback loop for β-catenin transcriptional activity, as E2F1 represses β-catenin transcription directly [90] or via upregulation of Siah1 [91]. Additionally, Lef1 activates E2F1 by attenuating the interaction between E2F1 and HDAC1 in a β-catenin-independent manner [92].

β-catenin downregulates miR-15, miR-16, miR-122a and miR-375 expression

Wnt/β-catenin signaling regulates miR-15/16 maturation rather than its transcription, as overexpression of β-catenin inhibits the expression of mature miR-15 and miR-16 isoforms. The mechanism of Wnt control of miR-15 and miR-16 maturation is unknown, but perhaps works through a protein complex controlled by or containing β-catenin. β-catenin control of the intensity and spatial pattern of Nodal responsiveness is thought to be regulated by miR-15 and miR-16 expression [93]. Upregulation of miR-122a expression in mutant APC cells induces a gain of wild type APC function, indicating that miR-122a works downstream of APC and suggests that miR-122a expression is lost or downregulated in APC-driven gastrointestinal cancers. Further, restoration of miR-122a expression significantly suppressed migration, invasion, anchorage-independent growth, and in vivo tumorigenicity of hepatocellular carcinoma cells. The mechanism by which miR-122a expression bypasses loss-of-function APC is unknown. Data suggests that miR-122a may be a novel target of APC/β-catenin signaling pathway, and that down-regulation of miR-122a mediated by aberrant APC/β-catenin signaling is important to the pathogenesis of gastrointestinal cancers [94]. Additionally, miR-375 has been demonstrated to be downregulated by β-catenin [95]. The function of miR-375 and the transcriptional mechanism that miR-375 regulated by β-catenin are not clear and for further investigation.

Anti-β-catenin agents

The β-catenin complex plays a critical role in tumorgenesis, angiogenesis, and progression of metastasis, making it an attractive therapeutic target for chemotherapy. Current drugs that target β-catenin include Quercetin, which inhibits the transcriptional activity of β-catenin by disrupting the binding of β-catenin and Tcf4 and suppressing their translocation to the nucleus [96, 97]. Similarly, Aspirin was demonstrated to increase phosphorylation of β-catenin but not decrease its nuclear translocation [77]. Neither quercetin nor aspirin, however, attenuate the protein level of total β-catenin. PKF118-310, PKF115-584, and CGP049090 reduce the binding of β-catenin and Tcf4, resulting in induction of G1/S phase arrest, inhibition of cell growth and activation of apoptosis [98]. 2,4-diaminoquinazolines and their analogues, a novel series of β-catenin antagonist, as well as multiple antagonists not presented here, have been demonstrated to inhibit the growth of colorectal cancer [99, 100], proving the value of β-catenin antagonists as potential therapeutics. Taken together, we believe that the potential for β-catenin antagonists as potent chemotherapeutics for Wnt-driven malignancies is great, and further study of these and related agents will yield effective therapies for human cancer.

Conclusion and Perspectives

Although Wnt/β-catenin transcriptional activity has been studied over the past several years, molecular regulation of Wnt/β-catenin pathway is complex and summarized in Fig. 5. Regulation of the expression of components of the Wnt/β-catenin pathway by microRNAs, newly discovered RNA sequences that modify gene expression profiles, has not only revealed increased complexity of β-catenin but enabled identification of increased crosstalk between Wnt/β-catenin signaling and other pathways. Accumulating data of microRNAs will likely identify even greater complexities. In conclusion, we believe that an improved understanding of the basic genetics and biology of β-catenin signaling will provide insights into the development of novel chemopreventive and therapeutic strategies for human cancers.

Acknowledgements

This work was supported by the China National Natural Scientific Fund (30971136, 30872657), the Tianjin Science and Technology Committee (09JCZDJC17600), a Program for New Century Excellent Talents in University (NCET-07-0615). The authors wish to express the gratitude to Shanghai Sensichip Co Ltd for bioinformatics analysis, and Dr. Jie Li (Department of Pathology, Yale University School of Medicine) and Jin-Quan Cheng (Departments of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute) for critical review and helpful discussion with the manuscript.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

KH, JXZ, LH and YPY drafted and wrote the manuscript. TJ and PYP revised the manuscript critically for important and intellectual content. CSK contributed to the writing of the manuscript and supervised the project. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Clevers H: Wnt/beta-catenin signaling in development and disease. Cell. 2006, 127: 469-480. 10.1016/j.cell.2006.10.018CrossRefPubMed Clevers H: Wnt/beta-catenin signaling in development and disease. Cell. 2006, 127: 469-480. 10.1016/j.cell.2006.10.018CrossRefPubMed
2.
Zurück zum Zitat Waltzer L, Bienz M: Drosophila CBP represses the transcription factor TCF to antagonize Wingless signalling. Nature. 1998, 395: 521-525. 10.1038/26785CrossRefPubMed Waltzer L, Bienz M: Drosophila CBP represses the transcription factor TCF to antagonize Wingless signalling. Nature. 1998, 395: 521-525. 10.1038/26785CrossRefPubMed
3.
Zurück zum Zitat Kemler R: Classical cadherins. Semin Cell Biol. 1992, 3: 149-155. 10.1016/S1043-4682(10)80011-XCrossRefPubMed Kemler R: Classical cadherins. Semin Cell Biol. 1992, 3: 149-155. 10.1016/S1043-4682(10)80011-XCrossRefPubMed
4.
Zurück zum Zitat Yost C, Torres M, Miller JR, Huang E, Kimelman D, Moon RT: The axis-inducing activity, stability, and subcellular distribution of beta-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3. Genes Dev. 1996, 10: 1443-1454. 10.1101/gad.10.12.1443CrossRefPubMed Yost C, Torres M, Miller JR, Huang E, Kimelman D, Moon RT: The axis-inducing activity, stability, and subcellular distribution of beta-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3. Genes Dev. 1996, 10: 1443-1454. 10.1101/gad.10.12.1443CrossRefPubMed
5.
Zurück zum Zitat Liu C, Li Y, Semenov M, Han C, Baeg GH, Tan Y, Zhang Z, Lin X, He X: Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell. 2002, 108: 837-847. 10.1016/S0092-8674(02)00685-2CrossRefPubMed Liu C, Li Y, Semenov M, Han C, Baeg GH, Tan Y, Zhang Z, Lin X, He X: Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell. 2002, 108: 837-847. 10.1016/S0092-8674(02)00685-2CrossRefPubMed
6.
Zurück zum Zitat Giles RH, van Es JH, Clevers H: Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta. 2003, 1653: 1-24.PubMed Giles RH, van Es JH, Clevers H: Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta. 2003, 1653: 1-24.PubMed
7.
Zurück zum Zitat Hart M, Concordet JP, Lassot I, Albert I, del los Santos R, Durand H, Perret C, Rubinfeld B, Margottin F, Benarous R, Polakis P: The F-box protein beta-TrCP associates with phosphorylated beta-catenin and regulates its activity in the cell. Curr Biol. 1999, 9: 207-210.CrossRefPubMed Hart M, Concordet JP, Lassot I, Albert I, del los Santos R, Durand H, Perret C, Rubinfeld B, Margottin F, Benarous R, Polakis P: The F-box protein beta-TrCP associates with phosphorylated beta-catenin and regulates its activity in the cell. Curr Biol. 1999, 9: 207-210.CrossRefPubMed
8.
Zurück zum Zitat Cadigan KM, Liu YI: Wnt signaling: complexity at the surface. J Cell Sci. 2006, 119: 395-402. 10.1242/jcs.02826CrossRefPubMed Cadigan KM, Liu YI: Wnt signaling: complexity at the surface. J Cell Sci. 2006, 119: 395-402. 10.1242/jcs.02826CrossRefPubMed
9.
Zurück zum Zitat Malbon CC, Wang HY: Dishevelled: a mobile scaffold catalyzing development. Curr Top Dev Biol. 2006, 72: 153-166. 10.1016/S0070-2153(05)72002-0CrossRefPubMed Malbon CC, Wang HY: Dishevelled: a mobile scaffold catalyzing development. Curr Top Dev Biol. 2006, 72: 153-166. 10.1016/S0070-2153(05)72002-0CrossRefPubMed
10.
