Next Article in Journal
FDG-PET Response Prediction in Pediatric Hodgkin’s Lymphoma: Impact of Metabolically Defined Tumor Volumes and Individualized SUV Measurements on the Positive Predictive Value
Next Article in Special Issue
Complex of MUC1, CIN85 and Cbl in Colon Cancer Progression and Metastasis
Previous Article in Journal / Special Issue
Extracellular Molecules Involved in Cancer Cell Invasion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Desmoglein 3: A Help or a Hindrance in Cancer Progression?

1
Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Center for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Blizard Building, London E1 2AT, UK
2
Department of Cellular and Molecular Physiology, University of Liverpool, Institute of Translational Medicine, Liverpool L69 3BX, UK
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2015, 7(1), 266-286; https://doi.org/10.3390/cancers7010266
Submission received: 26 November 2014 / Revised: 9 January 2015 / Accepted: 16 January 2015 / Published: 26 January 2015
(This article belongs to the Special Issue Cancer Cell Invasion)

Abstract

:
Desmoglein 3 is one of seven desmosomal cadherins that mediate cell-cell adhesion in desmosomes. Desmosomes are the intercellular junctional complexes that anchor the intermediate filaments of adjacent cells and confer strong cell adhesion thus are essential in the maintenance of tissue architecture and structural integrity. Like adherens junctions, desmosomes function as tumour suppressors and are down regulated in the process of epithelial-mesenchymal transition and in tumour cell invasion and metastasis. However, recently several studies have shown that various desmosomal components, including desmoglein 3, are up-regulated in cancer with increased levels of expression correlating with the clinical stage of malignancy, implicating their potentiality to serve as a diagnostic and prognostic marker. Furthermore, in vitro studies have demonstrated that overexpression of desmoglein 3 in cancer cell lines activates several signal pathways that have an impact on cell morphology, adhesion and locomotion. These additional signalling roles of desmoglein 3 may not be associated to its adhesive function in desmosomes but rather function outside of the junctions, acting as a key regulator in the control of actin based cellular processes. This review will discuss recent advances which support the role of desmoglein 3 in cancer progression.

1. Introduction

Desmosomal cadherins constitute the adhesive interface at the core domain of desmosome junctions (DSMs), which in principle function to anchor the intermediate filaments of neighbouring cells to sites of adhesion to form the desmosome-intermediate filament complex essential in the maintenance of tissue architecture and integrity [1]. Since cell-cell adhesion is a prerequisite in many cellular processes DSMs have been implicated in epithelial polarisation, proliferation, stratification, differentiation and morphogenesis as well as embryonic development through their ability to influence intercellular signal transduction [1,2]. A characteristic desmosome is essentially composed of three protein families arranged to form two symmetrical electron dense cytoplasmic plaques between adjacent cells that flank a shared central extracellular core domain [3,4]. Desmosomal cadherins, desmogleins (Dsg) 1–4 and desmocollins (Dsc) 1–3 mediate cell-cell adhesion and serve as a scaffold for the assembly of the cytoplasmic dense plaques. Armadillo proteins, plakoglobin (Pg) and plakophilins (Pkp), associate with the cytoplasmic tails of desmosomal cadherins to form the outer dense plaque which in turn associates with the N-terminal head domain of the Plakin family protein, desmoplakin (Dp), which links the stress-bearing intermediate filaments of the cytoskeleton to desmosome via its C-terminal domain, forming the inner dense plaque [1,5]. Another essential component is a p53 apoptosis effecter related to the PMP-22 (Perp). Although Perp has been proven to be critical in DSM assembly and maintenance its interacting partners currently remain unknown [6,7].
The tumour suppressor function of DSMs is believed to be achieved through mediating strong cell-cell adhesion and sequestering desmosomal components, some of which may have oncogenic potential, to enforce adhesion and to prevent epithelial to mesenchymal transition (EMT) and tumour development [8,9]. Paradoxically, evidence has emerged that shows an upregulation of some desmosomal components, such as Dsg2, Dsg3 and Pkp3 in cancer, in association with cancer progression and/or reduced survival [10,11,12,13,14,15]. There are several reports based on large cohort studies that suggest a pro-cancerous role for Dsg3 with a clear correlation between its expression levels and clinical stage and metastasis [11,15,16,17,18,19]. In line with this, in vitro studies based on overexpression of human Dsg3 in cancer cell lines have shed light on its signalling role that regulates a variety of cellular processes including cell cohesion and migratory behaviour. This review will focus on recent advances with regard to the role of Dsg3 in cancer progression and metastasis.

2. Desmosomes in Tumour Suppression

Intercellular junctions are conventionally accepted to function as tumour suppressors by restricting cell growth through contact-mediated inhibition of cell proliferation and locomotion [20,21]. Disruption of cell-cell adhesion has been shown to contribute significantly to uncontrolled cell proliferation and plays a causal role in EMT, cancer formation, progression and subsequent tumour dissemination (reviewed by [22]). In the process of EMT, dissolution of intercellular junctions including both DSMs and adherens junctions, in addition to other cellular and molecular events, occurs in epithelial cells that facilitates conversion from benign to metastatic tumours [9,23,24]. The tumour suppressor function of cell-cell adhesion is exemplified by the loss of E-cadherin, a hallmark of EMT, and its down-regulation is directly associated with the loss of contact inhibition and tumour development [25,26,27]. In accord, the expression of E-cadherin in cancer cells inhibits cell growth with a concomitant reduction of cell invasiveness [28,29,30]. Similarly the expression of desmosomal components, Dsc1a and Dsc1b, Dsg1 and Pg has been shown to provoke cell cohesion and inhibit motility and invasion [31,32,33]. Correspondingly, the loss of desmosomal components, Perp, Dsg1-3, Dsc2, Dsc3, Pg, Dp and Pkp1-3 is found in the development and progression of squamous cell carcinoma (SqCC), including that of the head and neck, skin, prostate and bladder [8].
The mechanisms by which DSMs suppress tumour formation are as diverse as the molecules that attribute to their function. Many studies suggest that this may be achieved by regulating the availability of desmosomal components with oncogenic potential, such as Pg and Pkps, to suppress tumour development [8,9]. Plakoglobin, a closely related homologue of β-catenin, is shown to exhibit β-catenin-like activity and modulate Wnt/β-catenin signalling by displacing adherens junction proteins [34,35]. This is believed to be achieved through its translocation to the nucleus, complex formation and activation with LEF/TCF transcription factors [35,36]. Indirectly Pg has also been theorised to modulate Wnt/β-catenin signalling by displace adherens junction-associated β-catenin or blocking its cytoplasmic degradation [9,37] to permit its cytoplasmic accumulation, subsequent nuclear translocation and activation of β-catenin-LEF/TCF target genes [36]. Thus it was reasoned that, by sequestering Pg to the plasma membrane DSMs, like adherens junctions, are able to influence Wnt/β-catenin signalling to suppress tumour development [8,9]. However, it is worth noting that Pg has also been shown in numerous studies to negatively regulate Wnt/β-catenin signalling by inhibiting TCF/LEF transcriptional activity through direct interaction [38,39,40] (discussed below).
The newly characterised desmosomal component Perp has emerged as a crucial mediator in tumour suppression through its function on DSM formation as well as its ability to induce apoptosis [8]. This is achieved through its transcriptional regulation by p63 during epithelial development and morphogenesis to promote cell-cell adhesion and also by the p53 tumour suppressor in response to DNA damage and oncogenic stress to induce apoptosis [6,41]. For more detail regarding DSMs in tumour suppression see [9,41].

2.1. Desmoglein 3 in Tumour Suppression

Evidence of the Dsg3 down-regulation in the context of cancer development and progression is comparatively less. However, a reduction of Dsg3 expression has been observed in moderately or poorly differentiated oral SqCC compared with normal oral epithelia [42] and immortalised oral SqCC cell lines compared to mortal oral SqCC and normal oral keratinocytes [43]. Down regulation of Dsg3 is also found to be associated with a loss of differentiation and enhanced metastasis in uterine endometrial and oral squamous cell carcinoma [44,45].

2.2. Lessons from Pemphigus Vulgaris

Our understanding of Dsg3 function is largely based on studies in Pemphigus Vulgaris (PV), a rare autoimmune blistering disorder. Characterised by the loss of keratinocyte cohesion in the oral mucosa and epidermis, PV is caused by cell surface binding of auto-antibodies (PV-IgGs) that target mainly the extracellular domain of Dsg3 and trigger a range of cellular and molecular events. Such studies have uncovered the fundamental role of Dsg3 in cell-cell adhesion as well as in signal transduction. The addition of PV-IgGs to keratinocyte culture has been shown to promote rapid re-organisation of cortical actin filaments [46,47] and to induce the phosphorylation of Dsg3 concomitant with its dissociation from DSMs and rapid endocytosis. Other events observed include increased intracellular calcium concentrations and activation of various signalling molecules such as Pg, PKC, p38 MAPK, heat shock protein p27, Src and c-Myc [46,48,49,50,51,52]. Taken together, these findings support the signalling role of Dsg3 which could potentially contribute to cancer development or suppression. Interestingly, an increased incidence of internal malignancy in the PV patients has been reported in the literature [53,54]. While this association may be attributed to the effect of immunosuppressive therapy administered to PV patients, or as an immunological consequence associated with autoimmune disease, it appears plausible that the development of cancer in these patients could be partly attributed to the loss of Dsg3 function. Further study in carcinogenesis using the active PV model may provide information about the link between PV and tumour development.

3. Desmoglein 3 in Cancer Progression

Despite the traditional view of DSMs as tumour suppressors and the contribution of Dsg3 in desmosomal adhesive function, recent studies have discovered that Dsg3 is upregulated in SqCC of the head and neck, lung, skin and oesophagus etc. [11,15,16,17,18,19]. Furthermore, a correlation between the expression levels of Dsg3 and tumour progression and metastasis has been reported [11,16,55], suggesting that in a given context Dsg3 may contribute to cancer progression. Adopting a gain-of-function approach, overexpression of wild type human Dsg3 in A431 and SqCC/Y1 carcinoma cell lines has been shown to significantly increase cell spreading, membrane protrusion and dynamics as well as cell migration and invasion, a phenotype that could be suppressed by Dsg3 silencing [56,57,58]. These findings are consistent with the reports adopting a loss-of function approach where Dsg3 silencing in oral SqCC cells results in suppression of tumour cell growth, migration and invasion in vitro and in vivo [11,43,59].

