Next Article in Journal
Studies in Cancer Epigenetics through a Sex and Gendered Lens: A Comprehensive Scoping Review
Previous Article in Journal
Predicting Severe Haematological Toxicity in Gastrointestinal Cancer Patients Undergoing 5-FU-Based Chemotherapy: A Bayesian Network Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Aberrant Expression of Claudins in Head and Neck Carcinomas and Their Prognostic and Therapeutic Value: A Narrative Review

by
Tarek Ziad Arabi
1,
Linah Abdulmohsen Algheryafi
1,
Nora A. Alodah
1,
Hamza M. Kossai Enabi
1,
Amjad Abdullah Alshehry
1 and
Abderrahman Ouban
1,2,*
1
College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
2
Department of Pathology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(17), 4208; https://doi.org/10.3390/cancers15174208
Submission received: 30 June 2023 / Revised: 3 August 2023 / Accepted: 11 August 2023 / Published: 22 August 2023

Abstract

:

Simple Summary

Head and neck cancer is one of the most common cancers worldwide and is associated with poor prognosis. Studies have demonstrated that claudins plays a significant role in the pathogenesis of cancers throughout the human body and highlights their prognostic role. The aberrant expression of various claudins has been strongly associated with carcinomas across the head and neck region. Additionally, the expression of these proteins predicts prognosis in several instances. Preclinical studies have highlighted that inhibiting the pathways which mediate altered claudins expression can halt neoplastic processes and minimize cellular invasion. Hence, understanding the role of claudins in head and neck carcinomas and their related mechanisms provides both prognostic and therapeutic value.

Abstract

Head and neck carcinomas have been associated with poor prognosis. Recent studies have highlighted the role of claudins’ expression in tumors throughout the body, and their prognostic and therapeutic role. Understanding the role of claudins and how their expression affects the progression of carcinomas in the head and neck region may allow for advances in the prognosis and management of this type of cancer. Several studies have highlighted the aberrant expression of the proteins in carcinomas in this region. Specifically, the overexpression of claudin-1 and downregulation of claudins-4, -7, and -17 have been linked with poor survival in oral squamous cell carcinoma patients. In laryngeal squamous cell carcinoma, increased levels of claudins-1 and reduced levels of claudins-3, -8, and -11 have been linked with poor outcomes. Targeting these proteins has shown promising outcomes as therapeutic in preclinical studies. However, studies remain extremely limited in nasal and hypopharyngeal carcinomas. In this review, we survey the available literature describing the aberrant expression of various claudins in carcinomas in this region, while highlighting their potential prognostic and therapeutic value. Then, we describe some molecular mechanisms involved in the aberrant expression of claudins and how they can be utilized as therapeutic targets.

1. Introduction

The barrier function of the cells is performed by the epithelia, which forms a protective layer between the body’s interior milieu and the outside environment [1,2]. At the heart of this barrier function are the tight junction strands, composed of the occludin, zona-occludens, desmosomes, and claudins proteins. Tight junctions regulate the permeability of the epithelial barrier and limit passage through the paracellular space [3]. In addition to their barrier role in protecting the underlying structures, tight junctions help regulate cellular proliferation and differentiation [4,5,6,7]. Tight junctions will also ensure the maintenance of cellular polarity, a critical element in maintaining proper cellular communication and signaling [8,9,10].
Recent mounting evidence points to signaling pathways which converge on tight junction positions and interact with the proteins forming those tight junctions [10,11]. Claudin proteins have also been noted to regulate the cell cycle via the Zonula occludens 1-associated nucleic acid-binding protein, p27kip1, and SP1 [12]. The aberrant expression of these tight junction proteins may play critical roles in establishing alternative signaling and activation of neoplastic pathways, which converge upon the cell and result in its transformation [13]. This was further supported by several studies which have shown that in cancer, claudins exhibits changing levels, with some showing increased and others showing decreased expression levels [5,14,15,16,17]. More evidence was presented with the manipulation of claudins levels in in vitro and animal studies, which resulted in the regression of aggressive tumor traits, especially cell motility, invasion, and metastases [5,14,15]. Additionally, claudin-6 has been shown to promote collective cell migration to the leading front of neoplastic cell lines [18]. In addition to their roles as tight junction proteins, the role of claudins in neoplastic development includes aberrant signaling [19,20,21], modulating autophagy [20,22,23,24], mediating the epithelial–mesenchymal transition phenomenon [19,25,26,27], mediating bacterial and viral oncogenesis [27,28,29], mediating the stemness of cancer cells [30], and the maintenance of the tumor microenvironment [30,31]. The above is added to the long-established role played by claudins’ knockdown in the development of cancer, allowing for the possible passage of toxins and carcinogens into the deeper layers of the epithelium. Collectively, the above evidence of claudins’ postulated roles in cancer development and progression suggests a great potential for these proteins in the fight against cancer and its complications.
Head and neck squamous cell carcinoma is the sixth most common tumor worldwide [32]. Neoplasms in this region present several challenges including old age at presentation, resulting in impaired functional status and comorbid medical conditions [33]; advanced stage at presentation [34]; significant toxicity associated with multimodality therapy [35]; multifocal disease; and the associated difficulty in mapping it, resulting in an increased likelihood of residual disease and recurrence [35]. Head and neck cancers are a challenging group of neoplasms which require additional strategies to counter, including the targets of molecular-era treatments.
Emerging evidence has highlighted the presence of irregular claudins expression in head and neck carcinomas. For example, Ouban et al. demonstrated that claudin-1 is overexpressed in carcinomas of several organs in the head and neck regions [17]. Additionally, overexpression of the same protein has been strongly associated with lymphatic invasion and poor overall survival in head and neck carcinoma patients [36]. Hence, studies have described the associations between head and neck carcinomas and claudins expression. Furthermore, studies have demonstrated that the level of expression of various claudins provides significant prognostic and clinicopathological value. Understanding the mechanisms underlying abnormal claudins expression in head and neck carcinomas may also provide the basis for therapeutic targets in future trials. This paper will provide a comprehensive review of the dysregulated expression of claudins in head and neck cancer and its possible utilization in the management of head and neck cancers. Then, we describe some of the molecular mechanisms resulting in irregular claudins expression and how they can be targeted in the management of such tumors.

2. Search Strategy

A literature search was conducted on the PubMed database using the following keywords: head and neck, mouth, tongue, lip, tonsil, salivary gland, larynx, nose, hypopharynx, nasopharynx, and carcinoma. Additionally, the references cited by the retrieved publications were manually reviewed. Irrelevant studies were excluded, and a narrative review was drafted accordingly.

3. Claudins Expression in the Normal Head and Neck Regions

The expression of claudins varies greatly between the different organs in the head and neck regions. For example, claudin-1 stains strongly in the junctional epithelium of the gingiva of rats [37]. Claudin-1 expression has been reported to be low in both tongue and palate squamous epithelium [17,38]. In salivary glands, claudin-1 can be seen in both acinar glands and ductal epithelial cells [17]. On the other hand, claudin-3 staining is strong in the granular and spinous layers of the gingival epithelium of rats [37]. Studies have also documented an intense expression of claudin-4 in the stratum germinativum and stratum spinosum, and a moderate expression in the stratum planocellulare in tissues from the hypopharynx, oropharynx, and larynx, while claudins-3, -8, and -10 are negative in the normal epithelium of these sites [39]. Understanding the expression of claudins proteins in normal epithelium across various regions of the head and neck is crucial to highlighting the role of aberrant claudins expressions.

