Next Article in Journal
Tannic Acid Mitigates Rotenone-Induced Dopaminergic Neurodegeneration by Inhibiting Inflammation, Oxidative Stress, Apoptosis, and Glutamate Toxicity in Rats
Next Article in Special Issue
Stability Characterization of the Novel Anti-Cancer HM-10/10 HDL-Mimetic Peptide
Previous Article in Journal
No Evidence for Myocarditis or Other Organ Affection by Induction of an Immune Response against Critical SARS-CoV-2 Protein Epitopes in a Mouse Model Susceptible for Autoimmunity
Previous Article in Special Issue
Mind the Curve: Dose–Response Fitting Biases the Synergy Scores across Software Used for Chemotherapy Combination Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of IL-17 in the Pathogenesis of Oral Squamous Cell Carcinoma

by
Nevena Ladjevac
1,2,
Marija Milovanovic
2,3,
Andra Jevtovic
2,4,
Dragana Arsenijevic
2,5,
Bojana Stojanovic
2,6,
Milica Dimitrijevic Stojanovic
2,7,
Bojan Stojanovic
2,8,
Nebojsa Arsenijevic
2,3,
Aleksandar Arsenijevic
2,3,* and
Jelena Milovanovic
2,9,*
1
Department of Otorhinolaryngology, General Hospital Uzice, 31000 Uzice, Serbia
2
Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
3
Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
4
Department of Otorhinolaryngology and Maxillofacial Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
5
Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
6
Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
7
Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
8
Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
9
Department of Histology end Embryology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(12), 9874; https://doi.org/10.3390/ijms24129874
Submission received: 12 May 2023 / Revised: 1 June 2023 / Accepted: 3 June 2023 / Published: 8 June 2023
(This article belongs to the Special Issue Cancer Treatment: New Drugs and Strategies)

Abstract

:
Elucidating the inflammatory mechanisms underlying formation and progression of oral squamous cell carcinoma (OSCC) is crucial for discovering new targeted therapeutics. The proinflammatory cytokine IL-17 has proven roles in tumor formation, growth, and metastasis. The presence of IL-17 is demonstrated in both in vitro and in vivo models, and in OSCC patients, is mostly accompanied by enhanced proliferation and invasiveness of cancer cells. Here we review the known facts regarding the role of IL-17 in OSCC pathogenesis, namely the IL-17 mediated production of proinflammatory mediators that mobilize and activate myeloid cells with suppressive and proangiogenic activities and proliferative signals that directly induce proliferation of cancer cells and stem cells. The possibility of a potential IL-17 blockade in OSCC therapy is also discussed.

1. Introduction

Among all head and neck carcinomas, including tumors arising from epithelial surfaces from the oral cavity, pharynx, larynx, and paranasal sinuses, and major and minor salivary glands, the most common are oral cell carcinomas representing almost 50% of such cases. The most frequent malignancy among oral cell carcinomas is oral squamous cell carcinoma (OSCC) accounting approximately 90% of these cases [1,2]. The OSCCs are tumors that arise in the oral cavity with localization on the lips, gums, lining of the cheeks and lips, front two-thirds of the tongue, floor of the mouth under the tongue, roof of the mouth, and oropharynx [3]. The primary approaches for the treatment of OSCC are traditional surgery, radiotherapy, and a combination of surgery and radiotherapy that, although it has been improved in recent years, has failed to increase the survival. The global 5-year survival rate is about 50% of all OSCC cases [4]. Prediction of survival in OSCC depends on classical parameters including tumor grade, depth of invasion, the time of diagnosis, and the inflammatory score, thus early-stage OSCC patients have a 5-year survival rate of about 75%, while patients with advanced stages of OSCC at diagnosis have only a 35% survival rate [5,6]. Immunotherapy has been recently introduced as an effective treatment option for OSCC and was firstly approved for recurrent/metastatic cases [7], and for preoperative neoadjuvant immunotherapy for untreated OSCC [8].
Discovering the mechanisms that promote malignant transformation, tumor progression, invasion, and metastasis are fundamental in the process of searching for new targeted therapeutics. There is plethora of evidence regarding an association between OSCC and chronic inflammation [9,10]. Chronic inflammation is a common feature of established OSCC [11]. Dysregulation of different genes whose products are involved in inflammation, wound healing, and angiogenesis was discovered in OSCC cell lines and tissue samples obtained from OSCC patients by microarray analysis [9]. Inflammatory and immune cells that are the main components of the tumor microenvironment affect the processes of OSCC proliferation, survival, invasiveness, and metastasis [12]. Furthermore, the progressive increase in density of inflammatory infiltration has been detected in parallel with increasing grades of oral malignant transformation process from non-dysplastic hyperkeratosis, epithelial dysplasia to OSCC [12,13]. Chronic oral inflammatory conditions, oral lichen planus, submucous fibrosis, and oral discoid lupus, are all predisposing for the development of OSCC [14]. Different cytokines, chemokines, prostaglandins, reactive oxygen species, and transcription factors present in the microenvironment of these chronic oral inflammatory conditions are known mediators of cell proliferation, epithelial-to-mesenchymal transition, and invasion [12,15].
The pathogenesis of OSCC is complex; it includes genetic predisposition, risk factors and interactions of all the components of the immune system. Interleukin-17 has been recently marked as a key link between inflammation, wound healing, and cancer [16] and its role in OSCC development and progression is the focus of this review.
The risk factors associated with OSCC include alcohol, marijuana, and tobacco (including cigarettes, pipes, smokeless tobacco, vaping) consumption, chewing of betel leaf and areca nut, human papillomavirus (HPV) infection, and prolonged oral dysbiosis [17]. Alcohol and tobacco consumption precede the development of 75% of all OSCC cases [18], with increased risk when both are used on a regular basis [19]. Cigarette smoke, by stimulation of interleukin-(IL-)17 mediated inflammation, induces genomic instability and thus can play an important role in the cancer development [20]. Moreover, it is known that hepatic steatosis, inflammation, fibrosis, and finally hepatocellular carcinoma (HCC), induced by alcohol are critically regulated by IL-17A [21].
About 15–20% of all OSCCs are related to high-risk HPV infection [22], with the most predilected localizations in the oropharynx, tonsils, and base of the tongue [23]. Almost 70% of OSCCs localized in the oropharynx are positive for high-risk HPVs [24]. The improved prognosis of HPV-positive OSCC has been shown to be associated with higher numbers of tumor infiltrating T helper (Th)17 cells and lower numbers of IL-17 producing non-T cells [25]. The broken balance between more than 700 bacterial species that are part of the bacterial flora in the oral cavity causes dysbiosis and leads to development of oral diseases such as periodontal disease [26]. Close links between the composition of the oral bacterial flora, presence of specific oral bacteria and OSCC have been reported [27,28,29]. Furthermore, several studies have shown that oral bacteria such as Porphyromonas gingivalis and Fusobacterium nucleatum alter the oral microbiota in a way that enhances the risk of OSCC development and OSCC progression by enhancing cell proliferation, inhibiting apoptosis, and improving tumor invasion [30,31,32]. These pathogens also activate monocytes resulting in increased IL-17 production by human immune cells, a process that precedes development of periodontal diseases [33] that can be a risk factor for OSCC.
A recent meta-analysis reported dominant overexpression of IL-17 and Th17 cells in the local inflammatory infiltrates in oral lichen planus, predisposing inflammatory condition to OSCC, higher concentration of IL-17 in the serum of these patients, and more intense IL-17 expression in erosive than in reticular oral lichen planus, suggesting a positive correlation between IL-17 levels and disease severity [34].
Various polymorphisms and combinations of specific genetic mutations have been reported to be associated with an increased risk for development of OSCC [35]. Interleukin 17A and IL-17F polymorphisms are associated with increased risk for OSCC and are related to tumor stage and differentiation. In addition, it has been shown that the IL-17A and IL-17F polymorphisms increase the risk of OSCC developing in a population exposed to two other risk factors, smoking and alcohol [36].
Development and progression of OSCC are strongly affected by different components of the immune system [37]. However, the impact of IL-17, an inflammatory cytokine that closely contributes to the development, progression and metastasis of various tumors and affects the sensitivity to chemotherapy and radiation therapy on OSCC formation and progression requires further elucidation. In this review, the possible roles of IL-17 in OSCC development will be discussed.