4. How Desmoglein 3 Overexpression Promotes Cancer Progression

4.1. Regulation of Rho GTPases and the Actin Cytoskeleton

Cell migration is a highly integrated multistep process initiated by actin based membrane morphological changes, specifically membrane protrusions [60]. The driving forces behind the formation and maintenance of pro-invasive and migratory structures require the intricate spatial and temporal regulation of the actin cytoskeleton [61,62]. Not surprisingly, aberrant modulation and organisation of the actin cytoskeleton plays a pivotal role in cancer progression and metastasis [62]. Mechanistic studies of PV have demonstrated that binding of PV-IgGs to cultured keratinocytes provokes cortical actin reorganisation [46,47,52] through Rho GTPase RhoA and to a lesser extent Rac1 and Cdc42 [46,47,50,51,52]. In the context of cancer development and progression, Rho GTPases are known to play a critical role in processes including tumourigenesis, cell-cycle control, invasion and metastasis [63]. The overexpression of Dsg3 in carcinoma cells has been shown to increase Rac1 and Cdc42 activities and to a lesser extent RhoA, accompanied by pronounced lamellipodia and filopodia and an enhanced rate of actin turnover [58]. These findings support the regulatory activity of Dsg3 in actin organisation and dynamics through the mechanism involved in Rho GTPases in cancer cell biology.

4.2. Organisation of Specialised Membrane Domains

The special regulation of actin based processes requires orchestration in order to achieve a global cellular response [64]. Ezrin, Radixin and Moesin or ERM family proteins are identified to play a key role in these processes through their cross-linking activity and recruitment of multi-protein complex to specific sub-cellular compartments [65]. Enriched in cell-surface structures, such as microvilli, filopodia, uropods and membrane ruffles [66,67,68,69], the ERM proteins have been shown to participate in the formation and maintenance of these structures to modulate cellular processes including polarity, adhesion, spreading and motility [70,71,72,73,74,75,76]. Furthermore, the ERM proteins are also found to be dynamically regulated during cancer progression, with Ezrin being upregulated during early metastatic progression and expansion but down regulated during establishment and survival in metastatic nodes [77,78]. Altered expression, phosphorylation and sub-cellular localisation of Ezrin is also correlating with enhanced migration, invasion and metastasis in a variety of cancers [79,80,81,82,83,84,85,86]. The potential of Ezrin as a downstream effecter of Dsg3 has recently been explored suggesting it plays a part in facilitating Dsg3-dependent regulation of pro-migratory and invasive structure in a malignant setting [56]. In this report, the overexpression of Dsg3 enhanced phosphorylation and localisation of Ezrin at basal plasma membrane domains [56,87], events indicative of the aberrant regulation of Ezrin in cancer progression [88].

4.3. Regulation of Src Signalling

The Src family of non-receptor tyrosine kinases, of which Src, Yes and Fyn are ubiquitously expressed, is a group of enzymes that propagate a diverse spectrum of receptor-induced biological activities through their ability to engage with different classes of cellular receptors and numerous cellular targets [89]. Activated following ligand-receptor engagement, Src is integral to the maintenance of normal cell homeostasis through the regulation of various functions including proliferation, adhesion, differentiation, gene transcription, cytoskeletal alterations and migration [89,90,91]. Not surprisingly, the aberrant modulation of Src activity is associated with abnormalities in specific cell types, tissues and physiological responses [89]. Moreover, the overexpression and/or hyper-activity of Src is associated with a variety of carcinomas, including colorectal, breast, head and neck and lung [90,91,92,93]. While its transforming ability is unlikely [94], the role of Src in tumour progression and metastasis is well established with increased Src activity being associated with hyper-proliferation, EMT, migration, invasion and metastasis [91,95,96]. The proposed mechanisms of the Src activation in a malignant setting are well documented [89,96]. As activating mutations and genomic amplifications in Src are rare, aberrant activation of Src is thought to be accomplished through upstream receptors (direct or indirect) and alteration in upstream kinases such as Csk and phosphatases that influence the intermolecular interactions, localisation and net phospho-status of Src [89,96]. Recent studies suggest that Dsg3 is able to act as a cell surface regulator in Src activation and this signalling pathway seems to be involved in E-cadherin assembly and adhesion. Increased expression of Dsg3 in cancer cell lines has been demonstrated to elicit a significant increase in the tyrosine phosphorylation of Src and its downstream targets of adherens junction proteins, E-cadherin, β-catenin and p120 with a consequence of decreased E-cadherin expression [57,97,98,99]. Src-mediated tyrosine phosphorylation of E-cadherin leads to its ubiquitination and lysosomal degradation [100]. The tyrosine phosphorylation of p120 has been reported to increases its affinity for Rho GTPases to potentially promote cell motility [101,102]. The tyrosine phosphorylation of β-catenin enhances the LEF/TCF transcriptional activity through Wnt/β-catenin signalling pathway.
Although it has been demonstrated that Dsg3 is capable of regulating Src activity it remains unclear how this could be achieved. Emerging evidence suggests that this regulatory pathway may involve caveolin-1, a major constituent of caveolae. Caveolae are a special type of lipid raft of the plasma membrane with a flask-like structure which functions as a regulator in endocytosis and exocytotic vesicle trafficking, cell adhesion and motility [103,104]. It is thought that caveolae modulate signal transduction through the compartmentalisation of specific signalling molecules and the regulation of their activity [105,106,107]. Caveolin-1 is shown to negatively regulate Src activation through an inhibitory interaction which prevents its auto-phosphorylation [106,107]. Amino acid sequence analysis of the Dsg subfamily identified a sequence in Dsg3 that potentially binds the scaffolding domain of caveolin-1 [108]. In support, the same region in Dsg2 has been demonstrated to bind directly to caveolin-1 [109]. It is likely that Dsg3 regulates Src activation by competing with inactive Src for a binding site on the scaffolding domain of caveolin-1 that in turn leads to release of Src followed by its auto-activation. Furthermore, the close association between Dsg3 and caveolin-1 has been demonstrated biochemically and microscopically by several independent studies [108,109,110].

4.4. Regulation of Wnt/β-catenin Signalling

In attempt to elucidate the molecular mechanism of Dsg3-dependent tumour cell migration a recent study by Chen et al. suggests that Dsg3 negatively regulates Wnt/β-catenin signalling in a Pg-dependent manor, most likely by sequestering Pg preventing its nuclear translocation and suppression of LEF/TCF transcriptional activity [59]. In support, other independent studies provide evidence suggesting an inhibitory role for Pg in Wnt/β-catenin/TCF pathway [38,39]. LEF/TCF transcription factors are activated through the Wnt signalling pathway which plays an essential role in development, cell proliferation, survival and migration. Aberrant signalling of Wnt/β-catenin/TCF pathway has been shown to result in defects in embryonic development and a wide range of adult pathologies, most prominently cancer [111,112,113,114]. The study by Chen et al. showed that Dsg3 silencing in head and neck SqCC increased the translocation of Pg to the nucleus where it interacts with and inhibits TCF/LEF transcription activity, to suppress tumour growth and invasion [59]. Correspondingly, increased Dsg3 levels were shown to be correlated with reduced nuclear Pg accompanied with elevated expression of the LEF/TCF transcriptional targets, cyclin D1, c-Myc and MMP7 in both the tissues of head and neck cancer patients and oral SqCC cell lines [59]. Thus, the upregulation of Dsg3 in oral SqCC could potentially tip the balance in the favour of the β-catenin-LEF/TCF interaction and activation. In support, the increased β-catenin expression, nuclear localisation and activity in oral SqCC has been reported in association with increased Wnt signalling [115]. In line with this notion, an in vitro study based on cancer cell lines shows that Dsg3 overexpression increases the tyrosine phosphorylation and activity of β-catenin as well as a reduction of E-cadherin [57,116]. Both the reduction of E-cadherin and tyrosine phosphorylation of β-catenin could promote the cytoplasmic accumulation of β-catenin and its subsequent translocation to the nucleus with a consequence of LEF/TCF transcription activation [117].

4.5. Regulation of the Transcription Factor, Activator Protein-1

Activator protein-1 (AP-1) is a dimeric transcription factor with an expansive transcriptional repertoire propagated by its diverse homo- and hetero-dimeric compositional array of Jun, Atf, Fos and Maf bZIP protein families. The activity of AP-1 is regulated at multiple levels; transcriptionally the abundance of AP-1 components can be influenced by various extracellular signals transduced through MAPK cascade. For example, the activation of JNK in turn activates ternary complex factors (TCFs) to induce the cFos expression [118] whereas the activated p38 induces the cJun expression [119]. Post-transcriptionally, the activity of both pre-existing and newly synthesized AP-1 components can be achieved through the protein phosphorylation. With respect to cJun, PKC-dependent phosphorylation of the N-terminal residues inhibits DNA binding of cJun homodimers [120], while the phosphorylation of cJun at serine 73 and to a lesser exstent serine 63, by JNK enhances its ability to activate transcription [121].
AP-1 is known to regulate a vast array of cellular processes including development, proliferation, apoptosis, immune and stress responses [122,123,124,125]. The deregulation of AP-1 has been associated with a variety of pathologies including inflammatory disease of the bone, skin and liver [126,127] and upregulated in numerous malignancies, including skin, breast, cervix and lung [87,128,129,130,131] through its ability to regulate genes associated with EMT and the migration-invasion programme [24,132,133,134,135,136,137]. Recently, it has been shown that Dsg3 overexpression enhances cJun phosphorylation at serine residues 63 and 73 [56,138] which translated into enhanced c-Jun:AP-1 transcriptional activity that could be abrogated by Dsg3, JNK, PKC, p38 and Src inhibition [56,138]. Furthermore, this MAPK-dependent mechanism appears to be universal as it can be demonstrated in various cell types, including Cos-1 fibroblastic-like cells [138]. In support, other study showed that cJun interacts with TCF4 to form a tertiary complex, containing c-Jun, TCF4 and β-catenin, that binds to cJun promoter thus conferring transcription activity, in a JNK-dependent manner, in intestinal tumourigenesis [139].