4. Claudins Expression in Oral Squamous Cell Carcinoma

Although the exact role of claudins is not very well understood in cancer progression, claudin-1 is a frequently studied protein in the literature. In oral squamous cell carcinoma (OSCC), claudin-1 expression levels ranged from high to no change [40]. Over-expression of claudin-1 correlates with higher histological grades, specifically when the protein is positioned intracellularly, compared to when it is membranous or cytoplasmically expressed [37]. Although several studies have found changes in claudin-1 expression in OSCC, its prognostic value appears to be limited (Table 1). One study by Upadhaya et al. found reduced survival rates among patients with increased claudin-1 levels; however, these findings were not statistically significant [37]. Another study reported that claudin-1 upregulates the activity of matrix metalloproteinases (MMP_, which in turn increases OSCC invasiveness [41]. The same study also found no linkage between claudin-1 expression and patient health outcome. Zejc et al. reported similar results, where claudin-1 expression was not correlated to tumor stage, expansion, histological differentiation, recurrence-free survival, and several other clinicopathological parameters [40]. On the other hand, reports have found significant positive associations between pathological grade, perineural and vascular invasion, nodal metastasis, and advanced tumor staging [42]. Monteiro et al. also found that the combined usage of claudin-1 and occludin expression levels independently predicts recurrence-free and cancer-specific survival in OSCC patients [43]. Additionally, it has been reported that concurrent detection of elevated claudin-1 and junctional adhesion molecule-A (JAM-A) levels is associated with poor survival rates in this patient population [37]. Hence, the prognostic utility of claudin-1 may be improved by combining it with the expression of other adhesion molecules.
Claudin-2 is significantly upregulated in OSCC, with evidence that overexpression is independently linked to poor recurrence-free survival, tumor stage, and lymphatics involvement [37]. However, it is worth noting that another study found that increased claudin-2 expression is associated with improved survival in patients with squamous cell carcinomas in the head and neck regions [39]. Therefore, future studies should focus on understanding the true prognostic value of claudin-2 in OSCC. Claudin-3, on the other hand, is not expressed in cancerous or normal epithelium [39].
In OSCC tissue, claudins-4, -5 and -7 are expressed significantly lower than normal tissue [39,40]. Specifically, downregulation of claudin-4 has been reported to predict reduced recurrence-free survival [40]. Positive expression of the same protein has been linked to decreased perineural infiltration [53], while other studies have found no correlation between its expression and clinicopathological parameters [42]. Claudin-7 expression has been found to be a good indicator for the prognosis and recurrence rate in OSCC patients [40]. The knock-down of this protein is associated with higher pathologic grade, advanced staging, increased perineural, vascular and lymphatic invasions, and reduced survival [40,56]. Li et al. studied the effects of the interferon regulatory factor-2 (IRF2) on claudin-17 expression in human OSCC cell lines [57]. IRF2-induced claudin-7 transcription resulted in reduced proliferation, invasion, and migration of the cell lines [57]. These findings may demonstrate the possible utility of IRF2 as a therapeutic target in the future. Lastly, claudin-17 gene expression has been shown to decrease in OSCC and reduced levels have been associated with poor TNM staging and survival [58]. Collectively, claudins-1, -2, -4, -7, and -17 have proven valuable in the prognosis of OSCC patients (Table 1 and Table 2).

5. Claudins Expression in Tongue Squamous Cell Carcinoma

Only a few claudins have been discussed with regards to their involvement in tongue squamous cell carcinoma (TSCC) (Table 1 and Table 2). Bello et al. analyzed the expression of claudins-1, -4, -5, and -7 in TSCC, where they observed a strong expression for claudins-1 and -7, medium immunoreactivity for claudin-4, and low immunoreactivity for claudin-5 [44]. The number of cells stained in the superficial parts of the tumors with claudins-4 and -7 correlated with the tumor stage. A moderate immunohistochemical score with claudin-7 in TSCC was associated with better survival. When they compared between the superficial versus the invasive fronts of the tumors, high staining intensity of claudin-7 in the invasive front of the tumors was associated with decreased patient survival. It has been documented in a study of 83 TSCC specimens that high cytoplasmic expression of claudin-1 is linked to cervical lymph node metastasis, but not with disease progression [45]. The study suggested that the ectopic, intracellular location of claudin-1 at the invasive front may be an additional and promising diagnostic marker of TSCC [45]. Although claudin-7 has shown promising results as a prognostic tool in TSCC, further studies are needed to confirm its role and the role of other claudins in the neoplasm.

6. Claudins Expression in Lip Squamous Cell Carcinoma

Studies reporting claudins expression in lower lip squamous cell carcinomas (LLSCC) are limited (Table 1). De Aquino et al. found that claudin-1 exhibited a stronger expression in metastatic LLSCC than in nonmetastatic tumors and a higher expression in advanced stages of LLSCC (stages III and IV) than in the lower stages (I and II) [51]. To the best of our knowledge, this is the only available study analyzing claudins expression in LLSCC.

7. Claudins Expression in Tonsillar Squamous Cell Carcinoma

Among the claudins, only claudins-1 and -7 have been identified in tonsillar squamous cell carcinoma, with claudin-1 being highly expressed and claudin-7 exhibiting low levels of expression in all stages of cancer development [46] (Table 1 and Table 2). These changes were seen as independent of human papilloma virus infection.

8. Claudins Expression in Salivary Gland Carcinomas

The relationship between claudins expression and the clinical–pathological parameters has been assessed in salivary gland carcinomas. Arruda et al. examined the effect of epidermal growth factor (EGF) on the gene expression of claudins in salivary gland carcinomas [47]. Most mucoepidermoid carcinomas showed highly expressed claudins-1, -3, -4, -5, and -7 (Table 1). Patients older than 40 and Caucasians had higher levels of claudin-1 expression. In vitro tests showed a propensity for claudins gene expression to rise in response to EGF stimuli. In line with these findings, concurrent treatment with EGF and vascular endothelial growth factor antagonists has been shown to inhibit neoplastic growth and metastasis in mice models with adenoid cystic carcinoma [63]. The biological understanding of these proteins’ expression and associated signaling pathways will serve as a foundation for future research into potential target therapies.
In a second study, Abd El-Ghani et al. assessed the claudin-4 gene and protein expression via real-time PCR, along with immunohistochemistry in 30 specimens containing normal salivary tissue, pleomorphic adenomas, Warthin tumors, mucoepidermoid carcinomas, and adenoid carcinomas [60]. Their study revealed that claudin-4 is strongly expressed in normal salivary glands and benign salivary tumors. On the contrary, malignant cells’ claudin-4 protein expression levels were moderate to weak [45], contrary to the study by Arruda et al. [47]. Therefore, further studies are needed to confirm the levels of expression of claudin-4 in salivary gland carcinomas.
A third study reported claudin-3 modulation via the tumor necrosis factor (TNF)-α through extracellular signal-regulated kinase (ERK) 1/2/slug signaling in the submandibular gland [64]. The TNF-α-induced downregulation of claudin-3 was identified in the submandibular gland, whereas the signaling pathway had no effect on claudins-1 and -4. Claudin-4 levels are also significantly increased in the irradiated submandibular glands of rat models [65]. Radiation-induced alterations, however, can be attenuated via pilocarpine administration, which heals the damage to tight junctions and restores claudin-4 levels [65].
A study investigating claudin-7 expressions in salivary adenoid cystic carcinomas revealed decreased claudin-7 expression via both immunohistochemistry and Western blot [61]. The reduced expression of the protein correlated well with lymph node metastases, recurrence, and gender. Through the analysis of a stably transfected claudin-7 knockdown of an adenoid cystic carcinoma cell line and a subcutaneous tumor formation model, it was evident that the tumorgenicity of the aforementioned cell lines was due to regulation of an epithelial–mesenchymal transition via the Wnt/β-catenin signaling pathway. Claudins in salivary gland carcinomas have been largely studied, leading to the discovery of signaling pathways to be used as potential therapeutic targets.