Zurück zum Zitat Kimelman D, Xu W: beta-catenin destruction complex: insights and questions from a structural perspective. Oncogene. 2006, 25: 7482-7491. 10.1038/sj.onc.1210055CrossRefPubMed Kimelman D, Xu W: beta-catenin destruction complex: insights and questions from a structural perspective. Oncogene. 2006, 25: 7482-7491. 10.1038/sj.onc.1210055CrossRefPubMed
11.
Zurück zum Zitat He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzler KW: Identification of c-MYC as a target of the APC pathway. Science. 1998, 281: 1509-1512. 10.1126/science.281.5382.1509CrossRefPubMed He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzler KW: Identification of c-MYC as a target of the APC pathway. Science. 1998, 281: 1509-1512. 10.1126/science.281.5382.1509CrossRefPubMed
12.
Zurück zum Zitat Tetsu O, McCormick F: Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 1999, 398: 422-426. 10.1038/18884CrossRefPubMed Tetsu O, McCormick F: Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 1999, 398: 422-426. 10.1038/18884CrossRefPubMed
13.
Zurück zum Zitat Caspi M, Zilberberg A, Eldar-Finkelman H, Rosin-Arbesfeld R: Nuclear GSK-3beta inhibits the canonical Wnt signalling pathway in a beta-catenin phosphorylation-independent manner. Oncogene. 2008, 27: 3546-3555. 10.1038/sj.onc.1211026CrossRefPubMed Caspi M, Zilberberg A, Eldar-Finkelman H, Rosin-Arbesfeld R: Nuclear GSK-3beta inhibits the canonical Wnt signalling pathway in a beta-catenin phosphorylation-independent manner. Oncogene. 2008, 27: 3546-3555. 10.1038/sj.onc.1211026CrossRefPubMed
14.
Zurück zum Zitat Yang LH, Xu HT, Han Y, Li QC, Liu Y, Zhao Y, Yang ZQ, Dong QZ, Miao Y, Dai SD, Wang EH: Axin downregulates TCF-4 transcription via beta-catenin, but not p53, and inhibits the proliferation and invasion of lung cancer cells. Mol Cancer. 2010, 9: 25- 10.1186/1476-4598-9-25PubMedCentralCrossRefPubMed Yang LH, Xu HT, Han Y, Li QC, Liu Y, Zhao Y, Yang ZQ, Dong QZ, Miao Y, Dai SD, Wang EH: Axin downregulates TCF-4 transcription via beta-catenin, but not p53, and inhibits the proliferation and invasion of lung cancer cells. Mol Cancer. 2010, 9: 25- 10.1186/1476-4598-9-25PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Ying Y, Tao Q: Epigenetic disruption of the WNT/beta-catenin signaling pathway in human cancers. Epigenetics. 2009, 4: 307-312. 10.4161/epi.4.5.9371CrossRefPubMed Ying Y, Tao Q: Epigenetic disruption of the WNT/beta-catenin signaling pathway in human cancers. Epigenetics. 2009, 4: 307-312. 10.4161/epi.4.5.9371CrossRefPubMed
16.
Zurück zum Zitat Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW: APC mutations occur early during colorectal tumorigenesis. Nature. 1992, 359: 235-237. 10.1038/359235a0CrossRefPubMed Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW: APC mutations occur early during colorectal tumorigenesis. Nature. 1992, 359: 235-237. 10.1038/359235a0CrossRefPubMed
17.
Zurück zum Zitat Sareddy GR, Panigrahi M, Challa S, Mahadevan A, Babu PP: Activation of Wnt/beta-catenin/Tcf signaling pathway in human astrocytomas. Neurochem Int. 2009, 55: 307-317. 10.1016/j.neuint.2009.03.016CrossRefPubMed Sareddy GR, Panigrahi M, Challa S, Mahadevan A, Babu PP: Activation of Wnt/beta-catenin/Tcf signaling pathway in human astrocytomas. Neurochem Int. 2009, 55: 307-317. 10.1016/j.neuint.2009.03.016CrossRefPubMed
18.
Zurück zum Zitat Pu P, Zhang Z, Kang C, Jiang R, Jia Z, Wang G, Jiang H: Downregulation of Wnt2 and beta-catenin by siRNA suppresses malignant glioma cell growth. Cancer Gene Ther. 2009, 16: 351-361. 10.1038/cgt.2008.78CrossRefPubMed Pu P, Zhang Z, Kang C, Jiang R, Jia Z, Wang G, Jiang H: Downregulation of Wnt2 and beta-catenin by siRNA suppresses malignant glioma cell growth. Cancer Gene Ther. 2009, 16: 351-361. 10.1038/cgt.2008.78CrossRefPubMed
20.
Zurück zum Zitat Jean C, Blanc A, Prade-Houdellier N, Ysebaert L, Hernandez-Pigeon H, Al Saati T, Haure MJ, Coluccia AM, Charveron M, Delabesse E, Laurent G: Epidermal growth factor receptor/beta-catenin/T-cell factor 4/matrix metalloproteinase 1: a new pathway for regulating keratinocyte invasiveness after UVA irradiation. Cancer Res. 2009, 69: 3291-3299. 10.1158/0008-5472.CAN-08-1909CrossRefPubMed Jean C, Blanc A, Prade-Houdellier N, Ysebaert L, Hernandez-Pigeon H, Al Saati T, Haure MJ, Coluccia AM, Charveron M, Delabesse E, Laurent G: Epidermal growth factor receptor/beta-catenin/T-cell factor 4/matrix metalloproteinase 1: a new pathway for regulating keratinocyte invasiveness after UVA irradiation. Cancer Res. 2009, 69: 3291-3299. 10.1158/0008-5472.CAN-08-1909CrossRefPubMed
21.
Zurück zum Zitat Hoschuetzky H, Aberle H, Kemler R: Beta-catenin mediates the interaction of the cadherin-catenin complex with epidermal growth factor receptor. J Cell Biol. 1994, 127: 1375-1380. 10.1083/jcb.127.5.1375CrossRefPubMed Hoschuetzky H, Aberle H, Kemler R: Beta-catenin mediates the interaction of the cadherin-catenin complex with epidermal growth factor receptor. J Cell Biol. 1994, 127: 1375-1380. 10.1083/jcb.127.5.1375CrossRefPubMed
22.
Zurück zum Zitat Roura S, Miravet S, Piedra J, Garcia de Herreros A, Dunach M: Regulation of E-cadherin/Catenin association by tyrosine phosphorylation. J Biol Chem. 1999, 274: 36734-36740. 10.1074/jbc.274.51.36734CrossRefPubMed Roura S, Miravet S, Piedra J, Garcia de Herreros A, Dunach M: Regulation of E-cadherin/Catenin association by tyrosine phosphorylation. J Biol Chem. 1999, 274: 36734-36740. 10.1074/jbc.274.51.36734CrossRefPubMed
23.
Zurück zum Zitat Lee CH, Hung HW, Hung PH, Shieh YS: Epidermal growth factor receptor regulates beta-catenin location, stability, and transcriptional activity in oral cancer. Mol Cancer. 2010, 9: 64- 10.1186/1476-4598-9-64PubMedCentralCrossRefPubMed Lee CH, Hung HW, Hung PH, Shieh YS: Epidermal growth factor receptor regulates beta-catenin location, stability, and transcriptional activity in oral cancer. Mol Cancer. 2010, 9: 64- 10.1186/1476-4598-9-64PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Zeng G, Apte U, Micsenyi A, Bell A, Monga SP: Tyrosine residues 654 and 670 in beta-catenin are crucial in regulation of Met-beta-catenin interactions. Exp Cell Res. 2006, 312: 3620-3630. 10.1016/j.yexcr.2006.08.003PubMedCentralCrossRefPubMed Zeng G, Apte U, Micsenyi A, Bell A, Monga SP: Tyrosine residues 654 and 670 in beta-catenin are crucial in regulation of Met-beta-catenin interactions. Exp Cell Res. 2006, 312: 3620-3630. 10.1016/j.yexcr.2006.08.003PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Blume-Jensen P, Janknecht R, Hunter T: The kit receptor promotes cell survival via activation of PI 3-kinase and subsequent Akt-mediated phosphorylation of Bad on Ser136. Curr Biol. 1998, 8: 779-782. 10.1016/S0960-9822(98)70302-1CrossRefPubMed Blume-Jensen P, Janknecht R, Hunter T: The kit receptor promotes cell survival via activation of PI 3-kinase and subsequent Akt-mediated phosphorylation of Bad on Ser136. Curr Biol. 1998, 8: 779-782. 10.1016/S0960-9822(98)70302-1CrossRefPubMed
26.