4.6. Role of Desmoglein 3 in Carcinogenesis

Although Dsg3 is shown to play a positive role in oncogenesis evidence suggests that Dsg3 may not function as a main driver in cancer formation but rather as a factor that promotes cancer progression. The miss-expression of Dsg3 in the epidermis of transgenic mice driven by the K1 promoter provokes hyper-proliferation, abnormal differentiation and parakeratosis of the epidermis [140]. While these characteristics are also observed in diseases associated with increased cell turnover including SqCC, no evidence of tumour development was reported in this study [140]. Using two models of skin carcinogenesis a recent study investigated the role of Dsg3 in cancer suppression [141]. In the first approach the tumourigenic potential of transformed Dsg3−/− and Dsg3+/− keratinocytes was examined in immune-compromised scid mice. A reduction in tumour formation and growth was observed in mice injected with Dsg3−/− keratinocytes compared with control group injected with Dsg3+/− keratinocytes [141]. In the second approach UVB-induced SqCC formation was analysed in Dsg3/ mice alongside their wild type counterpart. This approach revealed no significant difference in tumour latency, size and multiplicity between the two cohorts [141]. Collectively, these studies suggest that the ablation of Dsg3 does not appear to promote tumour development, implying Dsg3 does not possess tumour suppressive ability. Taken together, these studies are consistent with the notion that Dsg3 plays a positive role in cancer progression and metastasis [11,18,42,55,142].

4.7. The Diagnostic, Prognostic and Therapeutic Potential of Desmoglein 3

With more than 90% of cancer related deaths attributed to metastasis rather than the primary tumour [143], the importance of tackling this aspect of cancer cell behaviour is evident. Squamous cell carcinoma represent the most common cancer capable of metastasis [144]. Despite advances in its diagnosis and treatment [145], the four most common SqCCs, non-melanoma skin cancer, head and neck, oesophageal and non-small cell lung cancer [145], carry among the lowest 5-year survival rates (with the exception of non-melanoma skin cancer) which have not seen significant increases in 30 years [146]. This is in part believed to be attributed to late diagnosis and lack of biological markers which are essential for diagnosis, prognosis and monitoring tumour response to therapy. As the upregulation of Dsg3 has been identified in SqCCs of the head and neck, oesophagus and lung [11,15,16,18,147,148] its potential as a diagnostic and prognostic marker has been explored.
With respect to head and neck SqCC regional lymph node metastasis is common and represents the strongest prognostic factor pertinent to disease staging and treatment strategy selection [149,150,151,152]. With patient survival correlating with the level of nodal involvement [153] and the inherent oversight of micrometastases during clinical staging protocols [154,155], elective nodal dissection (END) followed by pathological examination are carried out as standard procedure. END significantly improves regional recurrence-free survival and lowers the incidences of distal metastasis [156,157,158,159]. Even so, 7%–15% of node-negative patients suffer from recurrence and 50%–70% of patients are over-treated [157,158,160,161,162,163,164]. Sentinel node biopsy presents a feasible and accurate means of avoiding unnecessary END by identifying node negative patients. However, its potential and application is limited by the lack of rapid and accurate methods and markers in the detection of metastatic nodes. Dsg3 has emerged as an accurate biomarker for the detection of lymph node metastasis in head and neck SqCC, discriminating between positive and benign nodes with ~100% accuracy [55,164,165,166]. Furthermore, by adopting the analyses of the Dsg3 expression in sentinel node biopsy using qRT-PCR and microfluidic immunoarray platforms the identification of positive and negative nodes can be achieved within an intraoperative timeframe [55,165,167]. These advances imply that Dsg3 could potentially be used as a maker for clinicians to reduce unnecessary lymph node dissection [168,169] and the frequency of false-negatives to improve diagnostic accuracy and to allow tailoring of treatment strategies for patients with head and neck SqCC.
In addition to head and neck SqCC, Dsg3 has also been shown to be of value in the diagnosis of pulmonary SqCC. Treatment selection for patients diagnosed with lung cancer is predominantly guided by separating lung carcinomas into small cell and non-small cell lung cancers (includes squamous cell carcinoma, adenocarcinoma and large cell carcinoma) and the stage of diseases [170,171,172]. Non-small lung cancers represent a heterogeneous group of cancers that most likely attribute to how patients respond to therapy [173,174]. The current combination of markers including cytokeratin 5/6 and p63 exhibits relatively low sensitivity and/or specificity [18]. Dsg3 has been shown not only to be highly expressed in pulmonary SqCC [17,18,175] but also appears to be a useful ancillary marker to separate pulmonary SqCC from other non-small cell lung cancers [18].
Although Dsg3 has been proposed as a potential therapeutic target in oral SqCC currently limited studies has been reported in the literature. Nevertheless, it was demonstrated by a group of researchers that the RNAi mediated Dsg3 silencing in head and neck SqCC lines reduces tumour growth and metastasis in xenograft studies in BALB/C nude mice [11,59] indicating the therapeutic potential of targeting Dsg3 in preventing cancer progression.

5. Future Perspectives

Although a large body of study regarding Dsg3/PVA has been documented in the literature in pemphigus research highlighting its critical role in cell cohesion, recent advances in the last decade have discovered Dsg3 as a potential oncogene and a positive biomarker of SqCC. There is no doubt that Dsg3 functions as a cell-cell adhesion receptor in DSMs. However, accumulating data suggest that Dsg3 also acts as an important surface regulator for various signal pathways that are involved in actin based cell adhesion, morphological change and locomotion. Like Src and some other signalling molecules, Dsg3 seems to act as a pleiotropic gene having influence on both cell-cell adhesion and cell locomotion depending on the context in study. In normal epithelial cells Dsg3 may function to facilitate cell-cell adhesion but in transformed cells where DSM function is impaired, the overexpression of Dsg3 may favour its oncogenic signalling activity that causes pronounced membrane protrusions and accelerated cell locomotion. However, there are still many questions unanswered. For example, it remains unclear the precise function of Dsg3 in tumour cell biology and whether it acts as a driver in tumour formation? What mechanism regulates its gene expression in normal and cancer cells? Are there any other environmental factors/cues in addition to calcium? Does the overexpression of Dsg3 in cancer involve post-translational regulation, like E-cadherin? As for the Dsg3 signalling, it remains unclear what triggers its activation? Does Dsg3 signalling occur on the cell surface and also in the cytoplasm and if it is on the surface, is it triggered by homophilic ligation, such as trans- and/or cis-binding? It might well be that certain transitional structural conformation of Dsg3 or protein dimerisation on the surface is required for its signalling action and when DSMs are established this structural conformation has undergone a different rearrangement. While this review outlines the potential participation of Dsg3 in cancer cell migration and invasion in a non-adhesive fashion, this by no means negates its role of cell-cell adhesion in collective cell migration and invasion which is an important aspect of the metastatic process [176,177]. Nevertheless, the consistent findings in SqCC have placed Dsg3 as a promising biomarker in cancer and a potential therapeutic target in preventing cancer progression and metastasis. The clarification of this additional function for Dsg3 will advance our understanding the role of Dsg3 in tumour cell biology and may have implication in the development of anti-cancer therapy.

6. Conclusions

It is well established that Dsg3 is targeted by pemphigus autoantibodies and plays a crucial role in pemphigus pathogenesis. Recent studies have shed light that Dsg3 also acts as a key regulator in various intracellular signal pathways that are likely hijacked in cancer and promote cancer cell invasion and dissemination. However, our understanding on the functions of this gene in tumour cell biology remains limited. Further study of Dsg3 in cancer cells and their microenvironment is required to advance our knowledge. Furthermore, in vivo investigation in transgenic model with Dsg3 overexpression will particularly benefit by providing new insights into how exactly this gene is involved in cancer development and progression.

Acknowledgments

The authors would like to thank Medical Research Council, British Skin Foundation and Institute of Dentistry, School of Medicine and Dentistry, Queen Mary University of London for support to the work in the authors’ lab.

