3,210
Views
36
CrossRef citations to date
0
Altmetric
REVIEW ARTICLE

150th Anniversary Series: Desmosomes and the Hallmarks of Cancer

&
Pages 15-28 | Received 04 Aug 2014, Accepted 07 Apr 2015, Published online: 02 Jul 2015

Abstract

Desmosomes represent adhesive, spot-like intercellular junctions that in association with intermediate filaments mechanically link neighboring cells and stabilize tissue architecture. In addition to this structural function, desmosomes also act as signaling platforms involved in the regulation of cell proliferation, differentiation, migration, morphogenesis, and apoptosis. Thus, deregulation of desmosomal proteins has to be considered to contribute to tumorigenesis. Proteolytic fragmentation and downregulation of desmosomal cadherins and plaque proteins by transcriptional or epigenetic mechanisms were observed in different cancer entities suggesting a tumor-suppressive role. However, discrepant data in the literature indicate that context-dependent differences based on alternative intracellular, signal transduction lead to altered outcome. Here, modulation of Wnt/β-catenin signaling by plakoglobin or desmoplakin and of epidermal growth factor receptor signaling appears to be of special relevance. This review summarizes current evidence on how desmosomal proteins participate in carcinogenesis, and depicts the molecular mechanisms involved.

Abbreviations
ADAM=

a disintegrin and metalloproteinase

AJ=

adherens junction

Dsc=

desmocollin

Dsg=

desmoglein

DP=

desmoplakin

GFR=

growth factor receptor

IF=

intermediate filament

MMP=

matrix metalloproteinase

PKP=

plakophilin

INTRODUCTION

The locally and timely coordinated establishment of cell–cell junctions is a driving force for cell positioning, morphogenesis, and differentiation during development. In the adult organism, maintenance of tissue integrity or repair of tissue defects after wounding in a similar way depends on specific cell–cell contacts. In contrast, a loss of cell–cell contacts in response to either deregulated protein expression or the expression of mutated variants of the corresponding junctional molecules is associated with diseases. Especially Ca2+-dependent cell–cell adhesion molecules of the cadherin family both in adherens junctions (AJs) and desmosomes play a pivotal role in this respect. With their single transmembrane domain, the cadherins are integrated into the plasma membrane and interact with cadherins on the surface of opposing cells (Citationvan Roy, 2014). A specific set of proteins associated with the cadherin cytosolic domains tethers them to actin or intermediate filaments (IFs), respectively, thus establishing a mechanically stable network of connections between cells in epithelial and endothelial cell layers (CitationWu & Yap, 2013). In addition to their adhesive function, some of these cadherin-associated proteins are involved in signaling. Specifically, members of the armadillo repeat family of proteins (β-catenin, p120ctn, plakoglobin, plakophilins [PKPs]) adopt a second function in the regulation of gene transcription and thus were classified as NACos (Nuclear and Adhesion Complexes) proteins (CitationBalda & Matter, 2003). In this context, cell–cell contacts do not only represent sites where cells just stick together but also have to be considered as highly dynamic platforms for outside-in and inside-out signaling. Disturbance of these processes can be linked to tumor formation.

In mammals, the desmosomal cadherins desmoglein1–4 (Dsg1–4) and desmocollin1–3 (Dsc1–3) through interactions of their extracellular domains form spot-like adhesion sites between opposing cell membranes. To establish and maintain desmosome-mediated mechanical stability of tissues it is essential that desmosomal cadherins are associated with IFs (CitationNekrasova & Green, 2013). This requires assembly of a characteristic complex of proteins binding to their cytosolic domains () detectable as an electron-dense plaque in electron micrographs. Within this protein complex, plakoglobin and PKPs directly associate with the desmosomal cadherins and thereby link desmoplakin (DP) or other plakins including plectin, envoplakin, periplakin, and epiplakin to the complex (for an overview on plakins see CitationBouameur et al., 2014). The desmosomal armadillo-repeat proteins, in addition to their role in clustering of desmosomal components within the adhesive complexes, exhibit signaling activities in the cytosol and nucleus. The specific role of plakoglobin and PKPs in this respect is summarized in recent reviews (CitationAktary & Pasdar, 2012; CitationHatzfeld et al., 2014). From these facts it is clear that impairment of desmosomal components by mutations or autoantibodies dramatically affects desmosome functions becoming especially prominent in skin and cardiac muscle. Readers interested in these desmosomal diseases are referred to recent exemplary reviews (CitationAl-Jassar et al., 2013; CitationAmagai & Stanley, 2012; CitationBrooke et al., 2012; CitationCirillo, 2014; CitationCirillo & Al-Jandan, 2013; CitationPatel & Green, 2014; CitationSimpson et al., 2011; CitationSpindler & Waschke, 2014).

Figure 1. Desmosomal proteins contribute to tumorigenesis. In normal epithelial cells, AJs (cadherin–catenin complex) linked to the actin cytoskeleton and desmosomes (desmosomal cadherins: Dsg and Dsc, and plaque proteins) associated with IFs form stable cell–cell contacts. The tetraspanin protein PERP appears to be essential for desmosome formation, but its role is not fully understood. Activation of GFR tyrosine kinases disrupts desmosomes and induces endocytosis of desmosomal cadherins and dissociation of desmoplakin from the desmosomal plaque. Moreover, MAPK family members are activated including ERK1/2 which normally can be kept inactive by Erbin. Cytosolic tyrosine kinases of the Src-family also destabilize desmosomes. In targeting specific residues in plakoglobin, they define its binding to specific interaction partners. Plakoglobin can be included into AJs or compete with β-catenin degradation and transcriptional activity in association with TCF/LEF transcription factors. Probably, plakoglobin like β-catenin can also regulate the transcriptional activity of further transcription factors. Plakophilin released from the desmosome is involved in the regulation of translation (not shown) or has nuclear functions in association with β-catenin or RNA-polymerase III (not shown, see CitationHatzfeld et al. 2014). α, α-catenin; β, β-catenin; DP, desmoplakin; Dsg, desmoglein; Dsc, desmocollin; E-cad, E-cadherin; GFR, growth factor receptor tyrosine kinase; IFs, intermediate filaments; LEF, lymphocyte enhancer factor; p120, p120-catenin; PERP, p53 apoptosis effector related to PMP-22; Pg, plakoglobin; PKP, plakophilin; TCF, T-cell factor; TF, transcription factor.

Figure 1. Desmosomal proteins contribute to tumorigenesis. In normal epithelial cells, AJs (cadherin–catenin complex) linked to the actin cytoskeleton and desmosomes (desmosomal cadherins: Dsg and Dsc, and plaque proteins) associated with IFs form stable cell–cell contacts. The tetraspanin protein PERP appears to be essential for desmosome formation, but its role is not fully understood. Activation of GFR tyrosine kinases disrupts desmosomes and induces endocytosis of desmosomal cadherins and dissociation of desmoplakin from the desmosomal plaque. Moreover, MAPK family members are activated including ERK1/2 which normally can be kept inactive by Erbin. Cytosolic tyrosine kinases of the Src-family also destabilize desmosomes. In targeting specific residues in plakoglobin, they define its binding to specific interaction partners. Plakoglobin can be included into AJs or compete with β-catenin degradation and transcriptional activity in association with TCF/LEF transcription factors. Probably, plakoglobin like β-catenin can also regulate the transcriptional activity of further transcription factors. Plakophilin released from the desmosome is involved in the regulation of translation (not shown) or has nuclear functions in association with β-catenin or RNA-polymerase III (not shown, see CitationHatzfeld et al. 2014). α, α-catenin; β, β-catenin; DP, desmoplakin; Dsg, desmoglein; Dsc, desmocollin; E-cad, E-cadherin; GFR, growth factor receptor tyrosine kinase; IFs, intermediate filaments; LEF, lymphocyte enhancer factor; p120, p120-catenin; PERP, p53 apoptosis effector related to PMP-22; Pg, plakoglobin; PKP, plakophilin; TCF, T-cell factor; TF, transcription factor.

Desmosomes are composed of a tissue-specific set of cadherin transmembrane proteins and are especially abundant in tissues subjected to heavy mechanical and contractile stress. Dsg2 and Dsc2 are common to all desmosome-containing tissues, whereas the other Dsgs and Dscs are expressed in a differentiation-dependent graded pattern in certain stratified epithelia (CitationKljuic et al., 2003; CitationMahoney et al., 2006; CitationNorth et al., 1996; CitationNuber et al., 1995). In human skin, Dsg1, Dsg4, and Dsc1 exhibit most prominent expression in the granular differentiated layer, whereas Dsg3, Dsc2, and Dsc3 show the highest expression in the basal proliferative layer (for more detail see previous reviews in this series CitationBerika & Garrod, 2014; CitationHarmon & Green, 2013). In cells expressing an overlapping pattern of desmosomal cadherins, different Dsgs and Dscs can also be detected in individual desmosomes (CitationNorth et al., 1996; CitationNuber et al., 1996). In general, there is evidence that all desmosomes in vivo contain at least one member of the Dsgs and Dscs, and only in the presence of one cadherin of each family strong adhesion occurs (CitationGetsios et al., 2004). In this context, different studies provided evidence, that both homo- and heterophilic interactions between desmosomal cadherins can exist (CitationChitaev & Troyanovsky, 1997; CitationGetsios et al., 2004; CitationMarcozzi et al., 1998; CitationNie et al., 2011; CitationRunswick et al., 2001; CitationSyed et al., 2002; CitationWaschke et al., 2007). Interestingly, a Ca2+-independent “hyperadhesive” state was reported for mature desmosomes, which is characterized by a complete resistance to chelation of Ca2+ ions with ethylene glycol tetraacetic acid (EGTA) (CitationGarrod & Tabernero, 2014; CitationGarrod, 2013; CitationGarrod et al., 2005).

Electron tomography of desmosomes from neonatal mouse skin embedded in plastic revealed that the N-terminal extracellular cadherin (EC) domains of desmosomal cadherins form a series of 10–20 disordered knot-like assemblies with an architecture of desmosomal cadherin extracellular domains highly similar to classical cadherins (CitationHe et al., 2003). A more recent cryo-electron tomographic analysis of vitreous sections of human skin desmosomes revealed a highly ordered zipper-like assembly of alternating cis- and trans-interacting desmosomal cadherins without interdigitations (CitationAl-Amoudi et al., 2007). Both approaches were not able to differentiate between homophilic and heterophilic interactions of desmosomal cadherins. Despite these structural similarities with E-cadherin, the contribution of desmosomal cadherins to tumorigenesis is by far less clear.

In two seminal papers, Hanahan and Weinberg defined characteristic hallmarks of cancer (CitationHanahan & Weinberg, 2000; CitationHanahan & Weinberg, 2011). Here, we want to discuss how the known functions of desmosomes or desmosomal components can be classified into these hallmarks of cancer and how they may be involved in cancer. This overview does not have the intention to be exhaustive.

