Interactions between transcription and signaling are fundamentally important for understanding both the structure and function of genetic pathways and their role in diseases such as cancer. The finding that β-catenin/TCF4 and JNK/c-Jun cooperate has important implications in carcinogenesis. Previously, we found that binding of c-Jun and β-catenin/TCF4 to the c-jun promoter is dependent upon JNK activity, thus one role for this complex is to contribute to the repression and/or activation of genes that may mediate cell maintenance, proliferation, differentiation, and death, whereas deregulation of these signals may contribute to carcinogenesis. Here we address the functional links reported between activated β-catenin/JNK signaling pathways, their component genes, and their common targets, and discuss how alterations in the properties of these genes lead to the development of cancer. (Mol Cancer Res 2009;7(8):1189–96)

Cancer is a genomic disease and is associated with genetic and epigenetic alterations. Accumulation of these alterations during carcinogenesis activates proto-oncogenes and inactivates tumor suppressors (1, 2). The mammalian Wnt gene family consists of 19 members and encodes secreted glycoproteins. These glycoproteins control many cellular processes such as cell proliferation, apoptosis, and differentiation in the adult and during embryonic development (refs. 3-5, and see the Wnt signaling site: http://www.stanford.edu/~rnusse/wntwindow.html). Wnt signaling is important because several of its signal transduction target genes are implicated in various cancer types and developmental disorders.

Wnt signaling has traditionally been classified as either canonical or noncanonical, with the former more extensively characterized. Here the binding of Wnts to Frizzled and LRP5/6 receptors stabilizes the cytoplasmic β-catenin that subsequently becomes translocated to the nucleus, where it interacts with transcription factors regulating the target gene expression. In fact, some Wnt pathway mutations, such as loss of the adenomatous polyposis coli (APC) gene is sufficient to give rise to a tumor. This mutation may, by abnormally activating Wnt signaling, also switch on associated pathways: Notch, Eph/ephrin, BMP, Hedgehog, and MAPK, as observed in the APCMin mouse model of intestinal carcinogenesis (1, 2).

c-Jun NH2-terminal kinase (JNK) is a member of the mitogen-activated protein kinases (MAPK) and is involved in noncanonical Wnt signaling and planar cell polarity. It is activated in response to growth factors, inhibition of DNA and protein synthesis, environmental stress and inflammatory cytokines, all of which regulate cell proliferation, differentiation, and apoptosis (6, 7).

Previously we described the link between canonical (β-catenin/TCF4) and noncanonical (JNK/c-Jun) Wnt-signaling pathways in intestinal tumorigenesis (8). This relationship has been supported by recent studies using alternative approaches (9-13). However, the links between the β-catenin/TCF4 and JNK/c-Jun pathways are still poorly understood, and the identities of the downstream targets and/or effectors of JNK/Wnt remain largely unknown. Moreover, little is known about the effect of phosphorylation by JNK on the functions of these targets in the development of cancer.

Activation of the Wnt-signaling pathway stimulates growth and mediates developmental and carcinogenic signaling between cells (2, 14). In the absence of the Wnt signal, β-catenin levels are regulated by a multiprotein complex that phosphorylates β-catenin targeting its degradation (Fig. 1; ref. 15). This β-catenin degradation complex consists of the APC tumor-suppressor protein Axin and GSK-3β (16). Upon docking of Wnt to Frizzled and LRP5/6 receptors, a cascade of events (canonical Wnt signaling) is relayed that destabilizes the degradation complex. β-catenin levels accumulate and translocate to the nucleus where β-catenin functions as a coactivator for the lymphoid-enhancing factor/T-cell factor (LEF/TCF) family of transcription factors (17). However, the components of this HMG box family, LEF-1, TCF1, TCF3, and TCF4, have a virtually identical DNA-binding domain and a β-catenin interaction domain (18), and this family of nuclear transcription factors remain the predominant partners of β-catenin.

FIGURE 1.

Schematic representation of canonical and noncanonical Wnt pathway components. The canonical signaling is initiated (left) by association of Wnt with its coreceptors Lrp5/6 and Frizzled (Fz) at the plasma membrane, leading to the inhibition of β-catenin (β-Ctnn) degradation complex (Axin/APC/GSK3-β), which permits the accumulation of β-catenin and its translocation to the nucleus to activate target gene transcription by associating with Lef/Tcf transcription factors. The heterotrimeric protein phosphates 2A (PP2A) may also regulate β-catenin stability as antagonists of the serine kinases and are required for the elevation of β-catenin levels, which are dependent on Wnt. In colorectal cancer, mutations of β-catenin itself, inactivation of the APC protein, or Wnt pathway inhibition of GSK3-β kinase lead to the inappropriate formation of β-catenin/Tcf4 complexes. Fz family receptors are also able to trigger the noncanonical Wnt pathways. The noncanonical Wnt pathways are: the Wnt/Ca2+ pathway (middle) and the planar cell polarity pathway (right). The Wnt/Ca2+ pathway, most likely, signals through G-heterotrimeric proteins to assemble intracellular Ca2+, and to stimulate PKC. In the planar cell polarity pathway, two independent pathways, which are initiated by dishevelled (DVL), result in the activation of Rho and Rac GTPases. Activation of Rho involves Daam-1, which results in the activation of the Rho-associated kinase ROCK. However, Rac activation is Daam-1 independent and stimulates JNK activity and consequently phosphorylates AP-1 transcription factors.

FIGURE 1.

Schematic representation of canonical and noncanonical Wnt pathway components. The canonical signaling is initiated (left) by association of Wnt with its coreceptors Lrp5/6 and Frizzled (Fz) at the plasma membrane, leading to the inhibition of β-catenin (β-Ctnn) degradation complex (Axin/APC/GSK3-β), which permits the accumulation of β-catenin and its translocation to the nucleus to activate target gene transcription by associating with Lef/Tcf transcription factors. The heterotrimeric protein phosphates 2A (PP2A) may also regulate β-catenin stability as antagonists of the serine kinases and are required for the elevation of β-catenin levels, which are dependent on Wnt. In colorectal cancer, mutations of β-catenin itself, inactivation of the APC protein, or Wnt pathway inhibition of GSK3-β kinase lead to the inappropriate formation of β-catenin/Tcf4 complexes. Fz family receptors are also able to trigger the noncanonical Wnt pathways. The noncanonical Wnt pathways are: the Wnt/Ca2+ pathway (middle) and the planar cell polarity pathway (right). The Wnt/Ca2+ pathway, most likely, signals through G-heterotrimeric proteins to assemble intracellular Ca2+, and to stimulate PKC. In the planar cell polarity pathway, two independent pathways, which are initiated by dishevelled (DVL), result in the activation of Rho and Rac GTPases. Activation of Rho involves Daam-1, which results in the activation of the Rho-associated kinase ROCK. However, Rac activation is Daam-1 independent and stimulates JNK activity and consequently phosphorylates AP-1 transcription factors.