9. Claudins Expression in Laryngeal Squamous Cell Carcinoma

The expression of several claudins has been studied in laryngeal squamous cell carcinomas (LSCC) (Table 1 and Table 2). The mRNA and protein expression of claudins-3 and -8 has been found to be overexpressed in LSCC, while claudins-1 and -7 are significantly downregulated [52]. Similar results can also be seen using immunohistochemistry [52]. Upregulation of claudin-3 has been associated with distant metastasis, while claudin-8 is associated with histological grade and Ki-67 expression [52]. On the other hand, downregulation of claudin-7 correlates with distant metastasis [52]. Moreover, analyses have been conducted on the relationships between claudins expression and LSCC patient survival. Expression of claudins-1 and -7 is significantly associated with improved survival times, while the opposite can be seen in regards to claudins-3 and -8 expression [52]. It is worth noting, however, that reports of claudin-7 in LSCC are conflicting. Kapral et al. found no significant difference between the LSCC and normal specimens in regard to claudin-7’s transcriptional activity [66]. Hence, it is important to interpret the results of claudin-7 in LSCC with caution until further reports are revealed.
Claudin-4’s role in LSCC remains unclear. Liu et al. demonstrated that the expression of claudin-4 is downregulated in LSCC and its expression is negatively correlated with the methyl-CpG-binding protein 2 (MeCP2), a prototype transcriptional repressor [59]. Additionally, neoplastic cell migration and invasion of LSCC cell lines are inhibited when claudin-4 DNA is demethylated via 5-aza-2′-deoxycytidine (a DNA methyltransferase inhibitor), but are recovered when its DNA is silenced [59]. The study by Liu et al. demonstrates that DNA methylation, likely through MeCP2 expression, is key for claudin-4 downregulation and tumor expansion. On the contrary, Xu et al. studied the effects of circular RNA SERPINA3 (circSERPINA3) on LSCC invasion and migration [54]. First, the authors found that circSERPINA3 is significantly upregulated in LSCC cell lines and tissues and downregulates microRNA (miR)-855-5p expression. Interestingly, the study then demonstrated that the silencing of circSERPINA3 and overexpressing of miR-855-5p suppresses the epithelial–mesenchymal transition by inhibiting claudin-4, snail, and vimentin expression and increasing E-cadherin expression [54]. Hence, further studies are needed to clarify whether claudin-4 plays a pro-oncogenic or anti-oncogenic role in LSCC.
Lastly, Shen et al. studied the clinical significance of claudin-11 promoter hypermethylation in LSCC using a quantitative methylation-specific polymerase chain reaction [62]. They reported that claudin-11 promoter methylation increased in neoplastic LSCC tissues compared to adjacent non-neoplastic tissues. The authors also found that increases in claudin-11 promoter methylation are associated with lymph node metastasis, advanced clinical stages, increased T classifications, and poor overall survival [62]. As a result, claudin-11 methylation plays a major role in the neoplastic processes of LSCC.

10. Claudins Expression in Nasal Squamous Cell Carcinoma

To the best of our knowledge, studies have yet to establish a relationship between nasal squamous cell carcinoma and claudins expression. Studies are urgently needed to identify the prognostic and therapeutic value of claudins in the neoplasm, as the 5-year overall survival rate is 50% [67].

11. Claudins Expression in Hypopharyngeal Squamous Cell Carcinoma

Immunohistochemical studies have demonstrated that claudin-1 is significantly upregulated in hypopharyngeal squamous cell carcinoma (HSCC) compared to non-neoplastic cells [49,50] (Table 1). Additionally, claudin-1 overexpression is significantly correlated with poor differentiation, lymph node metastasis, and survival rates among HSCC patients [49,50]. It has been hypothesized that claudin-1 induces tumor lymphatic vessel generation, thereby enhancing the spread of the tumor [50]. To the best of the authors’ knowledge, the role of other claudins proteins in HSCC has not yet been investigated.

12. Claudins Expression in Nasopharyngeal Squamous Cell Carcinoma

In a study by Hsueh et al., an increased expression of claudins-1 and -4 and a low expression of claudin-7 were seen via immunohistochemistry in nasopharyngeal carcinoma (NPC) [48] (Table 1 and Table 2). Low claudin-4 expression and high claudin-7 expression were correlated with distant metastases. Increased claudin-7 was also correlated with high tumor stages. Furthermore, decreased claudin-4 and increased claudin-7 expressions were independent risk factors for reduced distant metastases-free survival among these patients [48]. In a second study by Lee et al. assessing the role of claudin-1 in NPC, the authors reported an increased expression of claudin-1 in NPC cell lines when treated by serum-deprivation or with fluorouracil [68]. Increases in claudin-1 were associated with significant anti-apoptotic effects. Restoring E-cadherin significantly reduced claudin-1 levels and reversed the associated anti-apoptotic effects of the protein, suggesting a negative regulatory role of E-cadherin on claudin-1 in NPC.
Kojima et al. further assessed the expression of claudins in Epstein–Barr virus-associated non-keratinizing NPC [55]. The authors reaffirmed the overexpression of claudin-1 seen in NPC. The authors also demonstrated that claudin-4 is expressed in all specimens, claudin-2 was not expressed in any, and claudin-3 showed a variable expression among the samples. Given the fact that claudins-3 and 4 are receptors for cytotoxic Clostridium perfringens enterotoxin (CPE), CPE has drawn attention for malignant tumors expressing the two proteins [55]. CPE damages cells through osmotic disruption, leading to cell lysis [69]. The enterotoxin has shown promising results in claudins, inducing rapid cytolysis in claudin-3 and claudin-4-positive breast cancer cells [69]. Studies are needed to determine whether such effects can be reproduced in NPCs.