DESMOSOMES IN THE CONTEXT OF THE CANCER HALLMARKS

Activation of invasion and metastasis

A critical step during cancer progression is local invasion of primary tumor cells into nearby blood and lymphatic vessels, dissemination throughout the body, and finally extravasation into distant tissues. Loss of contacts between cells of the primary tumor definitely represents an essential step during malignant neoplasia. This includes that not only AJ but also all other cell–cell contacts including desmosomes have to be resolved or modulated (CitationBoyer et al., 1989; CitationSavagner et al., 1997). Early findings showed that expression of desmosomal cadherins inhibits invasive behavior of nonadhesive fibroblasts (CitationTselepis et al., 1998). Reduced expression of desmosomal cadherins later on was observed in different tumors such as skin (CitationChen et al., 2012b; CitationHarada et al., 1996; CitationTada et al., 2000; CitationXin et al., 2014), head and neck (CitationShinohara et al., 1998; CitationWong et al., 2008), lung (CitationCui et al., 2012a; CitationCui et al., 2012b), breast (CitationKlus et al., 2001; CitationOshiro et al., 2005), prostate (CitationBarber et al., 2014; CitationPan et al., 2014), cervix (CitationAlazawi et al., 2003), uterus (CitationNei et al., 1996), pancreas (CitationHamidov et al., 2011), liver (CitationSchüle et al., 2014), gastric (CitationBiedermann et al., 2005; CitationYashiro et al., 2006), and colon (CitationCui et al., 2011; CitationFunakoshi et al., 2008; CitationKamekura et al., 2013; CitationKhan et al., 2006; CitationKnösel et al., 2012; CitationKolegraff et al., 2011). Taken together, these data suggest a role of desmosomal cadherins as suppressors of tumor metastasis.

However, the situation is not as clear. Other studies report enhanced expression of desmosomal cadherins in cancer (CitationBrennan & Mahoney, 2009; CitationBrown et al., 2014; CitationChen et al., 2007; CitationDe Bruin et al., 1999; CitationHuang et al., 2010; CitationKurzen et al., 2003; CitationSavci-Heijink et al., 2009). Therefore, both loss and overexpression of desmosomal cadherin expression frequently correlate with advanced tumor grading, increased invasion and metastasis, and/or poor prognosis/survival. It is interesting to note that especially the expression of Dsgs appears to be upregulated in tumor cells.

Genetic mouse models represent interesting tools to address the role of desmosomal cadherins in vivo in more detail. Overexpression of Dsg2 or Dsg3 in suprabasal skin layers in a transgenic mouse model increased keratinocyte proliferation, anchorage-independent cell survival, and development of skin tumors (CitationBrennan et al., 2007; CitationMerritt et al., 2002). Expression of an N-terminally deleted Dsg3 in skin also resulted in hyperproliferation (CitationAllen et al., 1996), whereas Dsg3 knockout mice revealed no abnormalities (CitationKoch et al., 1997). In contrast, in a Dsc1 knockout situation, epidermis is hyperproliferative (CitationChidgey et al., 2001). When Dsg3−/− or Dsg3+/− keratinocytes transformed through activated H-Ras and p53 inactivation where assayed for tumor growth in Scid mice, tumors derived from Dsg3+/− keratinocytes showed significantly higher tumor volume compared with Dsg3−/−-derived tumors. This suggests that Dsg3 facilitates tumor growth (CitationBaron et al., 2012). In an autochthonous squamous cell carcinoma model with chronic ultraviolet B (UVB)-treated Dsg3−/− mice no enhanced tumorigenesis was detectable probably due to compensatory upregulation of Dsg1 and Dsg2 (CitationBaron et al., 2012). Consistently, Dsg1 was reported to compensate the genetic loss of Dsg3 in a transgenic mouse model (CitationHanakawa et al., 2002).

Currently, the molecular mechanisms for these desmosomal cadherin-specific differences in proliferative behavior are not understood. Apparently, mutations in desmosomal cadherin genes are not of major relevance in cancer (CitationBiedermann et al., 2005). Recent studies indicate that individual desmosomal cadherins define distinct variations in adhesiveness, migration, differentiation, and/or morphogenesis. Here, the ratio and expression of specific desmosomal cadherins, Ca2+-dependent or -independent state of adhesion, and distinct mechanism of incorporation into desmosomes appear to play a role (CitationGetsios et al., 2004; CitationHardman et al., 2005; CitationHartlieb et al., 2013; CitationIshii et al., 2001; CitationLowndes et al., 2014).

Regulation of desmosomal cadherin levels

According to the observations reported above, changes in desmosomal cadherin levels apparently contribute to tumorigenesis. Three major mechanisms have to be considered that can lead to altered desmosomal cadherin expression in cancer.

Transcriptional regulation of desmosomal cadherins

In general, little is known about the mechanisms controlling transcription of desmosomal cadherin genes, although promoter regions of genes have been identified early after cloning of the corresponding coding genes (CitationAdams et al., 1998; CitationJohns et al., 2005; CitationMarsden et al., 1997; CitationSilos et al., 1996). Recent years identified a number of transcriptional factors that modulate desmosomal cadherin expression including Stat6, Klf5, Smad4, grainy head-like 1, Foxn1, Cdx1/Cdx2, c-Rel, lymphocyte enhancer factor/transcription factor (LEF/TCF), and p53/p63 (CitationFerone et al., 2013; CitationFunakoshi et al., 2008; CitationJohnson et al., 2014; CitationKenchegowda et al., 2012; CitationMlacki et al., 2014; CitationOmori-Miyake et al., 2014; CitationOshiro et al., 2003; CitationOwens et al., 2008; CitationTokonzaba et al., 2013; CitationWilanowski et al., 2008). Interestingly, plakoglobin in a complex with LEF-1 is able to bind to the proximal promoter regions of both Dsc2 and Dsc3, however with opposite effects. Dsc2 expression is activated, whereas Dsc3 transcription is repressed resulting in a shift in isoform expression (CitationTokonzaba et al., 2013). This effect shows some similarity to the classical cadherin switch from E- to N-cadherin during epithelial–mesenchymal transition (EMT). In this context, it has to be noted that expression of the EMT-inducing transcription factor Snail was reported to induce degradation of Dsg2 (CitationKume et al., 2013). In addition, epigenetic alterations turned out to play a dominant role in the regulation of desmosomal cadherin expression in cancer. Promoter methylation (CitationCui et al., 2012a; CitationCui et al., 2011; CitationOshiro et al., 2005; CitationPan et al., 2014; CitationWang et al., 2014) as well as histone methylation (CitationKe et al., 2009) and histone acetylation (CitationGould et al., 2010; CitationJohnson et al., 2014) have been reported to modulate desmosomal cadherin expression. Similar observations have been made for the desmosomal plaque proteins plakoglobin (CitationBreault et al., 2005; CitationShafiei et al., 2008; CitationShiina et al., 2005; CitationWinn et al., 2002), PKPs (CitationOka et al., 2009), and DP (CitationYang et al., 2012) and have been summarized in recent reviews (CitationAktary & Pasdar, 2012; CitationBouameur et al., 2014; CitationHatzfeld et al., 2014).

Impaired transport, targeting, and assembly into mature desmosomes

Impaired desmosome transport and assembly is a highly complex process that requires multiple coordinated steps. According to the present view, desmosomal cadherins in association with plakoglobin are transported by kinesin-mediated vesicular traffic (CitationBurdett & Sullivan, 2002). Interestingly, vesicular transport of different desmosomal cadherins depends on distinct kinesins (Dsg2—kinesin-1; Dsc2—kinesin-2) (CitationNekrasova et al., 2011). DP and PKP2 play an important role in desmosomal cadherin association with IFs and assembly into cell–cell contact sites (CitationBass-Zubek et al., 2008; CitationGodsel et al., 2005). Impaired DP expression or function affects desmosome assembly at the cell surface and thus may contribute to tumorigenesis. In line with this, genetic deletion of DP in a pancreatic neuroendocrine tumor model resulted in increased local invasion (CitationChun & Hanahan, 2010). Finally, at the cell surface specific factors may stabilize desmosomal cadherins as shown for Dsg2 in association with the lectin galectin-3 (Gal3). In the absence of Gal3, Dsg2 is internalized (CitationJiang et al., 2014). However, it cannot be excluded that half-desmosomes pre-assemble in the membrane and attach with counterparts on the opposing cell surface to establish a mature desmosome (CitationDemlehner et al., 1995).

Moreover, the actin cytoskeleton has been shown to play a role for desmosome assembly and desmosomal cadherin dynamics (CitationRoberts et al., 2011). In this context, it was reported that the actin-organizing protein adducin regulates Dsg3 membrane incorporation and mobility in a phosphorylation- and RhoA-dependent manner and its knockdown impairs cohesion (CitationRötzer et al., 2014; CitationWaschke et al., 2006). On the other side, Dsg3 acts as a regulator of Rac-1 and Cdc42 activity, thereby modulating the structure of the actin cytoskeleton (CitationTsang et al., 2012a). For more details on desmosome assembly and dynamics see CitationHarmon & Green (2013) and CitationNekrasova & Green (2013). However, the impact of these mechanisms on tumor formation and progression has to be analyzed in more detail.

Inactivation by proteolytic cleavage

Active depletion of desmosomal cadherins in cancer by proteolytic degradation can either occur directly at the cell surface by shedding proteases or via the endosomal– lysosomal degradation pathway. Shedding proteases are frequently upregulated in tumor cells and promote the release of growth factors and cytokines but also target a number of adhesion molecules. Proteomic analyses indeed could detect released extracellular fragments of desmosomal cadherins in tumor secretomes (CitationBech-Serra et al., 2006; CitationGreening et al., 2013; CitationShi et al., 2009). Different proteinases have been identified that release desmosomal cadherin extracellular domains from cells including a disintegrin and metalloproteinase (ADAM), matrix metalloproteinases (MMPs), or kallikreins (CitationBech-Serra et al., 2006; CitationBorgono et al., 2007; CitationBrooke et al., 2014; CitationCirillo et al., 2007; CitationJiang et al., 2011; CitationKlessner et al., 2009; CitationWeiske et al., 2001). There is evidence that cleavage is cadherin- and/or cell-type-specific. It is currently not clear, if this proteolytic shedding of extracellular domains only reduces the number of molecules available for adhesion, or whether these soluble fragments themselves have a further physiological function, for example, in interference with intercellular adhesion (CitationNoe et al., 2001; CitationWheelock et al., 1987).

Deregulated endocytosis was defined as multifaceted characteristic of cancer cells (CitationMosesson et al., 2008) and represents another efficient route to reduce the levels of cell surface proteins. It is now well accepted that tumor-associated growth factors induce endocytic internalization of junctional molecules (also see “Sustainment of proliferative signaling” section). Recent studies indicate that cadherins can choose different routes of internalization depending on the cellular environment, growth factors, and migratory needs of the respective cells (CitationDelva & Kowalczyk, 2009). Dsg3 uses a lipid raft-mediated pathway for internalization in cross talk with epidermal growth factor receptor (EGFR) (CitationBektas et al., 2013; CitationCalkins et al., 2006; CitationDelva et al., 2008; CitationStahley et al., 2014). Interestingly, tail–tail interactions of the Dsg2 C-terminal unique region inhibit internalization (CitationChen et al., 2012a). In binding to a specific motif in Dsg2, a role for caveolin-1 in the process of endocytosis was recently reported (CitationBrennan et al., 2012).

Future studies have to further unravel the regulatory signals that induce endocytosis. In cancer cells, GFR-induced posttranslational modifications may contribute to destabilization of desmosomes in releasing desmosomal cadherins from mature junctional complexes and subsequent uptake into endocytotic vesicles (CitationAoyama et al., 2010; CitationJennings et al., 2011).

One important consequence of changes in desmosomal cadherin expression in cancer is that the signaling functions of the desmosomal plaque-associated armadillo repeat proteins are modulated. The interrelation of desmosomal plaque proteins with β-catenin signaling is of specific interest in this context. Changes in the localization of the desmosomal plaque proteins, their expression levels or stability, for example, in response to posttranslational modifications, have been shown to modulate TCF/LEF-dependent transcription in a context-specific manner, with plakoglobin mainly acting as negative regulator suppressing oncogenic β-catenin–TCF/LEF signaling. For more details see recent comprehensive reviews (CitationAktary & Pasdar, 2012; CitationChidgey & Dawson, 2007; CitationDusek & Attardi, 2011; CitationHatzfeld, 2007; CitationHatzfeld et al., 2014).