Close modal

Wnt signaling can also activate noncanonical, β-catenin-independent pathways (Fig. 1). Several noncanonical pathways have been identified, including activation of JNK (19, 20). JNKs are serine/threonine kinases that belong to the group of MAP kinases activated by phosphorylation of the threonine and tyrosine residues in the conserved Thr-X-Tyr motif (21). This phosphorylation is catalyzed by the MAPK kinases (MKK), which are in turn activated by a serine/threonine phosphorylation catalyzed by the MAPKK kinases (MEKK). JNK activation, which can occur via many types of cellular stresses or extracellular signals, plays an essential role in organogenesis during mouse development by regulating cell proliferation, survival, and apoptosis. JNK activation is also involved in mRNA stabilization, cell migration, and cytoskeleton integrity (22-26).

Recent reports proposed Wnt signaling output is dictated by receptor complement present and is dependent on the cell type (27, 28). However, more evidences of crosstalk between Wnt downstream signals through the β-catenin and JNK pathways have now appeared. For example, it is well known that abnormal stimulation of the Wnt pathway through, for instance, transgenic expression of Wnt1 (int-1) or Wnt10b triggers tumorigenesis in mice (29, 30). Wnt5a inhibits the canonical Wnt pathway by promoting β-catenin degradation in a GSK3β-independent manner (31). Also, Wnt5a induces JNK activation in a Ror2-dependent manner to regulate cell migration (22). However, Wnt7a promotes JNK activation and c-Jun phosphorylation in lung cancer cells (23). Moreover, in adult bone marrow mononuclear cells, Wnt3a promotes “stemness,” proliferation, and hematopoietic commitment, and Wnt11 induces cardiomyogenic differentiation in a JNK-dependent manner (24).

On the other hand, Dsh/Dvl and Axin are essential elements in both canonical and noncanonical pathways, because they activate the Wnt/JNK signaling pathway and interact with β-catenin. These interactions take place through different domains in the protein sequence (25). Axin induces JNK activity through its MEKK1 binding and self-association domains (26), but these domains are not required for the degradation of β-catenin in Xenopus axis determination (32). Moreover, the DIX domain of Dsh is essential for β-catenin signaling, but it is not required for the planar cell polarity pathway in Drosophila (33). This suggests that Wnt cascade might switch at the Axin and Dsh/Dvl level and the dual role of these transducers might indicate cross-signaling interaction between the Wnt pathways, as well as other signaling pathways. However, the significance of this interaction in human development and disease remains largely unknown.

Wnt and JNK pathways can also coordinate to activate certain genes that are common targets for both pathways. For example, transgenic mice with high levels of c-myc or cyclin D1, both Wnt targets, also develop tumorigenesis (34, 35). This activation could take place, at least in part, through the union of β-catenin/TCF4 and c-Jun/AP-1, which form a transcriptional complex that induce the expression of these common target genes (Table 1). These targets will be discussed further in this article.

Table 1.

Summary of β-catenin/TCF4 and JNK/c-Jun Common Target Genes with Promoters Contained in Binding Sites of AP-1 and TCFs, Which Were Analyzed and Transcriptionally Induced in Cancers

β-catenin/JNK Target GenesReference
c-myc Yochum et al. (110) 
c-jun Nateri et al. (8)  
cyclin-D1 Toualbi et al. (100)  
Mmp7 Crawford et al. (116)  
Mmp26 Marchenko et al. (119)  
Osteopontin(Eta-1) El-Tanani et al. (112)  
Cd44 Nateri et al. (8), Van der Flier et al. (108)  
Wnt2 Le Floch et al. (10)  
β-catenin/JNK Target GenesReference
c-myc Yochum et al. (110) 
c-jun Nateri et al. (8)  
cyclin-D1 Toualbi et al. (100)  
Mmp7 Crawford et al. (116)  
Mmp26 Marchenko et al. (119)  
Osteopontin(Eta-1) El-Tanani et al. (112)  
Cd44 Nateri et al. (8), Van der Flier et al. (108)  
Wnt2 Le Floch et al. (10)  

NOTE: These studies show JNK/β-catenin pathways cooperatively enhance their transcriptional activities including c-jun promoter itself, c-myc, cyclin D1, Mmp7, Mmp26, Wnt2, Eta-1, and Cd44.

Among the three JNK isoenzymes, JNK3 is restricted to expression in brain whereas the JNK1 and JNK2 proteins are ubiquitously expressed (21). JNK family members are involved in diverse biological phenomena through the phosphorylation and regulation of many types of proteins, among them, transcription factors, such as ATF2, Elk-1, stat3 (6, 36, 37), and c-Myc (38), as well as members of the Bcl-2 family (Bcl-2, Bcl-xL, Bim, and BAD), IRS-1, Itch, and Tau (39-43). Inactivation of single jnk genes in mice failed to exert marked developmental defects, whereas the double knockout of jnk1 and jnk2 resulted in embryonic lethality because of neural tube defects (44, 45). Moreover, mice embryos with the jnk1−/+; jnk2−/+ genotypes showed delayed epithelial development in the epidermis, intestines, and lungs accompanied by decreased EGF signaling (46). Although jnk3 has been shown to be mutated in brain tumors (47), its role in carcinogenesis has not been extensively studied (48).

The diversity of JNK target genes has defined differential functions for JNK, and the analyses of pathways regulated by JNK have shown that JNK is indispensable for both cell proliferation and apoptosis. It seems that, depending on the stimulus and the cell type, the activation of JNKs might give rise to cell proliferation or apoptosis (6, 36).

Recently, there are accumulating data supporting a key role of JNK activity in cell survival, proliferation, and carcinogenesis. JNK activity is induced by oncogenes in certain tumor types (49-51). Ras-transformed MEFs lacking both jnk1 and jnk2 had a reduced growth rate (52). Growth inhibition was also reported using myeloma and breast cancer cells treated with JNK inhibitors, and antisense oligonucleotides (53, 54). In addition, the small GTPase Ral, a putative proximal regulator of the JNK pathway, is required for murine skin carcinogenesis (55). Other studies showed that JNK1 is required for BCR/Abl-mediated transformation of pre-B cells in vitro and in vivo (56). Thus, there is growing evidence supporting a protumorigenic role for the JNKs, and the idea that blocking JNK signaling may be of therapeutic benefit in treating cancer led to the development of small molecule JNK inhibitors (57, 58).

Several studies provide information on the molecular targets through which JNKs may promote cell transformation. TGFβ1 can activate JNK1, which in turn phosphorylates cyclin-dependent kinase inhibitor p21/WAF1, a transcriptional target of p53 and cell cycle regulator, and increases its stability through a SMAD-independent mechanism (59, 60). Also, JNK is implicated in androgen-independent prostate cancer in which, after leptin stimulation, JNK mediates STAT3 activation through Ser-727 phosphorylation (37).