13. Mechanisms of Aberrant Claudins Expressions in Head and Neck Cancers and Their Therapeutic Value

The mechanisms underlying the aberrant expression of claudins in head and neck carcinomas are unclear. Although several studies have highlighted such processes in other cancers, studies remain limited in the head and neck regions in this regard. Chang et al. found that claudin-1 overexpression was negatively associated with AMP-activated protein kinase (AMPK) and is positively associated with the transforming growth factor (TGF)-β (Figure 1) [70]. Accordingly, AMPK activation overrides claudin-1-induced tumor invasion, suggesting a role of a claudin-1/AMPK/TGF-β axis in these tumors [70]. Indeed, it has been previously demonstrated that TGF-β promotes epithelial–mesenchymal transition and suppresses anti-neoplastic immune responses [71], and patients with elevated TGF-β mRNA levels have poor overall survival in silico [72]. Additionally, vactosertib, a TGF-β receptor inhibitor, exhibits significant antineoplastic effects on head and neck cell lines [72]. Clinically, bintrafusp alfa, a bifunctional function protein targeting TGF-β and programmed death-ligand-1, has shown clinical efficacy and a safe profile in patients with advanced head and neck squamous cell carcinomas in a phase I cohort study [73]. Hence, targeting the claudin-1/AMPK/TGF-β may prove as a plausible therapeutic target for head and neck cancer patients overexpressing claudin-1 and TGF-β.
As previously stated, TNF-α plays a key role in claudin-3 downregulation in submandibular gland neoplasms (Figure 1) [64]. TNF-α increases ERK1/2 phosphorylation and transcriptional factor slug expression [64]. Slug knockdown reduces claudin-3 expression in the submandibular glands [64]. TGF-β has been shown to result in slug-mediated increases in MMP-9 in OSCC cells, thereby enhancing tumor invasion [74]. Additionally, overexpression of the transcriptional factor correlates with poor disease recurrence and disease-free survival in LSCC patients and chemo- and radiotherapy resistance [75,76]. Interestingly, head and neck carcinoma patients with slug overexpression have a 3.3-times better chance of survival with upfront surgery compared to primary chemo- or radiotherapy [76]. Collectively, these results demonstrate that the slug transcriptional factor plays a critical role in the pathogenesis of head and neck carcinomas.
EGF and its accompanying receptor have also been a highlight of studies on head and neck carcinomas (Figure 1) [77]. EGF promotes aberrant claudins expression and neoplastic invasion [47]. EGF has been shown to regulate claudin-2 and -4 expressions through Src and STAT3 pathways in kidney cells [78]. Accordingly, the targeted inhibition of STAT3 in tumor-associated myeloid cells sensitizes squamous cell carcinomas to radiotherapy and T-cell-mediated immunity in human-to-mice xenotransplantation models [79]. In head and neck squamous cell carcinoma cell lines, treatment with EGF promotes cell invasion via ERK1/2- and PI3K-mediated epithelial–mesenchymal transitions and E-cadherin degradation via MMP-9 [80]. Overall, targeting TGF-β, TNF-α, and EGF pathways has shown promising results in preclinical studies. Clinical studies are needed, however, to determine whether these findings can be translated to patients.

14. Conclusions

Carcinomas of the head and neck remain a therapeutic challenge to this day, as a result of late age at presentation and rapid spread of the tumor. Recently, the role of claudin proteins in the development and progression of the neoplastic has fallen under the spotlight of the scientific community across various tumors of the human body. Studies have highlighted the aberrant expression of proteins that are extensively in the head and neck regions. Specifically, overexpression of claudin-1 (in combination with other adhesion molecules) and downregulation of claudins-4, -7, and -17 have been linked with poor survival in OSCC patients. In LSCC, increased levels of claudin-1 and reduced levels of claudins-3, -8, and -11 have been linked with poor outcomes.
We also summarize key molecular mechanisms regulating claudins expression in head and neck squamous cell carcinomas. The claudin-1/AMPK/TGF-β axis appears to be a key mediator of the epithelial–mesenchymal transition and neoplastic invasion. Both preclinical and clinical studies have demonstrated positive results. Additionally, TNF-α/slug pathways promote neoplastic processes by enhancing the epithelial–mesenchymal transition and increasing MMP-9 release. Lastly, EGF has been shown to modulate claudins expression via Src and STAT3 pathways and promote TNF-α-like effects through ERK1/2 and PI3K signaling.
However, our manuscript also highlights a lack of studies in some types of squamous cell carcinoma, especially nasal carcinomas and HSCC. Therefore, future studies are needed to demonstrate the expression of claudins and their role in the pathogenesis of these neoplasms. Emerging evidence has also identified several signaling pathways involving claudins with promising therapeutic results. Clinical studies are needed to confirm whether these outcomes reflect true clinical settings.