2. Interleukin-17, IL-17R, and Signal Transduction

Interleukin-17 is an inflammatory cytokine with proven roles in chronic inflammatory and autoimmune diseases [38] and in the formation and progression of cancers of the colon [39], stomach [40], pancreas [41], liver [21,42,43], skin [44], lung [45], and myeloma [46]. Interleukin-17as an inflammatory cytokine helps in establishing the tumor stroma that supports tumor formation and growth. Interleukin-17is a key cytokine of CD4+ T helper 17 (Th17) cells [47]. In addition to Th17 cells, a number of immune cells also produce IL-17, including γδ T cells [48], cytotoxic T cells (CD8+ αβ T cells [49] natural killer (NK) cells [50], invariant (i)NKT cells [51], innate lymphoid cells [52], neutrophils [53], eosinophils [54], and macrophages [55]. These cells are collectively named type 17 cells [56]. The transforming growth factor (TGF)-β together with IL-6 drives the differentiation of Th17 cells [47]. Most type 17 cells produce IL-17 after stimulation with IL-1 and IL-23 [57,58,59,60], which activates the transcriptional factors Signal transducer and activator of transcription (STAT)3 [61] and RAR-related orphan receptor (ROR)γt [62]. In γδT cells, the expression of IL-17 is controlled by the transcription factor c-Maf [63]. All these type 17 cells play spatially and temporally specific roles in physiological responses, but in chronic inflammatory conditions and cancer, all of them produce IL-17 that has roles in the maintenance of pathological processes [64].
The family of IL-17 cytokines consists of six cytokines: IL-17A (the prototype of IL-17), IL-17B, IL-17C, IL-17D, IL-17E (also known as IL-25), and IL-17F. The most studied cytokines of the IL-17 family are IL-17A and IL-17F, the two cytokines with the highest homology that are usually co-produced [65]. Interleukin-17A and IL-17F exist either as homodimers or as a heterodimer, and all forms of the cytokine signal through a heterodimeric dimeric receptor complex consisting of the IL-17 receptor A (IL-17RA) and IL-17RC [66]. All types of IL-17R molecules contain a conserved cytoplasmic motif known as the similar expression of fibroblast growth factor and IL-17R (SEFIR) domain [67]. The cytosolic adaptor Act1 contains a SEFIR domain and interacts with IL-17RA and IL-17RC through homotypic SEFIR interactions leading to the activation of all IL-17-dependent signaling pathways [68,69]. In addition to the SEFIR domain, IL-17RA also contains a non-conserved region that extends ~100 residues beyond the SEFIR, termed a “SEFIR-Extension” (SEFEX) that is required for IL-17RA signaling [70,71], and with SEFIR comprises a single composite structural motif [72]. Interleukin-17 upregulates the expression of signature genes (inflammatory cytokines, chemokines, antimicrobial peptides, and matrix metalloproteinases) either by inducing de novo gene transcription or by stabilizing target mRNA transcripts. The earliest event after IL-17 receptor engagement is the association of the IL-17R with Act1, a key adaptor molecule required for both the transcriptional and post-transcriptional changes induced by IL-17 (Figure 1) [73]. The Act1 is a nonredundant activator of IL-17RA dependent signals. It functions as a Lysine-63 (K63) E3 ubiquitin ligase, which recruits and ubiquitinates TNF receptor associated factor 6 (TRAF6), leading to the recruitment and activation of the transforming growth factor β-activated kinase (TAK)1 and the inhibitor of nuclear factor (NF)-κB kinase (IKK) complex [74,75]. The IKK then phosphorylates the IκB subunit of the NF-κB:IκB complex and marks it for proteasomal degradation, exposing thus a nuclear localization signal on NF-κB, and allowing the rapid nuclear translocation and consequent inflammatory gene transcription [76]. Additionally, IL-17 is able to activate the spleen tyrosine kinase (Syk) tyrosine kinase associated with IL-17RA, Act1 and TRAF6 in keratinocytes, resulting in activation of NF-κB [77]. The C/EBPs CCAAT/enhancer-binding protein (C/EBP) transcription factors are additional transcriptional regulators activated by IL-17 [78]. The TRAF6 activated by IL-17 binding to IL-17 receptor promotes activation of mitogen-activated protein kinase (MAPK) pathways: extracellular signal-regulated kinase (ERK), p38 and JUN N-terminal kinase (JNK) [79], AP1 (activator protein 1) pathways, and the C/EBPβ transcription factors [74]. The IKK mediates p105 phosphorylation, releases TPL2 kinase from p105 and activates p38 and JNK [80]. Furthermore, IL-17 induces formation of a multi-protein signaling complex that comprises IL-17R-ACT1-TRAF4- mitogen-activated protein kinase kinase kinase (MEKK)3-MEK5, which activates extracellular signal-regulated kinase (ERK)5, but not NF-κB, p38, JNK, or ERK1/2, inducing expression of IL-17 target genes, which leads to keratinocyte proliferation and eventually tumor formation [81].
Transcriptional changes induced by IL-17 are relatively weak in contrast to less well-defined but more robust IL-17 induced posttranscriptional changes that include stabilization of specific mRNAs and protein translation. The mRNAs that encode inflammatory mediators are relatively unstable, enabling the fine-tuning of gene expression during inflammatory responses [82]. Interleukin-17 enhances the level of inflammatory mRNAs by protection of inflammatory mRNAs from degradation through the inhibition of Regnase-1, an endoribonuclease [83]. The adaptor for IL-17R, Act1, can also function as an RNA binding protein in the complex with TRAF2 and TRAF5, and thus interacts with target mRNAs, including C-X-C Motif Chemokine Ligand 1 (Cxcl1), Colony stimulating factor-2 (Csf2) and Tumor necrosis factor (Tnf) [83], playing a direct role in mRNA metabolism, stabilization, and translation. The SEFIR domain of Act1 recognizes and binds to SEFIR-binding elements in IL-17 target transcripts, enabling the Act1 direct formation of three compartmentally distinct protein–RNA complexes that prevent mRNA decay in the nucleus, inhibit mRNA decapping in P-bodies, and promote client mRNA translation in the polyribosomes [83]. Interleukin-17 also induces the interactions of Act1 and IKKi and TANK-Binding Kinase 1 (TBK1), which translocate to nucleus and phosphorylate splicing factor(SF)2 and diminishes SF2 mediated mRNA decay [83,84]. Furthermore, Act1 facilitates the binding of HuR to mRNA, enabling the movement of mRNAs into polyribosomes for translation [85]. Interleukin-17 also induces the expression of the RNA binding protein, Arid5a, which binds TRAF2 and stabilizes IL-17 induced transcripts by competing with Regnase-1 [84]. The post-transcriptional regulation of mRNA by IL-17 is part of a self-reinforcing mechanism that potentiates IL-17 activity [86]. Moreover, the ability of IL-17 to modulate the post-transcriptional mRNA metabolism can explain its strong proinflammatory actions in vivo in contrast to modest transcriptional activation in vivo [64].
The expression of IL-17 receptors is ubiquitous, but the main targets of IL-17 are non-hematopoietic cells [87]. Interleukin-17 signaling induces the production of proinflammatory cytokines (IL-1, IL-6, G-CSF, GM-CSF, and TNF-α), chemokines (CXCL1, CXCL2, CXCL5, CCL2, CCL7, CCL20, and IL-8), matrix metalloproteinases (MMP1, MMP3, MMP9, and MMP13), and anti-microbial peptides (β-defensins, S-100 proteins) [88]. The biological activities of IL-17 are often the result of synergistic or cooperative effects with other inflammatory cytokines such as TNF-α [89] leading to amplifying of inflammatory response. Interleukin-17 also cooperates with other cytokines IFN-γ, IL-13, TGF-β, and microbial products [90,91,92].