Resistance to cell death

Loss of cell–matrix or cell–cell contact in normal epithelial cells is a cell-death-inducing signal (CitationGilmore, 2005; CitationValentijn et al., 2004); however, tumor cells often develop specific properties to resist cell death. During apoptosis, a large set of substrate proteins including adhesion molecules including desmosomal cadherins, cytoskeletal proteins, and their regulators were identified as caspase substrates. Interestingly, after induction of apoptosis not only a caspase-mediated cleavage within the cytosolic domain but also a release of the extracellular domain of desmosomal cadherins was observed (CitationCirillo et al., 2007; CitationCirillo et al., 2008; CitationDusek et al., 2006; CitationLanza & Cirillo, 2007; CitationWeiske et al., 2001). In addition, proteins of the desmosomal plaque have been identified as caspase targets (CitationAho, 2004; CitationKalinin et al., 2005; CitationWeiske et al., 2001).

The interesting question in respect to tumor progression now is, do the observed changes in desmosomal cadherin expression contribute to regulation of cell death and how? Overexpression of Dsg2 increased anchorage-independent cell growth in an EGFR and nuclear factor kappa B (NFκB)-dependent pathway resulting in upregulation of c-myc and anti-apoptotic Bcl-XL (CitationBrennan et al., 2007). In contrast, knockdown of Dsg1 in keratinocytes or of Dsg2 in enterocytes was associated with decreased rates of apoptosis in response to UV irradiation (CitationDusek et al., 2006; CitationNava et al., 2007). Interestingly, analogous effects were not observed with Dsc2 in enterocytes (CitationNava et al., 2007). This again suggests that there are cell-type- and cadherin-specific effects. However, overexpression of the apoptotic Dsg2 intracellular domain fragment induced apoptosis and furthermore increased expression of endogenous Dsg2. This increase is specific for the Dsg2 intracellular domain fragment, since expression of full-length Dsg2 did not result in upregulated expression of endogenous Dsg2 (CitationNava et al., 2007). Thus, the intracellular Dsg2 fragment appears to feed forward the apoptotic signal. Currently, the role of an upregulation of endogenous Dsg2 is not clear. In line with a feed-forward mechanism, more Dsg2 will generate further substrate for cleavage. Taken together, desmosomal cadherins contribute to regulation of apoptosis in keeping cells sensitive to apoptosis and in further enhancing the process. In contrast, loss of Dsg in tumor cells may represent a mechanism to evade apoptosis.

However, it has to be kept in mind that expression levels of specific cadherins may define whether cells finally divide or die. In addition, there is evidence that plakoglobin and PKPs are involved in regulation of apoptosis. Changes in the levels of desmosomal cadherins may increase or reduce binding sites for plakoglobin or PKPs at sites of cell–cell contact thereby switching their signaling functions in apoptosis regulation off or on, respectively. In this context, plakoglobin-deficient keratinocytes isolated from plakoglobin knockout mice are impaired in induction of apoptosis (CitationDusek et al., 2007). An opposite situation was observed during analysis of mice with epidermal knockout of plakoglobin (CitationLi et al., 2012). This suggests that multiple factors such as the local environment or mechanic tension influence behavior of cells.

It is interesting to note that plakoglobin, PKP2, and DP expression itself is repressed by the EMT-inducing transcription factors Slug or Sip1/zinc finger E-box-binding homeobox-2 or ZEB2 (CitationBailey et al., 2012; CitationVandewalle et al., 2005). Re-expression of plakoglobin in human squamous carcinoma cells revealed dose-dependent differences with low expressing cells showing contact inhibition of growth, whereas in high expressors, apoptosis is inhibited correlating with an increased expression of the anti-apoptotic factor Bcl-2 (CitationHakimelahi et al., 2000). Plakoglobin apparently does not control Bcl-2 expression directly, but indirectly in modulating -catenin activity (CitationLi et al., 2007a) and this in consequence may regulate Bcl-2 expression via c-myc and E2F1 (CitationLi et al., 2007b). Both plakoglobin and β-catenin can bind to the c-myc promoter and act as repressors or activators, respectively (CitationHe et al., 1998; CitationMüller-Tidow et al., 2004; CitationWilliamson et al., 2006). Furthermore, it was recently shown that plakoglobin but not β-catenin forms a complex with p53 and increases p53 transcriptional activity on the promoter of the tumor suppressor 14-3-3σ (CitationAktary et al., 2013), and in addition can repress the oncogenic chromatin remodeling factor special AT-rich sequence-binding protein-1 or SATB1 (CitationAktary & Pasdar, 2013). Loss of Rnd3/RhoE leads to an enhanced expression of desmosomal proteins and protects keratinocytes from cisplatin-induced apoptosis in a plakoglobin-dependent manner (CitationRyan et al., 2012). A recent study suggested that PKP2 also is involved in regulation of apoptosis (CitationKim et al., 2013). Furthermore, re-expression of DP in cells with an epigenetic inactivation of the DP gene exhibited increased sensitivity to apoptosis by upregulation of plakoglobin and concomitant inhibition of β-catenin–TCF/LEF-dependent transcription (CitationYang et al., 2012). For further reading on desmosomes and apoptosis also see CitationHuber & Weiske (2009).

Sustainment of proliferative signaling

Continuous expression of growth factors and cytokines in tumors provides an environment promoting unrestricted proliferation predominantly by signaling through receptors with tyrosine kinase activity. In respect to desmosomes, the role of EGF and EGFR has been studied in most detail. Plakoglobin and β-catenin were identified as targets of EGFR. They are tyrosine phosphorylated within minutes after EGF treatment and both interact with EGFR (CitationGaudry et al., 2001; CitationHoschuetzky et al., 1994; CitationKanai et al., 1995). EGFR-dependent phosphorylation of tyrosine residue(s) in the C-terminus of plakoglobin shifts it to the Triton X-100-soluble fraction in association with Dsg and induces dissociation of DP resulting in decreased adhesive strength (CitationGaudry et al., 2001; CitationYin et al., 2005). In line with these findings, inhibition of EGFR activity induces an epithelial phenotype, promotes assembly of desmosomes, recruits IFs, and strengthens cell adhesion. There is evidence that in addition to plakoglobin, Dsg2 can be phosphorylated (CitationLorch et al., 2004). Another important consequence of EGFR activation is the upregulation of metalloproteinases involved in the shedding of desmosomal cadherins (CitationEllerbroek et al., 2001; CitationSantiago-Josefat et al., 2007), thus contributing to enhanced migratory and invasive capacity of cells (see “Inactivation by proteolytic cleavage” section). In this context, it was shown that EGFR signaling regulates Dsg2 shedding and endocytotic trafficking in cooperation with ADAMs (CitationKlessner et al., 2009). Moreover, the intramembrane serine protease iRhom2 is involved in maturation of ADAM17 and as a consequence in the activation of the EGF–EGFR axis and desmosomal cadherin processing (CitationBrooke et al., 2014).

Activation of EGFR induces multiple downstream signaling events. EGFR-dependent activation of Erk5 was shown to induce expression of the EMT promoting transcription factor Slug resulting in altered desmosomal organization and mobility of cells (CitationArnoux et al., 2008). The Ras–Raf–mitogen-activated protein kinase (MAPK) pathway was found to be necessary and sufficient for EGF-induced desmosome breakdown (CitationEdme et al., 2002). Interestingly, Dsg1 with an N-terminal deletion which switches off its adhesive function still was able to suppress EGFR–extracellular-signal-regulated kinase 1 (ERK1)/ERK2 signaling in recruiting the EGFR into more insoluble membrane fractions, indicating that Dsg1 has adhesion-independent signaling functions (CitationGetsios et al., 2009). The ability of Dsg1 to capture the ERK regulator Erbin to its cytosolic domain thereby inhibiting ERK activation was identified as a potential mechanism (CitationHarmon et al., 2013). Similarly, re-expression of Dsc3 in lung cancer cells reduced ERK1/2 phosphorylation (CitationCui et al., 2012a). Moreover, Dsg3 appears to be involved in the regulation of p38MAPK in a cross talk with EGFR (CitationBektas et al., 2013; CitationBerkowitz et al., 2005; CitationBerkowitz et al., 2006).

In addition to EGF, other growth factors and cytokines including tumor necrosis factor alpha (TNFα), hepatocyte growth factor (HGF), fibroblast growth factor (FGF), insulin-like growth factor-1 (IGF-1), interferon gamma (IFNγ), interleukin 1β (IL-1β), IL-6, IL-13, or IL-17 (CitationAsimaki et al., 2011; CitationKampf et al., 1999; CitationLi et al., 2001; CitationSavagner et al., 1997; CitationWolf et al., 2013; CitationZuckerman et al., 2008) have been reported to impair desmosomal structure and function. However, they were not studied in detail as reported for EGF.

In addition to GFR-associated tyrosine kinases, cytosolic tyrosine kinases are frequently upregulated in cancer. Keratinocytes from plakoglobin –/– mice exhibit increased Src activity correlating with enhanced cell motility. Both the Src inhibitor PP2 and a dominant-negative Src kinase attenuated cell migration in plakoglobin-negative cells (CitationYin et al., 2004). Src signaling in these plakoglobin-negative cells appears to be regulated through Rho and dependent on extracellular matrix components such as fibronectin and vitronectin (CitationFranzen et al., 2012; CitationTodorovic et al., 2010). Furthermore, Src is involved in a cross talk between Dsg3 and E-cadherin (CitationTsang et al., 2012a; CitationTsang et al., 2012b; CitationTsang et al., 2010). Plakoglobin was identified as a target of Src, Fyn, and Fer kinases. Interestingly, although plakoglobin and β-catenin are highly similar in structure, phosphorylation by these kinases or EGFR has opposing effects on the interaction of plakoglobin with common binding partners such as α-catenin, classical cadherins, and TCF/LEF as well as on association with DP. Even effects induced by the cytosolic tyrosine kinases Src and Fer, respectively, are different (CitationMiravet et al., 2003).

Evasion of growth suppressors

The retinoblastoma proteins Rb and p53 represent two central regulators of cell cycle and apoptosis (CitationSherr & McCormick, 2002). A link between p53 and its close relative p63 to desmosomal structure and function is definitely given. Both not only directly control expression of desmosomal genes including Dsc3, Dsg1, and DP (CitationCui et al., 2012a; CitationCui et al., 2011; CitationFerone et al., 2013; CitationOshiro et al., 2003), but also p53 apoptosis effector related to peripheral myelin protein-22 (PERP). PERP encodes a desmosome-located 21-kDa protein with four transmembrane domains that acts as an essential factor for epithelial integrity (CitationIhrie & Attardi, 2005; CitationIhrie et al., 2005) and in apoptosis regulation (CitationIhrie & Attardi, 2004). Knockout of PERP in mice results in a loss of desmosomes, impaired wound healing, and enhanced tumorigenesis (CitationBeaudry et al., 2010a; CitationBeaudry et al., 2010b; CitationDusek et al., 2012), emphasizing the role of desmosomes in cancer progression. Nevertheless, it always has to be kept in mind that the role of desmosomal proteins in cancer is context-dependent and can result in cell-cycle arrest or induction of proliferation (CitationChen et al., 2013).