Author Contributions

Conceptualization, A.O.; writing—original draft preparation, T.Z.A., H.M.K.E., L.A.A., A.A.A., N.A.A. and A.O.; writing—review and editing, T.Z.A. and A.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No data were generated in the drafting of this manuscript, and all original data may be found in the referenced articles.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marchiando, A.M.; Graham, W.V.; Turner, J.R. Epithelial barriers in homeostasis and disease. Annu. Rev. Pathol. 2010, 5, 119–144. [Google Scholar] [CrossRef] [PubMed]
  2. Shin, K.; Fogg, V.C.; Margolis, B. Tight junctions and cell polarity. Annu. Rev. Cell Dev. Biol. 2006, 22, 207–235. [Google Scholar] [CrossRef] [PubMed]
  3. Diamond, J.M. Tight and leaky junctions of epithelia: A perspective on kisses in the dark. Fed. Proc. 1974, 33, 2220–2224. [Google Scholar] [PubMed]
  4. Dhawan, P.; Ahmad, R.; Chaturvedi, R.; Smith, J.J.; Midha, R.; Mittal, M.K.; Krishnan, M.; Chen, X.; Eschrich, S.; Yeatman, T.J.; et al. Claudin-2 expression increases tumorigenicity of colon cancer cells: Role of epidermal growth factor receptor activation. Oncogene 2011, 30, 3234–3247. [Google Scholar] [CrossRef] [PubMed]
  5. Dhawan, P.; Singh, A.B.; Deane, N.G.; No, Y.; Shiou, S.R.; Schmidt, C.; Neff, J.; Washington, M.K.; Beauchamp, R.D. Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. J. Clin. Investig. 2005, 115, 1765–1776. [Google Scholar] [CrossRef] [PubMed]
  6. Singh, A.; Mishra, A.K.; Ylaya, K.; Hewitt, S.M.; Sharma, K.C.; Saxena, S. Wilms tumor-1, claudin-1 and ezrin are useful immunohistochemical markers that help to distinguish schwannoma from fibroblastic meningioma. Pathol. Oncol. Res. 2012, 18, 383–389. [Google Scholar] [CrossRef] [PubMed]
  7. Singh, A.B.; Sharma, A.; Smith, J.J.; Krishnan, M.; Chen, X.; Eschrich, S.; Washington, M.K.; Yeatman, T.J.; Beauchamp, R.D.; Dhawan, P. Claudin-1 up-regulates the repressor ZEB-1 to inhibit E-cadherin expression in colon cancer cells. Gastroenterology 2011, 141, 2140–2153. [Google Scholar] [CrossRef] [PubMed]
  8. Balda, M.S.; Matter, K. Tight junctions in health and disease. Semin. Cell Dev. Biol. 2014, 36, 147–148. [Google Scholar] [CrossRef]
  9. Farkas, A.E.; Capaldo, C.T.; Nusrat, A. Regulation of epithelial proliferation by tight junction proteins. Ann. N. Y. Acad. Sci. 2012, 1258, 115–124. [Google Scholar] [CrossRef]
  10. Zihni, C.; Balda, M.S.; Matter, K. Signalling at tight junctions during epithelial differentiation and microbial pathogenesis. J. Cell Sci. 2014, 127, 3401–3413. [Google Scholar] [CrossRef]
  11. Severson, E.A.; Parkos, C.A. Mechanisms of outside-in signaling at the tight junction by junctional adhesion molecule A. Ann. N. Y. Acad. Sci. 2009, 1165, 10–18. [Google Scholar] [CrossRef] [PubMed]
  12. Venugopal, S.; Anwer, S.; Szászi, K. Claudin-2: Roles beyond Permeability Functions. Int. J. Mol. Sci. 2019, 20, 5655. [Google Scholar] [CrossRef]
  13. Singh, A.B.; Uppada, S.B.; Dhawan, P. Claudin proteins, outside-in signaling, and carcinogenesis. Pflug. Arch. 2017, 469, 69–75. [Google Scholar] [CrossRef] [PubMed]
  14. Oku, N.; Sasabe, E.; Ueta, E.; Yamamoto, T.; Osaki, T. Tight junction protein claudin-1 enhances the invasive activity of oral squamous cell carcinoma cells by promoting cleavage of laminin-5 gamma2 chain via matrix metalloproteinase (MMP)-2 and membrane-type MMP-1. Cancer Res. 2006, 66, 5251–5257. [Google Scholar] [CrossRef] [PubMed]
  15. Dos Reis, P.P.; Bharadwaj, R.R.; Machado, J.; Macmillan, C.; Pintilie, M.; Sukhai, M.A.; Perez-Ordonez, B.; Gullane, P.; Irish, J.; Kamel-Reid, S. Claudin 1 overexpression increases invasion and is associated with aggressive histological features in oral squamous cell carcinoma. Cancer 2008, 113, 3169–3180. [Google Scholar] [CrossRef]
  16. Ouban, A.; Ahmed, A.A. Claudins in human cancer: A review. Histol. Histopathol. 2010, 25, 83–90. [Google Scholar] [CrossRef] [PubMed]
  17. Ouban, A.; Hamdan, H.; Hakam, A.; Ahmed, A.A. Claudin-1 expression in squamous cell carcinomas of different organs: Comparative study of cancerous tissues and normal controls. Int. J. Surg. Pathol. 2012, 20, 132–138. [Google Scholar] [CrossRef]
  18. Kojima, M.; Sugimoto, K.; Kobayashi, M.; Ichikawa-Tomikawa, N.; Kashiwagi, K.; Watanabe, T.; Soeda, S.; Fujimori, K.; Chiba, H. Aberrant Claudin-6–Adhesion Signaling Promotes Endometrial Cancer Progression via Estrogen Receptor α. Mol. Cancer Res. 2021, 19, 1208–1220. [Google Scholar] [CrossRef]
  19. Zhu, J.; Jiang, Q. Twist1-mediated transcriptional activation of Claudin-4 promotes cervical cancer cell migration and invasion. Oncol. Lett. 2023, 26, 335. [Google Scholar] [CrossRef]
  20. Ahmad, R.; Kumar, B.; Tamang, R.L.; Talmon, G.A.; Dhawan, P.; Singh, A.B. P62/SQSTM1 binds with claudin-2 to target for selective autophagy in stressed intestinal epithelium. Commun. Biol. 2023, 6, 740. [Google Scholar] [CrossRef]
  21. Su, K.; Yao, X.; Guo, C.; Qian, C.; Wang, Y.; Ma, X.; Wang, X.; Yang, Y. Solasodine suppresses the metastasis of gastric cancer through claudin-2 via the AMPK/STAT3/NF-κB pathway. Chem. Biol. Interact. 2023, 379, 110520. [Google Scholar] [CrossRef] [PubMed]
  22. Huang, S.; Zhang, J.; Li, Y.; Xu, Y.; Jia, H.; An, L.; Wang, X.; Yang, Y. Downregulation of Claudin5 promotes malignant progression and radioresistance through Beclin1-mediated autophagy in esophageal squamous cell carcinoma. J. Transl. Med. 2023, 21, 379. [Google Scholar] [CrossRef] [PubMed]
  23. Dong, Y.; Jin, Q.; Sun, M.; Qi, D.; Qu, H.; Wang, X.; Quan, C. CLDN6 inhibits breast cancer metastasis through WIP-dependent actin cytoskeleton-mediated autophagy. J. Exp. Clin. Cancer Res. 2023, 42, 68. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, C.; Wu, N.; Pei, B.; Ma, X.; Yang, W. Claudin and pancreatic cancer. Front. Oncol. 2023, 13, 1136227. [Google Scholar] [CrossRef] [PubMed]
  25. Voutsadakis, I.A. EMT Features in Claudin-Low versus Claudin-Non-Suppressed Breast Cancers and the Role of Epigenetic Modifications. Curr. Issues Mol. Biol. 2023, 45, 6040–6054. [Google Scholar] [CrossRef] [PubMed]
  26. Pantia, S.; Kangsamaksin, T.; Janvilisri, T.; Komyod, W. Asiatic Acid Inhibits Nasopharyngeal Carcinoma Cell Viability and Migration via Suppressing STAT3 and Claudin-1. Pharmaceuticals 2023, 16, 902. [Google Scholar] [CrossRef]