JNKs phosphorylate c-Jun on Ser63, Ser73, Thr91, and Thr93 within the transcriptional activation domain. Phosphorylation of JNK was markedly increased in response to oncogene over-expression (49, 50, 61), and this phosphorylation is also required for activated Ras-induced skin tumorigenesis in vivo (62). Furthermore, c-Jun contributes to early stages of carcinogen-induced hepatocellular carcinoma by antagonizing the action of p53 (63), and transgenic mice expressing an oncogenic form of jun developed fibrosarcomas at sites of wound healing (64). Additionally, in human melanoma, the ERK signaling pathway upregulates JNK and activates c-Jun and its downstream targets, including cyclin D1 and RACK1, which in turn enables protein kinase C (PKC) to phosphorylate and enhance JNK activity (65). We have also identified TCF4 and β-catenin as novel partners of c-Jun. These three proteins form a complex in a JNK-dependent manner (Fig. 2), and if phosphorylation was prevented through mutation of the JNK phosphorylation sites the extent of intestinal tumorigenesis in the APCMin mouse model was delayed (8). Similarly, JNKs phosphorylate the androgen receptor in prostate cancer cells, leading to nuclear export and inhibition of androgen-receptor-dependent transcriptional responses (66). In lung cancer cells, the JNK pathway may contribute to cellular transformation by down-regulating tumor suppressors such as p53 or nuclear receptors that promote epithelial differentiation. Retinoic acid receptor α (RARα), involved in normal epithelial cell growth and differentiation, is phosphorylated on several residues by JNKs leading to its enhanced degradation (67). ATF2 transcription factor is phosphorylated by JNK, and its transcriptional activity depends on its heterodimerization with c-Jun (68). Inhibiting ATF2 interferes with, and slows the progression of, melanoma development (69), and over-expression of ATF2 is required for progression and growth of mouse skin tumors (70). However, other studies show that ATF2 can also elicit skin tumor suppressor function, and the loss of the transcriptional activity of ATF2 enhances the induced skin tumorigenesis that is associated with an increase in the expression of β-catenin, EGFR, phospho-JNK, phospho-c-Jun, and cyclin D (71). Additionally, DNA topoisomerase I, which is required for EGF receptor expression and cell proliferation of HT-1080 cells, has recently been identified as a JNK-dependent interaction partner with c-Jun (72). Also, JNKs have been shown to directly phosphorylate Smad2 and -3 on sites within their linker regions where phosphorylation is associated with increased nuclear localization of Smad3 and phospho-c-Jun. Moreover, the induction of JNK activity increased the progression and extension of colon cancer (73).

FIGURE 2.

Schematic representation of a JNK-dependent interaction on c-jun promoter between phosphorylated c-Jun and β-catenin/TCF4, which consequently activates c-Jun transcription. This activation requires both the TCF and AP-1 sites of the c-jun promoter acting in cis and depends on amino-terminal c-Jun serine residue phosphorylation (8).

FIGURE 2.

Schematic representation of a JNK-dependent interaction on c-jun promoter between phosphorylated c-Jun and β-catenin/TCF4, which consequently activates c-Jun transcription. This activation requires both the TCF and AP-1 sites of the c-jun promoter acting in cis and depends on amino-terminal c-Jun serine residue phosphorylation (8).

Close modal

We previously showed that induction of JNK activity is antagonized by an SCF-E3-Ligase FBXW7 tumor suppressor (74, 75). Moreover, Pdcd4 tumor suppressor inhibits the activity of JNK (76) and down-regulation of Pdcd4, which is frequently down-regulated in human colon cancer tissues, and in colon cancer HT29 cells, it produces activation of β-catenin/Tcf-dependent and AP-1-dependent transcription (77). Taken together this activity provides additional evidence for a role of JNKs and their targets in cell transformation and tumor development (51).

Many of the studies already discussed suggest that JNK signaling has a protumorigenic activity. This role is supported by some in vivo studies in which Jnk1−/− mutant mice exhibited a marked decrease in gastric tumor development caused by N-methyl-N-nitrosourea, in comparison with wild-type controls. Of note, p21 expression was similar between wild-type and JNK1−/− mice. The impaired carcinogenesis was associated with decreased cell transformation and proliferation (78). Moreover, JNK1−/− mice were much less susceptible to diethylnitrosamine-induced hepatocarcinogenesis, and the absence of JNK1 resulted in decreased expression of cyclin D and vascular endothelial growth factor, as well as reduced cell proliferation and tumor neovascularization (79).

In contrast to these findings, others found that JNK1−/− mice develop spontaneous intestinal tumors that were associated with down-regulation of p21 in intestinal epithelial cells (80), and recently the same group showed that JNK activity negatively regulates β-catenin signaling through GSK3β pathway and that the β-catenin alteration is probably responsible for the intestinal tumor formation in JNK1−/− mice (81). Furthermore, JNK1−/− mice developed more UVA-induced papillomas than either JNK+/+ or JNK2−/− mice, which was associated with suppressed Myt1 phosphorylation and decreased caspase-3 cleavage. This suggests that the JNK1-mediated phosphorylation of Myt1 plays an important role in UVA-induced apoptosis and the prevention of skin carcinogenesis (82). She and colleagues also described that Jnk1−/− mice are more susceptible to TPA-induced skin tumor development than wild-type mice (83); contrasting completely with JNK2, which primarily functions as a key regulator of carcinogenesis, as evidenced by the multiplicity of papillomas induced by TPA in Jnk2−/− mice compared with wild-type mice (84), which support a critical role for JNK2 in the tumor promotion process. Of relevance, fibroblasts lacking jnk2 unexpectedly show higher JNK activity and c-Jun phosphorylation (85, 86), and JNK2 can also phosphorylate and induce nuclear translocation of β-catenin (87).

These findings suggest that JNKs can have tumor-promoting or tumor-suppressing functions, depending on the cell type and/or organ or the stimuli, and provide mechanistic insights into the distinct roles of the different JNK isoforms.

However, it is of note that Jnk1−/− mice, reported by Tong and colleagues (80), are conventional knock-outs that lack JNK1 in all cell types, and it may be that the loss of JNK1 function in immune cells led to prolonged inflammation or reduced immune surveillance, thus contributing to tumor development. Indeed, jnk1−/− mice had reduced immunity, and jnk1-deficient T cells show abnormal T-helper cell differentiation and cytokine production (88, 89). As neither general JNK activity nor phosphorylation of its substrates (e.g., ATF2, c-Jun) have been analyzed in a conditionally ablated jnk1 null mouse, it remains unclear whether it has a functional role in tumor development. So far genetic changes in JNK1 have not been identified in human tumors. Therefore, to investigate the significance of JNK1 activation, a tissue-specific transgenic mouse allowing a constitutively active JNKK2-JNK1 fusion protein (90), thereby stimulating JNK and phosphorylating its substrates, is required.

TCF4 is a component of the HMG family of transcription factors, which is the predominant partner of β-catenin in the canonical wnt pathway. TCF4 plays an essential role in normal development (91). However, the proto-oncoprotein c-Jun belongs to the AP-1 group of transcription factors and is a key regulator of cellular proliferation, apoptosis, and tumorigenesis (51, 92). It also plays pivotal roles in bone, liver, heart, skin, hematopoietic, and neuronal development (51, 93-95).

c-Jun heterodimerizes and forms functional transcription factors with a number of interacting partners including all members of the Fos and ATF families, via a leucine zipper interaction interface (51). AP-1 activity is strongly induced in response to numerous signals including growth factors, cytokines, and extracellular stresses. AP-1 stimulation is mediated, in part, by the c-Jun N-terminal phosphorylation (JNP) and by the JNKs within its transactivation domain. It is thought that JNP increases the transcription of target genes, including the c-jun gene itself (21), which is a well-characterized Wnt target gene (96, 97). This observation suggests that high JNK activity may not only stimulate the expression of AP-1 target genes, but also increase the expression of Wnt downstream target genes. Chromatin immunoprecipitation (ChIP) analysis indicates that c-Jun binds several Wnt promoters, which are misregulated by activated-FOS expression, confirming that members of the Wnt pathway can be primary targets of AP-1 transcriptional regulation (12). Similarly, the use of a genome-wide ChIP-on-chip analysis identified the tcf4 promoter region as a target for phosphorylated c-Jun, which means that TCF4 is transcriptionally regulated through activated JNK signaling (11). In addition to TCF4, it has now been shown that JNK2, and to a much lesser extent JNK1, can phosphorylate and induce nuclear translocation of β-catenin (87). Moreover, molecular characterization of cultured keratinocytes and tumors indicates that c-Jun regulates the balance between the Wnt/β-catenin and Hedgehog signaling pathways through binding to several Wnt promoters (12). On the other hand, TCF/β-catenin strongly activates c-jun transcription in both hematopoietic and colon cancer cells (8, 96, 97). Moreover, GnRH stimulates the nuclear localization of β-catenin (98) and regulates, through a functional interaction between LEF/TCF and β-catenin, the expression of jun and c-Jun target genes in gonadotropes (99). Toualbi and colleagues (100) reported that c-Jun as well as c-Fos interact with β-catenin and activate the c-myc-luciferase reporter in a TCF-dependent manner.

Conditional ablation of c-Jun, for example deletion in the epidermis, causes an eye closure defect, affects keratinocyte proliferation in vitro, and delays skin tumor development invivo (101, 102). Moreover, genetic abrogation of JNP or gut-specific conditional c-jun inactivation reduced tumor number and size and prolonged lifespan in APCMin mice (8), which develop multiple intestinal neoplasias because of excessive canonical Wnt signaling (8, 103). Therefore, the phosphorylation-dependent interaction between c-Jun and TCF4 regulates tumorigenesis by integrating JNK and β-catenin, two distinct pathways activated by Wnt signaling (Fig. 3). However, JNK inhibition impairs the function of both AP-1 and TCF4 transcription factors and may have potentially beneficial therapeutic effects on colon cancer progression (77).

FIGURE 3.

Two distinct β-catenin/TCF4 and JNK/c-Jun pathways are physiologically cooperating in tumorigenesis. These interactions cooperatively enhance transcriptional activities of promoters containing AP-1 and TCF-binding sites. The resultant proteins can promote a positive feedback loop for many of Wnt-target genes, which is consistent with their observed role in human colorectal tumors.

FIGURE 3.

Two distinct β-catenin/TCF4 and JNK/c-Jun pathways are physiologically cooperating in tumorigenesis. These interactions cooperatively enhance transcriptional activities of promoters containing AP-1 and TCF-binding sites. The resultant proteins can promote a positive feedback loop for many of Wnt-target genes, which is consistent with their observed role in human colorectal tumors.

Close modal

In addition to colorectal cancer, this complex, alone or in combination with other transcription factors, may activate the transcription of different target genes; therefore, it could be involved in tumor formation in different organs.

A variety of Wnt target genes, including c-myc, c-jun, cyclinD1, cd44, WNTs, and MMPs are transcriptionally regulated through both TCF4 and c-Jun transcription factors (9-13, 51, 93, 104). It has been shown that c-jun expression is regulated by β-catenin/TCF4. ChIP analysis showed that binding of c-Jun and β-catenin/TCF4 to the c-jun promoter is dependent on JNK activity (8). Cyclin D1 is an important target gene through which c-Jun/AP-1 controls proliferation (105). Similarly, the expression of cyclin D1 is regulated by both JNK and Wnt signaling (104, 106) and AP-1/β-catenin/TCF4 cooperation is directly involved in the transcriptional induction of cyclin D1 (100). CD44 is both c-Jun and TCF4 target gene (8, 107, 108).

On the other hand, Gan and colleagues showed that binding of c-Jun, TCF4, β-catenin, and Dishevelled is required for full c-Myc expression (109). Moreover, association of c-Jun, β-catenin, and TCF4, specifically with the downstream enhancer, triggers c-Myc transcription in HCT116 human colorectal carcinoma cells after mitogen stimulation (110). Most notably, the use of a genome-wide ChIP-on-chip analysis identified that TCF4 (Tcf7l2) is transcriptionally regulated through activated JNK signaling (11) with three of the AP-1/c-Jun binding sites, at position −996 to −833 base pairs (bp) relative to the TATA box, of the tcf7l2 gene. In our current studies, we have shown TCF4/c-Jun colocalization in adenomas derived from APCMin mice and microarray data suggest a significant similarity in TCF4 transactivation in response to mutations in APC and/or β-catenin genes (111). However, in mammary epithelia, the scenario seems to be different: El-Tanani and colleagues (112) described that β-catenin-Lef-1 and c-Jun cooperate with Ets transcription factors family in regulation of osteopontin transcription, which plays a key role in neoplastic transformation, metastasis (113), and the prognosis of breast cancer (114). It was concluded that the presence of these transcription factors in human breast cancer is responsible for the over-expression of osteopontin (112). Similarly, AP-1, the Ets transcription factor PEA3 synergizes with β-catenin/Lef-1 in the up-regulation of the matrix metalloproteinase matrilysin (MMP7) in intestinal tumors (115, 116), itself predominantly expressed in the cells of gastrointestinal, breast, and lung carcinomas (117). Furthermore, Rivat and colleagues (118) corroborated that Src-mediated activation of the human MMP7 promoter requires the activation of AP-1 signaling and a cooperative interaction between c-Jun and LEF-1 transcription factors (118). Moreover, β-catenin, LEF/TCF, Ras, and c-Jun interact with, and synergistically activate, the MMP-26 promoter (119). MMP-26 is involved in hyperplastic, and malignant endometrial (120), and is over-expressed in skin cancer (121). Immunohistochemical analysis of human colorectal tumors showed nuclear expression of c-Jun, TCF4, and β-catenin (122).

Taken together, there is, therefore, significant crosstalk between JNK and the canonical Wnt pathways. The connection between these two pathways may result in co-expression of both c-Jun and TCF4 target genes. These genes, in turn, form a positive feedback loop increasing the strength of Wnt signaling (Fig. 3) and thereby maintaining tissue homeostasis and cancer development. However, the mechanisms through which β-catenin:TCF4/JNK:AP-1 cooperate to maintain a mechanistic basis for the stem cell phenotype in cancer remains to be elucidated.

No potential conflicts of interest were disclosed.

We thank K. Wang and A. Behrens for helpful comments on the manuscript.

1
Nowell
PC
. 
Chromosomes and cancer: the evolution of an idea
.
Adv Cancer Res
1993
;
62
:
1
17
.
2
Reya
T
,
Clevers
H
. 
Wnt signalling in stem cells and cancer
.
Nature
2005
;
434
:
843
50
.
3
Cadigan
KM
,
Nusse
R
. 
Wnt signaling: a common theme in animal development
.
Genes Dev
1997
;
11
:
3286
305
.
4
Moon
RT
,
Bowerman
B
,
Boutros
M
,
Perrimon
N
. 
The promise and perils of Wnt signaling through beta-catenin
.
Science
2002
;
296
:
1644
6
.
5
Wodarz
A
,
Nusse
R
. 
Mechanisms of Wnt signaling in development
.
Annu Rev Cell Dev Biol
1998
;
14
:
59
88
.
6
Liu
J
,
Lin
A
. 
Role of JNK activation in apoptosis: a double-edged sword
.
Cell Res
2005
;
15
:
36
42
.
7
Johnson
GL
,
Nakamura
K
. 
The c-jun kinase/stress-activated pathway: regulation, function and role in human disease
.
Biochim Biophys Acta
2007
;
1773
:
1341
8
.
8
Nateri
AS
,
Spencer-Dene
B
,
Behrens
A
. 
Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development
.
Nature
2005
;
437
:
281
5
.
9
Hwang
SG
,
Yu
SS
,
Lee
SW
,
Chun
JS
. 
Wnt-3a regulates chondrocyte differentiation via c-Jun/AP-1 pathway
.
FEBS Lett
2005
;
579
:
4837
42
.
10
Le Floch
N
,
Rivat
C
,
De Wever
O
, et al
. 
The proinvasive activity of Wnt-2 is mediated through a noncanonical Wnt pathway coupled to GSK-3beta and c-Jun/AP-1 signaling
.
FASEB J
2005
;
19
:
144
6
.
11
Hayakawa
J
,
Mittal
S
,
Wang
Y
, et al
. 
Identification of promoters bound by c-Jun/ATF2 during rapid large-scale gene activation following genotoxic stress
.
Mol Cell
2004
;
17
:
161
.
12
Gerdes
MJ
,
Myakishev
M
,
Frost
NA
, et al
. 
Activator protein-1 activity regulates epithelial tumor cell identity
.
Cancer Res
2006
;
66
:
7578
88
.
13
Igaki
T
,
Pagliarini
RA
,
Xu
T
. 
Loss of cell polarity drives tumor growth and invasion through JNK activation in Drosophila
.
Curr Biol
2006
;
16
:
1139
46
.
14
Bienz
M
,
Clevers
H
. 
Linking colorectal cancer to Wnt signaling
.
Cell
2000
;
103
:
311
20
.
15
Giles
RH
,
van Es
JH
,
Clevers
H
. 
Caught up in a Wnt storm: Wnt signaling in cancer
.
Biochim Biophys Acta
2003
;
1653
:
1
24
.
16
Behrens
J
,
Jerchow
BA
,
Würtele
M
, et al
. 
Functional interaction of an axin homolog, conductin, with beta-catenin, APC, and GSK3beta
.
Science
1998
;
280
:
596
9
.
17
Behrens
J
,
von Kries
JP
,
Kühl
M
, et al
. 
Functional interaction of beta-catenin with the transcription factor LEF-1
.
Nature
1996
;
382
:
638
42
.
18
Arce
L
,
Yokoyama
NN
,
Waterman
ML
. 
Diversity of LEF/TCF action in development and disease
.
Oncogene
2006
;
25
:
7492
504
.
19
Veeman
MT
,
Axelrod
JD
,
Moon
RT
. 
A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling
.
Dev Cell
2003
;
5
:
367
77
.
20
Kohn
AD
,
Moor
RT
. 
Wnt and calcium signaling: beta-catenin-independent pathways
.
Cell Calcium
2005
;
38
:
439
46
.
21
Davis
RJ
. 
Signal transduction by the JNK group of MAP kinases
.
Cell
2000
;
103
:
239
52
.
22
Nomachi
A
,
Nishita
M
,
Inaba
D
,
Enomoto
M
,
Hamasaki
M
,
Minami
Y
. 
Receptor tyrosine kinase Ror2 mediates Wnt5a-induced polarized cell migration by activating c-Jun N-terminal kinase via actin-binding protein filamin A
.
J Biol Chem
2008
;
283
:
27973
81
.
23
Heasley
LE
,
Winn
RA
. 
Analysis of Wnt7a-stimulated JNK activity and cJun phosphorylation in non-small cell lung cancer cells
.
Methods Mol Biol
2008
;
468
:
187
96
.
24
Flaherty
MP
,
Abdel-Latif
A
,
Li
Q
, et al
. 
Noncanonical Wnt11 signaling is sufficient to induce cardiomyogenic differentiation in unfractionated bone marrow mononuclear cells
.
Circulation
2008
;
117
:
2241
52
.
25
Klingensmith
J
,
Nusse
R
,
Perrimon
N
. 
The Drosophila segment polarity gene dishevelled encodes a novel protein required for response to the wingless signal
.
Genes Dev
1994
;
8
:
118
30
.
26
Zhang
Y
,
Neo
SY
,
Wang
X
,
Han
J
,
Lin
SC
. 
Axin forms a complex with MEKK1 and activates c-Jun NH(2)-terminal kinase/stress-activated protein kinase through domains distinct from Wnt signaling
.
J Biol Chem
1999
;
274
:
35247
54
.
27
Mikels
AJ
,
Nusse
R
. 
Purified Wnt5a protein activates or inhibits beta-catenin-TCF signalling depending on receptor context
.
PLoS Biol
2006
;
4
:
e115
.
28
van Amerongen
R
,
Mikels
A
,
Nusse
R
. 
Alternative wnt signaling is initiated by distinct receptors
.
Sci Signal
2008
;
1
:
re9
,
[Review]
.
29
Tsukamoto
AS
,
Grosschedl
R
,
Guzman
RC
,
Parslow
T
,
Varmus
HE
. 
Expression of the int-1 gene in transgenic mice is associated with mammary gland hyperplasia and adenocarcinomas in male and female mice
.
Cell
1988
;
55
:
619
25
.
30
Lane
TF
,
Leder
P
. 
Wnt-10b directs hypermorphic development and transformation in mammary glands of male and female mice
.
Oncogene
1997
;
15
:
2133
44
.
31
Topol
L
,
Jiang
X
,
Choi
H
,
Garrett-Beal
L
,
Carolan
PJ
,
Yang
Y
. 
Wnt-5a inhibits the canonical Wnt pathway by promoting GSK-3-independent beta-catenin degradation
.
J Cell Biol
2003
;
162
:
899
908
.
32
Fagotto
F
,
Jho
E
,
Zeng
L
, et al
. 
Domains of axin involved in protein-protein interactions, Wnt pathway inhibition, and intracellular localization
.
J Cell Biol
1999
;
145
:
741
56
.
33
Boutros
M
,
Paricio
N
,
Strutt
DI
,
Mlodzik
M
. 
Dishevelled activates JNK and discriminates between JNK pathways in planar polarity and wingless signaling
.
Cell
1998
;
94
:
109
18
.
34
Wang
TC
,
Cardiff
RD
,
Zukerberg
L
,
Lees
E
,
Arnold
A
,
Schmidt
EV
. 
Mammary hyperplasia and carcinoma in MMTV-cyclin D1 transgenic mice
.
Nature
1994
;
369
:
669
71
.
35
Sinn
E
,
Muller
W
,
Pattengale
P
,
Tepler
I
,
Wallace
R
,
Leder
P
. 
Coexpression of MMTV/v-Ha-ras and MMTV/c-myc genes in transgenic mice: synergistic action of oncogenes in vivo
.
Cell
1987
;
49
:
465
75
.
36
Whitmarsh
AJ
,
Davis
RJ
. 
Transcription factor AP-1 regulation by mitogen-activated protein kinase signal transduction pathways
.
J Mol Med
1996
;
74
:
589
607
,
[Review]
.
37
Miyazaki
T
,
Bub
JD
,
Iwamoto
Y
. 
c-Jun NH(2)-terminal kinase mediates leptin-stimulated androgen-independent prostate cancer cell proliferation via signal transducer and activator of transcription 3 and Akt
.
Biochim Biophys Acta
2008
;
1782
:
593
604
.
38
Noguchi
K
,
Kitanaka
C
,
Yamana
H
,
Kokubu
A
,
Mochizuki
T
,
Kuchino
Y
. 
Regulation of c-Myc through phosphorylation at Ser-62 and Ser-71 by c-Jun N-terminal kinase
.
J Biol Chem
1999
;
274
:
32580
7
.
39
Yamamoto
K
,
Ichijo
H
,
Korsmeyer
SJ
. 
BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-terminal protein kinase pathway normally activated at G(2)/M
.
Mol Cell Biol
1999
;
19
:
8469
78
.
40
Yu
C
,
Minemoto
Y
,
Zhang
J
, et al
. 
JNK suppresses apoptosis via phosphorylation of the proapoptotic Bcl-2 family protein BAD
.
Mol Cell
2004
;
13
:
329
40
.
41
Hiratani
K
,
Haruta
T
,
Tani
A
,
Kawahara
J
,
Usui
I
,
Kobayashi
M
. 
Roles of mTOR and JNK in serine phosphorylation, translocation, and degradation of IRS-1
.
Biochem Biophys Res Commun
2005
;
335
:
836
42
.
42
Gao
M
,
Labuda
T
,
Xia
Y
, et al
. 
Jun turnover is controlled through JNK-dependent phosphorylation of the E3 ligase Itch
.
Science
2004
;
306
:
271
5
.
43
Yoshida
H
,
Hastie
CJ
,
McLauchlan
H
,
Cohen
P
,
Goedert
M
. 
Phosphorylation of microtubule-associated protein tau by isoforms of c-Jun N-terminal kinase (JNK)
.
J Neurochem
2004
;
90
:
352
8
.
44
Kuan
CY
,
Yang
DD
,
Samanta Roy
DR
, et al
. 
The Jnk1 and Jnk2 protein kinases are required for regional specific apoptosis during early brain development
.
Neuron
1999
;
22
:
667
76
.
45
Sabapathy
K
,
Jochumm
W
,
Hochedlinger
K
, et al
. 
Defective neural tube morphogenesis and altered apoptosis in the absence of both JNK1 and JNK2
.
Mech Dev
1999
;
89
:
115
24
.
46
Weston
CR
,
Wong
A
,
Hall
JP
, et al
. 
The c-Jun NH2-terminal kinase is essential for epidermal growth factor expression during epidermal morphogenesis
.
Proc Natl Acad Sci U S A
2004
;
101
:
14114
9
.
47
Butterfield
L
,
Zentrich
E
,
Beekman
A
,
Heasley
LE
. 
Stress and cell type-dependent regulation of transfected cJun N-terminal kinase and mitogen-activated protein kinase kinase isoforms
.
Biochem J
1999
;
338
:
681
6
.
48
Yoshida
S
,
Fukino
K
,
Harada
H
, et al
. 
The c-Jun NH2-terminal kinase3 (JNK3) gene: genomic structure, chromosomal assignment, and loss of expression in brain tumors
.
J Hum Genet
2001
;
46
:
182
7
.
49
Dérijard
B
,
Hibi
M
,
Wu
IH
, et al
. 
JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain
.
Cell
1994
;
76
:
1025
37
.
50
Smeal
T
,
Binetruy
B
,
Mercola
DA
,
Birrer
M
,
Karin
M
. 
Oncogenic and transcriptional cooperation with Ha-Ras requires phosphorylation of c-Jun on serines 63 and 73
.
Nature
1991
;
354
:
494
6
.
51
Eferl
R
,
Wagner
EF
. 
AP-1: a double-edged sword in tumorigenesis
.
Nat Rev Cancer
2003
;
3
:
859
68
.
52
Kennedy
NJ
,
Sluss
HK
,
Jones
SN
, et al
. 
Suppression of Ras-stimulated transformation by the JNK signal transduction pathway
.
Genes Dev
2003
;
17
:
629
37
.
53
Hideshima
T
,
Hayashi
T
,
Chauhan
D
, et al
. 
Biologic sequelae of c-Jun NH(2)-terminal kinase (JNK) activation in multiple myeloma cell lines
.
Oncogene
2003
;
22
:
8797
801
.
54
Mingo-Sion
AM
,
Marietta
PM
,
Koller
E
,
Wolf
DM
,
Van Den Berg
CL
. 
Inhibition of JNK reduces G2/M transit independent of p53, leading to endoreduplication, decreased proliferation, and apoptosis in breast cancer cells
.
Oncogene
2004
;
23
:
596
604
.
55
Gonzalez-Garcia
A
,
Pritchard
CA
,
Paterson
HF
, et al
. 
RalGDS is required for tumor formation in a model of skin carcinogenesis
.
Cancer Cell
2005
;
7
:
219
26
.
56
Hess
P
,
Pihan
G
,
Sawyers
CL
,
Flavell
RA
,
Davis
RJ
. 
Survival signaling mediated by c-Jun NH(2)- terminal kinase in transformed B lymphoblasts
.
Nat Genet
2002
;
32
:
201
5
.
57
Karin
M
,
Gallagher
E
. 
From JNK to pay dirt: jun kinases, their biochemistry, physiology and clinical importance
.
IUBMB Life
2005
;
57
:
283
95
.
58
Manning
AM
,
Davis
RJ
. 
Targeting JNK for therapeutic benefit: from junk to gold?
Nat Rev Drug Discov
2003
;
2
:
554
65
.
59
Kim
GY
,
Mercer
SE
,
Ewton
DZ
,
Yan
Z
,
Jin
K
,
Friedman
E
. 
The stress-activated protein kinases p38 alpha and JNK1 stabilize p21(Cip1) by phosphorylation
.
J Biol Chem
2002
;
277
:
29792
802
.
60
Fan
Y
,
Chen
H
,
Qiao
B
, et al
. 
c-Jun NH2-terminal kinase decreases ubiquitination and promotes stabilization of p21(WAF1/CIP1) in K562 cell
.
Biochem Biophys Res Commun
2007
;
355
:
263
8
.
61
Smeal
T
,
Binetruy
B
,
Mercola
D
, et al
. 
Oncoprotein-mediated signalling cascade stimulates c-Jun activity by phosphorylation of serines 63 and 73
.
Mol Cell Biol
1992
;
12
:
3507
13
.
62
Behrens
A
,
Jochum
W
,
Sibilia
M
,
Wagner
EF
. 
Oncogenic transformation by ras and fos is mediated by c-Jun N-terminal phosphorylation
.
Oncogene
2000
;
19
:
2657
63
.
63
Eferl
R
,
Ricci
R
,
Kenner
L
, et al
. 
Liver tumor development. c-Jun antagonizes the proapoptotic activity of p53