Zurück zum Zitat Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA: Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995, 378: 785-789. 10.1038/378785a0CrossRefPubMed Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA: Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995, 378: 785-789. 10.1038/378785a0CrossRefPubMed
27.
Zurück zum Zitat Sekulic A, Hudson CC, Homme JL, Yin P, Otterness DM, Karnitz LM, Abraham RT: A direct linkage between the phosphoinositide 3-kinase-AKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells. Cancer Res. 2000, 60: 3504-3513.PubMed Sekulic A, Hudson CC, Homme JL, Yin P, Otterness DM, Karnitz LM, Abraham RT: A direct linkage between the phosphoinositide 3-kinase-AKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells. Cancer Res. 2000, 60: 3504-3513.PubMed
28.
Zurück zum Zitat Fang D, Hawke D, Zheng Y, Xia Y, Meisenhelder J, Nika H, Mills GB, Kobayashi R, Hunter T, Lu Z: Phosphorylation of beta-catenin by AKT promotes beta-catenin transcriptional activity. J Biol Chem. 2007, 282: 11221-11229. 10.1074/jbc.M611871200PubMedCentralCrossRefPubMed Fang D, Hawke D, Zheng Y, Xia Y, Meisenhelder J, Nika H, Mills GB, Kobayashi R, Hunter T, Lu Z: Phosphorylation of beta-catenin by AKT promotes beta-catenin transcriptional activity. J Biol Chem. 2007, 282: 11221-11229. 10.1074/jbc.M611871200PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat He XC, Yin T, Grindley JC, Tian Q, Sato T, Tao WA, Dirisina R, Porter-Westpfahl KS, Hembree M, Johnson T, Wiedemann LM, Barrett TA, Hood L, Wu H, Li L: PTEN-deficient intestinal stem cells initiate intestinal polyposis. Nat Genet. 2007, 39: 189-198. 10.1038/ng1928CrossRefPubMed He XC, Yin T, Grindley JC, Tian Q, Sato T, Tao WA, Dirisina R, Porter-Westpfahl KS, Hembree M, Johnson T, Wiedemann LM, Barrett TA, Hood L, Wu H, Li L: PTEN-deficient intestinal stem cells initiate intestinal polyposis. Nat Genet. 2007, 39: 189-198. 10.1038/ng1928CrossRefPubMed
30.
Zurück zum Zitat Liao G, Tao Q, Kofron M, Chen JS, Schloemer A, Davis RJ, Hsieh JC, Wylie C, Heasman J, Kuan CY: Jun NH2-terminal kinase (JNK) prevents nuclear beta-catenin accumulation and regulates axis formation in Xenopus embryos. Proc Natl Acad Sci USA. 2006, 103: 16313-16318. 10.1073/pnas.0602557103PubMedCentralCrossRefPubMed Liao G, Tao Q, Kofron M, Chen JS, Schloemer A, Davis RJ, Hsieh JC, Wylie C, Heasman J, Kuan CY: Jun NH2-terminal kinase (JNK) prevents nuclear beta-catenin accumulation and regulates axis formation in Xenopus embryos. Proc Natl Acad Sci USA. 2006, 103: 16313-16318. 10.1073/pnas.0602557103PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Lee MH, Koria P, Qu J, Andreadis ST: JNK phosphorylates beta-catenin and regulates adherens junctions. Faseb J. 2009, 23: 3874-3883. 10.1096/fj.08-117804PubMedCentralCrossRefPubMed Lee MH, Koria P, Qu J, Andreadis ST: JNK phosphorylates beta-catenin and regulates adherens junctions. Faseb J. 2009, 23: 3874-3883. 10.1096/fj.08-117804PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Hu D, Fang W, Han A, Gallagher L, Davis RJ, Xiong B, Yang W: c-Jun N-terminal kinase 1 interacts with and negatively regulates Wnt/beta-catenin signaling through GSK3beta pathway. Carcinogenesis. 2008, 29: 2317-2324. 10.1093/carcin/bgn239PubMedCentralCrossRefPubMed Hu D, Fang W, Han A, Gallagher L, Davis RJ, Xiong B, Yang W: c-Jun N-terminal kinase 1 interacts with and negatively regulates Wnt/beta-catenin signaling through GSK3beta pathway. Carcinogenesis. 2008, 29: 2317-2324. 10.1093/carcin/bgn239PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Wu X, Tu X, Joeng KS, Hilton MJ, Williams DA, Long F: Rac1 activation controls nuclear localization of beta-catenin during canonical Wnt signaling. Cell. 2008, 133: 340-353. 10.1016/j.cell.2008.01.052PubMedCentralCrossRefPubMed Wu X, Tu X, Joeng KS, Hilton MJ, Williams DA, Long F: Rac1 activation controls nuclear localization of beta-catenin during canonical Wnt signaling. Cell. 2008, 133: 340-353. 10.1016/j.cell.2008.01.052PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Nateri AS, Spencer-Dene B, Behrens A: Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development. Nature. 2005, 437: 281-285. 10.1038/nature03914CrossRefPubMed Nateri AS, Spencer-Dene B, Behrens A: Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development. Nature. 2005, 437: 281-285. 10.1038/nature03914CrossRefPubMed
35.
Zurück zum Zitat Fang W, Han A, Bi X, Xiong B, Yang W: Tumor inhibition by sodium selenite is associated with activation of c-Jun NH2-terminal kinase 1 and suppression of beta-catenin signaling. Int J Cancer. 2010, 127: 32-42.PubMedCentralCrossRefPubMed Fang W, Han A, Bi X, Xiong B, Yang W: Tumor inhibition by sodium selenite is associated with activation of c-Jun NH2-terminal kinase 1 and suppression of beta-catenin signaling. Int J Cancer. 2010, 127: 32-42.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Korpal M, Lee ES, Hu G, Kang Y: The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem. 2008, 283: 14910-14914. 10.1074/jbc.C800074200PubMedCentralCrossRefPubMed Korpal M, Lee ES, Hu G, Kang Y: The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem. 2008, 283: 14910-14914. 10.1074/jbc.C800074200PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, Brabletz T: A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008, 9: 582-589. 10.1038/embor.2008.74PubMedCentralCrossRefPubMed Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, Brabletz T: A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008, 9: 582-589. 10.1038/embor.2008.74PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ: The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008, 10: 593-601. 10.1038/ncb1722CrossRefPubMed Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ: The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008, 10: 593-601. 10.1038/ncb1722CrossRefPubMed
39.
Zurück zum Zitat Kong D, Li Y, Wang Z, Banerjee S, Ahmad A, Kim HR, Sarkar FH: miR-200 regulates PDGF-D-mediated epithelial-mesenchymal transition, adhesion, and invasion of prostate cancer cells. Stem Cells. 2009, 27: 1712-1721. 10.1002/stem.101PubMedCentralCrossRefPubMed Kong D, Li Y, Wang Z, Banerjee S, Ahmad A, Kim HR, Sarkar FH: miR-200 regulates PDGF-D-mediated epithelial-mesenchymal transition, adhesion, and invasion of prostate cancer cells. Stem Cells. 2009, 27: 1712-1721. 10.1002/stem.101PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Park SM, Gaur AB, Lengyel E, Peter ME: The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008, 22: 894-907. 10.1101/gad.1640608PubMedCentralCrossRefPubMed Park SM, Gaur AB, Lengyel E, Peter ME: The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008, 22: 894-907. 10.1101/gad.1640608PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Tryndyak VP, Beland FA, Pogribny IP: E-cadherin transcriptional down-regulation by epigenetic and microRNA-200 family alterations is related to mesenchymal and drug-resistant phenotypes in human breast cancer cells. Int J Cancer. 2010, 126: 2575-83.PubMed Tryndyak VP, Beland FA, Pogribny IP: E-cadherin transcriptional down-regulation by epigenetic and microRNA-200 family alterations is related to mesenchymal and drug-resistant phenotypes in human breast cancer cells. Int J Cancer. 2010, 126: 2575-83.PubMed
42.