  27. Hayashi, T.; Kobayashi, N.; Ushida, K.; Asai, N.; Nakano, S.; Fujii, K.; Ando, T.; Utsumi, T. Effect of eribulin on epithelial-mesenchymal transition plasticity in metastatic breast cancer: An exploratory, prospective study. Genes Cells 2023, 28, 364–373. [Google Scholar] [CrossRef]
  28. Choi, S.; Kim, N.; Park, J.H.; Nam, R.H.; Song, C.H.; Lee, H.S. Effect of Helicobacter pylori infection and its eradication on the expression of tight junction proteins in the gastric epithelium in relation to gastric carcinogenesis. Helicobacter 2022, 27, e12929. [Google Scholar] [CrossRef]
  29. Fukasawa, M. Claudin 1 as a target for anti-hepatitis C virus strategy. Yakugaku Zasshi 2014, 134, 635–640. [Google Scholar] [CrossRef]
  30. Fujiwara-Tani, R.; Mori, S.; Ogata, R.; Sasaki, R.; Ikemoto, A.; Kishi, S.; Kondoh, M.; Kuniyasu, H. Claudin-4: A New Molecular Target for Epithelial Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 5494. [Google Scholar] [CrossRef]
  31. Roehlen, N.; Muller, M.; Nehme, Z.; Crouchet, E.; Jühling, F.; Del Zompo, F.; Cherradi, S.; Duong, F.H.T.; Almeida, N.; Saviano, A.; et al. Treatment of HCC with claudin-1-specific antibodies suppresses carcinogenic signaling and reprograms the tumor microenvironment. J. Hepatol. 2023, 78, 343–355. [Google Scholar] [CrossRef] [PubMed]
  32. Parkin, D.M.; Bray, F.; Ferlay, J.; Pisani, P. Global cancer statistics, 2002. CA Cancer J. Clin. 2005, 55, 74–108. [Google Scholar] [CrossRef]
  33. de Rijke, J.M.; Schouten, L.J.; Schouten, H.C.; Jager, J.J.; Koppejan, A.G.; van den Brandt, P.A. Age-specific differences in the diagnostics and treatment of cancer patients aged 50 years and older in the province of Limburg, The Netherlands. Ann. Oncol. 1996, 7, 677–685. [Google Scholar] [CrossRef] [PubMed]
  34. Argiris, A.; Eng, C. Epidemiology, staging, and screening of head and neck cancer. Cancer Treat. Res. 2003, 114, 15–60. [Google Scholar] [CrossRef] [PubMed]
  35. VanderWalde, N.A.; Fleming, M.; Weiss, J.; Chera, B.S. Treatment of older patients with head and neck cancer: A review. Oncologist 2013, 18, 568–578. [Google Scholar] [CrossRef] [PubMed]
  36. Lee, J.; Kim, S.W.; Lee, S.H.; Jin, S.M.; Do, S.I.; Lee, H.J. Prognostic Significance of the Expression of Claudin-1 in Head and Neck Squamous Cell Carcinoma. Korean J. Otorhinolaryngol. Head. Neck Surg. 2021, 64, 336–343. [Google Scholar] [CrossRef]
  37. Upadhaya, P.; Barhoi, D.; Giri, A.; Bhattacharjee, A.; Giri, S. Joint detection of claudin-1 and junctional adhesion molecule-A as a therapeutic target in oral epithelial dysplasia and oral squamous cell carcinoma. J. Cell. Biochem. 2019, 120, 18117–18127. [Google Scholar] [CrossRef]
  38. Ouban, A.; Ahmed, A. Analysis of the Distribution and Expression of Claudin-1 Tight Junction Protein in the Oral Cavity. Appl. Immunohistochem. Mol. Morphol. 2015, 23, 444–448. [Google Scholar] [CrossRef]
  39. Nelhűbel, G.A.; Károly, B.; Szabó, B.; Lotz, G.; Kiss, A.; Tóvári, J.; Kenessey, I. The Prognostic Role of Claudins in Head and Neck Squamous Cell Carcinomas. Pathol. Oncol. Res. 2014, 20, 99–106. [Google Scholar] [CrossRef]
  40. Zejc, T.; Piontek, J.; Schulzke, J.-D.; Fromm, M.; Ervens, J.; Rosenthal, R. Clinical Significance of Claudin Expression in Oral Squamous Cell Carcinoma. Int. J. Mol. Sci. 2022, 23, 11234. [Google Scholar] [CrossRef]
  41. De Vicente, J.C.; Fernández-Valle, Á.; Vivanco-Allende, B.; Santamarta, T.R.; Lequerica-Fernández, P.; Hernández-Vallejo, G.; Allonca-Campa, E. The prognostic role of claudins-1 and -4 in oral squamous cell carcinoma. Anticancer Res. 2015, 35, 2949–2959. [Google Scholar] [PubMed]
  42. Sappayatosok, K.; Phattarataratip, E. Overexpression of Claudin-1 is Associated with Advanced Clinical Stage and Invasive Pathologic Characteristics of Oral Squamous Cell Carcinoma. Head Neck Pathol. 2015, 9, 173–180. [Google Scholar] [CrossRef] [PubMed]
  43. Monteiro, L.; Delgado, L.; Amaral, B.; Ricardo, S.; Fraga, M.; Lopes, C.; Warnakulasuriya, S. Occludin and claudin-1 are potential prognostic biomarkers in patients with oral squamous cell carcinomas: An observational study. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2022, 134, 588–598. [Google Scholar] [CrossRef] [PubMed]
  44. Bello, I.O.; Vilen, S.-T.; Niinimaa, A.; Kantola, S.; Soini, Y.; Salo, T. Expression of claudins 1, 4, 5, and 7 and occludin, and relationship with prognosis in squamous cell carcinoma of the tongue. Human Pathol. 2008, 39, 1212–1220. [Google Scholar] [CrossRef] [PubMed]
  45. Yamamoto, D.; Kayamori, K.; Sakamoto, K.; Tsuchiya, M.; Ikeda, T.; Harada, H.; Yoda, T.; Watabe, T.; Hara-Yokoyama, M. Intracellular claudin-1 at the invasive front of tongue squamous cell carcinoma is associated with lymph node metastasis. Cancer Sci. 2020, 111, 700–712. [Google Scholar] [CrossRef] [PubMed]
  46. Kondoh, A.; Takano, K.-I.; Kojima, T.; Ohkuni, T.; Kamekura, R.; Ogasawara, N.; Go, M.; Sawada, N.; Himi, T. Altered expression of claudin-1, claudin-7, and tricellulin regardless of human papilloma virus infection in human tonsillar squamous cell carcinoma. Acta Oto-Laryngol. 2011, 131, 861–868. [Google Scholar] [CrossRef] [PubMed]
  47. de Arruda, C.F.J.; Coutinho-Camillo, C.M.; Marques, M.M.; Nagano, C.P.; Bologna, S.B.; Bettim, B.B.; Germano, J.N.; Pinto, C.A.L.; Hsieh, R.; Lourenço, S.V. Claudin expression is maintained in mucoepidermoid carcinoma of the salivary gland. Pathol. Res. Pract. 2020, 216, 153161. [Google Scholar] [CrossRef]
  48. Hsueh, C.; Chang, Y.-S.; Tseng, N.-M.; Liao, C.-T.; Hsueh, S.; Chang, J.-H.; Wu, I.C.; Chang, K.-P. Expression pattern and prognostic significance of claudins 1, 4, and 7 in nasopharyngeal carcinoma. Human Pathol. 2010, 41, 944–950. [Google Scholar] [CrossRef]
  49. Li, W.; Dong, Q.; Li, L.; Zhang, Z.; Cai, X.; Pan, X. Prognostic significance of claudin-1 and cyclin B1 protein expression in patients with hypopharyngeal squamous cell carcinoma. Oncol. Lett. 2016, 11, 2995–3002. [Google Scholar] [CrossRef]
  50. Li, W.J.; Zhang, Z.L.; Yu, X.M.; Cai, X.L.; Pan, X.L.; Yang, X.Y. Expression of claudin-1 and its relationship with lymphatic microvessel generation in hypopharyngeal squamous cell carcinoma. Genet. Mol. Res. 2015, 14, 11814–11826. [Google Scholar] [CrossRef]
  51. de Aquino, A.R.L.; de Carvalho, C.H.P.; Nonaka, C.F.W.; Freitas, R.d.A.; de Souza, L.B.; Pinto, L.P. Immunoexpression of Claudin-1 and Nm23-H1 in Metastatic and Nonmetastatic Lower Lip Squamous-cell Carcinoma. Appl. Immunohistochem. Mol. Morphol. 2012, 20, 595–601. [Google Scholar] [CrossRef] [PubMed]