Induction of angiogenesis

Efficient formation of new vasculature is a prerequisite for tumor growth. Since endothelial cells do not possess desmosomes, only the desmosomal proteins plakoglobin, PKP4/p0071, and DP associated with the vascular endothelial (VE)-cadherin may contribute to angiogenesis in a desmosome-independent manner. Loss of DP results in impaired capillary formation and leakiness (CitationZhou et al., 2004). Vascular endothelial growth factor-A (VEGF-A) treatment of endothelial cells upregulates PKP suggesting an involvement in angiogenesis (CitationRennel et al., 2007). Studies with HUVEC cells indicated that plakoglobin is important for the mechanical stabilization of junctions under sheer stress and for regulation of endothelial cell migration, proliferation, and angiogenesis (CitationNagashima et al., 1997; CitationNottebaum et al., 2008; CitationSchnittler et al., 1997; CitationVenkiteswaran et al., 2002). Recently, it was shown that extracellular MMP inducer (EMMPRIN/basigin/CD147), an Ig superfamily member with strong relation to tumorigenesis contributes to angiogenesis in a complex with plakoglobin and Nm23, thereby regulating actomyosin contractility at endothelial junctions (CitationMoreno et al., 2014).

Genome instability and replicative immortality

Little is known if desmosomes or desmosomal proteins are involved in these processes. Overexpression of plakoglobin in HCT116 cells was reported to promote genomic instability in inducing aneuploidy (CitationPan et al., 2007). On the other side, it was reported that plakoglobin is subjected to loss of heterozygosity in certain cancers (CitationAberle et al., 1995). Loss of heterozygosity was also detected for DP; however, the patients show specific heart phenotypes (CitationMurray et al., 2013) with no specific evidence of enhanced tumorigenesis. It was recently shown that human telomerase is a direct target of β-catenin in association with TCF or Kruppel-like factor 4 (KLF4) (CitationHoffmeyer et al., 2012; CitationZhang et al., 2012); however, it is not known if plakoglobin or PKP in competing with β-catenin may modulate telomerase expression.

Tumor promoting inflammation, avoidance of destruction by the immune system, and deregulation of cellular energetics

During recent years, compelling evidence accumulated that reactions of the immune system and tumor progression are linked (CitationGrivennikov et al., 2010). Cancer cells have to evade anti-tumor activities of immune cells to avoid destruction. On the other side, immune cells, especially partially differentiated myeloid progenitors, appear to be the source of numerous growth factors and cytokines which augment tumor growth. Desmosomal protein expression is affected by these factors (see “Sustainment of proliferative signaling” section). In addition, it was observed that tumor cells frequently exhibit a switch in energy metabolism. Most well-known in this context is the Warburg effect describing the reprogramming of the glucose metabolism to aerobic glycolysis resulting in generation of increasing amounts of lactate (CitationDeBerardinis et al., 2008). These aspects were defined as the so-called “emerging” hallmarks of cancer by CitationHanahan and Weinberg (2011). The only minor hint to such a relation may be given by the observation that enhanced lipogenesis occurs in induced pluripotent stem cell or iPSC-derived cardiomyocytes which express a mutant form of PKP2 (CitationKim et al., 2013), a metabolic phenomenon also observed in tumor cells.

CONCLUSION

Despite many discrepant findings, current data provide substantial evidence that desmosomes or their protein components are involved in different processes during development and progression of cancer (). However, it always has to be kept in mind that desmosomes can differ in respect to their composition, expression levels of specific constituents, and their localization in a tissue- and context-dependent manner. Thus, changes of these conditions can have different outcomes. They either modulate adhesiveness or intracellular signaling pathways. According to our current understanding, plakoglobin and β-catenin signaling activities and their mutual regulation appear to be of central importance in this context. Currently, there is only limited evidence for a specific role of desmosomes or desmosomal proteins in the context of some of the hallmarks (see “Induction of angiogenesis,” “Genome instability and replicative immortality,” and “Tumor promoting inflammation, avoidance of destruction by the immune system, and deregulation of cellular energetics” sections) defined by Hanahan and Weinberg. The characterization of new players, the role of posttranslational modifications, and the identification of new cross talks will help to better understand the role of desmosomes in tissue homeostasis and malignant disease in the future.

Figure 2. Desmosomes and the hallmarks of cancer. The indicated connections may depend on the context such as cell type or tissue and expression of desmosomal cadherin family member(s). In many cases, the molecular mechanisms are not fully unraveled and it is not clear if the observed effects are direct or indirect. We also included links that are mediated by desmosomal proteins and do not depend on the presence of desmosome as is the case in respect to angiogenesis since endothelial cells in blood vessels under normal conditions do not express desmosomal cadherins and thus cannot assemble desmosomes.

Figure 2. Desmosomes and the hallmarks of cancer. The indicated connections may depend on the context such as cell type or tissue and expression of desmosomal cadherin family member(s). In many cases, the molecular mechanisms are not fully unraveled and it is not clear if the observed effects are direct or indirect. We also included links that are mediated by desmosomal proteins and do not depend on the presence of desmosome as is the case in respect to angiogenesis since endothelial cells in blood vessels under normal conditions do not express desmosomal cadherins and thus cannot assemble desmosomes.

ACKNOWLEDGEMENT

The authors apologize to all colleagues whose work we did not discuss or cite in this manuscript. We thank Dr. Laura Bloch for critical reading of the manuscript.

Declaration of interest: The authors declare no conflict of interest. The authors alone are responsible for the content and writing of the paper.