Author Contributions

Hong Wan and Louise Brown contributed equally in the preparation, writing and critical editing of this review. All authors read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Garrod, D.; Chidgey, M. Desmosome structure, composition and function. Biochim. Biophys. Acta 2008, 1778, 572–587. [Google Scholar] [CrossRef] [PubMed]
  2. Thomason, H.A.; Scothern, A.; McHarg, S.; Garrod, D.R. Desmosomes: Adhesive strength and signalling in health and disease. Biochem. J. 2010, 429, 419–433. [Google Scholar] [CrossRef] [PubMed]
  3. Farquhar, M.G.; Palade, G.E. Junctional complexes in various epithelia. J. Cell Biol. 1963, 17, 375–412. [Google Scholar] [CrossRef] [PubMed]
  4. Odland, G.F. The fine structure of the interrelationship of cells in the human epidermis. J. Biophys. Biochem. Cytol. 1958, 4, 529–538. [Google Scholar] [CrossRef] [PubMed]
  5. Delva, E.; Tucker, D.K.; Kowalczyk, A.P. The desmosome. Cold Spring Harb. Perspect. Biol. 2009, 1, a002543. [Google Scholar] [CrossRef] [PubMed]
  6. Ihrie, R.A.; Marques, M.R.; Nguyen, B.T.; Horner, J.S.; Papazoglu, C.; Bronson, R.T.; Mills, A.A.; Attardi, L.D. Perp is a p63-regulated gene essential for epithelial integrity. Cell 2005, 120, 843–856. [Google Scholar] [CrossRef] [PubMed]
  7. Marques, M.R.; Ihrie, R.A.; Horner, J.S.; Attardi, L.D. The requirement for perp in postnatal viability and epithelial integrity reflects an intrinsic role in stratified epithelia. J. Investig. Dermatol. 2006, 126, 69–73. [Google Scholar] [CrossRef] [PubMed]
  8. Dusek, R.L.; Attardi, L.D. Desmosomes: New perpetrators in tumour suppression. Nat. Rev. Cancer 2011, 11, 317–323. [Google Scholar] [CrossRef] [PubMed]
  9. Chidgey, M.; Dawson, C. Desmosomes: A role in cancer? Br. J. Cancer 2007, 96, 1783–1787. [Google Scholar] [CrossRef] [PubMed]
  10. Brennan, D.; Mahoney, M.G. Increased expression of Dsg2 in malignant skin carcinomas: A tissue-microarray based study. Cell Adh. Migr. 2009, 3, 148–154. [Google Scholar] [CrossRef] [PubMed]
  11. Chen, Y.J.; Chang, J.T.; Lee, L.; Wang, H.M.; Liao, C.T.; Chiu, C.C.; Chen, P.J.; Cheng, A.J. DSG3 is overexpressed in head neck cancer and is a potential molecular target for inhibition of oncogenesis. Oncogene 2007, 26, 467–476. [Google Scholar] [CrossRef] [PubMed]
  12. Kurzen, H.; Münzing, I.; Hartschuh, W. Expression of desmosomal proteins in squamous cell carcinomas of the skin. J. Cutan. Pathol. 2003, 30, 621–630. [Google Scholar] [CrossRef] [PubMed]
  13. Breuninger, S.; Reidenbach, S.; Sauer, C.G.; Ströbel, P.; Pfitzenmaier, J.; Trojan, L.; Hofmann, I. Desmosomal plakophilins in the prostate and prostatic adenocarcinomas: Implications for diagnosis and tumor progression. Am. J. Pathol. 2010, 176, 2509–2519. [Google Scholar] [CrossRef] [PubMed]
  14. Furukawa, C.; Daigo, Y.; Ishikawa, N.; Kato, T.; Ito, T.; Tsuchiya, E.; Sone, S.; Nakamura, Y. Plakophilin 3 oncogene as prognostic marker and therapeutic target for lung cancer. Cancer Res. 2005, 65, 7102–7110. [Google Scholar] [CrossRef] [PubMed]
  15. Huang, C.C.; Lee, T.J.; Chang, P.H.; Lee, Y.S.; Chuang, C.C.; Jhang, Y.J.; Chen, Y.W.; Chen, C.W.; Tsai, C.N. Desmoglein 3 is overexpressed in inverted papilloma and squamous cell carcinoma of sinonasal cavity. Laryngoscope 2010, 120, 26–29. [Google Scholar] [CrossRef] [PubMed]
  16. Fang, W.K.; Chen, B.; Xu, X.E.; Liao, L.D.; Wu, Z.Y.; Wu, J.Y.; Shen, J.; Xu, L.Y.; Li, E.M. Altered expression and localization of desmoglein 3 in esophageal squamous cell carcinoma. Acta Histochem. 2014, 116, 803–809. [Google Scholar] [CrossRef]
  17. Fukuoka, J.; Dracheva, T.; Shih, J.H.; Hewitt, S.M.; Fujii, T.; Kishor, A.; Mann, F.; Shilo, K.; Franks, T.J.; Travis, W.D.; et al. Desmoglein 3 as a prognostic factor in lung cancer. Hum. Pathol. 2007, 38, 276–283. [Google Scholar] [CrossRef]
  18. Savci-Heijink, C.D.; Kosari, F.; Aubry, M.C.; Caron, B.L.; Sun, Z.; Yang, P.; Vasmatzis, G. The role of desmoglein-3 in the diagnosis of squamous cell carcinoma of the lung. Am. J. Pathol. 2009, 174, 1629–1637. [Google Scholar] [CrossRef] [PubMed]
  19. Liu, Z.; Tian, Y.; Ma, F.; Zhu, L.; Hu, Y. Expression of desmoglein 3 in nasopharyngeal carcinoma: Research of 22 cases. Zhonghua Yi Xue Za Zhi 2007, 87, 2541–2543. [Google Scholar] [PubMed]
  20. Levine, E.M.; Becker, Y.; Boone, C.W.; Eagle, H. Contact inhibition, macromolecular synthesis, and polyribosomes in cultured human diploid fibroblasts. Proc. Natl. Acad. Sci. USA 1965, 53, 350–356. [Google Scholar] [CrossRef] [PubMed]
  21. Okegawa, T.; Li, Y.; Pong, R.C.; Hsieh, J.T. Cell adhesion proteins as tumor suppressors. J. Urol. 2002, 167, 1836–1843. [Google Scholar] [CrossRef] [PubMed]
  22. Moh, M.C.; Shen, S. The roles of cell adhesion molecules in tumor suppression and cell migration: A new paradox. Cell Adh. Migr. 2009, 3, 334–336. [Google Scholar] [CrossRef] [PubMed]
  23. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [PubMed]
  24. Davies, M.; Robinson, M.; Smith, E.; Huntley, S.; Prime, S.; Paterson, I. Induction of an epithelial to mesenchymal transition in human immortal and malignant keratinocytes by TGF-beta1 involves MAPK, Smad and AP-1 signalling pathways. J. Cell Biochem. 2005, 95, 918–931. [Google Scholar] [CrossRef] [PubMed]
  25. Pećina-Slaus, N. Tumor suppressor gene E-cadherin and its role in normal and malignant cells. Cancer Cell Int. 2003, 3, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Fanjul-Fernández, M.; Quesada, V.; Cabanillas, R.; Cadiñanos, J.; Fontanil, T.; Obaya, A.; Ramsay, A.J.; Llorente, J.L.; Astudillo, A.; Cal, S.; et al. Cell-cell adhesion genes CTNNA2 and CTNNA3 are tumour suppressors frequently mutated in laryngeal carcinomas. Nat. Commun. 2013, 4, 2531. [Google Scholar] [CrossRef]
  27. Yue, T.; Tian, A.; Jiang, J. The cell adhesion molecule echinoid functions as a tumor suppressor and upstream regulator of the Hippo signaling pathway. Dev. Cell 2012, 22, 255–267. [Google Scholar] [CrossRef] [PubMed]
  28. Frixen, U.H.; Behrens, J.; Sachs, M.; Eberle, G.; Voss, B.; Warda, A.; Löchner, D.; Birchmeier, W. E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. J. Cell Biol. 1991, 113, 173–185. [Google Scholar] [CrossRef] [PubMed]
  29. Vleminckx, K.; Vakaet, L.; 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. [Google Scholar] [CrossRef] [PubMed]
  30. Watabe, M.; Nagafuchi, A.; Tsukita, S.; Takeichi, M. Induction of polarized cell-cell association and retardation of growth by activation of the E-cadherin-catenin adhesion system in a dispersed carcinoma line. J. Cell Biol. 1994, 127, 247–256. [Google Scholar] [CrossRef] [PubMed]
  31. Tselepis, C.; Chidgey, M.; North, A.; Garrod, D. Desmosomal adhesion inhibits invasive behavior. Proc. Natl. Acad. Sci. USA 1998, 95, 8064–8069. [Google Scholar] [CrossRef] [PubMed]
  32. Yin, T.; Getsios, S.; Caldelari, R.; Godsel, L.M.; Kowalczyk, A.P.; Müller, E.J.; Green, K.J. Mechanisms of plakoglobin-dependent adhesion: Desmosome-specific functions in assembly and regulation by epidermal growth factor receptor. J. Biol. Chem. 2005, 280, 40355–40363. [Google Scholar] [CrossRef] [PubMed]
  33. Yin, T.; Getsios, S.; Caldelari, R.; Kowalczyk, A.P.; Müller, E.J.; Jones, J.C.; Green, K.J. Plakoglobin suppresses keratinocyte motility through both cell-cell adhesion-dependent and -independent mechanisms. Proc. Natl. Acad. Sci. USA 2005, 102, 5420–5425. [Google Scholar] [CrossRef] [PubMed]
  34. Maeda, O.; Usami, N.; Kondo, M.; Takahashi, M.; Goto, H.; Shimokata, K.; Kusugami, K.; Sekido, Y. Plakoglobin (gamma-catenin) has TCF/LEF family-dependent transcriptional activity in beta-catenin-deficient cell line. Oncogene 2004, 23, 964–972. [Google Scholar] [CrossRef] [PubMed]
  35. Conacci-Sorrell, M.E.; Ben-Yedidia, T.; Shtutman, M.; Feinstein, E.; Einat, P.; Ben-Ze’ev, A. Nr-CAM is a target gene of the beta-catenin/LEF-1 pathway in melanoma and colon cancer and its expression enhances motility and confers tumorigenesis. Genes Dev. 2002, 16, 2058–2072. [Google Scholar] [CrossRef] [PubMed]
  36. Zhurinsky, J.; Shtutman, M.; Ben-Ze’ev, A. Plakoglobin and beta-catenin: Protein interactions, regulation and biological roles. J. Cell Sci. 2000, 113, 3127–3139. [Google Scholar] [PubMed]
  37. Merriam, J.M.; Rubenstein, A.B.; Klymkowsky, M.W. Cytoplasmically anchored plakoglobin induces a wnt-like phenotype in xenopus. Dev. Biol. 1997, 185, 67–81. [Google Scholar] [CrossRef] [PubMed]