  52. Zhou, S.; Piao, X.; Wang, C.; Wang, R.; Song, Z. Identification of claudin-1, -3, -7 and -8 as prognostic markers in human laryngeal carcinoma. Mol. Med. Rep. 2019, 20, 393–400. [Google Scholar] [CrossRef] [PubMed]
  53. Lourenço, S.V.; Coutinho-Camillo, C.M.; Buim, M.E.C.; Pereira, C.M.; Carvalho, A.L.; Kowalski, L.P.; Soares, F.A. Oral squamous cell carcinoma: Status of tight junction claudins in the different histopathological patterns and relationship with clinical parameters. A tissue-microarray-based study of 136 cases. J. Clin. Pathol. 2010, 63, 609–614. [Google Scholar] [CrossRef] [PubMed]
  54. Xu, Q.; Yu, B.; Chen, W.; Li, W.; Sun, Y.; Fang, Y. CircSERPINA3 promoted cell proliferation, migration, and invasion of laryngeal squamous cell carcinoma by targeting miR-885-5p. Cell Biol. Int. 2022, 46, 1852–1863. [Google Scholar] [CrossRef] [PubMed]
  55. Kojima, F.; Ishida, M.; Takikita-Suzuki, M.; Hotta, M.; Katsura, K.; Nagata, A.; Enoki, Y.; Kato, G.I.; Okabe, H. Claudin expression profiles in Epstein-Barr virus-associated nasopharyngeal carcinoma. Oncol. Rep. 2010, 23, 927–931. [Google Scholar] [CrossRef] [PubMed]
  56. Phattarataratip, E.; Sappayatosok, K. Expression of claudin-5, claudin-7 and occludin in oral squamous cell carcinoma and their clinico-pathological significance. J. Clin. Exp. Dent. 2016, 8, e299–e306. [Google Scholar] [CrossRef] [PubMed]
  57. Li, X.; Yang, W. IRF2-induced Claudin-7 suppresses cell proliferation, invasion and migration of oral squamous cell carcinoma. Exp. Ther. Med. 2021, 23, 7. [Google Scholar] [CrossRef]
  58. Xu, Y.-N.; Deng, M.-S.; Liu, Y.-F.; Yao, J.; Xiao, Z.-Y. Tight junction protein CLDN17 serves as a tumor suppressor to reduce the invasion and migration of oral cancer cells by inhibiting epithelial-mesenchymal transition. Arch. Oral Biol. 2022, 133, 105301. [Google Scholar] [CrossRef]
  59. Liu, Y.; Chang, K.; Fu, K.; Dong, X.; Chen, X.; Liu, J.; Cui, N.; Ni, J. DNA demethylation of claudin-4 suppresses migration and invasion in laryngeal squamous carcinoma cells. Hum. Pathol. 2018, 75, 71–80. [Google Scholar] [CrossRef]
  60. Abd El-Ghani, S.F.; Kasem, R.F.; Ghallab, N.A.; Shaker, O.G. Detection of claudin-4 in salivary gland neoplasms (a study utilizing RT-PCR and immunohistochemistry). J. Oral Pathol. Med. 2013, 42, 781–787. [Google Scholar] [CrossRef]
  61. Ji, H.; Ding, X.; Zhang, W.; Zheng, Y.; Du, H.; Zheng, Y.; Song, H.; Li, M.; Jiang, Y.; Xie, J.; et al. Claudin-7 Inhibits Proliferation and Metastasis in Salivary Adenoid Cystic Carcinoma through Wnt/β-Catenin Signaling. Cell Transplant. 2020, 29, 096368972094358. [Google Scholar] [CrossRef] [PubMed]
  62. Shen, Z.; Cao, B.; Lin, L.; Zhou, C.; Ye, D.; Qiu, S.; Li, Q.; Cui, X. The Clinical Signification of Claudin-11 Promoter Hypermethylation for Laryngeal Squamous Cell Carcinoma. Med. Sci. Monit. 2017, 23, 3635–3640. [Google Scholar] [CrossRef] [PubMed]
  63. Younes, M.N.; Park, Y.W.; Yazici, Y.D.; Gu, M.; Santillan, A.A.; Nong, X.; Kim, S.; Jasser, S.A.; El-Naggar, A.K.; Myers, J.N. Concomitant inhibition of epidermal growth factor and vascular endothelial growth factor receptor tyrosine kinases reduces growth and metastasis of human salivary adenoid cystic carcinoma in an orthotopic nude mouse model. Mol. Cancer Ther. 2006, 5, 2696–2705. [Google Scholar] [CrossRef] [PubMed]
  64. Mei, M.; Xiang, R.-L.; Cong, X.; Zhang, Y.; Li, J.; Yi, X.; Park, K.; Han, J.-Y.; Wu, L.-L.; Yu, G.-Y. Claudin-3 is required for modulation of paracellular permeability by TNF-α through ERK1/2/slug signaling axis in submandibular gland. Cell Signal. 2015, 27, 1915–1927. [Google Scholar] [CrossRef] [PubMed]
  65. Yao, Q.-T.; Wu, Y.-H.; Liu, S.-H.; Song, X.-B.; Xu, H.; Li, J.; Shi, L. Pilocarpine improves submandibular gland dysfunction in irradiated rats by downregulating the tight junction protein claudin-4. Oral Dis. 2022, 28, 1528–1538. [Google Scholar] [CrossRef] [PubMed]
  66. Kapral, M.; Strzalka-Mrozik, B.; Kowalczyk, M.; Paluch, J.; Gola, J.; Gierek, T.; Weglarz, L. Transcriptional activities of histone H3, cyclin D1 and claudin 7 encoding genes in laryngeal cancer. Eur. Arch. Oto-Rhino-Laryngol. 2011, 268, 709–714. [Google Scholar] [CrossRef]
  67. Elgart, K.; Faden, D.L. Sinonasal Squamous Cell Carcinoma: Etiology, Pathogenesis, and the Role of Human Papilloma Virus. Curr. Otorhinolaryngol. Rep. 2020, 8, 111–119. [Google Scholar] [CrossRef]
  68. Lee, J.-W.; Hsiao, W.-T.; Chen, H.-Y.; Hsu, L.-P.; Chen, P.-R.; Lin, M.-D.; Chiu, S.-J.; Shih, W.-L.; Hsu, Y.-C. Upregulated claudin-1 expression confers resistance to cell death of nasopharyngeal carcinoma cells. Int. J. Cancer 2010, 126, 1353–1366. [Google Scholar] [CrossRef]
  69. McClane, B.A.; McDonel, J.L. Protective effects of osmotic stabilizers on morphological and permeability alterations induced in Vero cells by Clostridium perfringens enterotoxin. Biochim. Biophys. Acta 1981, 641, 401–409. [Google Scholar] [CrossRef]
  70. Chang, J.W.; Seo, S.T.; Im, M.A.; Won, H.-R.; Liu, L.; Oh, C.; Jin, Y.L.; Piao, Y.; Kim, H.J.; Kim, J.T.; et al. Claudin-1 mediates progression by regulating EMT through AMPK/TGF-β signaling in head and neck squamous cell carcinoma. Transl. Res. 2022, 247, 58–78. [Google Scholar] [CrossRef]
  71. Pang, X.; Tang, Y.L.; Liang, X.H. Transforming growth factor-β signaling in head and neck squamous cell carcinoma: Insights into cellular responses (Review). Oncol. Lett. 2018, 16, 4799–4806. [Google Scholar] [CrossRef] [PubMed]
  72. Jank, B.J.; Schnoell, J.; Kladnik, K.; Sparr, C.; Haas, M.; Gurnhofer, E.; Lein, A.L.; Brunner, M.; Kenner, L.; Kadletz-Wanke, L.; et al. Targeting TGF beta receptor 1 in head and neck squamous cell carcinoma. Oral Dis. 2023. [Google Scholar] [CrossRef] [PubMed]
  73. Cho, B.C.; Daste, A.; Ravaud, A.; Salas, S.; Isambert, N.; McClay, E.; Awada, A.; Borel, C.; Ojalvo, L.S.; Helwig, C.; et al. Bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in advanced squamous cell carcinoma of the head and neck: Results from a phase I cohort. J. Immunother. Cancer 2020, 8, e000664. [Google Scholar] [CrossRef] [PubMed]
  74. Joseph, M.J.; Dangi-Garimella, S.; Shields, M.A.; Diamond, M.E.; Sun, L.; Koblinski, J.E.; Munshi, H.G. Slug is a downstream mediator of transforming growth factor-beta1-induced matrix metalloproteinase-9 expression and invasion of oral cancer cells. J. Cell. Biochem. 2009, 108, 726–736. [Google Scholar] [CrossRef] [PubMed]