REFERENCES

  • Aberle H, Bierkamp C, Torchard D, Serova O, Wagner T, Natt E, Wirsching J, Heidkämper C, Montagna M, Lynch HT, Lenoir GM, Scherer G, Feunteun J, Kemler R (1995). The human plakoglobin gene localizes on chromosome 17q21 and is subjected to loss of heterozygosity in breast and ovarian cancers. Proc Natl Acad Sci USA. 92: 6384–6388.
  • Adams MJ, Reichel MB, King IA, Marsden MD, Greenwood MD, Thirlwell H, Arnemann J, Buxton RS, Ali RR (1998). Characterization of the regulatory regions in the human desmoglein genes encoding the pemphigus foliaceus and pemphigus vulgaris antigens. Biochem J. 329: 165–174.
  • Aho S (2004). Plakin proteins are coordinately cleaved during apoptosis but preferentially through the action of different caspases. Exp Dermatol. 13: 700–707.
  • Aktary Z, Kulak S, Mackey J, Jahroudi N, Pasdar M (2013). Plakoglobin interacts with the transcription factor p53 and regulates the expression of 14–3-3sigma. J Cell Sci. 126: 3031–3042.
  • Aktary Z, Pasdar M (2012). Plakoglobin: role in tumorigenesis and metastasis. Int J Cell Biol. 2012: 189521.
  • Aktary Z, Pasdar M (2013). Plakoglobin represses SATB1 expression and decreases in vitro proliferation, migration and invasion. PLoS One. 8: e78388.
  • Al-Amoudi A, Diez DC, Betts MJ, Frangakis AS (2007). The molecular architecture of cadherins in native epidermal desmosomes. Nature. 450: 832–839.
  • Al-Jassar C, Bikker H, Overduin M, Chigey M (2013). Mechanistic basis of desmosome-targeted disease. J Mol Biol. 425: 4006–4022.
  • Alazawi WO, Morris LS, Stanley MA, Garrod DR, Coleman N (2003). Altered expression of desmosomal components in high-grade squamous intraepithelial lesions of the cervix. Virchows Arch. 443: 51–56.
  • Allen E, Yu Q-C, Fuchs E (1996). Mice expressing a mutant desmosomal cadherin exhibit abnormalities in desmosomes, proliferation and epidermal differentiation. J Cell Biol. 135: 215–225.
  • Amagai M, Stanley JR (2012). Desmoglein as a target in skin disease and beyond. J Invest Dermatol. 132: 776–784.
  • Aoyama Y, Nagai M, Kitajima Y (2010). Binding of pemphigus vulgaris IgG to antigens in desmosome core domains excludes immune complexes rather than directly splitting desmosomes. Br J Dermatol. 162: 1049–1055.
  • Arnoux V, Nassour M, L’Helgoualc'h A, Hipskind RA, Savagner P (2008). Erk5 controls Slug expression and keratinocyte activation during wound healing. Mol Biol Cell. 19: 4738–4749.
  • Asimaki A, Tandri H, Duffy ER, Winterfield JR, Mackey-Bojack S, Picken MM, Cooper LT, Wilber DJ, Marcus FI, Basso C, Thiene G, Tsatsopoulou A, Protonotarios N, Stevenson WG, McKenna WJ, Gautam S, Remick DG, Calkins H, Saffitz JE (2011). Altered desmosomal proteins in granulomatous myocarditis and potential pathogenic links to arrhythmogenic right ventricular cardiomyopathy. Circ Arrhythm Electrophysiol. 4: 743–752.
  • Bailey CK, Mittal MK, Misra S, Chaudhuri G (2012). High motility of triple-negative breast cancer cells is due to repression of plakoglobin gene by metastasis modulator protein SLUG. J Biol Chem. 287: 19472–19486.
  • Balda MS, Matter K (2003). Epithelial cell adhesion and the regulation of gene expression. Trends Cell Biol. 13: 310–318.
  • Barber AG, Castillo-Martin M, Bonal DM, Rybicki BA, Christiano AM, Cordon-Cardo C (2014). Characterization of desmoglein expression in the normal prostatic gland. Desmoglein 2 is an independent prognostic factor for aggressive prostate cancer. PLoS One. 9: e98786.
  • Baron S, Hoang A, Vogel H, Attardi LD (2012). Unimpaired skin carcinogenesis in Desmoglein 3 knockout mice. PLoS One. 7: e50024.
  • Bass-Zubek AE, Hobbs RP, Amargo EV, Garcia NJ, Hsieh SN, Chen X, Wahl JK III, Denning MF, Green KJ (2008). Plakophilin 2: a critical scaffold for PKC alpha that regulates intercellular junction assembly. J Cell Biol. 181: 605–613.
  • Beaudry VG, Ihrie RA, Jacobs SB, Nguyen B, Pathak N, Park E, Attardi LD (2010a). Loss of the desmosomal component Perp impairs wound healing in vivo. Dermatol Res Pract. 2010: 759731.
  • Beaudry VG, Jiang D, Dusek RL, Park EJ, Knezevich S, Ridd K, Vogel H, Bastian BC, Attardi LD (2010b). Loss of the p53/p63 regulated desmosomal protein Perp promotes tumorigenesis. PLoS Genet. 6: e1001168.
  • Bech-Serra JJ, Santiago-Josefat B, Esselens C, Saftig P, Baselga J, Arribas J, Canals F (2006). Proteomic identification of desmoglein 2 and activated leukocyte cell adhesion molecule as substrates of ADAM17 and ADAM10 by difference gel electrophoresis. Mol Cell Biol. 26: 5086–5095.
  • Bektas M, Jolly PS, Berkowitz P, Amagai M, Rubenstein DS (2013). A pathophysiologic role for epidermal growth factor receptor in pemphigus acantholysis. J Biol Chem. 288: 9447–9456.
  • Berika M, Garrod D (2014). Desmosomal adhesion in vivo. Cell Commun Adhes. 21: 65–75.
  • Berkowitz P, Hu P, Liu Z, Diaz LA, Enghild JJ, Chua MP, Rubenstein DS (2005). Desmosome signaling. Inhibition of p38MAPK prevents pemphigus vulgaris IgG-induced cytoskeleton reorganization. J Biol Chem . 280: 23778–23784.
  • Berkowitz P, Hu P, Warren S, Liu Z, Diaz LA, Rubenstein DS (2006). p38MAPK inhibition prevents disease in pemphigus vulgaris mice. Proc Natl Acad Sci U S A. 103: 12855–12860.
  • Biedermann K, Vogelsang H, Becker I, Plaschke S, Siewert JR, Hofler H, Keller G (2005). Desmoglein 2 is expressed abnormally rather than mutated in familial and sporadic gastric cancer. J Pathol. 207: 199–206.
  • Borgono CA, Michael IP, Komatsu N, Jayakumar A, Kapadia R, Clayman GL, Sotiropoulou G, Diamandis EP (2007). A potential role for multiple tissue kallikrein serine proteases in epidermal desquamation. J Biol Chem. 282: 3640–3652.
  • Bouameur JE, Favre B, Borradori L (2014). Plakins, a versatile family of cytolinkers: roles in skin integrity and in human disease. J Invest Dermatol. 134: 885–894.
  • Boyer B, Tucker GC, Valles AM, Gavrilovic J, Thiery JP (1989). Reversible transition towards a fibroblastic phenotype in a rat carcinoma cell line. Int J Cancer Suppl. 4: 69–75.
  • Breault JE, Shiina H, Igawa M, Ribeiro-Filho LA, Deguchi M, Enokida H, Urakami S, Terashima M, Nakagawa M, Kane CJ, Carroll PR, Dahiya R (2005). Methylation of the γ-catenin gene is associated with poor prognosis of renal cell carcinoma. Clin Cancer Res. 11: 557–564.
  • Brennan D, Hu Y, Choi YW, Whitaker-Menezes D, O’Brian T, Uitto J, Rodeck U, Mahoney MG (2007). Suprabasal Dsg2 expression in transgenic mouse skin confers a hyperproliferative and apoptosis-resistant phenotype to keratinocytes. J Cell Sci. 120: 758–771.
  • Brennan D, Mahoney MG (2009). Increased expression of Dsg2 in malignant skin carcinomas: A tissue-microarray based study. Cell Adh Migr. 3: 148–154.
  • Brennan D, Peltonen S, Dowling A, Medhat W, Green KJ, Wahl JK III, Del Galdo F, Mahoney MG (2012). A role for caveolin-1 in desmoglein binding and desmosome dynamics. Oncogene. 31: 1636–1648.
  • Brooke MA, Etheridge SL, Kaplan N, Simpson C, O’Toole EA, Ishida-Yamamoto A, Marches O, Getsios S, Kelsell DP (2014). iRHOM2-dependent regulation of ADAM17 in cutaneous disease and epidermal barrier function. Hum Mol Genet. 23: 4064–4076.
  • Brooke MA, Nitoiu D, Kelsell DP (2012). Cell-cell connectivity: desmosomes and disease. J Pathol. 226: 158–171.
  • Brown L, Waseem A, Cruz IN, Szary J, Gunic E, Mannan T, Unadkat M, Yang M, Valderrama F, O’Toole EA, Wan H (2014). Desmoglein 3 promotes cancer cell migration and invasion by regulating activator protein 1 and protein kinase C-dependent-Ezrin activation. Oncogene. 33: 2363–2374.
  • Burdett ID, Sullivan KH (2002). Desmosome assembly in MDCK cells: transport of precursors to the cell surface occurs by two phases of vesicular traffic and involves major changes in centrosome and Golgi location during a Ca2+ shift. Exp Cell Res. 276: 296–309.
  • Calkins CC, Setzer SV, Jennings JM, Summers S, Tsunoda K, Amagai M, Kowalczyk AP (2006). Desmoglein endocytosis and desmosome disassembly are coordinated responses to pemphigus autoantibodies. J Biol Chem. 281: 7623–7634.
  • Chen J, Nekrasova OE, Patel DM, Klessner JL, Godsel LM, Koetsier JL, Amargo EV, Desai BV, Green KJ (2012a). The C-terminal unique region of desmoglein 2 inhibits its internalization via tail-tail interactions. J Cell Biol. 199: 699–711.
  • Chen J, O’Shea C, Fitzpatrick JE, Koster MI, Koch PJ (2012b). Loss of Desmocollin 3 in skin tumor development and progression. Mol Carcinog. 51: 535–545.
  • Chen YJ, Chang JT, Lee L, Wang HM, Liao CT, Chiu CC, Chen PJ, Cheng AJ (2007). DSG3 is overexpressed in head neck cancer and is a potential molecular target for inhibition of oncogenesis. Oncogene. 26: 467–476.
  • Chen YJ, Lee LY, Chao YK, Chang JT, Lu YC, Li HF, Chiu CC, Li YC, Li YL, Chiou JF, Cheng AJ (2013). DSG3 facilitates cancer cell growth and invasion through the DSG3-plakoglobin-TCF/LEF-Myc/cyclin D1/MMP signaling pathway. PLoS One. 8: e64088.
  • Chidgey M, Brakebusch C, Gustafsson E, Cruchley A, Hail C, Kirk S, Merritt A, North A, Tselepis C, Hewitt J, Byrne C, Fässler R, Garrod D (2001). Mice lacking desmocollin 1 show epidermal fragility accompanied by barrier defects and abnormal differentiation. J Cell Biol. 155: 821–832.
  • Chidgey M, Dawson C (2007). Desmosomes: a role in cancer? Brit J Cancer. 96: 1783–1787.
  • Chitaev NA, Troyanovsky SM (1997). Direct Ca2+-dependent heterophilic interaction between desmosomal cadherins, desmoglein and desmocollin, contributes to cell-cell adhesion. J Cell Biol. 138: 193–201.
  • Chun MG, Hanahan D (2010). Genetic deletion of the desmosomal component desmoplakin promotes tumor microinvasion in a mouse model of pancreatic neuroendocrine carcinogenesis. PLoS Genet. 6: e1001120.
  • Cirillo N (2014). 150th anniversary series: desmosomes in physiology and disease. Cell Commun Adhes. 21: 85–88.
  • Cirillo N, Al-Jandan BA (2013). Desmosomal adhesion and pemphigus vulgaris: the first half of the story. Cell Commun Adhes. 20: 1–10.
  • Cirillo N, Femiano F, Gombos F, Lanza A (2007). Metalloproteinase 9 is the outer executioner of desmoglein 3 in apoptotic keratinocytes. Oral Dis. 13: 341–345.
  • Cirillo N, Lanza M, De Rosa A, Cammarota M, La Gatta A, Gombos F, Lanza A (2008). The most widespread desmosomal cadherin, desmoglein 2, is a novel target of caspase 3-mediated apoptotic machinery. J Cell Biochem. 103: 598–606.
  • Cui T, Chen Y, Yang L, Knösel T, Huber O, Pacyna-Gengelbach M, Petersen I (2012a). The p53 target gene desmocollin 3 acts as a novel tumor suppressor through inhibiting EGFR/ERK pathway in human lung cancer. Carcinogenesis. 33: 2326–2333.
  • Cui T, Chen Y, Yang L, Knosel T, Zoller K, Huber O, Petersen I (2011). DSC3 expression is regulated by p53, and methylation of DSC3 DNA is a prognostic marker in human colorectal cancer. Br J Cancer. 104: 1013–1019.