  38. Miravet, S.; Piedra, J.; Miró, F.; Itarte, E.; García de Herreros, A.; Duñach, M. The transcriptional factor TCF-4 contains different binding sites for beta-catenin and plakoglobin. J. Biol. Chem. 2002, 277, 1884–1891. [Google Scholar] [CrossRef] [PubMed]
  39. Garcia-Gras, E.; Lombardi, R.; Giocondo, M.J.; Willerson, J.T.; Schneider, M.D.; Khoury, D.S.; Marian, A.J. Suppression of canonical Wnt/beta-catenin signaling by nuclear plakoglobin recapitulates phenotype of arrhythmogenic right ventricular cardiomyopathy. J. Clin. Investig 2006, 116, 2012–2021. [Google Scholar] [CrossRef] [PubMed]
  40. Williamson, L.; Raess, N.A.; Caldelari, R.; Zakher, A.; de Bruin, A.; Posthaus, H.; Bolli, R.; Hunziker, T.; Suter, M.M.; Müller, E.J. Pemphigus vulgaris identifies plakoglobin as key suppressor of c-Myc in the skin. EMBO J. 2006, 25, 3298–3309. [Google Scholar] [CrossRef] [PubMed]
  41. Attardi, L.D.; Reczek, E.E.; Cosmas, C.; Demicco, E.G.; McCurrach, M.E.; Lowe, S.W.; Jacks, T. PERP, an apoptosis-associated target of p53, is a novel member of the PMP-22/gas3 family. Genes Dev. 2000, 14, 704–718. [Google Scholar] [PubMed]
  42. Wang, L.; Liu, T.; Wang, Y.; Cao, L.; Nishioka, M.; Aguirre, R.L.; Ishikawa, A.; Geng, L.; Okada, N. Altered expression of desmocollin 3, desmoglein 3, and beta-catenin in oral squamous cell carcinoma: Correlation with lymph node metastasis and cell proliferation. Virchows Arch. 2007, 451, 959–966. [Google Scholar] [CrossRef] [PubMed]
  43. Teh, M.T.; Parkinson, E.K.; Thurlow, J.K.; Liu, F.; Fortune, F.; Wan, H. A molecular study of desmosomes identifies a desmoglein isoform switch in head and neck squamous cell carcinoma. J. Oral. Pathol. Med. 2011, 40, 67–76. [Google Scholar] [CrossRef] [PubMed]
  44. Nei, H.; Saito, T.; Tobioka, H.; Itoh, E.; Mori, M.; Kudo, R. Expression of component desmosomal proteins in uterine endometrial carcinoma and their relation to cellular differentiation. Cancer 1996, 78, 461–470. [Google Scholar] [CrossRef] [PubMed]
  45. Xin, Z.; Yamaguchi, A.; Sakamoto, K. Aberrant expression and altered cellular localization of desmosomal and hemidesmosomal proteins are associated with aggressive clinicopathological features of oral squamous cell carcinoma. Virchows Arch. 2014, 465, 35–47. [Google Scholar] [CrossRef] [PubMed]
  46. Berkowitz, P.; Hu, P.; Liu, Z.; Diaz, L.A.; Enghild, J.J.; Chua, M.P.; Rubenstein, D.S. Desmosome signaling. Inhibition of p38MAPK prevents pemphigus vulgaris IgG-induced cytoskeleton reorganization. J. Biol. Chem. 2005, 280, 23778–23784. [Google Scholar] [CrossRef] [PubMed]
  47. Gliem, M.; Heupel, W.M.; Spindler, V.; Harms, G.S.; Waschke, J. Actin reorganization contributes to loss of cell adhesion in pemphigus vulgaris. Am. J. Physiol. Cell Physiol. 2010, 299, C606–C613. [Google Scholar] [CrossRef] [PubMed]
  48. Kawasaki, Y.; Aoyama, Y.; Tsunoda, K.; Amagai, M.; Kitajima, Y. Pathogenic monoclonal antibody against desmoglein 3 augments desmoglein 3 and p38 MAPK phosphorylation in human squamous carcinoma cell line. Autoimmunity 2006, 39, 587–590. [Google Scholar] [CrossRef] [PubMed]
  49. Kitajima, Y.; Aoyama, Y.; Seishima, M. Transmembrane signaling for adhesive regulation of desmosomes and hemidesmosomes, and for cell-cell datachment induced by pemphigus IgG in cultured keratinocytes: Involvement of protein kinase C. J. Investig. Dermatol. Symp. Proc. 1999, 4, 137–144. [Google Scholar] [CrossRef] [PubMed]
  50. Spindler, V.; Waschke, J. Role of Rho GTPases in desmosomal adhesion and pemphigus pathogenesis. Ann. Anat. 2011, 193, 177–180. [Google Scholar] [CrossRef] [PubMed]
  51. Spindler, V.; Endlich, A.; Hartlieb, E.; Vielmuth, F.; Schmidt, E.; Waschke, J. The extent of desmoglein 3 depletion in pemphigus vulgaris is dependent on Ca2+-induced differentiation a role in suprabasal epidermal skin splitting? Am. J. Pathol. 2011, 179, 1905–1916. [Google Scholar] [CrossRef] [PubMed]
  52. Waschke, J.; Spindler, V.; Bruggeman, P.; Zillikens, D.; Schmidt, G.; Drenckhahn, D. Inhibition of Rho A activity causes pemphigus skin blistering. J. Cell Biol. 2006, 175, 721–727. [Google Scholar] [CrossRef] [PubMed]
  53. Krain, L.S.; Bierman, S.M. Pemphigus vulgaris and internal malignancy. Cancer 1974, 33, 1091–1099. [Google Scholar] [CrossRef] [PubMed]
  54. Ogawa, H.; Sakuma, M.; Morioka, S.; Kitamura, K.; Sasai, Y.; Imamura, S.; Inaba, Y. The incidence of internal malignancies in pemphigus and bullous pemphigoid in Japan. J. Dermatol. Sci. 1995, 9, 136–141. [Google Scholar] [CrossRef] [PubMed]
  55. Patel, V.; Martin, D.; Malhotra, R.; Marsh, C.A.; Doçi, C.L.; Veenstra, T.D.; Nathan, C.A.; Sinha, U.K.; Singh, B.; Molinolo, A.A.; et al. DSG3 as a biomarker for the ultrasensitive detection of occult lymph node metastasis in oral cancer using nanostructured immunoarrays. Oral. Oncol. 2013, 49, 93–101. [Google Scholar] [CrossRef] [PubMed]
  56. Brown, L.; Waseem, A.; Cruz, I.N.; Szary, J.; Gunic, E.; Mannan, T.; Unadkat, M.; Yang, M.; Valderrama, F.; O’Toole, E.A.; et al. Desmoglein 3 promotes cancer cell migration and invasion by regulating activator protein 1 and protein kinase C-dependent-Ezrin activation. Oncogene 2014, 33, 2363–2374. [Google Scholar] [CrossRef] [PubMed]
  57. Tsang, S.M.; Liu, L.; Teh, M.T.; Wheeler, A.; Grose, R.; Hart, I.R.; Garrod, D.R.; Fortune, F.; Wan, H. Desmoglein 3, via an interaction with E-cadherin, is associated with activation of Src. PLOS ONE 2010, 5, e14211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Tsang, S.M.; Brown, L.; Gadmor, H.; Gammon, L.; Fortune, F.; Wheeler, A.; Wan, H. Desmoglein 3 acting as an upstream regulator of Rho GTPases, Rac-1/Cdc42 in the regulation of actin organisation and dynamics. Exp. Cell Res. 2012, 318, 2269–2283. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, Y.J.; Lee, L.Y.; Chao, Y.K.; Chang, J.T.; Lu, Y.C.; Li, H.F.; Chiu, C.C.; Li, Y.C.; Li, Y.L.; Chiou, J.F.; et al. DSG3 facilitates cancer cell growth and invasion through the DSG3-plakoglobin-TCF/LEF-Myc/cyclin D1/MMP signaling pathway. PLOS ONE 2013, 8, e64088. [Google Scholar] [CrossRef] [PubMed]
  60. Bailly, M.; Condeelis, J. Cell motility: Insights from the backstage. Nat. Cell Biol. 2002, 4, E292–E294. [Google Scholar] [CrossRef] [PubMed]
  61. Pollard, T.D.; Borisy, G.G. Cellular motility driven by assembly and disassembly of actin filaments. Cell 2003, 112, 453–465. [Google Scholar] [CrossRef] [PubMed]
  62. Sahai, E. Mechanisms of cancer cell invasion. Curr. Opin. Genet. Dev. 2005, 15, 87–96. [Google Scholar] [CrossRef] [PubMed]
  63. Sahai, E.; Marshall, C.J. Rho-GTPases and cancer. Nat. Rev. Cancer 2002, 2, 133–142. [Google Scholar] [CrossRef] [PubMed]
  64. Rafelski, S.M.; Theriot, J.A. Crawling toward a unified model of cell mobility: Spatial and temporal regulation of actin dynamics. Annu. Rev. Biochem. 2004, 73, 209–239. [Google Scholar] [CrossRef] [PubMed]
  65. Arpin, M.; Chirivino, D.; Naba, A.; Zwaenepoel, I. Emerging role for ERM proteins in cell adhesion and migration. Cell Adh. Migr. 2011, 5, 199–206. [Google Scholar] [CrossRef] [PubMed]
  66. Bretscher, A. Regulation of cortical structure by the ezrin-radixin-moesin protein family. Curr. Opin. Cell Biol. 1999, 11, 109–116. [Google Scholar] [CrossRef] [PubMed]
  67. Sato, N.; Yonemura, S.; Obinata, T.; Tsukita, S. Radixin, a barbed end-capping actin-modulating protein, is concentrated at the cleavage furrow during cytokinesis. J. Cell Biol. 1991, 113, 321–330. [Google Scholar] [CrossRef] [PubMed]
  68. Amieva, M.R.; Furthmayr, H. Subcellular localization of moesin in dynamic filopodia, retraction fibers, and other structures involved in substrate exploration, attachment, and cell-cell contacts. Exp. Cell Res. 1995, 219, 180–196. [Google Scholar] [CrossRef] [PubMed]
  69. Serrador, J.M.; Alonso-Lebrero, J.L.; del Pozo, M.A.; Furthmayr, H.; Schwartz-Albiez, R.; Calvo, J.; Lozano, F.; Sánchez-Madrid, F. Moesin interacts with the cytoplasmic region of intercellular adhesion molecule-3 and is redistributed to the uropod of T lymphocytes during cell polarization. J. Cell Biol. 1997, 138, 1409–1423. [Google Scholar] [CrossRef] [PubMed]
  70. Paglini, G.; Kunda, P.; Quiroga, S.; Kosik, K.; Cáceres, A. Suppression of radixin and moesin alters growth cone morphology, motility, and process formation in primary cultured neurons. J. Cell Biol. 1998, 143, 443–455. [Google Scholar] [CrossRef]