Zurück zum Zitat Saydam O, Shen Y, Wurdinger T, Senol O, Boke E, James MF, Tannous BA, Stemmer-Rachamimov AO, Yi M, Stephens RM, Fraefel C, Gusella JF, Krichevsky AM, Breakefield XO: Downregulated microRNA-200a in meningiomas promotes tumor growth by reducing E-cadherin and activating the Wnt/beta-catenin signaling pathway. Mol Cell Biol. 2009, 29: 5923-5940. 10.1128/MCB.00332-09PubMedCentralCrossRefPubMed Saydam O, Shen Y, Wurdinger T, Senol O, Boke E, James MF, Tannous BA, Stemmer-Rachamimov AO, Yi M, Stephens RM, Fraefel C, Gusella JF, Krichevsky AM, Breakefield XO: Downregulated microRNA-200a in meningiomas promotes tumor growth by reducing E-cadherin and activating the Wnt/beta-catenin signaling pathway. Mol Cell Biol. 2009, 29: 5923-5940. 10.1128/MCB.00332-09PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Hashimi ST, Fulcher JA, Chang MH, Gov L, Wang S, Lee B: MicroRNA profiling identifies miR-34a and miR-21 and their target genes JAG1 and WNT1 in the coordinate regulation of dendritic cell differentiation. Blood. 2009, 114: 404-414. 10.1182/blood-2008-09-179150PubMedCentralCrossRefPubMed Hashimi ST, Fulcher JA, Chang MH, Gov L, Wang S, Lee B: MicroRNA profiling identifies miR-34a and miR-21 and their target genes JAG1 and WNT1 in the coordinate regulation of dendritic cell differentiation. Blood. 2009, 114: 404-414. 10.1182/blood-2008-09-179150PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Chen Y, Liu W, Chao T, Zhang Y, Yan X, Gong Y, Qiang B, Yuan J, Sun M, Peng X: MicroRNA-21 down-regulates the expression of tumor suppressor PDCD4 in human glioblastoma cell T98G. Cancer Lett. 2008, 272: 197-205. 10.1016/j.canlet.2008.06.034CrossRefPubMed Chen Y, Liu W, Chao T, Zhang Y, Yan X, Gong Y, Qiang B, Yuan J, Sun M, Peng X: MicroRNA-21 down-regulates the expression of tumor suppressor PDCD4 in human glioblastoma cell T98G. Cancer Lett. 2008, 272: 197-205. 10.1016/j.canlet.2008.06.034CrossRefPubMed
45.
Zurück zum Zitat Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T: MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007, 133: 647-658. 10.1053/j.gastro.2007.05.022PubMedCentralCrossRefPubMed Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T: MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 2007, 133: 647-658. 10.1053/j.gastro.2007.05.022PubMedCentralCrossRefPubMed
46.
Zurück zum Zitat Yao Q, Xu H, Zhang QQ, Zhou H, Qu LH: MicroRNA-21 promotes cell proliferation and down-regulates the expression of programmed cell death 4 (PDCD4) in HeLa cervical carcinoma cells. Biochem Biophys Res Commun. 2009, 388: 539-542. 10.1016/j.bbrc.2009.08.044CrossRefPubMed Yao Q, Xu H, Zhang QQ, Zhou H, Qu LH: MicroRNA-21 promotes cell proliferation and down-regulates the expression of programmed cell death 4 (PDCD4) in HeLa cervical carcinoma cells. Biochem Biophys Res Commun. 2009, 388: 539-542. 10.1016/j.bbrc.2009.08.044CrossRefPubMed
47.
Zurück zum Zitat Zhang Z, Li Z, Gao C, Chen P, Chen J, Liu W, Xiao S, Lu H: miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab Invest. 2008, 88: 1358-1366. 10.1038/labinvest.2008.94CrossRefPubMed Zhang Z, Li Z, Gao C, Chen P, Chen J, Liu W, Xiao S, Lu H: miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab Invest. 2008, 88: 1358-1366. 10.1038/labinvest.2008.94CrossRefPubMed
48.
Zurück zum Zitat Thatcher EJ, Paydar I, Anderson KK, Patton JG: Regulation of zebrafish fin regeneration by microRNAs. Proc Natl Acad Sci USA. 2008, 105: 18384-18389. 10.1073/pnas.0803713105PubMedCentralCrossRefPubMed Thatcher EJ, Paydar I, Anderson KK, Patton JG: Regulation of zebrafish fin regeneration by microRNAs. Proc Natl Acad Sci USA. 2008, 105: 18384-18389. 10.1073/pnas.0803713105PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Kennell JA, Gerin I, MacDougald OA, Cadigan KM: The microRNA miR-8 is a conserved negative regulator of Wnt signaling. Proc Natl Acad Sci USA. 2008, 105: 15417-15422. 10.1073/pnas.0807763105PubMedCentralCrossRefPubMed Kennell JA, Gerin I, MacDougald OA, Cadigan KM: The microRNA miR-8 is a conserved negative regulator of Wnt signaling. Proc Natl Acad Sci USA. 2008, 105: 15417-15422. 10.1073/pnas.0807763105PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Nagel R, le Sage C, Diosdado B, van der Waal M, Oude Vrielink JA, Bolijn A, Meijer GA, Agami R: Regulation of the adenomatous polyposis coli gene by the miR-135 family in colorectal cancer. Cancer Res. 2008, 68: 5795-5802. 10.1158/0008-5472.CAN-08-0951CrossRefPubMed Nagel R, le Sage C, Diosdado B, van der Waal M, Oude Vrielink JA, Bolijn A, Meijer GA, Agami R: Regulation of the adenomatous polyposis coli gene by the miR-135 family in colorectal cancer. Cancer Res. 2008, 68: 5795-5802. 10.1158/0008-5472.CAN-08-0951CrossRefPubMed
51.
Zurück zum Zitat Silver SJ, Hagen JW, Okamura K, Perrimon N, Lai EC: Functional screening identifies miR-315 as a potent activator of Wingless signaling. Proc Natl Acad Sci USA. 2007, 104: 18151-18156. 10.1073/pnas.0706673104PubMedCentralCrossRefPubMed Silver SJ, Hagen JW, Okamura K, Perrimon N, Lai EC: Functional screening identifies miR-315 as a potent activator of Wingless signaling. Proc Natl Acad Sci USA. 2007, 104: 18151-18156. 10.1073/pnas.0706673104PubMedCentralCrossRefPubMed
52.
Zurück zum Zitat Arce L, Pate KT, Waterman ML: Groucho binds two conserved regions of LEF-1 for HDAC-dependent repression. BMC Cancer. 2009, 9: 159- 10.1186/1471-2407-9-159PubMedCentralCrossRefPubMed Arce L, Pate KT, Waterman ML: Groucho binds two conserved regions of LEF-1 for HDAC-dependent repression. BMC Cancer. 2009, 9: 159- 10.1186/1471-2407-9-159PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Brannon M, Brown JD, Bates R, Kimelman D, Moon RT: XCtBP is a XTcf-3 co-repressor with roles throughout Xenopus development. Development. 1999, 126: 3159-3170.PubMed Brannon M, Brown JD, Bates R, Kimelman D, Moon RT: XCtBP is a XTcf-3 co-repressor with roles throughout Xenopus development. Development. 1999, 126: 3159-3170.PubMed
54.