3. IL-17 Dependent Inflammation and OSCC

Dysregulated IL-17 is marked as a major pathogenic factor involved in both the early and late development stages of various cancers [16]. The role of IL-17 in potentiation of OSCC development has been shown in the mouse model. Expression of IL-17 and IL-17-induced inflammatory molecules has been significantly upregulated during progression from normal mucosa to hyperplasia and tumor formation, while inhibition of IL-17 delayed the development of precancerous and cancerous lesions in mice treated with 4-Nitroquinoline 1-oxide (4NQO), and prolonged their survival [93].
In support of results obtained in animal studies, the serum levels of IL-17A [94] and IL-17F [95] were significantly higher in OSCC patients when compared to controls. One study has reported a lower concentration of IL-17F in the serum of OSCC patients compared to controls that was positively associated with the numbers of CD3+CD4+ T cells, indicating that CD4+ T cells are the main source of IL-17F during the development of OSCC [96]. Furthermore, concentrations of IL-17A, IL-17F, and TNF-α have been reported to be significantly higher in saliva of patients with cancer of the oral cavity and oropharynx and are strongly associated with disease advancement [97]. Polymorphisms of IL-17 and IL-17F have been associated with oral squamous cell carcinoma risk, and are related to tumor stage and differentiation, and potentiate the protumorogenic effects of tobacco and alcohol, enhancing thus the risk of OSCC development [36]. Recent analysis has revealed that the most significant differentially expressed genes in OSCC are, among others, genes encoding molecules involved in the IL-17 signaling pathway [98].
Several studies have reported the presence of IL-17 and IL-17 producing cells in the peripheral blood of OSCC patients. Significantly higher frequency of Th17 cells in the peripheral blood of OSCC patients compared to controls has been reported [99]. These cells were found to express markers of activation and CCR6 chemokine receptor. The cytokine profiling of these cells revealed three Th17 subsets (Th17/1 (IL17A+IFNγ+), Th17/inflammatory (IL17A+IL8+), and Th17/2 (IL17A+IL4+)), which all were elevated in OSCC patients compared to controls [99]. A shift toward the Th17/1 cell type was observed in the early stage OSCC patients [99]. In line with this, an increase in the Th17/Tregs ratio in early stages of OSCC without lymph node involvement and a decrease in this ratio in higher clinical stages and lymph node involvement has been reported, indicating Th17 and Tregs cells as significant prognostic factors in OSCC patients [100]. Additionally, significantly higher frequencies of Th17 and IL-17 producing CD8+ cells (Tc17) were found in the peripheral blood of head and neck cancer patients that were positively correlated with the disease stage, suggesting the role of IL-17 in the creation of the inflammatory pro-tumor environment [101].
A significantly higher expression of IL-17 in OSCC tissue and tumor margins compared to normal tissue, detected by immunohistochemical staining, has been reported [102]. Furthermore, a significant correlation between IL-17 positive tumor budding (insulated single or small clusters of cancer cells (no more than five cells) that indicates the loss of cellular cohesion and the presence of active invasive movement [103]) and tumor classification, lymph node metastasis, distant metastasis, clinical stage, and OSCC recurrence was found [102]. Tumor budding at the tumor invasion front of OSCC is related to local metastasis and poor prognosis [104]. The presence of IL-17 in OSCC budding suggests that IL-17 takes a role in tumor invasion and promotes OSCC progression. In another study, increased expression of IL-17 in the OSCC tumoral islands, the tumor-stroma interface, and more distant stroma was observed [105]. Helper T cells, cytotoxic T cells, and macrophages were identified as the main cellular source of IL-17 in this study and there was no IL-17 in supernatants of the OSCC cell lines [105]. As tumor cells in OSCC tissue express IL-17R [101], it is very possible that IL-17 released by immune cells in the OSCC microenvironment directly stimulates OSCC tumor cells and maybe induces their proliferation because it was shown that IL-17 stimulates proliferation of OSCC cell lines in vitro [101]. Analysis of tumor infiltrating lymphocytes in OSCC revealed higher amounts of Th17, Tc17, and Tregs in tumor tissue, ones higher in comparison with their frequency in peripheral blood, and a correlation of high Th17/Treg ratio and overall survival [106]. Furthermore, it has been found that the frequency of IL-17+ T cells was inversely correlated with tumor size, while the frequency of Foxp3+T cell in tumor infiltrates was positively correlated with the TNM stage [106]. Lee et al. reported increased prevalence of IL-17 producing FOXP3+CD4+ tumor infiltrating lymphocytes in oral squamous cell carcinoma that showed suppressive capacity [107]. These cells express CCR6 and suppress the proliferation of autologous CD4+CD25- responder T-cells in vitro [107]. In fact, both Th17 and Tregs were accumulated in the tumor microenvironment at early stages of the OSCC development, but in parallel with tumor progression, the numbers of Th17 cells in the infiltrates gradually decreased while the Treg numbers increased in infiltrates as the disease progresses. The reason for these opposite results may be the high plasticity of the Th17 cells that can convert into Th1, Treg, or Th2 cells in response to various microenvironments and thus gain various contrary activities [108]. Recently it has been reported that in the late stage of 4NQO induced oral tumors in mice, ablation of Treg cells triggers an increase in the number of both CD4+ and CD8+ effector T cells within oral lesions [109]. Interestingly, this manipulation does not induce tumor regression, instead it induces the effector T cell dependent rapid emergence of invasive OSCC [109].
Chronic IL-17 activity induces the production of proinflammatory cytokines and chemokines and thus stimulates accumulation of neutrophils in the blood and tissues and induces inflammation, it mediates the release of pro-angiogenic cytokines from fibroblasts stimulating wound-healing pathways [16] and may be associated with tissue destruction through matrix metalloproteinases [110,111]. All these processes may play a role in OSCC formation and progression. Dysregulated IL-17 production can be triggered by a pathogenic microbiota, which induces continuous activity of the immune system to limit invasive colonization. It has been recently shown that experimental periodontitis promotes the formation of OSCC [112]. The oral microbiota in periodontitis directly activates IL-17+γδ T cells, stimulates signal transducer and activator of transcription 3 (STAT3) pathway, and promotes infiltration of oral carcinoma tissue with M2-tumor-associated macrophages [112]. Inhibition of γδ T cells led to decreases in the concentration of IL-17A, the phosphorylation level of STAT3, and the size of tumors [112]. Moreover, the proportion of IL-17+ γδ T cells and the phosphorylation of STAT3 were higher in the tissues obtained from OSCC patients with periodontitis group compared to OSCC patients without periodontitis [112]. This study provides experimental evidence regarding cross talk among the microbiota, IL-17 signaling, inflammation, and oral carcinoma cells.
The significance of IL-17 in OSCC progression can be indirectly confirmed by the results of the study that showed overexpression of Akt1, nonredundant activator of IL-17RA-dependent signals in OSCC tissue, association of genetic alterations of Akt1 with a poor clinical outcome in OSCC, and decreased expression of proteins regulating cell survival leading to decreased OSCC cell survival after silencing of Akt1 [113].
The IL-23R knockout mice had faster progression of premalignant oral lesions to cancer, compared to wild type mice, but both groups developed the same histological OSCC score 18 weeks after initiation of 4NQO treatment [114]. Although IL-23R KO mice had reduced inflammation (lower levels of IL-17 among other inflammatory cytokines) in the stage of premalignant lesions, conversion to the inhibitory phenotype of inflammatory cells (IL-10 producing) in the stage of oral cancer lacked inflammation. Results of this study are in line with previously listed results obtained from human samples and confirm the importance of IL-17 in conversion of premalignant lesion to oral cancer, but also show the importance of IL-23 signaling and, indirectly, Th17 cells in limiting OSCC growth [115]. Mice bearing premalignant oral lesions treated with a TGF-β type 1 receptor inhibitor plus IL-23 in order to sustain the Th17 phenotype also slowed the progression of premalignant lesion to cancer [116]. Protective roles of Th17 and Tc17 cells in eradicating already established tumors have been reported [116].
Due to the impact on mRNA metabolism, IL-17 is able to perform its activity synergistically with other cytokines and activates diverse signaling pathways. Interleukin-17 and TNF-α synergistically activate NF-κB, a transcriptional factor with known roles in the promotion of inflammation and OSCC progression [117,118,119]. Interleukin-17 signals cooperatively with the IFN-γ enhances activation of STAT1, a molecule involved in signaling pathways important for OSCC growth and metastasis [120,121]. Furthermore, IL-17 cooperates with TGF-β and activates SMAD signal transducers known for their modulation of OSCC microenvironment and promotion of OSCC invasion [122,123]. Further investigations are needed in order to determine the impact of different synergizing partners of IL-17 in driving the inflammatory pathways and the OSCC outcome.