  71. Lamb, R.F.; Ozanne, B.W.; Roy, C.; McGarry, L.; Stipp, C.; Mangeat, P.; Jay, D.G. Essential functions of ezrin in maintenance of cell shape and lamellipodial extension in normal and transformed fibroblasts. Curr. Biol. 1997, 7, 682–688. [Google Scholar] [CrossRef] [PubMed]
  72. Takeuchi, K.; Sato, N.; Kasahara, H.; Funayama, N.; Nagafuchi, A.; Yonemura, S.; Tsukita, S. Perturbation of cell adhesion and microvilli formation by antisense oligonucleotides to ERM family members. J. Cell Biol. 1994, 125, 1371–1384. [Google Scholar] [CrossRef] [PubMed]
  73. Mak, H.; Naba, A.; Varma, S.; Schick, C.; Day, A.; SenGupta, S.K.; Arpin, M.; Elliott, B.E. Ezrin phosphorylation on tyrosine 477 regulates invasion and metastasis of breast cancer cells. BMC Cancer 2012, 12, 82. [Google Scholar] [CrossRef] [PubMed]
  74. Baumgartner, M.; Sillman, A.L.; Blackwood, E.M.; Srivastava, J.; Madson, N.; Schilling, J.W.; Wright, J.H.; Barber, D.L. The Nck-interacting kinase phosphorylates erm proteins for formation of lamellipodium by growth factors. Proc. Natl. Acad. Sci. USA 2006, 103, 13391–13396. [Google Scholar] [CrossRef] [PubMed]
  75. Gandy, K.A.; Canals, D.; Adada, M.; Wada, M.; Roddy, P.; Snider, A.J.; Hannun, Y.A.; Obeid, L.M. Sphingosine 1-phosphate induces filopodia formation through S1PR2 activation of ERM proteins. Biochem. J. 2013, 449, 661–672. [Google Scholar] [CrossRef] [PubMed]
  76. Zhu, L.; Crothers, J., Jr.; Zhou, R.; Forte, J.G. A possible mechanism for ezrin to establish epithelial cell polarity. Am. J. Physiol. Cell Physiol. 2010, 299, C431–C443. [Google Scholar] [CrossRef] [PubMed]
  77. Ren, L.; Hong, S.H.; Cassavaugh, J.; Osborne, T.; Chou, A.J.; Kim, S.Y.; Gorlick, R.; Hewitt, S.M.; Khanna, C. The actin-cytoskeleton linker protein ezrin is regulated during osteosarcoma metastasis by PKC. Oncogene 2009, 28, 792–802. [Google Scholar] [CrossRef] [PubMed]
  78. Khanna, C.; Wan, X.; Bose, S.; Cassaday, R.; Olomu, O.; Mendoza, A.; Yeung, C.; Gorlick, R.; Hewitt, S.M.; Helman, L.J. The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis. Nat. Med. 2004, 10, 182–186. [Google Scholar] [CrossRef] [PubMed]
  79. Chen, Y.; Wang, D.; Guo, Z.; Zhao, J.; Wu, B.; Deng, H.; Zhou, T.; Xiang, H.; Gao, F.; Yu, X.; et al. Rho kinase phosphorylation promotes ezrin-mediated metastasis in hepatocellular carcinoma. Cancer Res. 2011, 71, 1721–1729. [Google Scholar] [CrossRef] [PubMed]
  80. Chuan, Y.C.; Pang, S.T.; Cedazo-Minguez, A.; Norstedt, G.; Pousette, A.; Flores-Morales, A. Androgen induction of prostate cancer cell invasion is mediated by ezrin. J. Biol. Chem. 2006, 281, 29938–29948. [Google Scholar] [CrossRef] [PubMed]
  81. Donatello, S.; Babina, I.S.; Hazelwood, L.D.; Hill, A.D.; Nabi, I.R.; Hopkins, A.M. Lipid raft association restricts CD44-ezrin interaction and promotion of breast cancer cell migration. Am. J. Pathol. 2012, 181, 2172–2187. [Google Scholar] [CrossRef] [PubMed]
  82. Elliott, B.E.; Meens, J.A.; SenGupta, S.K.; Louvard, D.; Arpin, M. The membrane cytoskeletal crosslinker ezrin is required for metastasis of breast carcinoma cells. Breast Cancer Res. 2005, 7, R365–R373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Heiska, L.; Melikova, M.; Zhao, F.; Saotome, I.; McClatchey, A.I.; Carpén, O. Ezrin is key regulator of Src-induced malignant phenotype in three-dimensional environment. Oncogene 2011, 30, 4953–4962. [Google Scholar] [CrossRef] [PubMed]
  84. Ma, L.; Jiang, T. Clinical implications of ezrin and CD44 co-expression in breast cancer. Oncol. Rep. 2013, 30, 1899–1905. [Google Scholar] [PubMed]
  85. Meng, Y.; Lu, Z.; Yu, S.; Zhang, Q.; Ma, Y.; Chen, J. Ezrin promotes invasion and metastasis of pancreatic cancer cells. J. Transl. Med. 2010, 8, 61. [Google Scholar] [CrossRef] [PubMed]
  86. Xie, J.J.; Xu, L.Y.; Wu, Z.Y.; Zhao, Q.; Xu, X.E.; Wu, J.Y.; Huang, Q.; Li, E.M. Prognostic implication of ezrin expression in esophageal squamous cell carcinoma. J. Surg. Oncol. 2011, 104, 538–543. [Google Scholar] [CrossRef] [PubMed]
  87. Shen, Q.; Uray, I.P.; Li, Y.; Krisko, T.I.; Strecker, T.E.; Kim, H.T.; Brown, P.H. The AP-1 transcription factor regulates breast cancer cell growth via cyclins and E2F factors. Oncogene 2008, 27, 366–377. [Google Scholar] [CrossRef] [PubMed]
  88. Clucas, J.; Valderrama, F. ERM proteins in cancer progression. J. Cell Sci. 2014, 127, 267–275. [Google Scholar] [CrossRef] [PubMed]
  89. Thomas, S.M.; Brugge, J.S. Cellular functions regulated by Src family kinases. Annu. Rev. Cell Dev. Biol. 1997, 13, 513–609. [Google Scholar] [CrossRef] [PubMed]
  90. Yeatman, T.J. A renaissance for Src. Nat. Rev. Cancer 2004, 4, 470–480. [Google Scholar] [CrossRef] [PubMed]
  91. Irby, R.B.; Yeatman, T.J. Role of Src expression and activation in human cancer. Oncogene 2000, 19, 5636–5642. [Google Scholar] [CrossRef] [PubMed]
  92. Summy, J.M.; Gallick, G.E. Src family kinases in tumor progression and metastasis. Cancer Metastasis Rev. 2003, 22, 337–358. [Google Scholar] [CrossRef] [PubMed]
  93. Shor, A.C.; Keschman, E.A.; Lee, F.Y.; Muro-Cacho, C.; Letson, G.D.; Trent, J.C.; Pledger, W.J.; Jove, R. Dasatinib inhibits migration and invasion in diverse human sarcoma cell lines and induces apoptosis in bone sarcoma cells dependent on Src kinase for survival. Cancer Res. 2007, 67, 2800–2808. [Google Scholar] [CrossRef] [PubMed]
  94. Biscardi, J.S.; Tice, D.A.; Parsons, S.J. C-Src, receptor tyrosine kinases, and human cancer. Adv. Cancer Res. 1999, 76, 61–119. [Google Scholar] [PubMed]
  95. Guarino, M. Src signaling in cancer invasion. J. Cell Physiol. 2010, 223, 14–26. [Google Scholar] [PubMed]
  96. Sen, B.; Johnson, F.M. Regulation of Src family kinases in human cancers. J. Signal. Transduct. 2011, 2011, 865819. [Google Scholar] [PubMed]
  97. Tsang, S.M.; Brown, L.; Lin, K.; Liu, L.; Piper, K.; O’Toole, E.A.; Grose, R.; Hart, I.R.; Garrod, D.R.; Fortune, F.; et al. 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. 2012, 227, 81–93. [Google Scholar] [CrossRef] [PubMed]
  98. McLachlan, R.W.; Kraemer, A.; Helwani, F.M.; Kovacs, E.M.; Yap, A.S. E-cadherin adhesion activates C-Src signaling at cell-cell contacts. Mol. Biol. Cell 2007, 18, 3214–3223. [Google Scholar] [CrossRef] [PubMed]
  99. McLachlan, R.W.; Yap, A.S. Not so simple: The complexity of phosphotyrosine signaling at cadherin adhesive contacts. J. Mol. Med. (Berl.) 2007, 85, 545–554. [Google Scholar] [CrossRef]
  100. Shen, Y.; Hirsch, D.S.; Sasiela, C.A.; Wu, W.J. Cdc42 regulates E-cadherin ubiquitination and degradation through an epidermal growth factor receptor to Src-mediated pathway. J. Biol. Chem. 2008, 283, 5127–5137. [Google Scholar] [CrossRef] [PubMed]
  101. Grosheva, I.; Shtutman, M.; Elbaum, M.; Bershadsky, A.D. 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. [Google Scholar] [PubMed]
  102. Castaño, J.; Solanas, G.; Casagolda, D.; Raurell, I.; Villagrasa, P.; Bustelo, X.R.; García de Herreros, A.; Duñach, M. Specific phosphorylation of p120-catenin regulatory domain differently modulates its binding to RhoA. Mol. Cell Biol. 2007, 27, 1745–1757. [Google Scholar] [CrossRef] [PubMed]
  103. Cohen, A.W.; Hnasko, R.; Schubert, W.; Lisanti, M.P. Role of caveolae and caveolins in health and disease. Physiol. Rev. 2004, 84, 1341–1379. [Google Scholar] [CrossRef] [PubMed]
  104. Navarro, A.; Anand-Apte, B.; Parat, M.O. A role for caveolae in cell migration. FASEB J. 2004, 18, 1801–1811. [Google Scholar] [CrossRef] [PubMed]
  105. Lisanti, M.P.; Scherer, P.E.; Tang, Z.; Sargiacomo, M. Caveolae, caveolin and caveolin-rich membrane domains: A signalling hypothesis. Trends Cell Biol. 1994, 4, 231–235. [Google Scholar] [CrossRef] [PubMed]
  106. Li, S.; Couet, J.; Lisanti, M.P. Src tyrosine kinases, Galpha subunits, and H-Ras share a common membrane-anchored scaffolding protein, caveolin. Caveolin binding negatively regulates the auto-activation of Src tyrosine kinases. J. Biol. Chem. 1996, 271, 29182–29190. [Google Scholar] [CrossRef] [PubMed]
  107. Okamoto, T.; Schlegel, A.; Scherer, P.E.; Lisanti, M.P. Caveolins, a family of scaffolding proteins for organizing “preassembled signaling complexes” at the plasma membrane. J. Biol. Chem. 1998, 273, 5419–5422. [Google Scholar] [CrossRef] [PubMed]
  108. Tsang, S.M.; Wan, H. DSG3 regulates Src activity through caveolin-1. Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Clinical and Diagnostic Oral Sciences: London, UK, Unpublished data. 2015. [Google Scholar]
  109. Brennan, D.; Peltonen, S.; Dowling, A.; Medhat, W.; Green, K.J.; Wahl, J.K.; del Galdo, F.; Mahoney, M.G. A role for caveolin-1 in desmoglein binding and desmosome dynamics. Oncogene 2012, 31, 1636–1648. [Google Scholar] [CrossRef] [PubMed]
  110. Stahley, S.N.; Saito, M.; Faundez, V.; Koval, M.; Mattheyses, A.L.; Kowalczyk, A.P. Desmosome assembly and disassembly are membrane raft-dependent. PLOS ONE 2014, 9, e87809. [Google Scholar] [CrossRef] [PubMed]
  111. Arce, L.; Yokoyama, N.N.; Waterman, M.L. Diversity of LEF/TCF action in development and disease. Oncogene 2006, 25, 7492–7504. [Google Scholar] [CrossRef] [PubMed]
  112. Anastas, J.N.; Moon, R.T. WNT signalling pathways as therapeutic targets in cancer. Nat. Rev. Cancer 2013, 13, 11–26. [Google Scholar] [CrossRef] [PubMed]
  113. Clevers, H.; Nusse, R. WNT/β-catenin signaling and disease. Cell 2012, 149, 1192–1205. [Google Scholar] [CrossRef] [PubMed]
  114. Schuijers, J.; Mokry, M.; Hatzis, P.; Cuppen, E.; Clevers, H. WNT-induced transcriptional activation is exclusively mediated by TCF/LEF. EMBO J. 2014, 33, 146–156. [Google Scholar] [CrossRef]
  115. Uraguchi, M.; Morikawa, M.; Shirakawa, M.; Sanada, K.; Imai, K. Activation of WNT family expression and signaling in squamous cell carcinomas of the oral cavity. J. Dent. Res. 2004, 83, 327–332. [Google Scholar] [CrossRef] [PubMed]
  116. Brown, L.; Wan, H. Desmoglein 3 regulates beta catenin. Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Clinical and Diagnostic Oral Sciences: London, UK, Unpublished data. 2015. [Google Scholar]
  117. Brembeck, F.H.; Schwarz-Romond, T.; Bakkers, J.; Wilhelm, S.; Hammerschmidt, M.; Birchmeier, W. Essential role of BCL9-2 in the switch between beta-catenin’s adhesive and transcriptional functions. Genes Dev. 2004, 18, 2225–2230. [Google Scholar] [CrossRef] [PubMed]
  118. Cavigelli, M.; Dolfi, F.; Claret, F.X.; Karin, M. Induction of c-fos expression through JNK-mediated TCF/ELK-1 phosphorylation. EMBO J. 1995, 14, 5957–5964. [Google Scholar] [PubMed]
  119. Wei, X.; Sun, W.; Fan, R.; Hahn, J.; Joetham, A.; Li, G.; Webb, S.; Garrington, T.; Dakhama, A.; Lucas, J.; et al. MEF2C regulates c-jun but not TNF-alpha gene expression in stimulated mast cells. Eur. J. Immunol. 2003, 33, 2903–2909. [Google Scholar] [CrossRef] [PubMed]
  120. Boyle, W.J.; Smeal, T.; Defize, L.H.; Angel, P.; Woodgett, J.R.; Karin, M.; Hunter, T. Activation of protein kinase C decreases phosphorylation of c-jun at sites that negatively regulate its DNA-binding activity. Cell 1991, 64, 573–584. [Google Scholar] [CrossRef] [PubMed]
  121. Smeal, T.; Hibi, M.; Karin, M. Altering the specificity of signal transduction cascades: Positive regulation of c-jun transcriptional activity by protein kinase A. EMBO J. 1994, 13, 6006–6010. [Google Scholar] [PubMed]
  122. Wagner, E.F. Functions of AP1 (Fos/jun) in bone development. Ann. Rheum. Dis. 2002, 61, ii40–ii42. [Google Scholar] [CrossRef] [PubMed]
  123. Shaulian, E.; Karin, M. AP-1 in cell proliferation and survival. Oncogene 2001, 20, 2390–2400. [Google Scholar] [CrossRef] [PubMed]
  124. Foletta, V.C.; Segal, D.H.; Cohen, D.R. Transcriptional regulation in the immune system: All roads lead to AP-1. J. Leukoc. Biol. 1998, 63, 139–152. [Google Scholar] [PubMed]
  125. Zhou, L.Z.; Johnson, A.P.; Rando, T.A. NF kappa B and AP-1 mediate transcriptional responses to oxidative stress in skeletal muscle cells. Free Radic. Biol. Med. 2001, 31, 1405–1416. [Google Scholar] [CrossRef] [PubMed]
  126. Zenz, R.; Eferl, R.; Scheinecker, C.; Redlich, K.; Smolen, J.; Schonthaler, H.B.; Kenner, L.; Tschachler, E.; Wagner, E.F. Activator protein 1 (Fos/jun) functions in inflammatory bone and skin disease. Arthritis Res. Ther. 2008, 10, 201. [Google Scholar] [CrossRef] [PubMed]
  127. Thomsen, M.K.; Bakiri, L.; Hasenfuss, S.C.; Hamacher, R.; Martinez, L.; Wagner, E.F. JUNB/AP-1 controls IFN-γ during inflammatory liver disease. J. Clin. Investig. 2013, 123, 5258–5268. [Google Scholar] [CrossRef] [PubMed]
  128. Eckert, R.L.; Adhikary, G.; Young, C.A.; Jans, R.; Crish, J.F.; Xu, W.; Rorke, E.A. AP1 transcription factors in epidermal differentiation and skin cancer. J. Skin. Cancer 2013, 2013, 537028. [Google Scholar] [CrossRef] [PubMed]
  129. Ouyang, X.; Jessen, W.J.; Al-Ahmadie, H.; Serio, A.M.; Lin, Y.; Shih, W.J.; Reuter, V.E.; Scardino, P.T.; Shen, M.M.; Aronow, B.J.; et al. Activator protein-1 transcription factors are associated with progression and recurrence of prostate cancer. Cancer Res. 2008, 68, 2132–2144. [Google Scholar] [CrossRef] [PubMed]
  130. Prusty, B.K.; Das, B.C. Constitutive activation of transcription factor AP-1 in cervical cancer and suppression of human papillomavirus (HPV) transcription and AP-1 activity in hela cells by curcumin. Int. J. Cancer 2005, 113, 951–960. [Google Scholar] [CrossRef] [PubMed]
  131. Yu, Z.; Sato, S.; Trackman, P.C.; Kirsch, K.H.; Sonenshein, G.E. Blimp1 activation by AP-1 in human lung cancer cells promotes a migratory phenotype and is inhibited by the lysyl oxidase propeptide. PLOS ONE 2012, 7, e33287. [Google Scholar] [CrossRef] [PubMed]
  132. Malliri, A.; Symons, M.; Hennigan, R.F.; Hurlstone, A.F.; Lamb, R.F.; Wheeler, T.; Ozanne, B.W. The transcription factor AP-1 is required for EGF-induced activation of Rho-like GTPases, cytoskeletal rearrangements, motility, and in vitro invasion of A431 cells. J. Cell Biol. 1998, 143, 1087–1099. [Google Scholar] [CrossRef] [PubMed]
  133. Dong, Z.; Crawford, H.C.; Lavrovsky, V.; Taub, D.; Watts, R.; Matrisian, L.M.; Colburn, N.H. A dominant negative mutant of jun blocking 12-o-tetradecanoylphorbol-13-acetate-induced invasion in mouse keratinocytes. Mol. Carcinog. 1997, 19, 204–212. [Google Scholar] [CrossRef] [PubMed]
  134. Yuspa, S.H. The pathogenesis of squamous cell cancer: Lessons learned from studies of skin carcinogenesis. J. Dermatol. Sci. 1998, 17, 1–7. [Google Scholar] [CrossRef] [PubMed]
  135. Saez, E.; Rutberg, S.E.; Mueller, E.; Oppenheim, H.; Smoluk, J.; Yuspa, S.H.; Spiegelman, B.M. C-fos is required for malignant progression of skin tumors. Cell 1995, 82, 721–732. [Google Scholar] [CrossRef] [PubMed]
  136. Young, M.R.; Li, J.J.; Rincón, M.; Flavell, R.A.; Sathyanarayana, B.K.; Hunziker, R.; Colburn, N. Transgenic mice demonstrate AP-1 (activator protein-1) transactivation is required for tumor promotion. Proc. Natl. Acad. Sci. USA 1999, 96, 9827–9832. [Google Scholar] [CrossRef] [PubMed]
  137. Nguyen, P.T.; Tsunematsu, T.; Yanagisawa, S.; Kudo, Y.; Miyauchi, M.; Kamata, N.; Takata, T. The FGFR1 inhibitor PD173074 induces mesenchymal-epithelial transition through the transcription factor AP-1. Br. J. Cancer 2013, 109, 2248–2258. [Google Scholar] [CrossRef] [PubMed]
  138. Brown, L.; Hama, S.; Wan, H. Desmoglein 3 regulates AP-1 transcriptional activity. Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Clinical and Diagnostic Oral Sciences: London, UK, Unpublished data. 2015. [Google Scholar]
  139. Nateri, A.S.; Spencer-Dene, B.; Behrens, A. Interaction of phosphorylated c-jun with TCF4 regulates intestinal cancer development. Nature 2005, 437, 281–285. [Google Scholar] [CrossRef] [PubMed]
  140. Merritt, A.J.; Berika, M.Y.; Zhai, W.; Kirk, S.E.; Ji, B.; Hardman, M.J.; Garrod, D.R. Suprabasal desmoglein 3 expression in the epidermis of transgenic mice results in hyperproliferation and abnormal differentiation. Mol. Cell Biol. 2002, 22, 5846–5858. [Google Scholar] [CrossRef] [PubMed]
  141. Baron, S.; Hoang, A.; Vogel, H.; Attardi, L.D. Unimpaired skin carcinogenesis in desmoglein 3 knockout mice. PLOS ONE 2012, 7, e50024. [Google Scholar] [CrossRef] [PubMed]
  142. Gordon, L.A.; Mulligan, K.T.; Maxwell-Jones, H.; Adams, M.; Walker, R.A.; Jones, J.L. Breast cell invasive potential relates to the myoepithelial phenotype. Int. J. Cancer 2003, 106, 8–16. [Google Scholar] [CrossRef] [PubMed]
  143. Gupta, G.P.; Massagué, J. Cancer metastasis: Building a framework. Cell 2006, 127, 679–695. [Google Scholar] [CrossRef] [PubMed]
  144. Marinkovich, M.P. Tumour microenvironment: Laminin 332 in squamous-cell carcinoma. Nat. Rev. Cancer 2007, 7, 370–380. [Google Scholar] [CrossRef] [PubMed]
  145. Yan, W.; Wistuba, I.I.; Emmert-Buck, M.R.; Erickson, H.S. Squamous cell carcinoma—Similarities and differences among anatomical sites. Am. J. Cancer Res. 2011, 1, 275–300. [Google Scholar] [CrossRef] [PubMed]
  146. Agada, F.O.; Patmore, H.; Alhamarneh, O.; Stafford, N.D.; Greenman, J. Genetic profile of head and neck squamous cell carcinoma: Clinical implications. J. Laryngol. Otol. 2009, 123, 266–272. [Google Scholar] [CrossRef] [PubMed]
  147. Fang, W.K.; Gu, W.; Liao, L.D.; Chen, B.; Wu, Z.Y.; Wu, J.Y.; Shen, J.; Xu, L.Y.; Li, E.M. Prognostic significance of desmoglein 2 and desmoglein 3 in esophageal squamous cell carcinoma. Asian Pac. J. Cancer Prev. 2014, 15, 871–876. [Google Scholar] [CrossRef] [PubMed]
  148. Chung, C.H.; Parker, J.S.; Karaca, G.; Wu, J.; Funkhouser, W.K.; Moore, D.; Butterfoss, D.; Xiang, D.; Zanation, A.; Yin, X.; et al. Molecular classification of head and neck squamous cell carcinomas using patterns of gene expression. Cancer Cell 2004, 5, 489–500. [Google Scholar] [CrossRef] [PubMed]
  149. Thekdi, A.A.; Ferris, R.L. Diagnostic assessment of laryngeal cancer. Otolaryngol. Clin. North Am. 2002, 35, 953–969. [Google Scholar] [CrossRef] [PubMed]
  150. Takes, R.P. Staging of the neck in patients with head and neck squamous cell cancer: Imaging techniques and biomarkers. Oral. Oncol. 2004, 40, 656–667. [Google Scholar] [CrossRef] [PubMed]
  151. Argiris, A.; Karamouzis, M.V.; Raben, D.; Ferris, R.L. Head and neck cancer. Lancet 2008, 371, 1695–1709. [Google Scholar] [CrossRef] [PubMed]
  152. Rhee, D.; Wenig, B.M.; Smith, R.V. The significance of immunohistochemically demonstrated nodal micrometastases in patients with squamous cell carcinoma of the head and neck. Laryngoscope 2002, 112, 1970–1974. [Google Scholar] [CrossRef] [PubMed]
  153. Le Tourneau, C.; Velten, M.; Jung, G.M.; Bronner, G.; Flesch, H.; Borel, C. Prognostic indicators for survival in head and neck squamous cell carcinomas: Analysis of a series of 621 cases. Head Neck 2005, 27, 801–808. [Google Scholar] [CrossRef] [PubMed]
  154. Sato, F.; Shimada, Y.; Li, Z.; Watanabe, G.; Maeda, M.; Imamura, M. Lymph node micrometastasis and prognosis in patients with oesophageal squamous cell carcinoma. Br. J. Surg. 2001, 88, 426–432. [Google Scholar] [CrossRef] [PubMed]
  155. Shoaib, T.; Soutar, D.S.; MacDonald, D.G.; Camilleri, I.G.; Dunaway, D.J.; Gray, H.W.; McCurrach, G.M.; Bessent, R.G.; MacLeod, T.I.; Robertson, A.G. The accuracy of head and neck carcinoma sentinel lymph node biopsy in the clinically N0 neck. Cancer 2001, 91, 2077–2083. [Google Scholar] [CrossRef] [PubMed]
  156. Kligerman, J.; Lima, R.A.; Soares, J.R.; Prado, L.; Dias, F.L.; Freitas, E.Q.; Olivatto, L.O. Supraomohyoid neck dissection in the treatment of T1/T2 squamous cell carcinoma of oral cavity. Am. J. Surg. 1994, 168, 391–394. [Google Scholar] [CrossRef] [PubMed]
  157. McGuirt, W.F.; Johnson, J.T.; Myers, E.N.; Rothfield, R.; Wagner, R. Floor of mouth carcinoma. The management of the clinically negative neck. Arch. Otolaryngol. Head Neck Surg. 1995, 121, 278–282. [Google Scholar] [CrossRef] [PubMed]
  158. Duvvuri, U.; Simental, A.A.; D’Angelo, G.; Johnson, J.T.; Ferris, R.L.; Gooding, W.; Myers, E.N. Elective neck dissection and survival in patients with squamous cell carcinoma of the oral cavity and oropharynx. Laryngoscope 2004, 114, 2228–2234. [Google Scholar] [CrossRef] [PubMed]
  159. Onishi, A.; Nakashiro, K.; Mihara, M.; Sumida, T.; Kawamata, H.; Shintani, S.; Aida, T.; Tachikawa, T.; Hamakawa, H. Basic and clinical studies on quantitative analysis of lymph node micrometastasis in oral cancer. Oncol. Rep. 2004, 11, 33–39. [Google Scholar] [PubMed]
  160. Greenberg, J.S.; el Naggar, A.K.; Mo, V.; Roberts, D.; Myers, J.N. Disparity in pathologic and clinical lymph node staging in oral tongue carcinoma. Implication for therapeutic decision making. Cancer 2003, 98, 508–515. [Google Scholar] [CrossRef] [PubMed]
  161. Ambrosch, P.; Brinck, U. Detection of nodal micrometastases in head and neck cancer by serial sectioning and immunostaining. Oncology (Williston Park) 1996, 10, 1221–1226. [Google Scholar]
  162. Alvi, A.; Johnson, J.T. Extracapsular spread in the clinically negative neck (N0): Implications and outcome. Otolaryngol. Head Neck Surg. 1996, 114, 65–70. [Google Scholar] [CrossRef] [PubMed]
  163. Pitman, K.T.; Johnson, J.T.; Myers, E.N. Effectiveness of selective neck dissection for management of the clinically negative neck. Arch. Otolaryngol. Head Neck Surg. 1997, 123, 917–922. [Google Scholar] [CrossRef] [PubMed]
  164. Ferris, R.L.; Xi, L.; Raja, S.; Hunt, J.L.; Wang, J.; Gooding, W.E.; Kelly, L.; Ching, J.; Luketich, J.D.; Godfrey, T.E. Molecular staging of cervical lymph nodes in squamous cell carcinoma of the head and neck. Cancer Res. 2005, 65, 2147–2156. [Google Scholar] [CrossRef] [PubMed]
  165. Ferris, R.L.; Xi, L.; Seethala, R.R.; Chan, J.; Desai, S.; Hoch, B.; Gooding, W.; Godfrey, T.E. Intraoperative qRT-PCR for detection of lymph node metastasis in head and neck cancer. Clin. Cancer Res. 2011, 17, 1858–1866. [Google Scholar] [CrossRef] [PubMed]
  166. Trivedi, S.; Mattos, J.; Gooding, W.; Godfrey, T.E.; Ferris, R.L. Correlation of tumor marker expression with nodal disease burden in metastatic head and neck cancer. Otolaryngol. Head Neck Surg. 2013, 149, 261–268. [Google Scholar] [CrossRef] [PubMed]
  167. Solassol, J.; Burcia, V.; Costes, V.; Lacombe, J.; Mange, A.; Barbotte, E.; de Verbizier, D.; Cartier, C.; Makeieff, M.; Crampette, L.; et al. Pemphigus vulgaris antigen mrna quantification for the staging of sentinel lymph nodes in head and neck cancer. Br. J. Cancer 2010, 102, 181–187. [Google Scholar] [CrossRef] [PubMed]
  168. Morton, D.L.; Cochran, A.J. The case for lymphatic mapping and sentinel lymphadenectomy in the management of primary melanoma. Br. J. Dermatol. 2004, 151, 308–319. [Google Scholar] [CrossRef] [PubMed]
  169. Cochran, A.J.; Huang, R.R.; Lee, J.; Itakura, E.; Leong, S.P.; Essner, R. Tumour-induced immune modulation of sentinel lymph nodes. Nat. Rev. Immunol. 2006, 6, 659–670. [Google Scholar] [CrossRef] [PubMed]
  170. Gadgeel, S.M.; Ramalingam, S.S.; Kalemkerian, G.P. Treatment of lung cancer. Radiol. Clin. North Am. 2012, 50, 961–974. [Google Scholar] [CrossRef] [PubMed]
  171. Meyerson, M.; Franklin, W.A.; Kelley, M.J. Molecular classification and molecular genetics of human lung cancers. Semin. Oncol. 2004, 31, 4–19. [Google Scholar] [CrossRef] [PubMed]
  172. Travis, W.D.; Travis, L.B.; Devesa, S.S. Lung cancer. Cancer 1995, 75, 191–202. [Google Scholar] [CrossRef] [PubMed]
  173. Johnson, D.H.; Fehrenbacher, L.; Novotny, W.F.; Herbst, R.S.; Nemunaitis, J.J.; Jablons, D.M.; Langer, C.J.; deVore, R.F.; Gaudreault, J.; Damico, L.A.; et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J. Clin. Oncol. 2004, 22, 2184–2191. [Google Scholar] [CrossRef] [PubMed]
  174. Sandler, A.; Gray, R.; Perry, M.C.; Brahmer, J.; Schiller, J.H.; Dowlati, A.; Lilenbaum, R.; Johnson, D.H. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N. Engl. J. Med. 2006, 355, 2542–2550. [Google Scholar] [CrossRef] [PubMed]
  175. Sun, Z.; Yang, P.; Aubry, M.C.; Kosari, F.; Endo, C.; Molina, J.; Vasmatzis, G. Can gene expression profiling predict survival for patients with squamous cell carcinoma of the lung? Mol. Cancer 2004, 3, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Ilina, O.; Friedl, P. Mechanisms of collective cell migration at a glance. J. Cell Sci. 2009, 122, 3203–3208. [Google Scholar] [CrossRef] [PubMed]
  177. Friedl, P.; Alexander, S. Cancer invasion and the microenvironment: Plasticity and reciprocity. Cell 2011, 147, 992–1009. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Brown, L.; Wan, H. Desmoglein 3: A Help or a Hindrance in Cancer Progression? Cancers 2015, 7, 266-286. https://doi.org/10.3390/cancers7010266

AMA Style

Brown L, Wan H. Desmoglein 3: A Help or a Hindrance in Cancer Progression? Cancers. 2015; 7(1):266-286. https://doi.org/10.3390/cancers7010266

Chicago/Turabian Style

Brown, Louise, and Hong Wan. 2015. "Desmoglein 3: A Help or a Hindrance in Cancer Progression?" Cancers 7, no. 1: 266-286. https://doi.org/10.3390/cancers7010266

Article Metrics

Back to TopTop