Zurück zum Zitat Cavallo RA, Cox RT, Moline MM, Roose J, Polevoy GA, Clevers H, Peifer M, Bejsovec A: Drosophila Tcf and Groucho interact to repress Wingless signalling activity. Nature. 1998, 395: 604-608. 10.1038/26982CrossRefPubMed Cavallo RA, Cox RT, Moline MM, Roose J, Polevoy GA, Clevers H, Peifer M, Bejsovec A: Drosophila Tcf and Groucho interact to repress Wingless signalling activity. Nature. 1998, 395: 604-608. 10.1038/26982CrossRefPubMed
55.
Zurück zum Zitat Yu X, Li P, Roeder RG, Wang Z: Inhibition of androgen receptor-mediated transcription by amino-terminal enhancer of split. Mol Cell Biol. 2001, 21: 4614-4625. 10.1128/MCB.21.14.4614-4625.2001PubMedCentralCrossRefPubMed Yu X, Li P, Roeder RG, Wang Z: Inhibition of androgen receptor-mediated transcription by amino-terminal enhancer of split. Mol Cell Biol. 2001, 21: 4614-4625. 10.1128/MCB.21.14.4614-4625.2001PubMedCentralCrossRefPubMed
56.
Zurück zum Zitat Chen G, Fernandez J, Mische S, Courey AJ: A functional interaction between the histone deacetylase Rpd3 and the corepressor groucho in Drosophila development. Genes Dev. 1999, 13: 2218-2230. 10.1101/gad.13.17.2218PubMedCentralCrossRefPubMed Chen G, Fernandez J, Mische S, Courey AJ: A functional interaction between the histone deacetylase Rpd3 and the corepressor groucho in Drosophila development. Genes Dev. 1999, 13: 2218-2230. 10.1101/gad.13.17.2218PubMedCentralCrossRefPubMed
57.
Zurück zum Zitat Daniels DL, Weis WI: Beta-catenin directly displaces Groucho/TLE repressors from Tcf/Lef in Wnt-mediated transcription activation. Nat Struct Mol Biol. 2005, 12: 364-371. 10.1038/nsmb912CrossRefPubMed Daniels DL, Weis WI: Beta-catenin directly displaces Groucho/TLE repressors from Tcf/Lef in Wnt-mediated transcription activation. Nat Struct Mol Biol. 2005, 12: 364-371. 10.1038/nsmb912CrossRefPubMed
58.
Zurück zum Zitat Belenkaya TY, Han C, Standley HJ, Lin X, Houston DW, Heasman J, Lin X: pygopus Encodes a nuclear protein essential for wingless/Wnt signaling. Development. 2002, 129: 4089-4101.PubMed Belenkaya TY, Han C, Standley HJ, Lin X, Houston DW, Heasman J, Lin X: pygopus Encodes a nuclear protein essential for wingless/Wnt signaling. Development. 2002, 129: 4089-4101.PubMed
59.
Zurück zum Zitat Kramps T, Peter O, Brunner E, Nellen D, Froesch B, Chatterjee S, Murone M, Zullig S, Basler K: Wnt/wingless signaling requires BCL9/legless-mediated recruitment of pygopus to the nuclear beta-catenin-TCF complex. Cell. 2002, 109: 47-60. 10.1016/S0092-8674(02)00679-7CrossRefPubMed Kramps T, Peter O, Brunner E, Nellen D, Froesch B, Chatterjee S, Murone M, Zullig S, Basler K: Wnt/wingless signaling requires BCL9/legless-mediated recruitment of pygopus to the nuclear beta-catenin-TCF complex. Cell. 2002, 109: 47-60. 10.1016/S0092-8674(02)00679-7CrossRefPubMed
60.
Zurück zum Zitat Takemaru KI, Moon RT: The transcriptional coactivator CBP interacts with beta-catenin to activate gene expression. J Cell Biol. 2000, 149: 249-254. 10.1083/jcb.149.2.249PubMedCentralCrossRefPubMed Takemaru KI, Moon RT: The transcriptional coactivator CBP interacts with beta-catenin to activate gene expression. J Cell Biol. 2000, 149: 249-254. 10.1083/jcb.149.2.249PubMedCentralCrossRefPubMed
61.
Zurück zum Zitat Billin AN, Thirlwell H, Ayer DE: Beta-catenin-histone deacetylase interactions regulate the transition of LEF1 from a transcriptional repressor to an activator. Mol Cell Biol. 2000, 20: 6882-6890. 10.1128/MCB.20.18.6882-6890.2000PubMedCentralCrossRefPubMed Billin AN, Thirlwell H, Ayer DE: Beta-catenin-histone deacetylase interactions regulate the transition of LEF1 from a transcriptional repressor to an activator. Mol Cell Biol. 2000, 20: 6882-6890. 10.1128/MCB.20.18.6882-6890.2000PubMedCentralCrossRefPubMed
62.
Zurück zum Zitat Ye F, Chen Y, Hoang T, Montgomery RL, Zhao XH, Bu H, Hu T, Taketo MM, van Es JH, Clevers H, Hsieh J, Bassel-Duby R, Olson EN, Lu QR: HDAC1 and HDAC2 regulate oligodendrocyte differentiation by disrupting the beta-catenin-TCF interaction. Nat Neurosci. 2009, 12: 829-838. 10.1038/nn.2333PubMedCentralCrossRefPubMed Ye F, Chen Y, Hoang T, Montgomery RL, Zhao XH, Bu H, Hu T, Taketo MM, van Es JH, Clevers H, Hsieh J, Bassel-Duby R, Olson EN, Lu QR: HDAC1 and HDAC2 regulate oligodendrocyte differentiation by disrupting the beta-catenin-TCF interaction. Nat Neurosci. 2009, 12: 829-838. 10.1038/nn.2333PubMedCentralCrossRefPubMed
63.
Zurück zum Zitat Yamaguchi M, Tonou-Fujimori N, Komori A, Maeda R, Nojima Y, Li H, Okamoto H, Masai I: Histone deacetylase 1 regulates retinal neurogenesis in zebrafish by suppressing Wnt and Notch signaling pathways. Development. 2005, 132: 3027-3043. 10.1242/dev.01881CrossRefPubMed Yamaguchi M, Tonou-Fujimori N, Komori A, Maeda R, Nojima Y, Li H, Okamoto H, Masai I: Histone deacetylase 1 regulates retinal neurogenesis in zebrafish by suppressing Wnt and Notch signaling pathways. Development. 2005, 132: 3027-3043. 10.1242/dev.01881CrossRefPubMed
64.
Zurück zum Zitat Godman CA, Joshi R, Tierney BR, Greenspan E, Rasmussen TP, Wang HW, Shin DG, Rosenberg DW, Giardina C: HDAC3 impacts multiple oncogenic pathways in colon cancer cells with effects on Wnt and vitamin D signaling. Cancer Biol Ther. 2008, 7: 1570-1580. 10.4161/cbt.7.10.6561PubMedCentralCrossRefPubMed Godman CA, Joshi R, Tierney BR, Greenspan E, Rasmussen TP, Wang HW, Shin DG, Rosenberg DW, Giardina C: HDAC3 impacts multiple oncogenic pathways in colon cancer cells with effects on Wnt and vitamin D signaling. Cancer Biol Ther. 2008, 7: 1570-1580. 10.4161/cbt.7.10.6561PubMedCentralCrossRefPubMed
65.
Zurück zum Zitat Li Y, Zhang X, Polakiewicz RD, Yao TP, Comb MJ: HDAC6 is required for epidermal growth factor-induced beta-catenin nuclear localization. J Biol Chem. 2008, 283: 12686-12690. 10.1074/jbc.C700185200PubMedCentralCrossRefPubMed Li Y, Zhang X, Polakiewicz RD, Yao TP, Comb MJ: HDAC6 is required for epidermal growth factor-induced beta-catenin nuclear localization. J Biol Chem. 2008, 283: 12686-12690. 10.1074/jbc.C700185200PubMedCentralCrossRefPubMed
66.