4. The Role of IL-17 Stimulated Microenvironmental Cells in OSCC Progression

Interleukin-17 activates transcriptional factors and stabilizes specific mRNAs resulting in the production of inflammatory mediators and various ligands that induce favorable microenvironment for OSCC progression (Figure 2). Extracellular vesicles (EVs) contain proteins and nucleic acids that efficiently mediate intercellular communication [124] and are considered as one of the main players in the communication between cells in the inflammatory tumor microenvironment [125], actively contributing to tumor growth, invasion, and metastasis [126]. Recently it has been shown that OSCC-derived EVs induce overactivation of the IL-17A-signaling pathway in tumor tissue, causing an inflammatory cytokine imbalance in the tumor microenvironment, and thus promote OSCC xenograft tumor growth [127].

4.1. Impact of IL-17 on Myeloid Cells

Recruitment of neutrophils is critically controlled by IL-17 and contributes to host defense [128], but a sustained IL-17 activity because of non-resolving inflammation and associated chronic wounding, persistent infection, or carcinogenesis, induces generation of pathogenic myeloid cells, and myeloid derived suppressor cells (MDSC) [129]. Interleukin-17 stimulates G-CSF production that promotes the expansion of granulocytes [130], and stimulates production of the proinflammatory cytokines IL-6 and TNF-α that play roles in inducing a suppressive phenotype in the recruited myeloid cells [131]. The MDSCs suppress the anti-tumor functions of T and natural killer (NK) cells and promote tumor cell proliferation, survival, invasiveness, and metastasis [132].
Two studies have reported higher levels of MDSC in the peripheral blood of OSCC patients compared to controls, with significant positive correlation with the tumor size and stage [133,134]. In addition, the levels of MDSC are shown to correlate with tumor progression in human head and neck cancer [135]. Moreover, granulocytic/polymorphonuclear MDSC has been detected in OSCC tissue accompanied with increased levels of Th17 cells in peripheral blood and tumor tissue [136]. Another study has reported higher percentages of peripheral blood MDSCs and Th17 cells, and the level of IL-17 in serum of OSCC patients compared to healthy controls [137]. Additionally, significant correlation was found between the number of MDSCs and the level of IL-17, while no correlation was found between the numbers of MDSCs and Th17 cells implying that other cells, not Th17, are the main source of IL-17 in OSCC [137]. Infiltration with neutrophils and elevated expression of TGF-β1 and IL-17A in OSCC tissues accompanied with increased expression of MMP9 and decreased expression of CCL3 in circulating neutrophils, has been reported [138]. The cooperative effects of TGF-β1 and IL-17A were also reported in this study. It was shown that neutrophils in vitro exposed to TGF-β1 and IL-17A have augmented protumor activities that induced cell migration, proliferation, invasion, stemness, and epithelial to mesenchymal transition in OSCC cells in vitro [138]. Combined positivity for tumor associated neutrophils, MMP-9, IL-17, and CD105 was reported to be associated with the metastasis-prone phenotype of OSCC [139].
In a mouse model of oral cancer induced by 4NQO, higher number of MDSCs in the peripheral blood and spleen during carcinogenesis was reported in two studies [140,141]. Depletion of MDSCs in the mouse model of 4NQO induced oral cancer significantly ameliorated carcinogenesis that was promoted by a high fat diet [142]. The importance of MDSCs in 4NQO carcinogenesis was confirmed by the study that shows the difference in tumor growth between CD24−/− and CD24+/− mice was blunted by immuno-depletion of MDSCs [142]. Furthermore, Porphyromonas gingivalis induced progression of oral cancer by generating a cancer-promoting microenvironment that contains increased number of MDSCs [143,144]. In addition, it has been shown that C. albicans promotes development of 4NQO-induced oral cancer via the IL-17A/IL-17RA induced tumor associated macrophages [145]. Reduction in 4NQO induced lesions upon treatment with anti-PD-1 monoclonal antibody in vivo was accompanied by reduction in MDSCs in the lesion-microenvironment and peripheral lymph nodes [146]. The therapeutic effects of anti-hypoxic agents in 4NQO oral cancer have been accompanied by reduced presence of MDSCs in the tumor microenvironment [147].
Collectively, these studies demonstrate that myeloid suppressor cells that are IL-17 dependent accumulate in oral squamous cancers and contribute to tumor progression.

4.2. Impact of IL-17 on Cancer Associated Fibroblasts

One of the constituents of the tumor microenvironment is cancer associated fibroblasts (CAFs) that play one of the main roles in cancer progression. An additional contribution of IL-17 to forming the microenvironment that supports tumor growth is tumor immune exclusion by affecting CAFs to increase deposition of extracellular matrix [148]. Interleukin-17 contributes to pathological fibrosis in many organs including the lung and liver, enhances pro-fibrotic phenotypes in synergy with TGF-β [149], and in synergy with IL-22 contributes to the epithelial-mesenchymal transition in primary human salivary gland epithelial cells that are isolated from healthy subjects [150]. Deletion of IL-17 signaling, specifically in CAFs in late-stage tumors, led to reduced proliferation and numbers of CAFs, and reduced collagen deposition in murine models of cutaneous squamous cell carcinoma [148]. This reduction in CAFs and extracellular matrix made the tumors susceptible to anti–PD-L1 therapy [148]. There are many studies on clinical specimens showing that CAFs play a role in OSCC proliferation, susceptibility to antitumor immune response, progression, invasiveness, resistance to therapeutics [151,152,153,154,155]. Therefore, future studies in animal models are needed to elucidate the role of IL-17 signaling in CAFs in OSCC.