.
Cell
2003
;
112
:
181
92
.
64
Schuh
AC
,
Keating
SJ
,
Monteclaro
FS
,
Vogt
PK
,
Breitman
ML
. 
Obligatory wounding requirement for tumorigenesis in v-jun transgenic mice
.
Nature
1990
;
346
:
756
60
.
65
Lopez-Bergami
P
,
Huang
C
,
Goydos
JS
, et al
. 
Rewired ERK-JNK signaling pathways in melanoma
.
Cancer Cell
2007
;
11
:
447
60
.
66
Gioeli
D
,
Black
BE
,
Gordon
V
, et al
. 
Stress kinase signaling regulates androgen receptor phosphorylation, transcription, and localization
.
Mol Endocrinol
2006
;
20
:
503
15
.
67
Srinivas
H
,
Juroske
DM
,
Kalyankrishna
S
, et al
. 
c-Jun N-terminal kinase contributes to aberrant retinoid signaling in lung cancer cells by phosphorylating and inducing proteasomal degradation of retinoic acid receptor alpha
.
Mol Cell Biol
2005
;
25
:
1054
69
.
68
De Cesare
D
,
Vallone
D
,
Caracciolo
A
,
Sassone-Corsi
P
,
Nerlov
C
,
Verde
P
. 
Heterodimerization of c-Jun with ATF-2 and c-Fos is required for positive and negative regulation of the human urokinase enhancer
.
Oncogene
1995
;
11
:
365
76
.
69
Bhoumik
A
,
Jones
N
,
Ronai
Z
. 
Transcriptional switch by activating transcription factor 2-derived peptide sensitizes melanoma cells to apoptosis and inhibits their tumorigenicity
.
Proc Natl Acad Sci U S A
2004
;
101
:
4222
7
.
70
Papassava
P
,
Gorgoulis
VG
,
Papaevangeliou
D
,
Vlahopoulos
S
,
van Dam
H
,
Zoumpourlis
V
. 
Overexpression of activating transcription factor-2 is required for tumor growth and progression in mouse skin tumors
.
Cancer Res
2004
;
64
:
8573
84
.
71
Bhoumik
A
,
Fichtman
B
,
Derossi
C
, et al
. 
Suppressor role of activating transcription factor 2 (ATF2) in skin cancer
.
Proc Natl Acad Sci U S A
2008
;
105
:
1674
9
.
72
Mialon
A
,
Sankinen
M
,
Soderstrom
H
, et al
. 
DNA topoisomerase I is a cofactor for c-Jun in the regulation of epidermal growth factor receptor expression and cancer cell proliferation
.
Mol Cell Biol
2005
;
25
:
5040
51
.
73
Yamagata
H
,
Matsuzaki
K
,
Mori
S
, et al
. 
Acceleration of Smad2 and Smad3 phosphorylation via c-Jun NH(2)-terminal kinase during human colorectal carcinogenesis
.
Cancer Res
2005
;
65
:
157
65
.
74
Nateri
AS
,
Riera-Sans
L
,
Da Costa
C
,
Behrens
A
. 
The ubiquitin ligase SCFFbw7 antagonizes apoptotic JNK signaling
.
Science
2004
;
303
:
1374
8
.
75
Tan
Y
,
Sangfelt
O
,
Spruck
C
. 
The Fbxw7/hCdc4 tumor suppressor in human cancer
.
Cancer Lett
2008
;
271
:
1
12
.
76
Bitomsky
N
,
Bohm
M
,
Klempnauer
KH
. 
Transformation suppressor protein Pdcd4 interferes with JNK-mediated phosphorylation of c-Jun and recruitment of the coactivator p300 by c-Jun
.
Oncogene
2004
;
23
:
7484
93
.
77
Wang
Q
,
Sun
Z
,
Yang
HS
. 
Downregulation of tumor suppressor Pdcd4 promotes invasion and activates both beta-catenin/Tcf and AP-1-dependent transcription in colon carcinoma cells
.
Oncogene
2008
;
27
:
1527
35
.
78
Shibata
W
,
Maeda
S
,
Hikiba
Y
, et al
. 
c-Jun NH2-terminal kinase 1 is a critical regulator for the development of gastric cancer in mice
.
Cancer Res
2008
;
68
:
5031
9
.
79
Sakurai
T
,
Maeda
S
,
Chang
L
,
Karin
M
. 
Loss of hepatic NF- B activity enhances chemical hepatocarcinogenesis through sustained c-Jun N-terminal kinase 1 activation
.
Proc Natl Acad Sci U S A
2006
;
103
:
10544
51
.
80
Tong
C
,
Yin
Z
,
Song
Z
, et al
. 
c-Jun NH2-terminal kinase 1 plays a critical role in intestinal homeostasis and tumor suppression
.
Am J Pathol
2007
;
171
:
297
303
.
81
Hu
D
,
Fang
W
,
Han
A
, et al
. 
c-Jun N-terminal kinase 1 interacts with and negatively regulates Wnt/beta-catenin signaling through GSK3beta pathway
.
Carcinogenesis
2008
;
29
:
2317
24
.
82
Choi
HS
,
Bode
AM
,
Shim
JH
,
Lee
SY
,
Dong
Z
. 
c-Jun N-terminal kinase 1 phosphorylates Myt1 to prevent UVA-induced skin cancer
.
Mol Cell Biol
2009
;
29
:
2168
80
.
83
She
QB
,
Chen
N
,
Bode
AM
,
Flavell
RA
,
Dong
Z
. 
Deficiency of c-Jun-NH(2)-terminal kinase-1 in mice enhances skin tumor development by 12-O-tetradecanoylphorbol-13-acetate
.
Cancer Res
2002
;
62
:
1343
8
.
84
Chen
N
,
Nomura
M
,
She
QB
, et al
. 
Suppression of skin tumorigenesis in c-Jun NH(2)-terminal kinase-2-deficient mice
.
Cancer Res
2001
;
61
:
3908
12
.
85
Sabapathy
K
,
Hochedlinger
K
,
Nam
SY
,
Bauer
A
,
Karin
M
,
Wagner
EF
. 
Distinct roles for JNK1 and JNK2 in regulating JNK activity and c-Jun-dependent cell proliferation
.
Mol Cell
2004
;
15
:
713
25
.
86
Sabapathy
K
,
Wagner
EF
. 
JNK2: a negative regulator of cellular proliferation
.
Cell Cycle
2004
;
3
:
1520
3
.
87
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
53
.
88
Constant
SL
,
Dong
C
,
Yang
DD
,
Wysk
M
,
Davis
RJ
,
Flavell
RA
. 
JNK1 is required for T cell-mediated immunity against Leishmania major infection
.
J Immunol
2000
;
165
:
2671
6
.
89
Dong
C
,
Yang
DD
,
Wysk
M
,
Whitmarsh
AJ
,
Davis
RJ
,
Flavell
RA
. 
Defective T cell differentiation in the absence of Jnk1
.
Science
1998
;
282
:
2092
5
.
90
Zheng
C
,
Xiang
J
,
Hunter
T
,
Lin
A
. 
The JNKK2–1 fusion protein acts as a constitutively active c-Jun kinase that stimulates c-Jun transcription activity
.
JBiol Chem
1999
;
274
:
28966
71
.
91
Korinek
V
,
Barker
N
,
Moerer
P
, et al
. 
Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4
.
Nat Genet
1998
;
19
:
379
83
.
92
Shaulian
E
,
Karin
M
. 
AP-1 as a regulator of cell life and death
.
Nat Cell Biol
2002
;
4
:
E131
6
.
93
Jochum
W
,
Passegue
E
,
Wagner
EF
. 
AP-1 in mouse development and tumorigenesis
.
Oncogene
2001
;
20
:
2401
12
.
94
Wagner
EF
,
Eferl
R
. 
Fos/AP-1 proteins in bone and the immune system
.
Immunol Rev
2005
;
208
:
126
40
.
95
Hilberg
F
,
Aguzzi
A
,
Howells
N
,
Wagner
EF
. 
c-jun is essential for normal mouse development and hepatogenesis
.
Nature
1993
;
365
:
179
81
.
96
Mann
B
,
Gelos
M
,
Siedow
A
, et al
. 
Target genes of beta-catenin-T cell-factor/lymphoid-enhancer-factor signaling in human colorectal carcinomas
.
Proc Natl Acad Sci U S A
1999
;
96
:
1603
8
.
97
Staal
FJ
,
Weerkamp
F
,
Baert
MR
, et al
. 
Wnt target genes identified by DNA microarrays in immature CD34+ thymocytes regulate proliferation and cell adhesion
.
J Immunol
2004
;
172
:
1099
108
.
98
Gardner
S
,
Maudsley
S
,
Millar
RP
,
Pawson
AJ
. 
Nuclear stabilization of beta-catenin and inactivation of glycogen synthase kinase-3beta by gonadotropin-releasing hormone: targeting Wnt signaling in the pituitary gonadotrope
.
Mol Endocrinol
2007
;
21
:
3028
38
.
99
Salisbury
TB
,
Binder
AK
,
Grammer
JC
,
Nilson
JH
. 
GnRH-regulated expression of Jun and JUN target genes in gonadotropes requires a functional interaction between TCF/LEF family members and beta-catenin
.
Mol Endocrinol
2009
;
23
:
402
11
.
100
Toualbi
K
,
Güller
MC
,
Mauriz
JL
, et al
. 
Physical and functional cooperation between AP-1 and beta-catenin for the regulation of TCF-dependent genes
.
Oncogene
2007
;
26
:
3492
502
.
101
Zenz
R
,
Scheuch
H
,
Martin
P
, et al
. 
c-Jun regulates eyelid closure and skin tumor development through EGFR signaling
.
Dev Cell
2003
;
4
:
879
89
.
102
Li
G
,
Gustafson-Brown
C
,
Hanks
SK
, et al
. 
c-Jun is essential for organization of the epidermal leading edge
.
Dev Cell
2003
;
4
:
865
77
.
103
Moser
AR
,
Pitot
HC
,
Dove
WF
. 
A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse
.
Science
1990
;
247
:
322
4
.
104
Tetsu
O
,
McCormick
F
. 
Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells
.
Nature
1999
;
398
:
422
6
.
105
Wisdom
R
,
Johnson
RS
,
Moore
C
. 
c-Jun regulates cell cycle progression and apoptosis by distinct mechanisms
.
EMBO J
1999
;
18
:
188
97
.
106
Wulf
GM
,
Ryo
A
,
Wulf
GG
, et al
. 
Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcriptional activity of c-Jun toward cyclin D1
.
EMBO J
2001
;
20
:
3459
72
.
107
Raivich
G
,
Bohatschek
M
,
Da Costa
C
, et al
. 
The AP-1 transcription factor c-Jun is required for efficient axonal regeneration
.
Neuron
2004
;
43
:
57
67
.
108
Van der Flier
LG
,
Sabates-Bellver
J
,
Oving
I
, et al
. 
The intestinal Wnt/TCF signature
.
Gastroenterology
2007
;
132
:
628
32
.
109
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
100
.
110
Yochum
GS
,
Cleland
R
,
Goodman
RH
. 
A genome-wide screen for beta-catenin binding sites identifies a downstream enhancer element that controls c-Myc gene expression
.
Mol Cell Biol
2008
;
28
:
7368
79
.
111
Sancho
R
,
Nateri
AS
,
de Vinuesa
AG
, et al
. 
JNK signalling modulates intestinal homeostasis and tumourigenesis in mice
.
EMBO J
2009
;
28
:
1843
54
.
112
El-Tanani
M
,
Platt-Higgins
A
,
Rudland
PS
,
Campbell
FC
. 
Ets gene PEA3 cooperates with beta-catenin-Lef-1 and c-Jun in regulation of osteopontin transcription
.
J Biol Chem
2004
;
279
:
20794
806
.
113
Weber
GF
. 
The metastasis gene osteopontin: a candidate target for cancer therapy
.
Biochim Biophys Acta
2001
;
1552
:
61
85
,
[Review]
.
114
Patani
N
,
Jouhra
F
,
Jiang
W
,
Mokbel
K
. 
Osteopontin expression profiles predict pathological and clinical outcome in breast cancer
.
Anticancer Res
2008
;
28[6B]
:
4105
10
.
115
Crawford
HC
,
Fingleton
BM
,
Rudolph-Owen
LA
, et al
. 
The metalloproteinase matrilysin is a target of beta-catenin transactivation in intestinal tumors
.
Oncogene
1999
;
18
:
2883
91
.
116
Crawford
HC
,
Fingleton
B
,
Gustavson
MD
, et al
. 
The PEA3 subfamily of Ets transcription factors synergizes with beta-catenin-LEF-1 to activate matrilysin transcription in intestinal tumors
.
Mol Cell Biol
2001
;
21
:
1370
83
.
117
McDonnell
S
,
Navre
M
,
Coffey
RJ,
 Jr.
,
Matrisian
LM
. 
Expression and localization of the matrix metalloproteinase pump-1 (MMP-7) in human gastric and colon carcinomas
.
Mol Carcinog
1991
;
4
:
527
33
.
118
Rivat
C
,
Le Floch
N
,
Sabbah
M
, et al
. 
Synergistic cooperation between the AP-1 and LEF-1 transcription factors in activation of the matrilysin promoter by the src oncogene: implications in cellular invasion
.
FASEB J
2003
;
17
:
1721
3
.
119
Marchenko
ND
,
Marchenko
GN
,
Weinreb
RN
, et al
. 
Beta-catenin regulates the gene of MMP-26, a novel metalloproteinase expressed both in carcinomas and normal epithelial cells
.
Int J Biochem Cell Biol
2004
;
36
:
942
56
.
120
Pilka
R
,
Norata
GD
,
Domanski
H
, et al
. 
Matrix metalloproteinase-26 (matrilysin-2) expression is high in endometrial hyperplasia and decreases with loss of histological differentiation in endometrial cancer
.
Gynecol Oncol
2004
;
94
:
661
70
.
121
Ahokas
K
,
Skoog
T
,
Suomela
S
, et al
. 
Matrilysin-2 (matrix metalloproteinase-26) is upregulated in keratinocytes during wound repair and early skin carcinogenesis
.
J Invest Dermatol
2005
;
124
:
849
56
.
122
Takeda
K
,
Kinoshita
I
,
Shimizu
Y
, et al
. 
Clinicopathological significance of expression of p-c-Jun, TCF4 and beta-Catenin in colorectal tumors
.
BMC Cancer
2008
;
8
:
328
37
.