Zurück zum Zitat Mann B, Gelos M, Siedow A, Hanski ML, Gratchev A, Ilyas M, Bodmer WF, Moyer MP, Riecken EO, Buhr HJ, Hanski C: Target genes of beta-catenin-T cell-factor/lymphoid-enhancer-factor signaling in human colorectal carcinomas. Proc Natl Acad Sci USA. 1999, 96: 1603-1608. 10.1073/pnas.96.4.1603PubMedCentralCrossRefPubMed Mann B, Gelos M, Siedow A, Hanski ML, Gratchev A, Ilyas M, Bodmer WF, Moyer MP, Riecken EO, Buhr HJ, Hanski C: Target genes of beta-catenin-T cell-factor/lymphoid-enhancer-factor signaling in human colorectal carcinomas. Proc Natl Acad Sci USA. 1999, 96: 1603-1608. 10.1073/pnas.96.4.1603PubMedCentralCrossRefPubMed
67.
Zurück zum Zitat Toualbi K, Guller MC, Mauriz JL, Labalette C, Buendia MA, Mauviel A, Bernuau D: Physical and functional cooperation between AP-1 and beta-catenin for the regulation of TCF-dependent genes. Oncogene. 2007, 26: 3492-3502. 10.1038/sj.onc.1210133CrossRefPubMed Toualbi K, Guller MC, Mauriz JL, Labalette C, Buendia MA, Mauviel A, Bernuau D: Physical and functional cooperation between AP-1 and beta-catenin for the regulation of TCF-dependent genes. Oncogene. 2007, 26: 3492-3502. 10.1038/sj.onc.1210133CrossRefPubMed
68.
Zurück zum Zitat Gan XQ, Wang JY, Xi Y, Wu ZL, Li YP, Li L: Nuclear Dvl, c-Jun, beta-catenin, and TCF form a complex leading to stabilization of beta-catenin-TCF interaction. J Cell Biol. 2008, 180: 1087-1100. 10.1083/jcb.200710050PubMedCentralCrossRefPubMed Gan XQ, Wang JY, Xi Y, Wu ZL, Li YP, Li L: Nuclear Dvl, c-Jun, beta-catenin, and TCF form a complex leading to stabilization of beta-catenin-TCF interaction. J Cell Biol. 2008, 180: 1087-1100. 10.1083/jcb.200710050PubMedCentralCrossRefPubMed
69.
Zurück zum Zitat Hwang SG, Yu SS, Lee SW, Chun JS: Wnt-3a regulates chondrocyte differentiation via c-Jun/AP-1 pathway. FEBS Lett. 2005, 579: 4837-4842. 10.1016/j.febslet.2005.07.067CrossRefPubMed Hwang SG, Yu SS, Lee SW, Chun JS: Wnt-3a regulates chondrocyte differentiation via c-Jun/AP-1 pathway. FEBS Lett. 2005, 579: 4837-4842. 10.1016/j.febslet.2005.07.067CrossRefPubMed
70.
Zurück zum Zitat Mahmoudi T, Li VS, Ng SS, Taouatas N, Vries RG, Mohammed S, Heck AJ, Clevers H: The kinase TNIK is an essential activator of Wnt target genes. Embo J. 2009, 28: 3329-3340. 10.1038/emboj.2009.285PubMedCentralCrossRefPubMed Mahmoudi T, Li VS, Ng SS, Taouatas N, Vries RG, Mohammed S, Heck AJ, Clevers H: The kinase TNIK is an essential activator of Wnt target genes. Embo J. 2009, 28: 3329-3340. 10.1038/emboj.2009.285PubMedCentralCrossRefPubMed
71.
Zurück zum Zitat Stemmer V, de Craene B, Berx G, Behrens J: Snail promotes Wnt target gene expression and interacts with beta-catenin. Oncogene. 2008, 27: 5075-5080. 10.1038/onc.2008.140CrossRefPubMed Stemmer V, de Craene B, Berx G, Behrens J: Snail promotes Wnt target gene expression and interacts with beta-catenin. Oncogene. 2008, 27: 5075-5080. 10.1038/onc.2008.140CrossRefPubMed
72.
Zurück zum Zitat Evans PM, Chen X, Zhang W, Liu C: KLF4 interacts with beta-catenin/TCF4 and blocks p300/CBP recruitment by beta-catenin. Mol Cell Biol. 2010, 30: 372-381. 10.1128/MCB.00063-09PubMedCentralCrossRefPubMed Evans PM, Chen X, Zhang W, Liu C: KLF4 interacts with beta-catenin/TCF4 and blocks p300/CBP recruitment by beta-catenin. Mol Cell Biol. 2010, 30: 372-381. 10.1128/MCB.00063-09PubMedCentralCrossRefPubMed
73.
Zurück zum Zitat Hong CF, Chou YT, Lin YS, Wu CW: MAD2B, a novel TCF4-binding protein, modulates TCF4-mediated epithelial-mesenchymal transdifferentiation. J Biol Chem. 2009, 284: 19613-19622. 10.1074/jbc.M109.005017PubMedCentralCrossRefPubMed Hong CF, Chou YT, Lin YS, Wu CW: MAD2B, a novel TCF4-binding protein, modulates TCF4-mediated epithelial-mesenchymal transdifferentiation. J Biol Chem. 2009, 284: 19613-19622. 10.1074/jbc.M109.005017PubMedCentralCrossRefPubMed
74.
Zurück zum Zitat Gao H, Le Y, Wu X, Silberstein LE, Giese RW, Zhu Z: VentX, a novel lymphoid-enhancing factor/T-cell factor-associated transcription repressor, is a putative tumor suppressor. Cancer Res. 2010, 70: 202-211. 10.1158/0008-5472.CAN-09-2668CrossRefPubMed Gao H, Le Y, Wu X, Silberstein LE, Giese RW, Zhu Z: VentX, a novel lymphoid-enhancing factor/T-cell factor-associated transcription repressor, is a putative tumor suppressor. Cancer Res. 2010, 70: 202-211. 10.1158/0008-5472.CAN-09-2668CrossRefPubMed
75.
Zurück zum Zitat Young CS, Kitamura M, Hardy S, Kitajewski J: Wnt-1 induces growth, cytosolic beta-catenin, and Tcf/Lef transcriptional activation in Rat-1 fibroblasts. Mol Cell Biol. 1998, 18: 2474-2485.PubMedCentralCrossRefPubMed Young CS, Kitamura M, Hardy S, Kitajewski J: Wnt-1 induces growth, cytosolic beta-catenin, and Tcf/Lef transcriptional activation in Rat-1 fibroblasts. Mol Cell Biol. 1998, 18: 2474-2485.PubMedCentralCrossRefPubMed
76.
Zurück zum Zitat Hatzis P, van der Flier LG, van Driel MA, Guryev V, Nielsen F, Denissov S, Nijman IJ, Koster J, Santo EE, Welboren W, Versteeg R, Cuppen E, van de Wetering M, Clevers H, Stunnenberg HG: Genome-wide pattern of TCF7L2/TCF4 chromatin occupancy in colorectal cancer cells. Mol Cell Biol. 2008, 28: 2732-2744. 10.1128/MCB.02175-07PubMedCentralCrossRefPubMed Hatzis P, van der Flier LG, van Driel MA, Guryev V, Nielsen F, Denissov S, Nijman IJ, Koster J, Santo EE, Welboren W, Versteeg R, Cuppen E, van de Wetering M, Clevers H, Stunnenberg HG: Genome-wide pattern of TCF7L2/TCF4 chromatin occupancy in colorectal cancer cells. Mol Cell Biol. 2008, 28: 2732-2744. 10.1128/MCB.02175-07PubMedCentralCrossRefPubMed
77.
Zurück zum Zitat Dihlmann S, Kloor M, Fallsehr C, von Knebel Doeberitz M: Regulation of AKT1 expression by beta-catenin/Tcf/Lef signaling in colorectal cancer cells. Carcinogenesis. 2005, 26: 1503-1512. 10.1093/carcin/bgi120CrossRefPubMed Dihlmann S, Kloor M, Fallsehr C, von Knebel Doeberitz M: Regulation of AKT1 expression by beta-catenin/Tcf/Lef signaling in colorectal cancer cells. Carcinogenesis. 2005, 26: 1503-1512. 10.1093/carcin/bgi120CrossRefPubMed
78.
Zurück zum Zitat Han A, Song Z, Tong C, Hu D, Bi X, Augenlicht LH, Yang W: Sulindac suppresses beta-catenin expression in human cancer cells. Eur J Pharmacol. 2008, 583: 26-31. 10.1016/j.ejphar.2007.12.034PubMedCentralCrossRefPubMed Han A, Song Z, Tong C, Hu D, Bi X, Augenlicht LH, Yang W: Sulindac suppresses beta-catenin expression in human cancer cells. Eur J Pharmacol. 2008, 583: 26-31. 10.1016/j.ejphar.2007.12.034PubMedCentralCrossRefPubMed
79.
Zurück zum Zitat Behari J, Zeng G, Otruba W, Thompson MD, Muller P, Micsenyi A, Sekhon SS, Leoni L, Monga SP: R-Etodolac decreases beta-catenin levels along with survival and proliferation of hepatoma cells. J Hepatol. 2007, 46: 849-857. 10.1016/j.jhep.2006.11.017PubMedCentralCrossRefPubMed Behari J, Zeng G, Otruba W, Thompson MD, Muller P, Micsenyi A, Sekhon SS, Leoni L, Monga SP: R-Etodolac decreases beta-catenin levels along with survival and proliferation of hepatoma cells. J Hepatol. 2007, 46: 849-857. 10.1016/j.jhep.2006.11.017PubMedCentralCrossRefPubMed
80.
Zurück zum Zitat Rigas B, Kashfi K: Nitric-oxide-donating NSAIDs as agents for cancer prevention. Trends Mol Med. 2004, 10: 324-330. 10.1016/j.molmed.2004.05.004CrossRefPubMed Rigas B, Kashfi K: Nitric-oxide-donating NSAIDs as agents for cancer prevention. Trends Mol Med. 2004, 10: 324-330. 10.1016/j.molmed.2004.05.004CrossRefPubMed
81.
Zurück zum Zitat Wang Z, Havasi A, Gall JM, Mao H, Schwartz JH, Borkan SC: Beta-catenin promotes survival of renal epithelial cells by inhibiting Bax. J Am Soc Nephrol. 2009, 20: 1919-1928. 10.1681/ASN.2009030253PubMedCentralCrossRefPubMed Wang Z, Havasi A, Gall JM, Mao H, Schwartz JH, Borkan SC: Beta-catenin promotes survival of renal epithelial cells by inhibiting Bax. J Am Soc Nephrol. 2009, 20: 1919-1928. 10.1681/ASN.2009030253PubMedCentralCrossRefPubMed
82.
Zurück zum Zitat Li B, Kuriyama S, Moreno M, Mayor R: The posteriorizing gene Gbx2 is a direct target of Wnt signalling and the earliest factor in neural crest induction. Development. 2009, 136: 3267-3278. 10.1242/dev.036954PubMedCentralCrossRefPubMed Li B, Kuriyama S, Moreno M, Mayor R: The posteriorizing gene Gbx2 is a direct target of Wnt signalling and the earliest factor in neural crest induction. Development. 2009, 136: 3267-3278. 10.1242/dev.036954PubMedCentralCrossRefPubMed
83.
Zurück zum Zitat Delmas V, Beermann F, Martinozzi S, Carreira S, Ackermann J, Kumasaka M, Denat L, Goodall J, Luciani F, Viros A, Demirkan N, Bastian BC, Goding CR, Larue L: Beta-catenin induces immortalization of melanocytes by suppressing p16INK4a expression and cooperates with N-Ras in melanoma development. Genes Dev. 2007, 21: 2923-2935. 10.1101/gad.450107PubMedCentralCrossRefPubMed Delmas V, Beermann F, Martinozzi S, Carreira S, Ackermann J, Kumasaka M, Denat L, Goodall J, Luciani F, Viros A, Demirkan N, Bastian BC, Goding CR, Larue L: Beta-catenin induces immortalization of melanocytes by suppressing p16INK4a expression and cooperates with N-Ras in melanoma development. Genes Dev. 2007, 21: 2923-2935. 10.1101/gad.450107PubMedCentralCrossRefPubMed
84.
Zurück zum Zitat Savage J, Voronova A, Mehta V, Sendi-Mukasa F, Skerjanc IS: Canonical Wnt signaling regulates Foxc1/2 expression in P19 cells. Differentiation. 2010, 79: 31-40. 10.1016/j.diff.2009.08.008CrossRefPubMed Savage J, Voronova A, Mehta V, Sendi-Mukasa F, Skerjanc IS: Canonical Wnt signaling regulates Foxc1/2 expression in P19 cells. Differentiation. 2010, 79: 31-40. 10.1016/j.diff.2009.08.008CrossRefPubMed
85.
Zurück zum Zitat Rena V, Angeletti S, Panzetta-Dutari G, Genti-Raimondi S: Activation of beta-catenin signalling increases StarD7 gene expression in JEG-3 cells. Placenta. 2009, 30: 876-883. 10.1016/j.placenta.2009.07.010CrossRefPubMed Rena V, Angeletti S, Panzetta-Dutari G, Genti-Raimondi S: Activation of beta-catenin signalling increases StarD7 gene expression in JEG-3 cells. Placenta. 2009, 30: 876-883. 10.1016/j.placenta.2009.07.010CrossRefPubMed
86.
Zurück zum Zitat Hao J, Li TG, Qi X, Zhao DF, Zhao GQ: WNT/beta-catenin pathway up-regulates Stat3 and converges on LIF to prevent differentiation of mouse embryonic stem cells. Dev Biol. 2006, 290: 81-91. 10.1016/j.ydbio.2005.11.011CrossRefPubMed Hao J, Li TG, Qi X, Zhao DF, Zhao GQ: WNT/beta-catenin pathway up-regulates Stat3 and converges on LIF to prevent differentiation of mouse embryonic stem cells. Dev Biol. 2006, 290: 81-91. 10.1016/j.ydbio.2005.11.011CrossRefPubMed
87.
Zurück zum Zitat Yan S, Zhou C, Zhang W, Zhang G, Zhao X, Yang S, Wang Y, Lu N, Zhu H, Xu N: beta-Catenin/TCF pathway upregulates STAT3 expression in human esophageal squamous cell carcinoma. Cancer Lett. 2008, 271: 85-97. 10.1016/j.canlet.2008.05.035CrossRefPubMed Yan S, Zhou C, Zhang W, Zhang G, Zhao X, Yang S, Wang Y, Lu N, Zhu H, Xu N: beta-Catenin/TCF pathway upregulates STAT3 expression in human esophageal squamous cell carcinoma. Cancer Lett. 2008, 271: 85-97. 10.1016/j.canlet.2008.05.035CrossRefPubMed
88.
Zurück zum Zitat Kawada M, Seno H, Uenoyama Y, Sawabu T, Kanda N, Fukui H, Shimahara Y, Chiba T: Signal transducers and activators of transcription 3 activation is involved in nuclear accumulation of beta-catenin in colorectal cancer. Cancer Res. 2006, 66: 2913-2917. 10.1158/0008-5472.CAN-05-3460CrossRefPubMed Kawada M, Seno H, Uenoyama Y, Sawabu T, Kanda N, Fukui H, Shimahara Y, Chiba T: Signal transducers and activators of transcription 3 activation is involved in nuclear accumulation of beta-catenin in colorectal cancer. Cancer Res. 2006, 66: 2913-2917. 10.1158/0008-5472.CAN-05-3460CrossRefPubMed
89.
Zurück zum Zitat Abramova MV, Zatulovskiy EA, Svetlikova SB, Kukushkin AN, Pospelov VA: e2f1 Gene is a new member of Wnt/beta-catenin/Tcf-regulated genes. Biochem Biophys Res Commun. 2010, 391: 142-146. 10.1016/j.bbrc.2009.11.020CrossRefPubMed Abramova MV, Zatulovskiy EA, Svetlikova SB, Kukushkin AN, Pospelov VA: e2f1 Gene is a new member of Wnt/beta-catenin/Tcf-regulated genes. Biochem Biophys Res Commun. 2010, 391: 142-146. 10.1016/j.bbrc.2009.11.020CrossRefPubMed
90.
Zurück zum Zitat Morris EJ, Ji JY, Yang F, Di Stefano L, Herr A, Moon NS, Kwon EJ, Haigis KM, Naar AM, Dyson NJ: E2F1 represses beta-catenin transcription and is antagonized by both pRB and CDK8. Nature. 2008, 455: 552-556. 10.1038/nature07310PubMedCentralCrossRefPubMed Morris EJ, Ji JY, Yang F, Di Stefano L, Herr A, Moon NS, Kwon EJ, Haigis KM, Naar AM, Dyson NJ: E2F1 represses beta-catenin transcription and is antagonized by both pRB and CDK8. Nature. 2008, 455: 552-556. 10.1038/nature07310PubMedCentralCrossRefPubMed
91.
Zurück zum Zitat Xie W, Jin L, Mei Y, Wu M: E2F1 represses beta-catenin/TCF activity by direct upregulation of Siah1. J Cell Mol Med. 2009, 13: 1719-1727. 10.1111/j.1582-4934.2008.00423.xCrossRefPubMed Xie W, Jin L, Mei Y, Wu M: E2F1 represses beta-catenin/TCF activity by direct upregulation of Siah1. J Cell Mol Med. 2009, 13: 1719-1727. 10.1111/j.1582-4934.2008.00423.xCrossRefPubMed
92.
Zurück zum Zitat Zhou F, Zhang L, Gong K, Lu G, Sheng B, Wang A, Zhao N, Zhang X, Gong Y: LEF-1 activates the transcription of E2F1. Biochem Biophys Res Commun. 2008, 365: 149-153. 10.1016/j.bbrc.2007.10.138CrossRefPubMed Zhou F, Zhang L, Gong K, Lu G, Sheng B, Wang A, Zhao N, Zhang X, Gong Y: LEF-1 activates the transcription of E2F1. Biochem Biophys Res Commun. 2008, 365: 149-153. 10.1016/j.bbrc.2007.10.138CrossRefPubMed
93.
Zurück zum Zitat Martello G, Zacchigna L, Inui M, Montagner M, Adorno M, Mamidi A, Morsut L, Soligo S, Tran U, Dupont S, Cordenonsi M, Wessely O, Piccolo S: MicroRNA control of Nodal signalling. Nature. 2007, 449: 183-188. 10.1038/nature06100CrossRefPubMed Martello G, Zacchigna L, Inui M, Montagner M, Adorno M, Mamidi A, Morsut L, Soligo S, Tran U, Dupont S, Cordenonsi M, Wessely O, Piccolo S: MicroRNA control of Nodal signalling. Nature. 2007, 449: 183-188. 10.1038/nature06100CrossRefPubMed
94.
Zurück zum Zitat Wang X, Lam EK, Zhang J, Jin H, Sung JJ: MicroRNA-122a functions as a novel tumor suppressor downstream of adenomatous polyposis coli in gastrointestinal cancers. Biochem Biophys Res Commun. 2009, 387: 376-380. 10.1016/j.bbrc.2009.07.034CrossRefPubMed Wang X, Lam EK, Zhang J, Jin H, Sung JJ: MicroRNA-122a functions as a novel tumor suppressor downstream of adenomatous polyposis coli in gastrointestinal cancers. Biochem Biophys Res Commun. 2009, 387: 376-380. 10.1016/j.bbrc.2009.07.034CrossRefPubMed
95.
Zurück zum Zitat Ladeiro Y, Couchy G, Balabaud C, Bioulac-Sage P, Pelletier L, Rebouissou S, Zucman-Rossi J: MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology. 2008, 47: 1955-1963. 10.1002/hep.22256CrossRefPubMed Ladeiro Y, Couchy G, Balabaud C, Bioulac-Sage P, Pelletier L, Rebouissou S, Zucman-Rossi J: MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology. 2008, 47: 1955-1963. 10.1002/hep.22256CrossRefPubMed
96.
Zurück zum Zitat Park CH, Chang JY, Hahm ER, Park S, Kim HK, Yang CH: Quercetin, a potent inhibitor against beta-catenin/Tcf signaling in SW480 colon cancer cells. Biochem Biophys Res Commun. 2005, 328: 227-234. 10.1016/j.bbrc.2004.12.151CrossRefPubMed Park CH, Chang JY, Hahm ER, Park S, Kim HK, Yang CH: Quercetin, a potent inhibitor against beta-catenin/Tcf signaling in SW480 colon cancer cells. Biochem Biophys Res Commun. 2005, 328: 227-234. 10.1016/j.bbrc.2004.12.151CrossRefPubMed
97.
Zurück zum Zitat Shan BE, Wang MX, Li RQ: Quercetin inhibit human SW480 colon cancer growth in association with inhibition of cyclin D1 and survivin expression through Wnt/beta-catenin signaling pathway. Cancer Invest. 2009, 27: 604-612. 10.1080/07357900802337191CrossRefPubMed Shan BE, Wang MX, Li RQ: Quercetin inhibit human SW480 colon cancer growth in association with inhibition of cyclin D1 and survivin expression through Wnt/beta-catenin signaling pathway. Cancer Invest. 2009, 27: 604-612. 10.1080/07357900802337191CrossRefPubMed
98.
Zurück zum Zitat Wei W, Chua MS, Grepper S, So S: Small molecule antagonists of Tcf4/beta-catenin complex inhibit the growth of HCC cells in vitro and in vivo. Int J Cancer. 2010, 126: 2426-2436.PubMed Wei W, Chua MS, Grepper S, So S: Small molecule antagonists of Tcf4/beta-catenin complex inhibit the growth of HCC cells in vitro and in vivo. Int J Cancer. 2010, 126: 2426-2436.PubMed
99.
Zurück zum Zitat Takemaru KI, Ohmitsu M, Li FQ: An oncogenic hub: beta-catenin as a molecular target for cancer therapeutics. Handb Exp Pharmacol. 2008, 261-284. Takemaru KI, Ohmitsu M, Li FQ: An oncogenic hub: beta-catenin as a molecular target for cancer therapeutics. Handb Exp Pharmacol. 2008, 261-284.
100.
Zurück zum Zitat Dehnhardt CM, Venkatesan AM, Chen Z, Ayral-Kaloustian S, Dos Santos O, Delos Santos E, Curran K, Follettie MT, Diesl V, Lucas J, Geng Y, Dejoy SQ, Petersen R, Chaudhary I, Brooijmans N, Mansour TS, Arndt K, Chen L: Design and synthesis of novel diaminoquinazolines with in vivo efficacy for beta-catenin/T-cell transcriptional factor 4 pathway inhibition. J Med Chem. 2010, 53: 897-910.CrossRefPubMed Dehnhardt CM, Venkatesan AM, Chen Z, Ayral-Kaloustian S, Dos Santos O, Delos Santos E, Curran K, Follettie MT, Diesl V, Lucas J, Geng Y, Dejoy SQ, Petersen R, Chaudhary I, Brooijmans N, Mansour TS, Arndt K, Chen L: Design and synthesis of novel diaminoquinazolines with in vivo efficacy for beta-catenin/T-cell transcriptional factor 4 pathway inhibition. J Med Chem. 2010, 53: 897-910.CrossRefPubMed
Metadaten
Titel
MicroRNA roles in beta-catenin pathway
verfasst von
Kai Huang
Jun-Xia Zhang
Lei Han
Yong-Ping You
Tao Jiang
Pei-Yu Pu
Chun-Sheng Kang
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2010
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-9-252

Weitere Artikel der Ausgabe 1/2010

Molecular Cancer 1/2010 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.