5. Protective Roles of IL-17 in Oral Squamous Cell Carcinoma

Interleukin-17 was reported to play protective roles in several types of cancer, including oral, colon, and hepatocellular carcinoma [95,155,156]. Several articles reported protective roles of IL-17F in OSCC. Most cells derived extracellular IL-17F at the tumor invasion front, which was associated with better disease-specific survival among patients with all stages oral tongue squamous cell carcinoma [157]. Two studies reported decreased serum level of IL-17F in OSCC patients compared with healthy controls [95,96]. Antitumor effects of IL-17F against OSCC cells were observed in vitro. Interleukin-17F inhibited the proliferation and random migration of oral tongue squamous cell carcinoma cells, HSC-3 [158]. Furthermore, IL-17F suppressed the human umbilical vein endothelial cells tube formation suggesting antiangiogenic effects of this cytokine [158]. Additionally, IL-17F suppressed cancer associated fibroblasts mediated invasion of oral tongue squamous carcinoma cells in tumor spheroids [158]. Recently, the potential of IL-17F to inhibit the formation of vasculogenic mimicry structures, an alternative vasculogenic system made by aggressive tumor cells implicated in treatment failure and poor survival of cancer patients, was reported [159]. One study also reported the protective role of IL-17A in OSCC, mediated by promotion of Th17 differentiation associated with suppressed growth of implanted OSCC tumors in nude mice [160].

6. Direct Effects of IL-17 on Tumor Cells

It has been shown that IL-17 stimulation, by activating mitogenic signaling pathways, can directly promote the proliferation of keratinocytes and intestinal epithelial cells [161,162]. Interleukin-17 mediated signaling is critical for the maintenance and repair of tissue barrier function in the oral cavity [163]. The main producers of IL-17 in the oral mucosa are γδT cells that release this cytokine spontaneously as part of the process of maintaining epithelial integrity and also under inflammatory conditions [163]. Since recently, it is known that IL-17 can directly promote the proliferation of premalignant cells, that play a crucial role in the early stage of tumorigenesis. Interleukin-17 is a critical inflammatory signal that activates a group of Lrig1+ stem cells that normally reside in the hair follicle to participate in wound healing in the skin [164]. Interleukin-17 induced transactivation of EGFR, Src, and ERK5 in vivo that leads to expansion and migration of Lrig1+stem cells and their transformation, links IL-17 mediated wound healing and tumorigenesis [164]. The negative regulator of EGF, Lrig1, induced EGFR activation [165], but IL-17 is able to transactivate EGFR and activates cells in a state of inflammation or environmentally challenged. Oral epithelial stem cells that express Lrig1 are slow-cycling but are stress responsive [166], thus it is possible that IL-17, in the states of chronic inflammation in the oral cavity and precancerous lesions, such as lichen planus where IL-17 expression in the lesion correlates with disease severity [34], stimulates these cells for tissue repair and possibly induces tumorigenesis. Recently it was shown that extracellular vesicles obtained from mouse OSCC cell lines increased serum levels of IL-17 in mouse recipients and significantly increased xenograft tumor growth and invasion [167]. Furthermore, treatment with extracellular vesicles significantly enhanced the expression levels of crucial molecules in the IL-17A pathway, IL-17A, TRAF6 and c-FOS in tumor tissue and suppressed immune responses of CD8+ cells in vivo [167]. Interleukin-17A showed direct effects on OSCC cells in vitro, it enhanced cell migration and invasion in SCC15, a tongue squamous cell carcinoma cell line [168].

7. IL-17 and Cancer Therapy

Intratumoral inflammatory mechanisms are one of the most influential players in development of resistance to therapy [169]. Since IL-17 plays the main role in developing the protumorigenic inflammatory environment and drives tissue repair it is expected that it also contributes to tissue healing after chemotherapy and radiotherapy and its role in causing chemoresistance is being explored. Since IL-17 induces the activation, proliferation, and epithelial-to-mesenchymal transition of quiescent Lrig1 expressing epithelial stem cells [165], and Lrig1 can be induced in cancer cells, IL-17 signaling could provide better survival of tumor cells after chemo- or radio-therapy. The low-dose irradiation induces expression of IL-17 in tumor beds and enhances the growth of subsequently implanted tumor cells, while treatment with anti-IL-17 antibody abolished the acceleration of tumor growth, confirming the key role of IL-17 in enhancing tumor growth in pre-irradiated tumor beds [170]. Interleukin-17 plays a role in restoration of the damaged epithelia in radiation induced oral mucositis and attenuates epithelial damage [171], which imply that a similar IL-17 mediated process could contribute to better recovery of OSCC cells after radiotherapy and eventually chemotherapy and subsequently contribute to resistance of oral squamous cancer to therapy. Flavonoid induced better sensitivity of chemo-resistant OSCC cells to cisplatin is accompanied by the downregulation of IL-17 [171]. Moreover, in the mouse model of OSCC, it has been shown that inhibition of IL-17A combined with the PD-1 blockade delayed the development of precancerous and cancerous lesions and prolonged the survival of 4NQO-treated mice, suggesting the IL-17A blockade as a potential approach to augment the tumor eliminating effects of anti-PD-1 therapy [92].

8. Conclusions

In this review, the literature regarding the role of IL-17 in the promotion of OSCC tumorigenesis is summarized. Interleukin-17 can promote OSCC tumorigenesis by several pathways, such as provisioning the microenvironment that promotes cell transformation and tumor formation and inducing production of molecules (VEGF, MMP-9, MMP-13, CCR6, and PGE2) that promote invasiveness and angiogenesis; enhancement of immunosuppressive effects of MDSCs that promote tumor cell proliferation, survival, invasiveness and metastasis; direct proliferative effects on tumor cells; activation and transformation of stem cells. In contrast, the results of some studies mark IL-17 as a protective cytokine in OSCC. These opposite reports could be explained by different phases of the disease when the evaluation of cytokine was done. In addition, the cellular source of IL-17 could be related to its impact on tumor formation and proliferation. Therefore, further studies are needed for better elucidation of the role of IL-17 in OSCC tumorigenesis, exploring synergizing partners of IL-17 in driving the inflammatory pathways in OSCC, and the role of IL-17 stimulated CAFs in OSCC pathogenesis.
There is limited information regarding the role of IL-17 in regulating the response to checkpoint inhibitors or other immunomodulators and to chemotherapy and radiotherapy of OSCC. It is important to further explore whether blockade of IL-17 sensitizes resistant oral squamous cell carcinomas to chemo-, radio- and immuno-therapy, or can it be used in the prevention of OSCC.

Author Contributions

N.L. and A.J. were responsible for conceptualization and writing. D.A., M.D.S., B.S. (Bojana Stojanovic) and B.S. (Bojan Stojanovic) were responsible for analysis and the reviewing the literature. M.M. provided critical insights and interpretation of the reviewed data. N.A. was responsible for the overall project administration and supervision. A.A. and J.M. were responsible for supervision, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Science, Technological Development, and Innovation of the Republic of Serbia, contract number for 2023: 451-03-47/2023-01/200111), and the Faculty of Medical Sciences of the University of Kragujevac, Serbia (MP 01/19 and JP18/19, JP 19/19).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This study is a review article and does not generate any new datasets or involve the use of existing datasets. The authors have drawn upon and analyzed previously published research in the field, and all relevant sources have been cited within the manuscript. Readers interested in accessing the original data sources can refer to the cited references for further information.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nakashima, T.; Tomita, H.; Hirata, A.; Ishida, K.; Hisamatsu, K.; Hatano, Y.; Kanayama, T.; Niwa, A.; Noguchi, K.; Kato, K.; et al. Promotion of Cell Proliferation by the Proto-Oncogene DEK Enhances Oral Squamous Cell Carcinogenesis through Field Cancerization. Cancer Med. 2017, 6, 2424–2439. [Google Scholar] [CrossRef] [PubMed]
  2. Vigneswaran, N.; Williams, M.D. Epidemiologic Trends in Head and Neck Cancer and Aids in Diagnosis. Oral Maxillofac. Surg. Clin. North Am. 2014, 26, 123–141. [Google Scholar] [CrossRef] [PubMed]
  3. Daraei, P.; Moore, C.E. Racial Disparity among the Head and Neck Cancer Population. J. Cancer Educ. 2015, 30, 546–551. [Google Scholar] [CrossRef]
  4. Sathiasekar, A.C.; Mathew, D.G.; Jaish Lal, M.S.; Arul Prakash, A.A.; Goma Kumar, K.U. Oral Field Cancerization and Its Clinical Implications in the Management in Potentially Malignant Disorders. J. Pharm. Bioallied Sci. 2017, 9, S23–S25. [Google Scholar] [PubMed]
  5. Chin, D.; Boyle, G.M.; Porceddu, S.; Theile, D.R.; Parsons, P.G.; Coman, W.B. Head and Neck Cancer: Past, Present and Future. Expert Rev. Anticancer Ther. 2006, 6, 1111–1118. [Google Scholar] [CrossRef]
  6. Lee, S.; Kim, D.W.; Kwon, S.; Kim, H.J.; Cha, I.H.; Nam, W. Prognostic value of systemic inflammatory markers for oral cancer patients based on the 8th edition of AJCC staging system. Sci. Rep. 2020, 10, 12111. [Google Scholar] [CrossRef]
  7. Zandberg, D.P.; Algazi, A.P.; Jimeno, A.; Good, J.S.; Fayette, J.; Bouganim, N.; Ready, N.E.; Clement, P.M.; Even, C.; Jang, R.W.; et al. Durvalumab for recurrent or metastatic head and neck squamous cell carcinoma: Results from a single-arm, phase II study in patients with >/=25% tumour cell PD-L1 expression who have progressed on platinum-based chemotherapy. Eur. J. Cancer 2019, 107, 142–152. [Google Scholar] [CrossRef] [Green Version]
  8. Schoenfeld, J.D.; Hanna, G.J.; Jo, V.Y.; Rawal, B.; Chen, Y.H.; Catalano, P.S.; Lako, A.; Ciantra, Z.; Weirather, J.L.; Criscitiello, S.; et al. Neoadjuvant Nivolumab or Nivolumab Plus Ipilimumab in Untreated Oral Cavity Squamous Cell Carcinoma: A Phase 2 Open-Label Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1563–1570. [Google Scholar] [CrossRef]
  9. Rao, S.K.; Pavicevic, Z.; Du, Z.; Kim, J.G.; Fan, M.; Jiao, Y.; Rosebush, M.; Samant, S.; Gu, W.; Pfeffer, L.M.; et al. Pro-inflammatory genes as biomarkers and therapeutic targets in oral squamous cell carcinoma. J. Biol. Chem. 2010, 285, 32512–32521. [Google Scholar] [CrossRef] [Green Version]
  10. Wu, T.; Hong, Y.; Jia, L.; Wu, J.; Xia, J.; Wang, J.; Hu, Q.; Cheng, B. Modulation of IL-1β reprogrammes the tumor microenvironment to interrupt oral carcinogenesis. Sci. Rep. 2016, 6, 20208. [Google Scholar] [CrossRef] [Green Version]
  11. León, X.; Bothe, C.; García, J.; Parreño, M.; Alcolea, S.; Quer, M.; Vila, L.; Camacho, M. Expression of IL-1α correlates with distant metastasis in patients with head and neck squamous cell carcinoma. Oncotarget 2015, 6, 37398–37409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Niklander, S.E. Inflammatory Mediators in Oral Cancer: Pathogenic Mechanisms and Diagnostic Potential. Front. Oral Health 2021, 2, 642238. [Google Scholar] [CrossRef] [PubMed]