  • Cui T, Chen Y, Yang L, Mireskandari M, Knosel T, Zhang Q, Kohler LH, Kunze A, Presselt N, Petersen I (2012b). Diagnostic and prognostic impact of desmocollins in human lung cancer. J Clin Pathol. 65: 1100–1106.
  • De Bruin A, Muller E, Wurm S, Caldelari R, Wyder M, Wheelock MJ, Suter MM (1999). Loss of invasiveness in squamous cell carcinoma cells overexpressing desmosomal cadherins. Cell Adhes Commun. 7: 13–28.
  • DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB (2008). The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7: 11–20.
  • Delva E, Jennings JM, Calkins CC, Kottke MD, Faundez V, Kowalczyk AP (2008). Pemphigus vulgaris IgG-induced desmoglein-3 endocytosis and desmosomal disassembly are mediated by a clathrin- and dynamin-independent mechanism. J Biol Chem. 283: 18303–18313.
  • Delva E, Kowalczyk AP (2009). Regulation of cadherin trafficking. Traffic. 10: 259–267.
  • Demlehner MP, Schäfer S, Grund C, Franke WW (1995). Continual assembly of half-desmosomal structures in the absence of cell contacts and their frustrated endocytosis: a coordinated Sisyphus cycle. J Cell Biol. 131: 745–760.
  • Dusek RL, Attardi LD (2011). Desmosomes: new perpetuators in tumor suppression. Nat Rev Cancer. 11: 317–323.
  • Dusek RL, Bascom JL, Vogel H, Baron S, Borowsky AD, Bissell MJ, Attardi LD (2012). Deficiency of the p53/p63 target Perp alters mammary gland homeostasis and promotes cancer. Breast Cancer Res. 14: R65.
  • Dusek RL, Getsios S, Chen F, Park JK, Amargo EV, Cryns VL, Green KJ (2006). The differentiation-dependent desmosomal cadherin desmoglein 1 is a novel caspase-3 target that regulates apoptosis in keratinocytes. J Biol Chem. 281: 3614–3624.
  • Dusek RL, Godsel LM, Chen F, Strohecker AM, Getsios S, Harmon R, Müller EJ, Caldelari R, Cryns VL, Green KJ (2007). Plakoglobin deficiency protects keratinocytes from apoptosis. J Invest Dermatol. 127: 792–801.
  • Edme N, Downward J, Thiery JP, Boyer B (2002). Ras induces NBT-II epithelial cell scattering through the coordinate activities of Rac and MAPK pathways. J Cell Sci. 115: 2591–2601.
  • Ellerbroek SM, Halbleib JM, Benavidez M, Warmka JK, Wattenberg EV, Stack MS, Hudson LG (2001). Phosphatidylinositol 3-kinase activity in epidermal growth factor-stimulated matrix metalloproteinase-9 production and cell surface association. Cancer Res. 61: 1855–1861.
  • Ferone G, Mollo MR, Thomason HA, Antonini D, Zhou H, Ambrosio R, De Rosa L, Salvatore D, Getsios S, van Bokhoven H, Dixon J, Missero C (2013). p63 control of desmosome gene expression and adhesion is compromised in AEC syndrome. Hum Mol Genet. 22: 531–543.
  • Franzen CA, Todorovic V, Desai BV, Mirzoeva S, Yang XJ, Green KJ, Pelling JC (2012). The desmosomal armadillo protein plakoglobin regulates prostate cancer cell adhesion and motility through vitronectin-dependent Src signaling. PLoS One. 7: e42132.
  • Funakoshi S, Ezaki T, Kong J, Guo RJ, Lynch JP (2008). Repression of the desmocollin 2 gene expression in human colon cancer cells is relieved by the homeodomain transcription factors Cdx1 and Cdx2. Mol Cancer Res. 6: 1478–1490.
  • Garrod D, Tabernero L (2014). Hyper-adhesion: a unique property of desmosomes. Cell Commun Adhes. 30: 1–8.
  • Garrod DR (2013). The assay that defines desmosome hyperadhesion. J Invest Dermatol. 133: 576–577.
  • Garrod DR, Berika MY, Bardsley WF, Holmes D, Tabernero L (2005). Hyperadhesion in desmosomes: its regulation in wound healing and possible relationship to cadherin crystal structure. J Cell Sci. 118: 5743–5754.
  • Gaudry CA, Palka HL, Dusek RL, Huen AC, Khandekar MJ, Hudson LG, Green KJ (2001). Tyrosine-phosphorylated plakoglobin is associated with desmogleins but not desmoplakin after epidermal growth factor receptor activation. J Biol Chem. 276: 24871–24880.
  • Getsios S, Amargo EV, Dusek RL, Ishii K, Sheu L, Godsel LM, Green KJ (2004). Coordinated expression of desmoglein 1 and desmocollin 1 regulates intercellular adhesion. Differentiation. 72: 419–433.
  • Getsios S, Simpson CL, Kojima S, Harmon R, Sheu LJ, Dusek RL, Cornwell M, Green KJ (2009). Desmoglein 1-dependent suppression of EGFR signaling promotes epidermal differentiation and morphogenesis. J Cell Biol. 185: 1243–1258.
  • Gilmore AP (2005). Anoikis. Cell Death Diff. 12: 1473–1477.
  • Godsel LM, Hsieh SN, Amargo EV, Bass AE, Pascoe-McGillicuddy LT, Huen AC, Thorne ME, Gaudry CA, Park JK, Myung K, Goldman RD, Chew TL, Green KJ (2005). Desmoplakin assembly dynamics in four dimensions: multiple phases differentially regulated by intermediate filaments and actin. J Cell Biol. 171: 1045–1059.
  • Gould JJ, Kenney PA, Rieger-Christ KM, Silva Neto B, Wszolek MF, LaVoie A, Holway AH, Spurrier B, Austin J, Cammarata BK, Canes D, Libertino JA, Summerhayes IC (2010). Identification of tumor and invasion suppressor gene modulators in bladder cancer by different classes of histone deacetylase inhibitors using reverse phase protein arrays. J Urol. 183: 2395–2402.
  • Greening DW, Kapp EA, Ji H, Speed TP, Simpson RJ (2013). Colon tumour secretopeptidome: insights into endogenous proteolytic cleavage events in the colon tumour microenvironment. Biochim Biophys Acta. 1834: 2396–2407.
  • Grivennikov SI, Greten FR, Karin M (2010). Immunity, inflammation, and cancer. Cell. 140: 883–899.
  • Hakimelahi S, Parker HR, Gilchrist AJ, Barry M, Li Z, Bleackley RC, Pasdar M (2000). Plakoglobin regulates the expression of the anti-apoptotic protein Bcl-2. J Biol Chem. 275: 10905–10911.
  • Hamidov Z, Altendorf-Hofmann A, Chen Y, Settmacher U, Petersen I, Knosel T (2011). Reduced expression of desmocollin 2 is an independent prognostic biomarker for shorter patients survival in pancreatic ductal adenocarcinoma. J Clin Pathol. 64: 990–994.
  • Hanahan D, Weinberg RA (2000). The hallmarks of cancer. Cell. 100: 57–70.
  • Hanahan D, Weinberg RA (2011). Hallmarks of cancer: the next generation. Cell. 144: 646–674.
  • Hanakawa Y, Matsuyoshi N, Stanley JR (2002). Expression of desmoglein 1 compensates for genetic loss of desmoglein 3 in keratinocyte adhesion. J Invest Dermatol. 119: 27–31.
  • Harada H, Iwatsuki K, Ohtsuka M, Han GW, Kaneko F (1996). Abnormal desmoglein expression by squamous cell carcinoma cells. Acta Derm Venereol. 76: 417–420.
  • Hardman MJ, Liu K, Avilion AA, Merritt A, Brennan K, Garrod DR, Byrne C (2005). Desmosomal cadherin misexpression alters β-catenin stability and epidermal differentiation. Mol Cell Biol. 25: 969–978.
  • Harmon RM, Green KJ (2013). Structural and functional diversity of desmosomes. Cell Commun Adhes. 20: 171–187.
  • Harmon RM, Simpson CL, Johnson JL, Koetsier JL, Dubash AD, Najor NA, Sarig O, Sprecher E, Green KJ (2013). Desmoglein-1/Erbin interaction suppresses ERK activation to support epidermal differentiation. J Clin Invest. 123: 1556–1570.
  • Hartlieb E, Kempf B, Partilla M, Vigh B, Spindler V, Waschke J (2013). Desmoglein 2 is less important than desmoglein 3 for keratinocyte cohesion. PLoS One. 8: e53739.
  • Hatzfeld M (2007). Plakophilins: Multifunctional proteins or just regulators of desmosomes. Biochim Biophys Acta. 1773: 69–77.
  • Hatzfeld M, Wolf A, Keil R (2014). Plakophilins in desmosomal adhesion and signaling. Cell Commun Adhes. 21: 25–42.
  • He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzler KW (1998). Identification of c-myc as a target of the APC pathway. Science. 281: 1509–1512.
  • He W, Cowin P, Stokes DL (2003). Untangling desmosomal knots with electron tomography. Science. 302: 109–113.
  • Hoffmeyer K, Raggioli A, Rudloff S, Anton R, Hierholzer A, Del Valle I, Hein K, Vogt R, Kemler R (2012). Wnt/β-catenin signaling regulates telomerase in stem cells and cancer cells. Science. 336: 1549–1554.
  • Hoschuetzky H, Aberle H, Kemler R (1994). β-Catenin mediates the interaction of the cadherin-catenin complex with epidermal growth factor receptor. J Cell Biol. 127: 1375–1380.
  • Huang CC, Lee TJ, Chang PH, Lee YS, Chuang CC, Jhang YJ, Chen YW, Chen CW, Tsai CN (2010). Desmoglein 3 is overexpressed in inverted papilloma and squamous cell carcinoma of sinonasal cavity. Laryngoscope. 120: 26–29.
  • Huber O, Weiske J. (2009). Structure and function of desmosomes during apoptosis: Is there an active contribution? In: Cirillo N, Lanza A, Gombos F, (eds.). Pathophysiology of the Desmosme. Research Signpost, Kerala, India. pp. 63–86.
  • Ihrie RA, Attardi LD (2004). Perp-etrating p53-dependent apoptosis. Cell Cycle. 3: 267–269.
  • Ihrie RA, Attardi LD (2005). A new Perp in the lineup: linking p63 and desmosomal adhesion. Cell Cycle. 4: 873–876.
  • Ihrie RA, Marques MR, Nguyen BT, Horner JS, Papazoglu C, Bronson RT, Mills AA, Attardi LD (2005). Perp is a p63-regulated gene essential for epithelial integrity. Cell. 120: 843–856.
  • Ishii K, Norvell SM, Bannon LJ, Amargo EV, Pascoe LT, Green KJ (2001). Assembly of desmosomal cadherins into desmosomes is isoform dependent. J Invest Dermatol. 117: 26–35.
  • Jennings JM, Tucker DK, Kottke MD, Saito M, Delva E, Hanakawa Y, Amagai M, Kowalczyk AP (2011). Desmosome disassembly in response to pemphigus vulgaris IgG occurs in distinct phases and can be reversed by expression of exogenous Dsg3. J Invest Dermatol. 131: 706–718.
  • Jiang K, Rankin CR, Nava P, Sumagin R, Kamekura R, Stowell SR, Feng M, Parkos CA, Nusrat A (2014). Galectin-3 regulates desmoglein-2 and intestinal epithelial intercellular adhesion. J Biol Chem. 289: 10510–10517.
  • Jiang R, Shi Z, Johnson JJ, Liu Y, Stack MS (2011). Kallikrein-5 promotes cleavage of desmoglein-1 and loss of cell-cell cohesion in oral squamous cell carcinoma. J Biol Chem. 286: 9127–9135.
  • Johns SA, Soullier S, Rashbass P, Cunliffe VT (2005). Foxn1 is required for tissue assembly and desmosomal cadherin expression in the hair shaft. Dev Dyn. 232: 1062–1068.
  • Johnson JL, Koetsier JL, Sirico A, Agidi AT, Antonini D, Missero C, Green KJ (2014). The desmosomal protein desmoglein 1 aids recovery of epidermal differentiation after acute UV light exposure. J Invest Dermatol. 134: 2154–2162.
  • Kalinin AE, Kalinin AE, Aho M, Uitto J, Aho S (2005). Breaking the connection: caspase 6 disconnects intermediate filament-binding domain of periplakin from its actin-binding N-terminal region. J Invest Dermatol. 124: 46–55.
  • Kamekura R, Kolegraff KN, Nava P, Hilgarth RS, Feng M, Parkos CA, Nusrat A (2013). Loss of the desmosomal cadherin desmoglein-2 suppresses colon cancer cell proliferation through EGFR signaling. Oncogene. 33: 4531–4536.
  • Kampf C, Relova AJ, Sandler S, Roomans GM (1999). Effects of TNF-α, IFN-γ and IL-β on normal human bronchial epithelial cells. Eur Respir J. 14: 84–91.
  • Kanai Y, Ochiai A, Shibata T, Oyama T, Ushijima S, Akimoto S, Hirohashi S (1995). c-ErbB-2 gene product directly associates with β-catenin and plakoglobin. Biochem Biophys Res Commun. 208: 1067–1072.
  • Ke XS, Qu Y, Rostad K, Li WC, Lin B, Halvorsen OJ, Haukaas SA, Jonassen I, Petersen K, Goldfinger N, Rotter V, Akslen LA, Oyan AM, Kalland KH (2009). Genome-wide profiling of histone H3 lysine 4 and lysine 27 trimethylation reveals an epigenetic signature in prostate carcinogenesis. PLoS One. 4: e4687.
  • Kenchegowda D, Harvey SA, Swamynathan S, Lathrop KL, Swamynathan SK (2012). Critical role of Klf5 in regulating gene expression during post-eyelid opening maturation of mouse corneas. PLoS One. 7: e44771.
  • Khan K, Hardy R, Haq A, Ogunbiyi O, Morton D, Chidgey M (2006). Desmocollin switching in colorectal cancer. Br J Cancer. 95: 1367–1370.
  • Kim C, Wong J, Wen J, Wang S, Wang C, Spiering S, Kan NG, Forcales S, Puri PL, Leone TC, Marine JE, Calkins H, Kelly DP, Judge DP, Chen HS (2013). Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature. 494: 105–110.
  • Klessner JL, Desai BV, Amargo EV, Getsios S, Green KJ (2009). EGFR and ADAMs cooperate to regulate shedding and endocytic trafficking of the desmosomal cadherin desmoglein 2. Mol Biol Cell. 20: 328–337.
  • Kljuic A, Bazzi H, Sundberg JP, Martinez-Mir A, O'Shaugnessy R, Mahoney MG, Levy M, Montagutelli X, Ahmad W, Aita VM, Gordon D, Uitto J, Whiting D, Ott J, Fischer S, Gilliam TC, Jahoda CAB, Morris RJ, Panteleyev AA, Nguyen VT, Christiano AM (2003). Desmoglein 4 in hair follicle differentiation and epidermal adhesion: evidence from inherited hypotrichosis and acquired pemphigus vulgaris. Cell. 113: 249–260.
  • Klus GT, Rokaeus N, Bittner ML, Chen Y, Korz DM, Sukumar S, Schick A, Szallasi Z (2001). Down-regulation of the desmosomal cadherin desmocollin 3 in human breast cancer. Int J Oncol. 19: 169–174.
  • Knösel T, Chen Y, Hotovy S, Settmacher U, Altendorf-Hofmann A, Petersen I (2012). Loss of desmocollin 1–3 and homeobox genes PITX1 and CDX2 are associated with tumor progression and survival in colorectal carcinoma. Int J Colorectal Dis. 27: 1391–1399.
  • Koch PJ, Mahoney MG, Ishikawa H, Pulkkinen L, Uitto J, Shultz L, Murphy GF, Whitaker-Menezes D, Stanley J (1997). Targeted disruption of the pemphigus vulgaris antigen (desmoglein 3) gene in mice causes loss of keratinocyte cell adhesion with a phenotype similar to pemphigus vulgaris. J Cell Biol. 137: 1091–1102.
  • Kolegraff K, Nava P, Helms MN, Parkos CA, Nusrat A (2011). Loss of desmocollin-2 confers a tumorigenic phenotype to colonic epithelial cells through activation of Akt/β-catenin signaling. Mol Biol Cell. 22: 1121–1134.
  • Kume K, Haraguchi M, Hijioka H, Ishida T, Miyawaki A, Nakamura N, Ozawa M (2013). The transcription factor Snail enhanced the degradation of E-cadherin and desmoglein 2 in oral squamous cell carcinoma cells. Biochem Biophys Res Commun. 430: 889–894.
  • Kurzen H, Munzing I, Hartschuh W (2003). Expression of desmosomal proteins in squamous cell carcinomas of the skin. J Cutan Pathol. 30: 621–630.
  • Lanza A, Cirillo N (2007). Caspase-dependent cleavage of desmoglein 1 depends on the apoptotic stimulus. Br J Dermatol. 299: 400–402.
  • Li D, Zhang W, Liu Y, Haneline LS, Shou W (2012). Lack of plakoglobin in epidermis leads to keratoderma. J Biol Chem. 287: 10435–10443.
  • Li G, Schaider H, Satyamoorthy K, Hanakawa Y, Hashimoto K, Herlyn M (2001). Downregulation of E-cadherin and desmoglein 1 by autocrine hepatocyte growth factor during melanoma development. Oncogene. 20: 8125–8135.
  • Li L, Chapman K, Hu X, Wong A, Pasdar M (2007a). Modulation of the oncogenic potential of β-catenin by the subcellular distribution of plakoglobin. Mol Carinog. 46: 824–838.
  • Li Q, Dashwood WM, Zhong X, Nakagama H, Dashwood RH (2007b). Bcl-2 overexpression in PhIP-induced colon tumors: cloning of the rat Bcl-2 promoter and characterization of a pathway involving β-catenin, c-myc and E2F1. Oncogene. 26: 6194–6202.
  • Lorch JH, Klessner J, Park JK, Getsios S, Wu YL, Stack MS, Green KJ (2004). Epidermal growth factor receptor inhibition promotes desmosome assembly and strengthens intercellular adhesion in squamous cell carcinoma cells. J Biol Chem. 279: 37191–37200.
  • Lowndes M, Rakshit S, Shafraz O, Borghi N, Harmon RM, Green KJ, Sivasankar S, Nelson WJ (2014). Different roles of cadherins in the assembly and structural integrity of the desmosome complex. J Cell Sci. 127: 2339–2350.
  • Mahoney MG, Hu Y, Brennan D, Brazzi H, Christiano AM, Wahl JKI (2006). Delineation of diversified desmoglein distribution in stratified squamous epithelia: implications in disease. Exp Dermatol. 15: 101–109.
  • Marcozzi C, Burdett IDJ, Buxton RS, Magee AI (1998). Coexpression of both types of demosomal cadherin and plakoglobin confers strong intercellular adhesion. J Cell Sci. 111: 495–509.
  • Marsden MD, Collins JE, Greenwood MD, Adams MJ, Fleming TP, Magee AI, Buxton RS (1997). Cloning and transcriptional analysis of the promoter of the human type 2 desmocollin gene (DSC2). Gene. 186: 237–247.
  • Merritt AJ, Berika MY, Zhai W, Kirk SE, Ji B, Hardman MJ, Garrod DR (2002). Suprabasal desmoglein 3 expression in the epidermis of transgenic mice results in hyperproliferation and abnormal differentiation. Mol Cell Biol. 22: 5846–5858.
  • Miravet S, Piedra J, Castano J, Raurell I, Franci C, Dunach M, Garcia de Herreros A (2003). Tyrosine phosphorylation of plakoglobin causes contrary effects on its association with desmosomes and adherens junction components and modulates β-catenin-mediated transcription. Mol Cell Biol. 23: 7391–7402.
  • Mlacki M, Darido C, Jane SM, Wilanowski T (2014). Loss of Grainy head-like 1 is associated with disruption of the epidermal barrier and squamous cell carcinoma of the skin. PLoS One. 9: e89247.
  • Moreno V, Gonzalo P, Gomez-Escudero J, Pollan A, Acin-Perez R, Breckenridge M, Yanez-Mo M, Barreiro O, Orsenigo F, Kadomatsu K, Chen CS, Enriquez JA, Dejana E, Sanchez-Madrid F, Arroyo AG (2014). An EMMPRIN/γ-catenin/Nm23 complex drives ATP production and actomyosin contractility at endothelial junctions. J Cell Sci. 127: 3768–3781.
  • Mosesson Y, Mills GB, Yarden Y (2008). Derailed endocytosis: an emerging feature of cancer. Nat Rev Cancer. 8: 835–850.
  • Müller-Tidow C, Steffen B, Cauvet T, Tickenbrock L, Ji P, Diederichs S, Sargin B, Köhler G, Stelljes M, Puccetti E, Ruthardt M, deVos S, Hiebert SW, Koeffler HP, Berdel WE, Serve H (2004). Translocation products in acute myeloid leukemia activate the Wnt signaling pathway in hematopoietic cells. Mol Cell Biol. 24: 2890–2904.
  • Murray B, Wagle R, Amat-Alarcon N, Wilkens A, Stephens P, Zackai EH, Goldmuntz E, Calkins H, Deardorff MA, Judge DP (2013). A family with a complex clinical presentation characterized by arrhythmogenic right ventricular dysplasia/cardiomyopathy and features of branchio-oculo-facial syndrome. Am J Med Genet A. 161A: 371–376.
  • Nagashima H, Okada M, Hidai C, Hosoda S, Kasanuki H, Kawana M (1997). The role of cadherin-catenin-cytoskeleton complex in angiogenesis: antisense oligonucleotide of plakoglobin promotes angiogenesis in vitro, and protein kinase C (PKC) enhances angiogenesis through the plakoglobin signaling pathway. Heart Vessels. Suppl 12: 110–112.
  • Nava P, Laukoetter MG, Hopkins AM, Laur O, Gerner-Smidt K, Green KJ, Parkos CA, Nusrat A (2007). Desmoglein-2: a novel regulator of apoptosis in the intestinal epithelium. Mol Biol Cell. 18: 4565–4578.
  • Nei H, Saito T, Tobioka H, Itoh E, Mori M, Kudo R (1996). Expression of component desmosomal proteins in uterine endometrial carcinoma and their relation to cellular differentiation. Cancer. 78: 461–470.
  • Nekrasova O, Green KJ (2013). Desmosome assembly and dynamics. Trends Cell Biol. 23: 537–546.
  • Nekrasova OE, Amargo EV, Smith WO, Chen J, Kreitzer GE, Green KJ (2011). Desmosomal cadherins utilize distinct kinesins for assembly into desmosomes. J Cell Biol. 195: 1185–1203.
  • Nie Z, Merritt A, Rouhi-Parkouhi M, Tabernero L, Garrod D (2011). Membrane-impermeable cross-linking provides evidence for homophilic, isoform-specific binding of desmosomal cadherins in epithelial cells. J Biol Chem. 286: 2143–2154.
  • Noe V, Fingleton B, Jacobs K, Crawford HC, Vermeulen S, Steelant W, Brunynel E, Matrisian LM, Mareel M (2001). Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci. 114: 111–118.
  • North AJ, Chidgey MAJ, Clarke JP, Bardsley WG, Garrod DR (1996). Distinct desmocollin isoforms occur in the same desmosome and show reciprocally graded distributions in bovine nasal epidermis. Proc Natl Acad Sci USA. 93: 7701–7705.
  • Nottebaum AF, Cagna G, Winderlich M, Gamp AC, Linnepe R, Polaschegg C, Filippova K, Lyck R, Engelhardt B, Kamenyeva O, Bixel MG, Butz S, Vestweber D (2008). VE-PTP maintains the endothelial barrier via plakoglobin and becomes dissociated from VE-cadherin by leukocytes and by VEGF. J Exp Med. 205: 2929–2945.
  • Nuber UA, Schäfer S, Schmidt A, Koch PJ, Franke WW (1995). The widespread human desmocollin Dsc2 and tissue-specific patterns of synthesis of various desmocollin subtypes. Eur J Cell Biol. 66: 69–74.
  • Nuber UA, Schäfer S, Stehr S, Rackwitz HR, Franke WW (1996). Patterns of desmocollin synthesis in human epithelia: immunolocalization of desmocollins 1 and 3 in special epithelia and in cultured cells. Eur J Cell Biol. 71: 1–13.
  • Oka D, Yamashita S, Tomioka T, Nakanishi Y, Kato H, Kaminishi M, Ushijima T (2009). The presence of aberrant DNA methylation in noncancerous esophageal mucosae in association with smoking history: a target for risk diagnosis and prevention of esophageal cancers. Cancer. 115: 3412–3426.
  • Omori-Miyake M, Yamashita M, Tsunemi Y, Kawashima M, Yagi J (2014). In vitro assessment of IL-4- or IL-13-mediated changes in the structural components of keratinocytes in mice and humans. J Invest Dermatol. 134: 1342–1350.
  • Oshiro MM, Kim CJ, Wozniak RJ, Junk DJ, Munoz-Rodriguez JL, Burr JA, Fitzgerald M, Pawar SC, Cress AE, Domann FE, Futscher BW (2005). Epigenetic silencing of Dsc3 is a common event in human breast cancer. Breast Cancer Res. 7: R669–R680.
  • Oshiro MM, Watts GS, Wozniak RJ, Junk DJ, Munoz-Rodriguez JL, Domann FE, Futscher BW (2003). Mutant p53 and aberrant cytosine methylation cooperate to silence gene expression. Oncogene. 22: 3624–3634.
  • Owens P, Bazzi H, Engelking E, Han G, Christiano AM, Wang XJ (2008). Smad4-dependent desmoglein-4 expression contributes to hair follicle integrity. Dev Biol. 322: 156–166.
  • Pan H, Gao F, Papageorgis P, Abdolmaleky HM, Faller DV, Thiagalingam S (2007). Aberrant activation of gamma-catenin promotes genomic instability and oncogenic effects during tumor progression. Cancer Biol Ther. 6: 1638–1643.
  • Pan J, Chen Y, Mo C, Wang D, Chen J, Mao X, Guo S, Zhuang J, Qiu S (2014). Association of Dsc3 mRNA down-regulation in prostate cancer with promoter hypermethylation and poor prognosis. PLoS One. 9: e92815.
  • Patel DM, Green KJ (2014). Desmosomes in the heart: a review of clinical and mechanistic analyses. Cell Commun Adhes. 21: 109–128.
  • Rennel E, Mellberg S, Dimberg A, Petersson L, Botling J, Ameur A, Westholm JO, Komorowski J, Lassalle P, Cross MJ, Gerwins P (2007). Endocan is a VEGF-A and PI3K regulated gene with increased expression in human renal cancer. Exp Cell Res. 313: 1285–1294.
  • Roberts BJ, Pashaj A, Johnson KR, Wahl JK III (2011). Desmosome dynamics in migrating epithelial cells requires the actin cytoskeleton. Exp Cell Res. 317: 2814–2822.
  • Rötzer V, Breit A, Waschke J, Spindler V (2014). Adducin is required for desmosomal cohesion in keratinocytes. J Biol Chem. 289: 14925–14940.
  • Runswick SK, O'Hare MJ, Jones JL, Streuli C, Garrod DR (2001). Desmosomal adhesion regulates epithelial morphogenesis and cell positioning. Nat Cell Biol. 3: 823–830.
  • Ryan KR, Lock FE, Heath JK, Hotchin NA (2012). Plakoglobin-dependent regulation of keratinocyte apoptosis by Rnd3. J Cell Sci. 125: 3202–3209.
  • Santiago-Josefat B, Esselens C, Bech-Serra JJ, Arribas J (2007). Post-transcriptional up-regulation of ADAM17 upon epidermal growth factor receptor activation and in breast tumors. J Biol Chem. 282: 8325–8331.
  • Savagner P, Yamada KM, Thiery JP (1997). The zinc-finger protein slug causes desmosome dissociation, an initial and necessary step for growth factor-induced epithelial-mesenchymal transition. J Cell Biol. 137: 1403–1419.
  • Savci-Heijink CD, Kosari F, Aubry MC, Caron BL, Sun Z, Yang P, Vasmatzis G (2009). The role of desmoglein-3 in the diagnosis of squamous cell carcinoma of the lung. Am J Pathol. 174: 1629–1637.
  • Schnittler HJ, Puschel B, Drenckhahn D (1997). Role of cadherins and plakoglobin in interendothelial adhesion under resting conditions and shear stress. Am J Physiol. 273: H2396–2405.
  • Schüle S, Neuhauser C, Rauchfuss F, Knosel T, Settmacher U, Altendorf-Hofmann A (2014). The influence of desmocollin 1–3 expression on prognosis after curative resection of colorectal liver metastases. Int J Colorectal Dis. 29: 9–14.
  • Shafiei F, Rahnama F, Pawella L, Mitchell MD, Gluckman PD, Lobie PE (2008). DNMT3A and DNMT3B mediate autocrine hGH repression of plakoglobin gene transcription and consequent phenotypic conversion of mammary carcinoma cells. Oncogene. 27: 2602–2612.
  • Sherr CJ, McCormick F (2002). The Rb and p53 pathways in cancer. Cancer Cell. 2: 103–112.
  • Shi HJ, Stubbs R, Hood K (2009). Characterization of de novo synthesized proteins released from human colorectal tumour explants. Electrophoresis. 30: 2442–2453.
  • Shiina H, Breault JE, Basset WW, Enokida H, Urakami S, Li LC, Okino ST, Deguchi M, Kaneuchi M, Terashima M, Yoneda T, Shigeno K, Carroll PR, Igawa M, Dahiya R (2005). Functional loss of the γ-catenin gene through epigenetic and genetic pathways in human prostate cancer. Cancer Res. 65: 2130–2138.
  • Shinohara M, Hiraki A, Ikebe T, Nakamura S, Kurahara S, Shirasuna K, Garrod DR (1998). Immunohistochemical study of desmosomes in oral squamous cell carcinoma: correlation with cytokeratin and E-cadherin staining, and with tumour behaviour. J Pathol. 184: 369–381.
  • Silos SA, Tamai K, Li K, Kivirikko S, Kouba D, Chritiano AM, Uitto J (1996). Cloning of the gene for human pemphigus vulgaris antigen (desmoglein 3), a desmosomal cadherin. Characterization of the promoter region and identification of a keratinocyte-specific cis element. J Biol Chem. 271: 17504–17511.
  • Simpson DL, Patel DM, Green KJ (2011). Deconstructing the skin: cytoarchitectural determinants of epidermal morphogenesis. Nat Rev Mol Cell Biol. 12: 565–580.
  • Spindler V, Waschke J (2014). Desmosomal cadherins and signaling: lessons from autoimmune disease. Cell Commun Adhes. 21: 77–84.
  • Stahley SN, Saito M, Faundez V, Koval M, Mattheyses AL, Kowalczyk AP (2014). Desmosome assembly and disassembly are membrane raft-dependent. PLoS One. 9: e87809.
  • Syed S, Trinnaman B, Martin S, Major S, Hutchinson J, Magee AI (2002). Molecular interactions between desmosomal cadherins. Biochem J. 362: 317–327.
  • Tada H, Hatoko M, Tanaka A, Kuwahara M, Muramatsu T (2000). Expression of desmoglein I and plakoglobin in skin carcinomas. J Cutan Pathol. 27: 24–29.
  • Todorovic V, Desai BV, Patterson MJ, Amargo EV, Dubash AD, Yin T, Jones JC, Green KJ (2010). Plakoglobin regulates cell motility through Rho- and fibronectin-dependent Src signaling. J Cell Sci. 123: 3576–3586.
  • Tokonzaba E, Chen J, Cheng X, Den Z, Ganeshan R, Muller EJ, Koch PJ (2013). Plakoglobin as a regulator of desmocollin gene expression. J Invest Dermatol. 133: 2732–2740.
  • Tsang SM, Brown L, Gadmor H, Gammon L, Fortune F, Wheeler A, Wan H (2012a). Desmoglein 3 acting as an upstream regulator of Rho GTPases, Rac-1/Cdc42 in the regulation of actin organisation and dynamics. Exp Cell Res. 318: 2269–2283.
  • Tsang SM, Brown L, Lin K, Liu L, Piper K, O’Toole EA, Grose R, Hart IR, Garrod DR, Fortune F, Wan H (2012b). Non-junctional human desmoglein 3 acts as an upstream regulator of Src in E-cadherin adhesion, a pathway possibly involved in the pathogenesis of pemphigus vulgaris. J Pathol. 227: 81–93.
  • Tsang SM, Liu L, Teh MT, Wheeler A, Grose R, Hart IR, Garrod DR, Fortune F, Wan H (2010). Desmoglein 3, via an interaction with E-cadherin, is associated with activation of Src. PLoS One. 5: e14211.
  • Tselepis C, Chidgey M, North A, Garrod DR (1998). Desmosomal adhesion inhibits invasive behavior. Proc Natl Acad Sci USA. 95: 8064–8069.
  • Valentijn AJ, Zouq N, Gilmore AP (2004). Anoikis. Biochem Soc Trans. 32: 421–425.
  • van Roy F (2014). Beyond E-cadherin: roles of other cadherin superfamily members in cancer. Nat Rev Cancer. 14: 121–134.
  • Vandewalle C, Comijn J, De Craene B, Vermassen P, Bruyneel E, Andersen H, Tulchinsky E, Van Roy F, Berx G (2005). SIP1/ZEB2 induces EMT by repressing genes of different epithelial cell-cell junctions. Nucleic Acids Res. 33: 6566–6578.
  • Venkiteswaran K, Xiao K, Summers S, Calkins CC, Vincent PA, Pumiglia K, Kowalczyk AP (2002). Regulation of endothelial barrier function and growth by VE-cadherin, plakoglobin, and β-catenin. Am J Physiol Cell Physiol. 283: C811–821.
  • Wang Q, Peng D, Zhu S, Chen Z, Hu T, Soutto M, Saad R, Zhang S, Ei-Rifai W (2014). Regulation of desmocollin3 expression by promoter hypermethylation is associated with advanced esophageal adenocarcinomas. J Cancer. 5: 457–464.
  • Waschke J, Menendez-Castro C, Bruggman P, Koob R, Amagai M, Gruber HJ, Drenckhahn D, Baumgartner W (2007). Imaging and force spectroscopy on desmoglein 1 using atomic force microscopy reveal multivalent Ca2+-dependent, low-affintiy trans-interactions. J Membr Biol. 216: 83–92.
  • Waschke J, Spindler V, Bruggeman P, Zillikens D, Schmidt G, Drenckhahn D (2006). Inhibition of Rho A activity causes pemphigus skin blistering. J Cell Biol. 175: 721–727.
  • Weiske J, Schöneberg T, Schröder W, Hatzfeld M, Tauber R, Huber O (2001). The fate of desmosomal proteins in apoptotic cells. J Biol Chem. 276: 41175–41181.
  • Wheelock MJ, Buck CA, Bechtol KB, Damsky CH (1987). Soluble 80-kd fragment of cell-CAM 120/80 disrupts cell-cell adhesion. J Cell Biochem. 34: 187–202.
  • Wilanowski T, Caddy J, Ting SB, Hislop NR, Cerruti L, Auden A, Zhao LL, Asquith S, Ellis S, Sinclair R, Cunningham JM, Jane SM (2008). Perturbed desmosomal cadherin expression in grainy head-like 1-null mice. EMBO J. 27: 886–897.
  • Williamson L, Raess NA, Caldelari R, Zakher A, de Bruin A, Posthaus H, Bolli R, Hunziker T, Suter MM, Müller EJ (2006). Pemphigus vulgaris identifies plakoglobin as key suppressor of c-Myc in skin. EMBO J. 25: 3298–3309.
  • Winn RA, Bremnes RM, Bemis L, Franklin WA, Miller YE, Cool C, Heasley LE (2002). γ-Catenin expression is reduced or absent in a subset of human lung cancers and re-expression inhibits transformed cell growth. Oncogene. 21: 7497–7506.
  • Wolf A, Rietscher K, Glass M, Hüttelmeier S, Schutkowski M, Ihling C, Sinz A, Wingenfeld A, Mun A, Hatzfeld M (2013). Insulin signaling via Akt2 switches plakophilin 1 function from stabilizing cell adhesion to promoting cell proliferation. J Cell Sci. 126: 1832–1844.
  • Wong MP, Cheang M, Yorida E, Coldman A, Gilks CB, Huntsman D, Berean K (2008). Loss of desmoglein 1 expression associated with worse prognosis in head and neck squamous cell carcinoma patients. Pathology. 40: 611–616.
  • Wu SK, Yap AS (2013). Patterns in space: Coordinating adhesion and actomyosin contractility at E-cadherin junctions. Cell Commun Adhes. 20: 201–212.
  • Xin Z, Yamaguchi A, Sakamoto K (2014). Aberrant expression and altered cellular localization of desmosomal and hemidesmosomal proteins are associated with aggressive clinicopathological features of oral squamous cell carcinoma. Virchows Arch. 465: 35–47.
  • Yang L, Chen Y, Cui T, Knosel T, Zhang Q, Albring KF, Huber O, Petersen I (2012). Desmoplakin acts as a tumor suppressor by inhibition of the Wnt/β-catenin signaling pathway in human lung cancer. Carcinogenesis. 33: 1863–1870.
  • Yashiro M, Nishioka N, Hirakawa K (2006). Decreased expression of the adhesion molecule desmoglein-2 is associated with diffuse-type gastric carcinoma. Eur J Cancer. 42: 2397–2403.
  • Yin T, Getsios S, Caldelari R, Godsel LM, Kowalczyk AP, Muller EJ, Green KJ (2005). Mechanisms of plakoglobin-dependent adhesion: desmosome-specific functions in assembly and regulation by epidermal growth factor receptor. J Biol Chem. 280: 40355–40363.
  • Yin T, Getsios S, Caldelari R, Kowalczyk AP, Müller EJ, Jones JCR, Green KJ (2004). Plakoglobin suppresses keratinocyte motility through both cell-cell adhesion-dependent and -independent mechanisms. Proc Natl Acad Sci USA. 102: 5420–5425.
  • Zhang Y, Toh L, Lau P, Wang X (2012). Human telomerase reverse transcriptase (hTERT) is a novel target of the Wnt/β-catenin pathway in human cancer. J Biol Chem. 287: 32494–32511.
  • Zhou X, Stuart A, Dettin LE, Rodriguez G, Hoel B, Gallicano GI (2004). Desmoplakin is required for microvascular tube formation in culture. J Cell Sci. 117: 3129–3140.
  • Zuckerman JD, Lee WY, DelGaudio JM, Moore CE, Nava P, Nusrat A, Parkos CA (2008). Pathophysiology of nasal polyposis: the role of desmosomal junctions. Am J Rhinol. 22: 589–597.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.