  75. Cappellesso, R.; Marioni, G.; Crescenzi, M.; Giacomelli, L.; Guzzardo, V.; Mussato, A.; Staffieri, A.; Martini, A.; Blandamura, S.; Fassina, A. The prognostic role of the epithelial-mesenchymal transition markers E-cadherin and Slug in laryngeal squamous cell carcinoma. Histopathology 2015, 67, 491–500. [Google Scholar] [CrossRef] [PubMed]
  76. Riechelmann, H.; Steinbichler, T.B.; Sprung, S.; Santer, M.; Runge, A.; Ganswindt, U.; Gamerith, G.; Dudas, J. The Epithelial-Mesenchymal Transcription Factor Slug Predicts Survival Benefit of Up-Front Surgery in Head and Neck Cancer. Cancers 2021, 13, 772. [Google Scholar] [CrossRef] [PubMed]
  77. Smith, A.; Teknos, T.N.; Pan, Q. Epithelial to mesenchymal transition in head and neck squamous cell carcinoma. Oral Oncol. 2013, 49, 287–292. [Google Scholar] [CrossRef] [PubMed]
  78. García-Hernández, V.; Flores-Maldonado, C.; Rincon-Heredia, R.; Verdejo-Torres, O.; Bonilla-Delgado, J.; Meneses-Morales, I.; Gariglio, P.; Contreras, R.G. EGF regulates claudin-2 and -4 expression through Src and STAT3 in MDCK cells. J. Cell Physiol. 2015, 230, 105–115. [Google Scholar] [CrossRef]
  79. Moreira, D.; Sampath, S.; Won, H.; White, S.V.; Su, Y.-L.; Alcantara, M.; Wang, C.; Lee, P.; Maghami, E.; Massarelli, E.; et al. Myeloid cell–targeted STAT3 inhibition sensitizes head and neck cancers to radiotherapy and T cell–mediated immunity. J. Clin. Investig. 2021, 131, e137001. [Google Scholar] [CrossRef]
  80. Zuo, J.H.; Zhu, W.; Li, M.Y.; Li, X.H.; Yi, H.; Zeng, G.Q.; Wan, X.X.; He, Q.Y.; Li, J.H.; Qu, J.Q.; et al. Activation of EGFR promotes squamous carcinoma SCC10A cell migration and invasion via inducing EMT-like phenotype change and MMP-9-mediated degradation of E-cadherin. J. Cell. Biochem. 2011, 112, 2508–2517. [Google Scholar] [CrossRef]
Figure 1. TGF-β, TNF-α, and EGF play a crucial role in neoplastic processes through the modulation of claudins expression in head and neck carcinomas. This figure was generated using biorender.com accessed on July 31 2023. ↑: increased activity; ↓: decreased activity.
Figure 1. TGF-β, TNF-α, and EGF play a crucial role in neoplastic processes through the modulation of claudins expression in head and neck carcinomas. This figure was generated using biorender.com accessed on July 31 2023. ↑: increased activity; ↓: decreased activity.
Cancers 15 04208 g001
Table 1. Claudins showing overexpression in various carcinomas in the head and neck regions. (* conflicting data).
Table 1. Claudins showing overexpression in various carcinomas in the head and neck regions. (* conflicting data).
Claudins OverexpressionType of CancerClinical SignificanceMethodologyReference(s)
1OSCCPoor recurrence-free and cancer-specific survival *IHC and Western blot[37,40,41,42,43]
TSCCIncreased cervical lymph node metastasis IHC[44,45]
Tonsillar squamous cell carcinomaN/A IHC and PCR[46]
Mucoepidermoid carcinomasN/AIHC and PCR[47]
NPCN/AIHC[48]
HSCCPoor differentiation, lymph node metastasis, and reduced survival ratesIHC[49,50]
LLSCCAdvanced tumor stagingIHC[51]
2OSCCShorter recurrence-free survival *IHC[37,39]
3Mucoepidermoid carcinomas N/A IHC and PCR [47]
LSCCDistant metastasis and poorPCR and Western blot[52]
4OSCCIncreased perineural infiltration and poor recurrence-free survivalIHC[39,41,53]
Mucoepidermoid carcinomas *N/AIHC and PCR[47]
LSCC *N/AWestern blot[54]
NPCN/AIHC[48,55]
5Mucoepidermoid carcinomasN/AIHC and PCR[46]
7TSCCPoor survivalIHC and Western blot[44]
Mucoepidermoid carcinomasN/AIHC and PCR[47]
NPCDistant metastasis, increased tumor stages, and poor distant metastases-free survivalIHC[48]
8LSCCPoor histological grade and survivalPCR and Western blot[52]
OSCC: oral squamous cell carcinoma; TSCC: tongue squamous cell carcinoma; NPC: nasopharyngeal carcinoma; HSCC: hypopharyngeal squamous cell carcinoma; LLSCC: lower lip squamous cell carcinoma; LSCC: laryngeal squamous cell carcinoma; IHC: immunohistochemistry; PCR: polymerase chain reaction; N/A: not available.
Table 2. Claudins showing under-expression in various carcinomas of the head and neck. (* conflicting data).
Table 2. Claudins showing under-expression in various carcinomas of the head and neck. (* conflicting data).
Claudins UnderexpressionType of CancerClinical SignificanceMethodologyReference(s)
1LSCCPoor survivalIHC[52]
2NPCN/AIHC[55]
4OSCCPoor recurrence-free survival and increased perineural infiltration *Western blot and IHC[40,41,42,53]
NPCPoor distant metastases-free surivvalIHC[48]
LSCC *N/AIHC and PCR[59]
Salivary gland carcinomas *N/AIHC and PCR[60]
5OSCCN/AIHC and Western blot[39,40]
TSCCN/AIHC[44]
7OSCCHigh pathological grade, advanced staging, increased perineural, vascular, and lymphatic invasions, and poor survivalWestern blot and IHC[40,56]
Salivary adenoid cystic carcinomaIncreased lymph node metastasis and recurrenceIHC, PCR, Western blot, and IFC[61]
Tonsillar squamous cell carcinomaN/AIHC and PCR[46]
LSCC *Poor survival and distant metastasisPCR and Western blot[52]
11LSCCLymph node metastasis, advanced clinical stages, increased T classifications, and poor survival (correlates with claudin-11 promoter hypermethylation)PCR[62]
17OSCCPoor TNM staging and survivalPCR, IHC, and Western blot[58]
LSCC: laryngeal squamous cell carcinoma; NPC: nasopharyngeal carcinoma; OSCC: oral squamous cell carcinoma; TSCC: tongue squamous cell carcinoma; IHC: immunohistochemistry; PCR: polymerase chain reaction; IFC: immunofluorescence; N/A: not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Arabi, T.Z.; Algheryafi, L.A.; Alodah, N.A.; Enabi, H.M.K.; Alshehry, A.A.; Ouban, A. Aberrant Expression of Claudins in Head and Neck Carcinomas and Their Prognostic and Therapeutic Value: A Narrative Review. Cancers 2023, 15, 4208. https://doi.org/10.3390/cancers15174208

AMA Style

Arabi TZ, Algheryafi LA, Alodah NA, Enabi HMK, Alshehry AA, Ouban A. Aberrant Expression of Claudins in Head and Neck Carcinomas and Their Prognostic and Therapeutic Value: A Narrative Review. Cancers. 2023; 15(17):4208. https://doi.org/10.3390/cancers15174208

Chicago/Turabian Style

Arabi, Tarek Ziad, Linah Abdulmohsen Algheryafi, Nora A. Alodah, Hamza M. Kossai Enabi, Amjad Abdullah Alshehry, and Abderrahman Ouban. 2023. "Aberrant Expression of Claudins in Head and Neck Carcinomas and Their Prognostic and Therapeutic Value: A Narrative Review" Cancers 15, no. 17: 4208. https://doi.org/10.3390/cancers15174208

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop