Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Sep 1.
Published in final edited form as: Exp Cell Res. 2017 Apr 12;358(1):78–85. doi: 10.1016/j.yexcr.2017.04.006

A central role for cadherin signaling in cancer

Antonis Kourtidis 1, Ruifeng Lu 2, Lindy Pence 2, Panos Z Anastasiadis 2,*
PMCID: PMC5544584  NIHMSID: NIHMS868698  PMID: 28412244

Abstract

Cadherins are homophilic adhesion molecules with important functions in cell-cell adhesion, tissue morphogenesis, and cancer. In epithelial cells, E-cadherin accumulates at areas of cell-cell contact, coalesces into macromolecular complexes to form the adherens junctions (AJs), and associates via accessory partners with a subcortical ring of actin to form the apical zonula adherens (ZA). As a master regulator of the epithelial phenotype, E-cadherin is essential for the overall maintenance and homeostasis of polarized epithelial monolayers. Its expression is regulated by a host of genetic and epigenetic mechanisms related to cancer, and its function is modulated by mechanical forces at the junctions, by direct binding and phosphorylation of accessory proteins collectively termed catenins, by endocytosis, recycling and degradation, as well as, by multiple signaling pathways and developmental processes, like the epithelial to mesenchymal transition (EMT). Nuclear signaling mediated by the cadherin associated proteins β-catenin and p120 promotes growth, migration and pluripotency. Receptor tyrosine kinase, PI3K/AKT, Rho GTPase, and HIPPO signaling, are all regulated by E-cadherin mediated cell-cell adhesion. Finally, the recruitment of the microprocessor complex to the ZA by PLEKHA7, and the subsequent regulation of a small subset of miRNAs provide an additional mechanism by which the state of epithelial cell-cell adhesion affects translation of target genes to maintain the homeostasis of polarized epithelial monolayers. Collectively, the data indicate that loss of E-cadherin function, especially at the ZA, is a common and crucial step in cancer progression.

Keywords: Cell-cell adhesion, E-cadherin, β-catenin, p120 catenin, Kaiso, Rho GTPases, EMT, Cancer progression, miRNA, PLEKHA7

Introduction

It is increasingly clear that traditional signaling pathways and mechanical forces converge at the cell-cell junctions to regulate the behavior of epithelial monolayers. The fact that the majority of human solid tumors are epithelial in origin has focused attention to the adhesion molecules at the junctions of epithelial cells and the signaling pathways involved in the maintenance of the epithelial phenotype. Cadherins, and their associated proteins, have emerged as key players in epithelial homeostasis and cancer.

The cadherin-catenin complex

Cadherins are cell surface glycoproteins with important functions in cell-cell adhesion, tissue pattering and cancer (for review, see13). Classical cadherins, one of the five classes of proteins containing cadherin repeats4, are a prominent class of adhesion molecules. Through their extracellular domains, they interact with cadherins on adjacent cells in a Ca++ dependent, homophilic manner, to form cell-cell adhesions called adherens junctions (AJs)5. Mature AJs form at apical regions of polarized epithelia, at the zonula adherens (ZA)6 (Figure 1). E-cadherin is a key component of the apical ZA in epithelial monolayers, and is considered a master regulator of the epithelial phenotype, due in part to the association of the ZA with a sub-membrane acto-myosin circumferential ring, which stabilizes the epithelial architecture7.

Figure 1.

Figure 1

Schematic diagram illustrating the main components of the cadherin-catenin complex at mature adherens junctions, and catenin-mediated signaling events to the nucleus. Under conditions of strong cell-cell adhesion, nuclear signaling by catenins (either β-catenin or p120) is suppressed. Upon activation of Wnt signaling, or under conditions that deregulate E-cadherin mediated adhesion (i.e. phosphorylation, endocytosis, loss of E-cadherin expression, etc.), β-catenin and p120 are free to bind their nuclear effectors. With the exception of Glis2, binding of p120 to Kaiso or REST/COREST prevents DNA binding and allows activation of target genes. Binding of β-catenin to Tcf/LEF alone, or combined with loss of Kaiso repressive activity, promotes the expression of Wnt target genes.

Classical cadherins contain a highly conserved cytoplasmic domain, which interacts with proteins that are collectively termed catenins. The related armadillo repeat proteins β-catenin (CTNNB1; mammalian homologue of Drosophila “armadillo”), or γ-catenin (also known as plakoglobin; JUP) bind to the cadherin carboxy terminal catenin-binding domain (CBD). Similarly, the membrane proximal cadherin juxtamembrane domain (JMD) interacts with members of the p120 catenin family of armadillo proteins, including p120 catenin (CTNND1; herein p120), NPRAP/δ-catenin (CTNND2), ARVCF, and p0071 (also known as plakophilin 4; PKP4)(for review see4). Through these interactions catenins regulate AJ function and stability. For example, β-catenin links cadherins to α-catenin, to promote the re-organization of the actin cytoskeleton812. Whether this reorganization is due to direct binding of actin filaments via α-catenin, via the regulation of monomeric versus dimeric α-catenin pools, tension-induced activation of α-catenin and vinculin, and/or via other actin binding β-catenin partners, like EPLIN, or ZO1, is still a mater of active investigation. Binding of β-catenin to the CBD is essential for cadherin function and for the maturation of AJs at areas of cell-cell contact. Consistent with the significance of the CBD in cadherin function, phosphorylation of either E-cadherin or β-catenin regulates β-catenin binding to the CBD, while phosphorylation of β-catenin regulates binding of α-catenin to the cadherin-β-catenin complex. Additionally, the CBD is thought to overlap, at least in part, with E-cadherin binding sites for the type Iγ PI phosphate kinase (PIPKIγ), and of the phosphatase PTPm (for review see4). Binding to the lipid kinase PIPKIγ promoted intracellular E-cadherin trafficking by engaging clathrin adaptor proteins13, while association with PTPm stabilized cadherin complexes on the cell membrane14.

p120 catenin (p120) interacts with and stabilizes cadherins at areas of cell-cell contact1520. Binding of the JMD to the E3 ligase Hakai21 and subsequent ubiquitination, prevents further p120 association and induces E-cadherin endocytosis and degradation2123. Alternatively, binding of presenilin to the JMD instead of p120, promotes cleavage of the cadherin cytoplasmic domain24. Therefore, the JMD plays a crucial role in cadherin function by regulating cadherin stability through its association with p120 family proteins or by promoting cadherin degradation and/or cleavage via its association with alternative binding partners. Additionally, p120 binds to microtubules and kinesins, regulating cadherin and p120 localization25,26, and also associates with PLEKHA7 at the apical ZA, which mediates interaction of the cadherin complex with non-centrosomal microtubules through the PLEKHA7 interacting protein Nezha27. Interestingly, p120 family members are expressed in several isoforms and are subject to heavy phosphorylation in both tyrosine and serine/threonine residues. P120 phosphorylation is now linked to inside-out cadherin signaling, where adhesive function of the cadherin complex is affected by p120 phosphorylation events28.

Another important aspect of p120 function is its ability to regulate the cytoskeleton via RhoGTPases29. P120 can bind directly to RhoA and suppress its activation in the cytoplasm or at the cleavage furrow during cytokinesis30,31, or it can recruit p190RhoGAP to cadherin complexes and suppress RhoA following Rac1 activation32. Binding of cytoplasmic p120 to MPRIP inhibits MPRIP-mediated suppression of Rho/ROCK function, leading to increased ROCK signaling33, while binding of p120 to ROCK1 recruits ROCK1 and junctional actin to newly formed AJs34. Finally, association of p120 to the RhoGEF Vav2 induces the activation of Rac1 and Cdc4235, which may account for the essential role of the JMD in Rac1 activation following the ligation of E-cadherin extracellular domains at early stages of cell-cell contact36. Importantly, through their association/dissociation with the cadherin cytoplasmic domain, β-catenin and p120 mediate a variety of signaling events that functionally link the cadherin-catenin complex to the cytoskeleton and ultimately regulate the balance between cell-cell adhesion and cell differentiation on the one hand, and cell growth and motility on the other.

Beta-catenin signaling in pluripotency and cancer

Armadillo, the Drosophila homologue of vertebrate β-catenin, was originally characterized by its role in the Wingless signaling pathway, which controls changes in gene expression that specify segment polarity and cell fate during Drosophila development (reviewed in37). In contrast, β-catenin in vertebrates was initially identified as a binding partner to E-cadherin at the AJs. It is now clear that Armadillo/β-catenin proteins play dual roles in regulating cell-cell adhesion and gene expression.

β-catenin consists of multiple armadillo repeats at its central region and a transcriptional activator domain at the C-terminal. The central region serves as a platform for binding to E-cadherin and other partners. In epithelial cells, the majority of β-catenin associates closely with cadherin complexes at the AJs. Phosphorylation regulates this association, releasing β-catenin into the cytoplasm where it interacts with a protein complex consisting of Axin, adenomatous polyposis coli (APC), and the Ser/Thr kinase glycogen synthase 3β (GSK-3β)(reviewed in38). In this “so-called” degradation pathway, GSK-3β (as well as casein kinases I and II) phosphorylates β-catenin, thereby inducing its ubiquitination and degradation in the 26S proteasome. The degradation pathway functions to keep cytoplasmic (and hence nuclear) levels of β-catenin low in the absence of Wnt signaling. Wnt (the vertebrate homolog of Wingless) binds its receptor Frizzled and activates Dishevelled (Dsh), which inhibits the Axin/APC/GSK-3β complex, thus preventing degradation of β-catenin. Intact cytoplasmic β-catenin can then translocate to the nucleus, where it binds to and activates transcription factors of the T cell factor/lymphoid enhancer factor (Tcf/Lef) family, thereby increasing transcription of target genes3941 (see figure 1).

Numerous studies indicate that nuclear β-catenin signaling is a major contributor to human cancer (reviewed in42). Initially, mutations in the APC gene were recognized as the predominant cause of hereditary colon cancer, by activating nuclear β-catenin signaling via the disruption of the β-catenin degradation pathway4345. Deregulation of Wnt/β-catenin signaling by genetic alterations of APC, β-catenin or Axin was shown to lead to colorectal cancer4649. Wnt/β-catenin signaling is also implicated in other cancers, such as leukemia, melanoma, hepatocellular, meduloblastoma, lymphoma, pancreas, and breast cancer (reviewed in42,50). Finally, elevated expression of Wnt proteins, or decreased expression of sFRP1 or DKK1, soluble proteins that inhibit Wnt function, is common in many human cancers, indicating that activation of Wnt/β-catenin signaling is a major pro-tumorigenic pathway in human disease42.

In addition to playing key roles in cancer progression, the β-catenin nuclear signaling pathway is crucial for the pluripotent phenotype and self-renewal of both normal and cancer stem cells51,52. Cancer stem cells are resistant to classic chemotherapy and radiotherapy, and as such, they are thought to contribute to the reappearance of cancer following treatment. While the mechanism of action is still unclear, nuclear β-catenin binds to the Telomerase reverse transcriptase (TERT) promoter and enhances TERT expression53,54. TERT maintains long telomeres and its up-regulation is a hallmark of stemness.

p120 nuclear signaling

Similar to β-catenin, p120 can also localize to the nucleus55 and effect changes in nuclear signaling (for review see56). Daniel and Reynolds originally reported the association of p120 with the transcription factor Kaiso57. More recently, additional interactions of p120 with the transcription factor Glis2, and the REST/CoREST complex have also been reported58,59.

Kaiso (ZBTB33) is a member of the BTB/POZ family of zinc finger transcription factors. p120 binds to a C-terminal region of Kaiso that overlaps the zinc finger domains, preventing association of Kaiso to DNA60. It is generally thought that Kaiso binds promoter regions via two different mechanisms: a specific DNA consensus sequence61, or through binding to methylated CpG dinucleotides62. Consistent with reported roles of the BTB/POZ family of transcription factors in repressing transcription, Kaiso represses transcription of Siamois, Matrilysin, Wnt11, through the recruitment of transcriptional co-repressors, like N-CoR56,60. While massively understudied compared to nuclear β-catenin signaling, available data suggest that Kaiso acts primarily as a transcriptional repressor and that p120 binding activates transcription of Kaiso targets via de-repression. In agreement, p120 overexpression reversed Kaiso-mediated reduction in Siamois reporter activity in Xenopus, while p120 depletion had the opposite effect63.

Increasing data support an overlap between the nuclear signaling roles of p120 and β-catenin56. Through binding to Kaiso, p120 is able to regulate gene targets, such as Siamois, CCND1, and MMP7, which are also regulated by β-catenin signaling. In this model, the nuclear translocation and transcriptional activation function of β-catenin is not sufficient to induce expression of its target genes, due to the repressive function of nearby Kaiso binding sites. Therefore, binding of p120 to Kaiso and subsequent de-repression of target genes maybe essential for the expression of at least some nuclear β-catenin targets. Interestingly, while p120 is not rapidly degraded when uncoupled from E-cadherin, Wnt signaling promotes p120 stability, via inhibition of G3SKβ-induced phosphorylation and degradation of p120 isoform 164, or via direct association of p120 with Frodo, a downstream target of Dsh65. Indeed, the increased binging of p120 to Kaiso may also lead to de-repression of the Kaiso target Wnt11, and promote further signaling through the non-canonical Wnt signaling pathway66.

In addition to Kaiso, p120 was shown to interact with Gli-similar 2 (Glis2), a Kruppel-like protein known to repress transcription67. p120 binding induced cleavage of Glis2, an effect that was enhanced by co-expression of p120 with the tyrosine kinase SRC and abrogated by co-expression of p120 with E-cadherin58. This suggests that p120 is able to bind and promote cleavage of Glis2 when it is not bound to E-cadherin. However, unlike the p120-Kaiso interaction, binding of p120 to Glis2 did not alter its ability to bind DNA58. At present, the functional significance of p120 binding to Glis2 is largely unknown, although this binding promotes nuclear translocation of p12058. Finally, p120 also associates with REST and CoREST, a complex involved in transcriptional repression of target genes and embryonic stem cell differentiation59. As with Kaiso, p120 binds to the DNA binding zinc finger region of REST and displaces the REST-CoREST complex from its gene targets. The interaction of p120 with REST is mediated by p120’s central Arm repeat region, and is exclusive of its association with E-cadherin. Additionally, p120 depletion resulted in increased expression of the pluripotency factors Nanog, Oct4 and Sox2, and a reduction of neuronal differentiation markers59, possibly through the REST-CoREST complex59.

While the mechanistic details are still under investigation, the data argue that E-cadherin binding to p120 prevents p120 from binding to Kaiso, Glis2, or to the REST-CoREST complex, and therefore blocks nuclear p120 functions58,59 (see figure 1). In addition to Wnt signals, it is likely that other mechanisms also regulate nuclear p120 signaling. The increased phosphorylation, cytoplasmic mislocalization, loss, or nuclear translocation of p120 in cancer, may therefore affect nuclear signaling, and play crucial roles in cancer progression.

Cadherin dysfunction in cancer

Normal cells inhibit their growth and migration when they adhere to each other. These properties are progressively lost in tumor cells, contributing to increased rates of cell proliferation and migration. The processes imply that adhesion-triggered signaling events regulate both cell growth and motility. Not surprisingly, E-cadherin loss is relatively common in cancers of epithelial origin.

Studies in a variety of cancers have documented E-cadherin’s role as a tumor suppressor68. Cancer-associated germline mutations in the E-cadherin gene have been reported in patients with hereditary diffuse gastric cancer69. Some of these mutations reportedly affect the adhesive state of the cadherin by interfering with inside-out cadherin signaling, a mechanism whereby binding of intracellular catenins and their phosphorylation status28 affects the overall conformation of the cadherin complex to induce strong adhesion70. Additionally, loss of heterozygosity in 16q22.1, the chromosomal region encoding E-cadherin (CDH1), is frequent in lobular carcinoma of the breast, a subtype characterized by single cell invasion71. A tumor suppressor role for E-cadherin has been established in many other epithelial malignancies, including hepatocellular carcinoma, squamous cell carcinomas of the skin, head and neck, esophagus, and melanoma (for review see4,68,72). In most of these cases, E-cadherin expression is downregulated by promoter hypermethylation73, while in others (i.e. lung cancer) E-cadherin is degraded as a result of the frequent loss of p120 expression74,75.

In experimental models, E-cadherin depletion leads to mesenchymal morphology and increased cell migration and invasion76,77. Conversely, ectopic expression of E-cadherin in cell lines lacking endogenous expression leads to reversal of undifferentiated, highly invasive carcinoma phenotypes7880. These effects may be mediated by many distinct functions of the E-cadherin complex, including the organization of epithelial cell architecture and polarity, and the regulation of signaling events driven by changes in the state of cell-cell adhesion.

As mentioned earlier, a major pathway related to tumor progression and pluripotency is the nuclear β-catenin/TCF signaling pathway. However, the relationship of E-cadherin mediated cell-cell adhesion and β-catenin signaling appears to be complex. E-cadherin loss by itself is insufficient to promote β-catenin nuclear signaling in the presence of an intact β-catenin degradation pathway. In contrast, when the degradation pathway is disrupted, increased expression of E-cadherin can provide a “sink” that affects nuclear β-catenin levels and counteracts β-catenin signaling (for review see72). This model is consistent with the early loss of APC function in colon cancer associated with increased β-catenin nuclear signaling and the formation of colon adenomas, followed by the later loss of E-cadherin, which is associated with carcinoma transition and metastatic spread. In pancreatic cancer, depletion of E-cadherin using the Rip-Tag system was associated with the transition from an adenoma to a carcinoma without involvement of β-catenin nuclear signaling77. It is likely that the role of E-cadherin in maintaining the proper structure and polarity of epithelial monolayers contributes to this transition, in addition to any effects on intracellular signaling.

In addition to β-catenin nuclear signaling, a number of oncogenic signaling pathways are known to be regulated by E-cadherin mediated cell-cell adhesion. These include, regulation of the overall levels of the cyclin kinase inhibitor p2781,82, activation of mitogen activated kinase (MAPK), rat sarcoma viral oncogene (Ras) and ras-related C3 botulinum toxin substrate (Rac1) signaling83,84, phosphatidylinositol-3-kinase (PI3K)/AKT signaling8587, as well as regulation of Hippo signaling88. While several potential mechanisms may underlie these effects, one likely explanation is that E-cadherin engagement during cell-cell adhesion suppresses signaling from a variety of receptor tyrosine kinases (RTKs), including EGF receptor (EGFR) and c-Met89, resulting in decreased rates of cell growth. This inhibition is thought to be mediated by direct binding of the E-cadherin extracellular domain to RTKs and the subsequent segregation of RTKs away from their ligands. Binding of cadherin complexes to phosphatases, i.e. PTPm, PTPj (otherwise known as density-enhanced phosphatase1), or PTPN14 is also thought to contribute to these events, by stabilizing cadherin complexes14, dephosphorylating catenins90,91, or by affecting YAP1 localization92 during cell adhesion. In the case of PI3K, direct recruitment of the p85 subunit to the cadherin complexes was demonstrated following phosphorylation by Src85,86, leading to AKT activation and adhesion-mediated cell survival85. Under normal conditions, cadherin-mediated PI3K signaling was required for calcium-induced phospholipase-C-γ1 activation and epidermal keratinocyte differentiation93. In ovarian cancer, the increased levels of E-cadherin expression directly contributed to the increased PI3K/AKT signaling of ovarian carcinoma cells87.

An important aspect of cadherin-mediated cell-cell adhesion is the regulation of Rho family GTPases94. p120 and the p120-binding JMD domain of E-cadherin are thought to mediate these effects29,94. Rho GTPases, including RhoA, Rac1 and Cdc42, act to reorganize the actin cytoskeleton and play crucial roles in cell-cell adhesion and cell migration95. Consistent with this, p120 was essential for the increased migration of cancer cells lacking E-cadherin expression and expressing mesenchymal cadherins, by regulating the activity of Rho family GTPases79,9698. Combined, the data argue that E-cadherin mediated AJs are hubs of intracellular signaling that regulate growth and migration of epithelial cells and are commonly deregulated in cancer.

Cadherins and EMT

Loss of E-cadherin expression during cancer progression is often the result of a phenomenon known as the epithelial-to-mesenchymal transition (EMT). EMT normally occurs during embryogenesis and development, enabling processes such as gastrulation, neural crest development, placenta formation and others. A key event during EMT is the loss of strong cell-cell adhesion due to suppression of E-cadherin expression and the subsequent transformation of epithelial cells into a more migratory and mesenchymal phenotype characterized by loose cell-cell contacts and by the expression of other types of cadherins, such as N-cadherin, R-cadherin, P-cadherin, or Cadherin-1199. This event is widely known as “cadherin switch” and is primarily the result of transcriptional regulation of cadherin expression through a number of factors, such as SNAI1, SLUG, TWIST, ZEB1, ZEB299,100. The main signaling cascades leading to transcriptional activation of EMT are the Wnt/β-catenin, TGF-β, and Hedgehog signaling pathways, which can act in response to a series of endocrine or paracrine cues99102. A number of post-transcriptional mechanisms are also involved in the induction of EMT, such as the proteasomal regulation of factors like SNAI1, or miRNA-mediated regulation103105. For example, the miR-200 and miR-205 miRNAs are key suppressors of ZEB1 and ZEB2 expression and thus negative regulators of EMT, whereas their downregulation is observed in tumors that have undergone EMT106,107. Several environmental factors can also induce EMT, such as increased hypoxia and subsequent activation of HIF-1a, which can upregulate TWIST and ZEB1108,109. Furthermore, the role of the extracellular matrix in the activation of EMT has been well-documented, mainly involving integrin signaling upstream or downstream of TGF-β and through signaling molecules such as ILK, FAK and SRC110.

Although an essential process during development or wound healing, the occurrence of EMT in mature epithelial tissues has been considered for a long time as a strong driver of tumor progression and metastasis. Indeed, downregulation of E-cadherin and upregulation of mesenchymal cadherins is frequent in tumors of epithelial origin and promotes cell migration and tumor invasiveness2,84,111113. Loss of E-cadherin can alleviate suppression of ligand-dependent RTK signaling89,114 and results in upregulation of the JUN oncogene via a so far undocumented post-transcriptional mechanism115. It also results in p120-mediated induction of Rac1 and cellular transformation83. Overall, p120 catenin has been recognized as an important rheostat in the regulation of downstream signaling and cell behavior upon a cadherin switch. Indeed, although p120 binding to E-cadherin is essential for maintenance of the epithelial phenotype and its tumor-suppressing functions18,116,117, its association with mesenchymal cadherins promotes p120’s Rac1-activating functions and subsequent induction of cell invasiveness118. In agreement, P-cadherin overexpression in pancreatic cancer cells induces p120-dependent activation of Rac1 and Cdc42119. Interestingly, p120 seems to be preferentially sequestered to ectopically overexpressed N-cadherin or R-cadherin, which results in subsequent endocytosis and degradation of endogenous E-cadherin and increased cell migration120122. E-cadherin and N-cadherin also associate with different p120 isoforms with critical functional consequences: N-cadherin associates with the long phosphorylated p120 isoform 1 that can promote invasiveness, whereas E-cadherin binds predominantly to the small non-phosphorylated p120 isoform 3 that lacks this function123,124.

Beyond the important role of p120 in mediating cadherin signaling, several other mechanisms of action have been reported downstream of mesenchymal cadherins during EMT that promote cellular transformation and pro-tumorigenic phenotypes. Cadherin-11 promotes lamellipodia and filopodia formation and cell migration via activation of the Trio GEF125. N-cadherin promotes anchorage-independent growth via loss of contact inhibition126 and protects from bile acid-induced apoptosis in hepatocellular carcinomas127. N-cadherin also facilitates FGF receptor and PDGF receptor activities, thus promoting cell motility and metastatic behavior128,129. Interestingly, N-cadherin enables stronger epithelial-endothelial cell interactions, which suggests for a role in promoting invasion and metastasis via the vasculature130. Similarly, an E-cadherin to N-cadherin switch in melanocytes results in reduced contacts between melanocytes and keratinocytes and increased melanocyte-fibroblast interactions that enable increased melanocytic growth and migration and decreased apoptosis via AKT activation131. P-cadherin overexpression leads to induction of MMP activity, which in turn results in P-cadherin cleavage and ectodomain fragments that promote cell invasion132. R-cadherin promotes cell motility via Rho GTPase activity133. Notably, the expression of mesenchymal cadherins is often sufficient for the induction of pro-tumorigenic cell behavior, even in the presence of E-cadherin122,132,134. Cumulatively, the role of cadherins is central during EMT and significantly determines intracellular and intercellular signaling, overall cell behavior, and tumor progression (see Figure 2).

Figure 2.

Figure 2

Schematic summarizing the role of cadherin complexes during pro-tumorigenic transformation. Normal epithelial cells primarily express E-cadherin, which is essential for mature monolayer formation. A number of cues can trigger EMT, which results in downregulation of E-cadherin, loss of the epithelial phenotype and upregulation of mesenchymal cadherins that promote migratory and invasive cell behavior. Alternatively, E-cadherin expression is maintained in many tumors and is required for their progression. This is due to the deregulation of the fine balance between an apical E-cadherin complex at the zonula adherens (ZA) that suppresses pro-tumorigenic signaling via miRNA processing, and a basolateral complex that promotes this signaling. Disruption of the ZA-specific complex in cancer results in miRNA deregulation and increased anchorage-independent growth, which depends on the presence and activity of the basolateral E-cadherin complex.

Beyond EMT: cadherin complexes and the RNAi machinery

The loss of E-cadherin during EMT is generally considered a major driver of cancer progression and metastasis. However, recent studies argue that EMT is actually not required for tumorigenic transformation or metastasis135,136. In addition, E-cadherin expression is robust in many cancer types, even at metastatic sites137142. Strikingly, the presence of an E-cadherin-based junctional complex is essential in these cancer types for: a) the transmission of pro-tumorigenic signals stemming from oncogenes such as SRC, Rac1, EGFR, ERBB2143146, b) collective cell migration147,148, c) cell proliferation under conditions of high confluence146, and d) increased anchorage-independent growth and chemoresistance in Ewing sarcomas149. Paradoxically, E-cadherin is essential for the progression of some very aggressive tumors, such as inflammatory breast cancer and a subtype of glioblastoma139,150. These results indicated that in addition to its well-established tumor suppressive functions, E-cadherin can also promote tumor progression in many cases140.

To resolve this conundrum, a recent study tested the hypothesis that the presence or absence of accessory junctional components eventually determines E-cadherin function. Indeed, it appears that E-cadherin forms two distinct complexes at the junctions of non-transformed polarized epithelial cells: one restricted to the apical junctions at the ZA, and another along the basolateral membrane in these cells151,152. The apical complex is characterized by the presence of the p120-binding partner PLEKHA7, as well as by non-phosphorylated p120 and SRC, and activated RhoA, whereas the basolateral lacks PLEKHA7 and contains phosphorylated p120, activated Rac1 and SRC, p190RhoGAP and p130CAS152. The presence of PLEKHA7 at the apical cadherin complex confers the anti-tumorigenic properties of E-cadherin152. More specifically, loss of PLEKHA7 promoted anchorage-independent growth, induced activation of SRC and phosphorylation of p120, and increased the expression of a series of pro-tumorigenic markers, such as SNAI1, Cadherin-11, MYC and CCND1152. Notably, these events were not followed by an EMT, since E-cadherin levels remained unaltered. Furthermore, the presence of the basolateral E-cadherin complex was required for the induction of these pro-tumorigenic signals, via SRC activation and p120 phosphorylation152. Consistent with these results, this and other studies have shown extensive and early loss of PLEKHA7 in breast and kidney tumors, whereas E-cadherin was still expressed to a large extend and p120 phosphorylation was also increased in tumors152154. Collectively, these findings provided an explanation for the conflicting reports regarding the role of E-cadherin in anti- vs. pro-tumorigenic signaling.

Unexpectedly, mechanistic studies revealed that the apical E-cadherin complex suppresses pro-tumorigenic signals by recruiting the microprocessor complex, one of the main components of the RNA interference (RNAi) machinery152,155. In particular, PLEKHA7 mediates the association of non-nuclear fractions of the core members of the microprocessor DROSHA and DGCR8, as well as of a set of primary miRNAs (pri-miRNAs), which are the substrates of the complex, at the apical ZA152 (Figure 2). As a result, the apical cadherin complex possesses miRNA processing activity and regulates the levels of a set of miRNAs with tumor suppressing function, such as miR-30b, which eventually mediates the PLEKHA7-driven suppression of SNAI1152. This was the first demonstration of any interaction of junctional complexes with the RNAi machinery and particularly with the microprocessor complex. Previously thought to reside solely in the nucleus156,157, these findings revealed a non-nuclear microprocessor at the junctions of polarized epithelial cells155, as part of the E-cadherin complex A possible reason that the junctional presence of the microprocessor wasn’t previously noticed is that all microprocessor-related studies were performed in cells that have undergone some degree of tumorigenic transformation and therefore lack mature AJs. PLEKHA7 is required for the recruitment of DROSHA and DGCR8 to the junctions and SRC activity negatively affects localization of PLEKHA7, DROSHA and DGCR8 to the ZA152, which is consistent with the well-established function of SRC in perturbing strong adhesion and mature junction formation158,159. Therefore, the above suggest that the microprocessor may be junctional only in non-transformed epithelial cells that normally localize PLEKHA7 at the ZA and form mature AJs (Figure 2). Further investigation of the localization patterns of DROSHA and DGCR8 in a broad range of cells and tissues would be important to confirm this notion and further investigate this new role of AJs in tumor progression.

Apart from the association with the microprocessor complex, proteomics analysis revealed a preferential enrichment of the apical cadherin complex with several additional RNA-binding proteins152. The data implied that the functional crosstalk of the apical cadherin complex with mechanisms involved in RNA-processing and expression may be extensive and going beyond the relationship with the microprocessor. This new direction of investigation could define the functional relevance of RNA regulation at the junctions, shed more light on the complicated roles of cadherin complexes in cancer, and potentially provide new targets for therapeutic intervention155.

Summary

Combined, existing data indicate that the E-cadherin/catenin complex is essential for the homeostasis and maintenance of epithelial monolayers. As most solid tumors are epithelial in origin, deregulation of E-cadherin complexes has long been thought to play important roles in cancer progression. E-cadherin loss, mutation, or destabilization through loss of p120 binding, all contribute to cancer progression. Similarly, nuclear β-catenin and p120 signaling promote cell growth, migration and pluripotency. EMT provides one mechanism of overcoming the tumor-suppressing function of E-cadherin complexes. Another mechanism is the disruption of the crosstalk between mature AJs and the RNAi machinery. Indeed, loss of mature AJs at the apical ZA, through disruption of PLEKHA7 binding to cadherin complexes, uncovers a tumor promoting function of E-cadherin. Therefore, it is not simply the presence but also the association of E-cadherin with key accessory proteins and the formation of mature junctions that underlie its ability to act as a tumor suppressor. Collectively, it is clear that loss or disruption of mature AJs at the apical ZA occurs by a multitude of mechanisms, and the resulting loss of anti-tumorigenic E-cadherin signaling combined with the gain of nuclear catenin signaling and the activation of various additional pathways (RTK, Rho GTPases, PI3K, Hippo), are central events in tumor progression and metastasis.

Highlights.

  • General review of the cadherin-catenin complex

  • Nuclear signaling events regulated by β-catenin, p120, and E-cadherin

  • Cadherin dysfunction in cancer

  • Crosstalk between cadherin complexes and the RNAi machinery

Acknowledgments

Funding: This work was supported by the National Institutes of Health (R01 CA100467, R01 NS069753, P50 CA116201) and the Mayo Clinic Center for Biomedical Discovery (PZA). AK is supported by the Abney Foundation Scholarship Award and by pilot research funding from an American Cancer Society Institutional Research Grant awarded to the Hollings Cancer Center, Medical University of South Carolina.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Takeichi M. Morphogenetic roles of classic cadherins. Curr Opin in Cell Biol. 1995;7:619–627. doi: 10.1016/0955-0674(95)80102-2. [DOI] [PubMed] [Google Scholar]
  • 2.Nollet F, Berx G, van Roy F. The role of the E-cadherin/catenin adhesion complex in the development and progression of cancer. Mol Cell Biol Res Commun. 1999;2:77–85. doi: 10.1006/mcbr.1999.0155. [DOI] [PubMed] [Google Scholar]
  • 3.Yap AS. The morphogenetic role of cadherin cell adhesion molecules in human cancer: a thematic review. Cancer Invest. 1998;16:252–261. doi: 10.3109/07357909809039774. [DOI] [PubMed] [Google Scholar]
  • 4.van Roy F, Berx G. The cell-cell adhesion molecule E-cadherin. Cellular and molecular life sciences: CMLS. 2008;65:3756–3788. doi: 10.1007/s00018-008-8281-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Takeichi M. Cadherin cell adhesion receptors as a morphogenetic regulator. Science. 1991;251:1451–1455. doi: 10.1126/science.2006419. [DOI] [PubMed] [Google Scholar]
  • 6.Nishimura T, Takeichi M. Remodeling of the adherens junctions during morphogenesis. Curr Top Dev Biol. 2009;89:33–54. doi: 10.1016/S0070-2153(09)89002-9. [DOI] [PubMed] [Google Scholar]
  • 7.Miyoshi J, Takai Y. Structural and functional associations of apical junctions with cytoskeleton. Biochim Biophys Acta. 2008;1778:670–691. doi: 10.1016/j.bbamem.2007.12.014. [DOI] [PubMed] [Google Scholar]
  • 8.Herrenknecht K, et al. The uvomorulin-anchorage protein alpha catenin is a vinculin homologue. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:9156–9160. doi: 10.1073/pnas.88.20.9156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nagafuchi A, Takeichi M, Tsukita S. The 102 kd cadherin-associated protein: similarity to vinculin and posttranscriptional regulation of expression. Cell. 1991;65:849–857. doi: 10.1016/0092-8674(91)90392-c. [DOI] [PubMed] [Google Scholar]
  • 10.Rimm DL, Koslov ER, Kebriaei P, Cianci CD, Morrow JS. Alpha 1(E)-catenin is an actin-binding and -bundling protein mediating the attachment of F-actin to the membrane adhesion complex. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:8813–8817. doi: 10.1073/pnas.92.19.8813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Drees F, Pokutta S, Yamada S, Nelson WJ, Weis WI. Alpha-catenin is a molecular switch that binds E-cadherin-beta-catenin and regulates actin-filament assembly. Cell. 2005;123:903–915. doi: 10.1016/j.cell.2005.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bertocchi C, et al. Nanoscale architecture of cadherin-based cell adhesions. Nat Cell Biol. 2017;19:28–37. doi: 10.1038/ncb3456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ling K, et al. Type I gamma phosphatidylinositol phosphate kinase modulates adherens junction and E-cadherin trafficking via a direct interaction with mu 1B adaptin. The Journal of cell biology. 2007;176:343–353. doi: 10.1083/jcb.200606023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Brady-Kalnay SM, Rimm DL, Tonks NK. Receptor Protein Tyrosine Phosphatase PTPm Associates with Cadherins and Catenins In Vivo. J Cell Biol. 1995;130:977–986. doi: 10.1083/jcb.130.4.977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ireton RC, et al. A novel role for p120 catenin in E-cadherin function. The Journal of cell biology. 2002;159:465–476. doi: 10.1083/jcb.200205115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Davis MA, Ireton RC, Reynolds AB. A core function for p120-catenin in cadherin turnover. The Journal of cell biology. 2003;163:525–534. doi: 10.1083/jcb.200307111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Xiao K, et al. Cellular levels of p120 catenin function as a set point for cadherin expression levels in microvascular endothelial cells. The Journal of cell biology. 2003;163:535–545. doi: 10.1083/jcb.200306001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Davis MA, Reynolds AB. Blocked acinar development, E-cadherin reduction, and intraepithelial neoplasia upon ablation of p120-catenin in the mouse salivary gland. Dev Cell. 2006;10:21–31. doi: 10.1016/j.devcel.2005.12.004. [DOI] [PubMed] [Google Scholar]
  • 19.Ireton RC, et al. A novel role for p120 catenin in E-cadherin function. The Journal of cell biology. 2002;159:465–476. doi: 10.1083/jcb.200205115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kourtidis A, Ngok SP, Anastasiadis PZ. p120 catenin: an essential regulator of cadherin stability, adhesion-induced signaling, and cancer progression. Prog Mol Biol Transl Sci. 2013;116:409–432. doi: 10.1016/B978-0-12-394311-8.00018-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fujita Y, et al. Hakai, a c-Cbl-like protein, ubiquitinates and induces endocytosis of the E-cadherin complex. Nat Cell Biol. 2002;4:222–231. doi: 10.1038/ncb758. [DOI] [PubMed] [Google Scholar]
  • 22.Hartsock A, Nelson WJ. Competitive regulation of E-cadherin juxtamembrane domain degradation by p120-catenin binding and Hakai-mediated ubiquitination. PloS one. 2012;7:e37476. doi: 10.1371/journal.pone.0037476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Shrestha H, et al. Hakai, an E3-ligase for E-cadherin, stabilizes delta-catenin through Src kinase. Cell Signal. 2017;31:135–145. doi: 10.1016/j.cellsig.2017.01.009. [DOI] [PubMed] [Google Scholar]
  • 24.Baki L, et al. Presenilin-1 binds cytoplasmic epithelial cadherin, inhibits cadherin/p120 association, and regulates stability and function of the cadherin/catenin adhesion complex. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:2381–2386. doi: 10.1073/pnas.041603398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yanagisawa M, et al. A novel interaction between kinesin and p120 modulates p120 localization and function. The Journal of biological chemistry. 2004;279:9512–9521. doi: 10.1074/jbc.M310895200. [DOI] [PubMed] [Google Scholar]
  • 26.Chen X, Kojima S, Borisy GG, Green KJ. p120 catenin associates with kinesin and facilitates the transport of cadherin-catenin complexes to intercellular junctions. The Journal of cell biology. 2003;163:547–557. doi: 10.1083/jcb.200305137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Meng W, Mushika Y, Ichii T, Takeichi M. Anchorage of microtubule minus ends to adherens junctions regulates epithelial cell-cell contacts. Cell. 2008;135:948–959. doi: 10.1016/j.cell.2008.09.040. [DOI] [PubMed] [Google Scholar]
  • 28.Petrova YI, Spano MM, Gumbiner BM. Conformational epitopes at cadherin calcium-binding sites and p120-catenin phosphorylation regulate cell adhesion. Mol Biol Cell. 2012;23:2092–2108. doi: 10.1091/mbc.E11-12-1060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Anastasiadis PZ. p120-ctn: A nexus for contextual signaling via Rho GTPases. Biochim Biophys Acta. 2007;1773:34–46. doi: 10.1016/j.bbamcr.2006.08.040. [DOI] [PubMed] [Google Scholar]
  • 30.Anastasiadis PZ, et al. Inhibition of RhoA by p120 catenin. Nat Cell Biol. 2000;2:637–644. doi: 10.1038/35023588. [DOI] [PubMed] [Google Scholar]
  • 31.van de Ven RA, et al. p120-catenin prevents multinucleation through control of MKLP1-dependent RhoA activity during cytokinesis. Nature communications. 2016;7:13874. doi: 10.1038/ncomms13874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wildenberg GA, et al. p120-catenin and p190RhoGAP regulate cell-cell adhesion by coordinating antagonism between Rac and Rho. Cell. 2006;127:1027–1039. doi: 10.1016/j.cell.2006.09.046. [DOI] [PubMed] [Google Scholar]
  • 33.Schackmann RC, et al. Cytosolic p120-catenin regulates growth of metastatic lobular carcinoma through Rock1-mediated anoikis resistance. The Journal of clinical investigation. 2011;121:3176–3188. doi: 10.1172/JCI41695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Smith AL, Dohn MR, Brown MV, Reynolds AB. Association of Rho-associated protein kinase 1 with E-cadherin complexes is mediated by p120-catenin. Mol Biol Cell. 2012;23:99–110. doi: 10.1091/mbc.E11-06-0497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Noren NK, Liu BP, Burridge K, Kreft B. p120 catenin regulates the actin cytoskeleton via Rho family GTPases. The Journal of cell biology. 2000;150:567–580. doi: 10.1083/jcb.150.3.567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Goodwin M, Kovacs EM, Thoreson MA, Reynolds AB, Yap AS. Minimal mutation of the cytoplasmic tail inhibits the ability of E-cadherin to activate Rac but not phosphatidylinositol 3-kinase: direct evidence of a role for cadherin-activated Rac signaling in adhesion and contact formation. The Journal of biological chemistry. 2003;278:20533–20539. doi: 10.1074/jbc.M213171200. [DOI] [PubMed] [Google Scholar]
  • 37.Peifer M. Cell adhesion and signal transduction: the Armadillo connection. Trends Cell Biol. 1995;5:224–229. doi: 10.1016/s0962-8924(00)89015-7. [DOI] [PubMed] [Google Scholar]
  • 38.Polakis P. Wnt signaling and cancer. Genes Dev. 2000;14:1837–1851. [PubMed] [Google Scholar]
  • 39.MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009;17:9–26. doi: 10.1016/j.devcel.2009.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Heuberger J, Birchmeier W. Interplay of cadherin-mediated cell adhesion and canonical Wnt signaling. Cold Spring Harbor perspectives in biology. 2010;2:a002915. doi: 10.1101/cshperspect.a002915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127:469–480. doi: 10.1016/j.cell.2006.10.018. [DOI] [PubMed] [Google Scholar]
  • 42.Polakis P. Wnt signaling in cancer. Cold Spring Harbor perspectives in biology. 2012;4 doi: 10.1101/cshperspect.a008052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kinzler KW, et al. Identification of a gene located at chromosome 5q21 that is mutated in colorectal cancers. Science. 1991;251:1366–1370. doi: 10.1126/science.1848370. [DOI] [PubMed] [Google Scholar]
  • 44.Kinzler KW, et al. Identification of FAP locus genes from chromosome 5q21. Science. 1991;253:661–665. doi: 10.1126/science.1651562. [DOI] [PubMed] [Google Scholar]
  • 45.Nakamura Y, et al. Mutations of the adenomatous polyposis coli gene in familial polyposis coli patients and sporadic colorectal tumors. Princess Takamatsu Symp. 1991;22:285–292. [PubMed] [Google Scholar]
  • 46.Morin PJ, et al. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science. 1997;275:1787–1790. doi: 10.1126/science.275.5307.1787. [DOI] [PubMed] [Google Scholar]
  • 47.Clements WM, et al. beta-Catenin mutation is a frequent cause of Wnt pathway activation in gastric cancer. Cancer Res. 2002;62:3503–3506. [PubMed] [Google Scholar]
  • 48.Korinek V, et al. Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC−/− colon carcinoma. Science. 1997;275:1784–1787. doi: 10.1126/science.275.5307.1784. [DOI] [PubMed] [Google Scholar]
  • 49.Liu W, et al. Mutations in AXIN2 cause colorectal cancer with defective mismatch repair by activating beta-catenin/TCF signalling. Nat Genet. 2000;26:146–147. doi: 10.1038/79859. [DOI] [PubMed] [Google Scholar]
  • 50.Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene. 2016 doi: 10.1038/onc.2016.304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Miki T, Yasuda SY, Kahn M. Wnt/beta-catenin signaling in embryonic stem cell self-renewal and somatic cell reprogramming. Stem cell reviews. 2011;7:836–846. doi: 10.1007/s12015-011-9275-1. [DOI] [PubMed] [Google Scholar]
  • 52.Yang K, et al. The evolving roles of canonical WNT signaling in stem cells and tumorigenesis: implications in targeted cancer therapies. Laboratory investigation; a journal of technical methods and pathology. 2016;96:116–136. doi: 10.1038/labinvest.2015.144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Peifer M, et al. Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature. 2015;526:700–704. doi: 10.1038/nature14980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Park JI, et al. Telomerase modulates Wnt signalling by association with target gene chromatin. Nature. 2009;460:66–72. doi: 10.1038/nature08137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.van Hengel J, Vanhoenacker P, Staes K, van Roy F. Nuclear localization of the p120(ctn) armadillo-like catenin is counteracted by a nuclear export signal and by E-cadherin expression. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:7980–7985. doi: 10.1073/pnas.96.14.7980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.McCrea PD, Maher MT, Gottardi CJ. Nuclear signaling from cadherin adhesion complexes. Curr Top Dev Biol. 2015;112:129–196. doi: 10.1016/bs.ctdb.2014.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Daniel JM, Reynolds AB. The catenin p120(ctn) interacts with Kaiso, a novel BTB/POZ domain zinc finger transcription factor. Mol Cell Biol. 1999;19:3614–3623. doi: 10.1128/mcb.19.5.3614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hosking CR, et al. The transcriptional repressor Glis2 is a novel binding partner for p120 catenin. Mol Biol Cell. 2007;18:1918–1927. doi: 10.1091/mbc.E06-10-0941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lee M, Ji H, Furuta Y, Park JI, McCrea PD. p120-catenin regulates REST and CoREST, and modulates mouse embryonic stem cell differentiation. J Cell Sci. 2014;127:4037–4051. doi: 10.1242/jcs.151944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Daniel JM. Dancing in and out of the nucleus: p120(ctn) and the transcription factor Kaiso. Biochim Biophys Acta. 2007;1773:59–68. doi: 10.1016/j.bbamcr.2006.08.052. [DOI] [PubMed] [Google Scholar]
  • 61.Daniel JM, Spring CM, Crawford HC, Reynolds AB, Baig A. The p120(ctn)-binding partner Kaiso is a bi-modal DNA-binding protein that recognizes both a sequence-specific consensus and methylated CpG dinucleotides. Nucleic Acids Res. 2002;30:2911–2919. doi: 10.1093/nar/gkf398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Prokhortchouk A, et al. The p120 catenin partner Kaiso is a DNA methylation-dependent transcriptional repressor. Genes Dev. 2001;15:1613–1618. doi: 10.1101/gad.198501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Park JI, et al. Kaiso/p120-catenin and TCF/beta-catenin complexes coordinately regulate canonical Wnt gene targets. Dev Cell. 2005;8:843–854. doi: 10.1016/j.devcel.2005.04.010. [DOI] [PubMed] [Google Scholar]
  • 64.Hong JY, et al. Shared molecular mechanisms regulate multiple catenin proteins: canonical Wnt signals and components modulate p120-catenin isoform-1 and additional p120 subfamily members. J Cell Sci. 2010;123:4351–4365. doi: 10.1242/jcs.067199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Park JI, et al. Frodo links Dishevelled to the p120-catenin/Kaiso pathway: distinct catenin subfamilies promote Wnt signals. Dev Cell. 2006;11:683–695. doi: 10.1016/j.devcel.2006.09.022. [DOI] [PubMed] [Google Scholar]
  • 66.Kim SW, et al. Non-canonical Wnt signals are modulated by the Kaiso transcriptional repressor and p120-catenin. Nat Cell Biol. 2004;6:1212–1220. doi: 10.1038/ncb1191. [DOI] [PubMed] [Google Scholar]
  • 67.Zhang F, et al. Characterization of Glis2, a novel gene encoding a Gli-related, Kruppel-like transcription factor with transactivation and repressor functions. Roles in kidney development and neurogenesis. J Biol Chem. 2002;277:10139–10149. doi: 10.1074/jbc.M108062200. [DOI] [PubMed] [Google Scholar]
  • 68.Berx G, van Roy F. Involvement of members of the cadherin superfamily in cancer. Cold Spring Harbor perspectives in biology. 2009;1:a003129. doi: 10.1101/cshperspect.a003129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Pinheiro H, et al. Allele-specific CDH1 downregulation and hereditary diffuse gastric cancer. Hum Mol Genet. 2010;19:943–952. doi: 10.1093/hmg/ddp537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Petrova YI, Schecterson L, Gumbiner BM. Roles for E-cadherin cell surface regulation in cancer. Mol Biol Cell. 2016;27:3233–3244. doi: 10.1091/mbc.E16-01-0058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Berx G, Van Roy F. The E-cadherin/catenin complex: an important gatekeeper in breast cancer tumorigenesis and malignant progression. Breast cancer research: BCR. 2001;3:289–293. doi: 10.1186/bcr309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jeanes A, Gottardi CJ, Yap AS. Cadherins and cancer: how does cadherin dysfunction promote tumor progression? Oncogene. 2008;27:6920–6929. doi: 10.1038/onc.2008.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Winter JM, et al. Absence of E-cadherin expression distinguishes noncohesive from cohesive pancreatic cancer. Clin Cancer Res. 2008;14:412–418. doi: 10.1158/1078-0432.CCR-07-0487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Liu Y, et al. Abnormal expression of p120-catenin, E-cadherin, and small GTPases is significantly associated with malignant phenotype of human lung cancer. Lung Cancer. 2009;63:375–382. doi: 10.1016/j.lungcan.2008.12.012. [DOI] [PubMed] [Google Scholar]
  • 75.Mortazavi F, An J, Dubinett S, Rettig M. p120-catenin is transcriptionally downregulated by FOXC2 in non-small cell lung cancer cells. Mol Cancer Res. 2010;8:762–774. doi: 10.1158/1541-7786.MCR-10-0004. [DOI] [PubMed] [Google Scholar]
  • 76.Vleminckx K, Vakaet L, Jr, Mareel M, Fiers W, van Roy F. Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 1991;66:107–119. doi: 10.1016/0092-8674(91)90143-m. [DOI] [PubMed] [Google Scholar]
  • 77.Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G. A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature. 1998;392:190–193. doi: 10.1038/32433. [DOI] [PubMed] [Google Scholar]
  • 78.Frixen UH, et al. E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. The Journal of cell biology. 1991;113:173–185. doi: 10.1083/jcb.113.1.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Yanagisawa M, Anastasiadis PZ. p120 catenin is essential for mesenchymal cadherin-mediated regulation of cell motility and invasiveness. The Journal of cell biology. 2006;174:1087–1096. doi: 10.1083/jcb.200605022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wong AS, Gumbiner BM. Adhesion-independent mechanism for suppression of tumor cell invasion by E-cadherin. The Journal of cell biology. 2003;161:1191–1203. doi: 10.1083/jcb.200212033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Motti ML, et al. Reduced E-cadherin expression contributes to the loss of p27kip1-mediated mechanism of contact inhibition in thyroid anaplastic carcinomas. Carcinogenesis. 2005;26:1021–1034. doi: 10.1093/carcin/bgi050. [DOI] [PubMed] [Google Scholar]
  • 82.St Croix B, et al. E-Cadherin-dependent growth suppression is mediated by the cyclin- dependent kinase inhibitor p27(KIP1) The Journal of cell biology. 1998;142:557–571. doi: 10.1083/jcb.142.2.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Soto E, et al. p120 catenin induces opposing effects on tumor cell growth depending on E-cadherin expression. The Journal of cell biology. 2008;183:737–749. doi: 10.1083/jcb.200805113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wijnhoven BP, Dinjens WN, Pignatelli M. E-cadherin-catenin cell-cell adhesion complex and human cancer. Br J Surg. 2000;87:992–1005. doi: 10.1046/j.1365-2168.2000.01513.x. [DOI] [PubMed] [Google Scholar]
  • 85.Pece S, Chiariello M, Murga C, Gutkind JS. Activation of the protein kinase Akt/PKB by the formation of E-cadherin-mediated cell-cell junctions. Evidence for the association of phosphatidylinositol 3-kinase with the E-cadherin adhesion complex. The Journal of biological chemistry. 1999;274:19347–19351. doi: 10.1074/jbc.274.27.19347. [DOI] [PubMed] [Google Scholar]
  • 86.Pang JH, Kraemer A, Stehbens SJ, Frame MC, Yap AS. Recruitment of phosphoinositide 3-kinase defines a positive contribution of tyrosine kinase signaling to E-cadherin function. The Journal of biological chemistry. 2005;280:3043–3050. doi: 10.1074/jbc.M412148200. [DOI] [PubMed] [Google Scholar]
  • 87.De Santis G, Miotti S, Mazzi M, Canevari S, Tomassetti A. E-cadherin directly contributes to PI3K/AKT activation by engaging the PI3K-p85 regulatory subunit to adherens junctions of ovarian carcinoma cells. Oncogene. 2009;28:1206–1217. doi: 10.1038/onc.2008.470. [DOI] [PubMed] [Google Scholar]
  • 88.Kim NG, Koh E, Chen X, Gumbiner BM. E-cadherin mediates contact inhibition of proliferation through Hippo signaling-pathway components. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:11930–11935. doi: 10.1073/pnas.1103345108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Qian X, Karpova T, Sheppard AM, McNally J, Lowy DR. E-cadherin-mediated adhesion inhibits ligand-dependent activation of diverse receptor tyrosine kinases. Embo J. 2004;23:1739–1748. doi: 10.1038/sj.emboj.7600136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Holsinger LJ, Ward K, Duffield B, Zachwieja J, Jallal B. The transmembrane receptor protein tyrosine phosphatase DEP1 interacts with p120(ctn) Oncogene. 2002;21:7067–7076. doi: 10.1038/sj.onc.1205858. [DOI] [PubMed] [Google Scholar]
  • 91.Wadham C, Gamble JR, Vadas MA, Khew-Goodall Y. The protein tyrosine phosphatase Pez is a major phosphatase of adherens junctions and dephosphorylates beta-catenin. Mol Biol Cell. 2003;14:2520–2529. doi: 10.1091/mbc.E02-09-0577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wang W, et al. PTPN14 is required for the density-dependent control of YAP1. Genes Dev. 2012;26:1959–1971. doi: 10.1101/gad.192955.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Xie Z, Bikle DD. The recruitment of phosphatidylinositol 3-kinase to the E-cadherin-catenin complex at the plasma membrane is required for calcium-induced phospholipase C-gamma1 activation and human keratinocyte differentiation. The Journal of biological chemistry. 2007;282:8695–8703. doi: 10.1074/jbc.M609135200. [DOI] [PubMed] [Google Scholar]
  • 94.Ratheesh A, Priya R, Yap AS. Coordinating Rho and Rac: the regulation of Rho GTPase signaling and cadherin junctions. Progress in molecular biology and translational science. 2013;116:49–68. doi: 10.1016/B978-0-12-394311-8.00003-0. [DOI] [PubMed] [Google Scholar]
  • 95.Hall A. Rho GTPases and the control of cell behaviour. Biochemical Society transactions. 2005;33:891–895. doi: 10.1042/BST20050891. [DOI] [PubMed] [Google Scholar]
  • 96.Molina-Ortiz I, Bartolome RA, Hernandez-Varas P, Colo GP, Teixido J. Overexpression of E-cadherin on melanoma cells inhibits chemokine-promoted invasion involving p190RhoGAP/p120ctn-dependent inactivation of RhoA. The Journal of biological chemistry. 2009;284:15147–15157. doi: 10.1074/jbc.M807834200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Grosheva I, Shtutman M, Elbaum M, Bershadsky AD. p120 catenin affects cell motility via modulation of activity of Rho-family GTPases: a link between cell-cell contact formation and regulation of cell locomotion. J Cell Sci. 2001;114:695–707. doi: 10.1242/jcs.114.4.695. [DOI] [PubMed] [Google Scholar]
  • 98.Bellovin DI, Bates RC, Muzikansky A, Rimm DL, Mercurio AM. Altered localization of p120 catenin during epithelial to mesenchymal transition of colon carcinoma is prognostic for aggressive disease. Cancer research. 2005;65:10938–10945. doi: 10.1158/0008-5472.CAN-05-1947. [DOI] [PubMed] [Google Scholar]
  • 99.Wheelock MJ, Shintani Y, Maeda M, Fukumoto Y, Johnson KR. Cadherin switching. J Cell Sci. 2008;121:727–735. doi: 10.1242/jcs.000455. [DOI] [PubMed] [Google Scholar]
  • 100.Tam WL, Weinberg RA. The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat Med. 2013;19:1438–1449. doi: 10.1038/nm.3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Katoh Y, Katoh M. Hedgehog signaling, epithelial-to-mesenchymal transition and miRNA (review) International journal of molecular medicine. 2008;22:271–275. [PubMed] [Google Scholar]
  • 102.Scheel C, et al. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell. 2011;145:926–940. doi: 10.1016/j.cell.2011.04.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Serrano-Gomez SJ, Maziveyi M, Alahari SK. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Molecular cancer. 2016;15:18. doi: 10.1186/s12943-016-0502-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Abba ML, Patil N, Leupold JH, Allgayer H. MicroRNA Regulation of Epithelial to Mesenchymal Transition. J Clin Med. 2016;5 doi: 10.3390/jcm5010008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Gregory PA, Bracken CP, Bert AG, Goodall GJ. MicroRNAs as regulators of epithelial-mesenchymal transition. Cell cycle (Georgetown, Tex) 2008;7:3112–3118. doi: 10.4161/cc.7.20.6851. [DOI] [PubMed] [Google Scholar]
  • 106.Gregory PA, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nature cell biology. 2008;10:593–601. doi: 10.1038/ncb1722. [DOI] [PubMed] [Google Scholar]
  • 107.Gibbons DL, et al. Contextual extracellular cues promote tumor cell EMT and metastasis by regulating miR-200 family expression. Genes & development. 2009;23:2140–2151. doi: 10.1101/gad.1820209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Sun S, et al. Hypoxia-inducible factor-1alpha induces Twist expression in tubular epithelial cells subjected to hypoxia, leading to epithelial-to-mesenchymal transition. Kidney Int. 2009;75:1278–1287. doi: 10.1038/ki.2009.62. [DOI] [PubMed] [Google Scholar]
  • 109.Joseph JV, et al. Hypoxia enhances migration and invasion in glioblastoma by promoting a mesenchymal shift mediated by the HIF1alpha-ZEB1 axis. Cancer letters. 2015;359:107–116. doi: 10.1016/j.canlet.2015.01.010. [DOI] [PubMed] [Google Scholar]
  • 110.Cichon MA, Radisky DC. Extracellular matrix as a contextual determinant of transforming growth factor-beta signaling in epithelial-mesenchymal transition and in cancer. Cell adhesion & migration. 2014;8:588–594. doi: 10.4161/19336918.2014.972788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pishvaian MJ, et al. Cadherin-11 is expressed in invasive breast cancer cell lines. Cancer research. 1999;59:947–952. [PubMed] [Google Scholar]
  • 112.Sarrio D, et al. Cytoplasmic localization of p120ctn and E-cadherin loss characterize lobular breast carcinoma from preinvasive to metastatic lesions. Oncogene. 2004;23:3272–3283. doi: 10.1038/sj.onc.1207439. [DOI] [PubMed] [Google Scholar]
  • 113.Gravdal K, Halvorsen OJ, Haukaas SA, Akslen LA. A switch from E-cadherin to N-cadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer. Clin Cancer Res. 2007;13:7003–7011. doi: 10.1158/1078-0432.CCR-07-1263. [DOI] [PubMed] [Google Scholar]
  • 114.Perrais M, Chen X, Perez-Moreno M, Gumbiner BM. E-cadherin homophilic ligation inhibits cell growth and epidermal growth factor receptor signaling independently of other cell interactions. Molecular biology of the cell. 2007;18:2013–2025. doi: 10.1091/mbc.E06-04-0348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Knirsh R, et al. Loss of E-cadherin-mediated cell-cell contacts activates a novel mechanism for up-regulation of the proto-oncogene c-Jun. Mol Biol Cell. 2009;20:2121–2129. doi: 10.1091/mbc.E08-12-1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Smalley-Freed WG, et al. p120-catenin is essential for maintenance of barrier function and intestinal homeostasis in mice. The Journal of clinical investigation. 120:1824–1835. doi: 10.1172/JCI41414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Stairs DB, et al. Deletion of p120-catenin results in a tumor microenvironment with inflammation and cancer that establishes it as a tumor suppressor gene. Cancer cell. 2011;19:470–483. doi: 10.1016/j.ccr.2011.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Yanagisawa M, Anastasiadis PZ. p120 catenin is essential for mesenchymal cadherin-mediated regulation of cell motility and invasiveness. J Cell Biol. 2006;174:1087–1096. doi: 10.1083/jcb.200605022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Taniuchi K, et al. Overexpressed P-cadherin/CDH3 promotes motility of pancreatic cancer cells by interacting with p120ctn and activating rho-family GTPases. Cancer research. 2005;65:3092–3099. doi: 10.1158/0008.5472.CAN-04-3646. [DOI] [PubMed] [Google Scholar]
  • 120.Maeda M, et al. Expression of inappropriate cadherins by epithelial tumor cells promotes endocytosis and degradation of E-cadherin via competition for p120(ctn) Oncogene. 2006;25:4595–4604. doi: 10.1038/sj.onc.1209396. [DOI] [PubMed] [Google Scholar]
  • 121.Islam S, Carey TE, Wolf GT, Wheelock MJ, Johnson KR. Expression of N-cadherin by human squamous carcinoma cells induces a scattered fibroblastic phenotype with disrupted cell-cell adhesion. J Cell Biol. 1996;135:1643–1654. doi: 10.1083/jcb.135.6.1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nieman MT, Prudoff RS, Johnson KR, Wheelock MJ. N-cadherin promotes motility in human breast cancer cells regardless of their E-cadherin expression. The Journal of cell biology. 1999;147:631–644. doi: 10.1083/jcb.147.3.631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Yanagisawa M, et al. A p120 catenin isoform switch affects Rho activity, induces tumor cell invasion, and predicts metastatic disease. The Journal of biological chemistry. 2008;283:18344–18354. doi: 10.1074/jbc.M801192200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Seidel B, Braeg S, Adler G, Wedlich D, Menke A. E- and N-cadherin differ with respect to their associated p120ctn isoforms and their ability to suppress invasive growth in pancreatic cancer cells. Oncogene. 2004;23:5532–5542. doi: 10.1038/sj.onc.1207718. [DOI] [PubMed] [Google Scholar]
  • 125.Kashef J, et al. Cadherin-11 regulates protrusive activity in Xenopus cranial neural crest cells upstream of Trio and the small GTPases. Genes & development. 2009;23:1393–1398. doi: 10.1101/gad.519409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Ozawa M. The N-cadherin cytoplasmic domain confers anchorage-independent growth and the loss of contact inhibition. Sci Rep. 2015;5:15368. doi: 10.1038/srep15368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Gwak GY, et al. Anti-apoptotic N-cadherin signaling and its prognostic implication in human hepatocellular carcinomas. Oncol Rep. 2006;15:1117–1123. [PubMed] [Google Scholar]
  • 128.Theisen CS, Wahl JK, 3rd, Johnson KR, Wheelock MJ. NHERF links the N-cadherin/catenin complex to the platelet-derived growth factor receptor to modulate the actin cytoskeleton and regulate cell motility. Mol Biol Cell. 2007;18:1220–1232. doi: 10.1091/mbc.E06-10-0960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Suyama K, Shapiro I, Guttman M, Hazan RB. A signaling pathway leading to metastasis is controlled by N-cadherin and the FGF receptor. Cancer cell. 2002;2:301–314. doi: 10.1016/s1535-6108(02)00150-2. [DOI] [PubMed] [Google Scholar]
  • 130.Hazan RB, Phillips GR, Qiao RF, Norton L, Aaronson SA. Exogenous expression of N-cadherin in breast cancer cells induces cell migration, invasion, and metastasis. The Journal of cell biology. 2000;148:779–790. doi: 10.1083/jcb.148.4.779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Li G, Satyamoorthy K, Herlyn M. N-cadherin-mediated intercellular interactions promote survival and migration of melanoma cells. Cancer research. 2001;61:3819–3825. [PubMed] [Google Scholar]
  • 132.Ribeiro AS, et al. Extracellular cleavage and shedding of P-cadherin: a mechanism underlying the invasive behaviour of breast cancer cells. Oncogene. 2010;29:392–402. doi: 10.1038/onc.2009.338. [DOI] [PubMed] [Google Scholar]
  • 133.Johnson E, Theisen CS, Johnson KR, Wheelock MJ. R-cadherin influences cell motility via Rho family GTPases. The Journal of biological chemistry. 2004;279:31041–31049. doi: 10.1074/jbc.M400024200. [DOI] [PubMed] [Google Scholar]
  • 134.Paredes J, et al. Breast carcinomas that co-express E- and P-cadherin are associated with p120-catenin cytoplasmic localisation and poor patient survival. Journal of clinical pathology. 2008;61:856–862. doi: 10.1136/jcp.2007.052704. [DOI] [PubMed] [Google Scholar]
  • 135.Fischer KR, et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527:472–476. doi: 10.1038/nature15748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Zheng X, et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015;527:525–530. doi: 10.1038/nature16064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Rakha EA, et al. Further evidence that E-cadherin is not a tumour suppressor gene in invasive ductal carcinoma of the breast: an immunohistochemical study. Histopathology. 2013;62:695–701. doi: 10.1111/his.12066. [DOI] [PubMed] [Google Scholar]
  • 138.Chao YL, Shepard CR, Wells A. Breast carcinoma cells re-express E-cadherin during mesenchymal to epithelial reverting transition. Molecular cancer. 2010;9:179. doi: 10.1186/1476-4598-9-179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Lewis-Tuffin LJ, et al. Misregulated E-cadherin expression associated with an aggressive brain tumor phenotype. PloS one. 2010;5:e13665. doi: 10.1371/journal.pone.0013665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Rodriguez FJ, Lewis-Tuffin LJ, Anastasiadis PZ. E-cadherin’s dark side: Possible role in tumor progression. Biochim Biophys Acta. 2012;1826:23–31. doi: 10.1016/j.bbcan.2012.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kuphal S, Bosserhoff AK. E-cadherin cell-cell communication in melanogenesis and during development of malignant melanoma. Arch Biochem Biophys. 2012;524:43–47. doi: 10.1016/j.abb.2011.10.020. [DOI] [PubMed] [Google Scholar]
  • 142.Kleer CG, van Golen KL, Braun T, Merajver SD. Persistent E-cadherin expression in inflammatory breast cancer. Mod Pathol. 2001;14:458–464. doi: 10.1038/modpathol.3880334. [DOI] [PubMed] [Google Scholar]
  • 143.Mariner DJ, Davis MA, Reynolds AB. EGFR signaling to p120-catenin through phosphorylation at Y228. J Cell Sci. 2004;117:1339–1350. doi: 10.1242/jcs.01001. [DOI] [PubMed] [Google Scholar]
  • 144.Johnson E, et al. HER2/ErbB2-induced breast cancer cell migration and invasion require p120 catenin activation of Rac1 and Cdc42. J Biol Chem. 2010;285:29491–29501. doi: 10.1074/jbc.M110.136770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Dohn MR, Brown MV, Reynolds AB. An essential role for p120-catenin in Src- and Rac1-mediated anchorage-independent cell growth. J Cell Biol. 2009;184:437–450. doi: 10.1083/jcb.200807096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Liu WF, Nelson CM, Pirone DM, Chen CS. E-cadherin engagement stimulates proliferation via Rac1. J Cell Biol. 2006;173:431–441. doi: 10.1083/jcb.200510087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Shamir ER, et al. Twist1-induced dissemination preserves epithelial identity and requires E-cadherin. The Journal of cell biology. 2014;204:839–856. doi: 10.1083/jcb.201306088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Campbell K, Casanova J. A role for E-cadherin in ensuring cohesive migration of a heterogeneous population of non-epithelial cells. Nat Commun. 2015;6:7998. doi: 10.1038/ncomms8998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kang HG, et al. E-cadherin cell-cell adhesion in ewing tumor cells mediates suppression of anoikis through activation of the ErbB4 tyrosine kinase. Cancer research. 2007;67:3094–3105. doi: 10.1158/0008-5472.CAN-06-3259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Silvera D, et al. Essential role for eIF4GI overexpression in the pathogenesis of inflammatory breast cancer. Nat Cell Biol. 2009;11:903–908. doi: 10.1038/ncb1900. [DOI] [PubMed] [Google Scholar]
  • 151.Kourtidis A, Anastasiadis PZ. PLEKHA7 defines an apical junctional complex with cytoskeletal associations and miRNA-mediated growth implications. Cell Cycle. 2016;15:498–505. doi: 10.1080/15384101.2016.1141840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Kourtidis A, et al. Distinct E-cadherin-based complexes regulate cell behaviour through miRNA processing or Src and p120 catenin activity. Nat Cell Biol. 2015;17:1145–1157. doi: 10.1038/ncb3227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Tille JC, et al. The Expression of the Zonula Adhaerens Protein PLEKHA7 Is Strongly Decreased in High Grade Ductal and Lobular Breast Carcinomas. PloS one. 2015;10:e0135442. doi: 10.1371/journal.pone.0135442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kourtidis A, Yanagisawa M, Huveldt D, Copland JA, Anastasiadis PZ. Pro-Tumorigenic Phosphorylation of p120 Catenin in Renal and Breast Cancer. PloS one. 2015;10:e0129964. doi: 10.1371/journal.pone.0129964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Kourtidis A, Anastasiadis PZ. Bringing together cell-to-cell adhesion and miRNA biology in cancer research. Future Oncol. 2016 doi: 10.2217/fon-2016-0012. [DOI] [PubMed] [Google Scholar]
  • 156.Gregory RI, et al. The Microprocessor complex mediates the genesis of microRNAs. Nature. 2004;432:235–240. doi: 10.1038/nature03120. [DOI] [PubMed] [Google Scholar]
  • 157.Ha M, Kim VN. Regulation of microRNA biogenesis. Nature reviews. Molecular cell biology. 2014;15:509–524. doi: 10.1038/nrm3838. [DOI] [PubMed] [Google Scholar]
  • 158.Irby RB, Yeatman TJ. Increased Src activity disrupts cadherin/catenin-mediated homotypic adhesion in human colon cancer and transformed rodent cells. Cancer Res. 2002;62:2669–2674. [PubMed] [Google Scholar]
  • 159.Ozawa M, Ohkubo T. Tyrosine phosphorylation of p120(ctn) in v-Src transfected L cells depends on its association with E-cadherin and reduces adhesion activity. J Cell Sci. 2001;114:503–512. doi: 10.1242/jcs.114.3.503. [DOI] [PubMed] [Google Scholar]

RESOURCES