  13. Goertzen, C.; Mahdi, H.; Laliberte, C.; Meirson, T.; Eymael, D.; Gil-Henn, H.; Magalhaes, M. Oral inflammation promotes oral squamous cell carcinoma invasion. Oncotarget. 2018, 9, 29047–29063. [Google Scholar] [CrossRef] [Green Version]
  14. Speight, P.M.; Khurram, S.A.; Kujan, O. Oral potentially malignant disorders: Risk of progression to malignancy. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2018, 125, 612–627. [Google Scholar] [CrossRef] [Green Version]
  15. Lee, C.H.; Chang, J.S.; Syu, S.H.; Wong, T.S.; Chan, J.Y.; Tang, Y.C.; Yang, Z.P.; Yang, W.C.; Chen, C.T.; Lu, S.C.; et al. IL-1β promotes malignant transformation and tumor aggressiveness in oral cancer. J. Cell Physiol. 2015, 230, 875–884. [Google Scholar] [CrossRef] [PubMed]
  16. Zhao, J.; Chen, X.; Herjan, T.; Li, X. The role of interleukin-17 in tumor development and progression. J. Exp. Med. 2020, 217, e20190297. [Google Scholar] [CrossRef]
  17. Cohen, N.; Fedewa, S.; Chen, A.Y. Epidemiology and Demographics of the Head and Neck Cancer Population. Oral Maxillofac. Surg. Clin. N. Am. 2018, 30, 381–395. [Google Scholar] [CrossRef]
  18. Sheikh, M.N.; Hanif, S.; Zia, M.; Qayyum, Z. Effects of Nicotine on an In Vitro Reconstituted Model Oral Mucosa in Terms of Cytokine Production. J. Ayub. Med. Coll. Abbottabad 2011, 23, 80–84. [Google Scholar]
  19. Hashibe, M.; Brennan, P.; Chuang, S.C.; Boccia, S.; Castellsague, X.; Chen, C.; Curado, M.P.; Dal Maso, L.; Daudt, A.W.; Fabianova, E.; et al. Interaction between Tobacco and Alcohol Use and the Risk of Head and Neck Cancer: Pooled Analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol. Biomarkers Prev. 2009, 18, 541–550. [Google Scholar] [CrossRef] [Green Version]
  20. Cao, C.; Tian, B.; Geng, X.; Zhou, H.; Xu, Z.; Lai, T.; Wu, Y.; Bao, Z.; Chen, Z.; Li, W.; et al. IL-17-Mediated Inflammation Promotes Cigarette Smoke-Induced Genomic Instability. Cells 2021, 10, 1173. [Google Scholar] [CrossRef]
  21. Ma, H.Y.; Yamamoto, G.; Xu, J.; Liu, X.; Karin, D.; Kim, J.Y.; Alexandrov, L.B.; Koyama, Y.; Nishio, T.; Benner, C.; et al. IL-17 signaling in steatotic hepatocytes and macrophages promotes hepatocellular carcinoma in alcohol-related liver disease. J. Hepatol. 2020, 72, 946–959. [Google Scholar] [CrossRef] [PubMed]
  22. Kreimer, A.R.; Clifford, G.M.; Boyle, P.; Franceschi, S. Human Papillomavirus Types in Head and Neck Squamous Cell Carcinomas Worldwide: A Systematic Review. Cancer Epidemiol. Biomarkers Prev. 2005, 14, 467–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gillison, M.L.; Koch, W.M.; Capone, R.B.; Spafford, M.; Westra, W.H.; Wu, L.; Zahurak, M.L.; Daniel, R.W.; Viglione, M.; Symer, D.E.; et al. Evidence for a Causal Association between Human Papillomavirus and a Subset of Head and Neck Cancers. J. Natl. Cancer Inst. 2000, 92, 709–720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Jelihovschi, I.; Bidescu, A.C.; Tucaliuc, S.E.; Iancu, L.S. Detection of Human Papilloma Virus in Head and Neck Squamous Cell Carcinomas: A Literature Review. Rev. Med. Chir. Soc. Med. Nat. Iasi 2015, 119, 502–509. [Google Scholar]
  25. Punt, S.; Dronkers, E.A.; Welters, M.J.; Goedemans, R.; Koljenović, S.; Bloemena, E.; Snijders, P.J.; Gorter, A.; van der Burg, S.H.; Baatenburg de Jong, R.J.; et al. A beneficial tumor microenvironment in oropharyngeal squamous cell carcinoma is characterized by a high T cell and low IL-17(+) cell frequency. Cancer Immunol. Immunother. 2016, 65, 393–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Baker, J.L.; Bor, B.; Agnello, M.; Shi, W.; He, X. Ecology of the Oral Microbiome: Beyond Bacteria. Trends Microbiol. 2017, 25, 362–374. [Google Scholar] [CrossRef] [Green Version]
  27. Zhang, W.L.; Wang, S.S.; Wang, H.F.; Tang, Y.J.; Tang, Y.L.; Liang, X.H. Who is Who in Oral Cancer? Exp. Cell Res. 2019, 384, 111634. [Google Scholar] [CrossRef] [PubMed]
  28. Lafuente Ibáñez de Mendoza, I.; Maritxalar Mendia, X.; García de la Fuente, A.M.; Quindós Andrés, G.; Aguirre Urizar, J.M. Role of Porphyromonas Gingivalis in Oral Squamous Cell Carcinoma Development: A Systematic Review. J. Periodontal Res. 2020, 55, 13–22. [Google Scholar] [CrossRef]
  29. Guerrero-Preston, R.; Godoy-Vitorino, F.; Jedlicka, A.; Rodríguez-Hilario, A.; González, H.; Bondy, J.; Folawiyo, O.; Michailidi, C.; Dziedzic, A.; Thangavel, R.; et al. 16s rRNA Amplicon Sequencing Identifies Microbiota Associated with Oral Cancer, Human Papilloma Virus Infection and Surgical Treatment. Oncotarget 2016, 7, 51320–51334. [Google Scholar] [CrossRef] [Green Version]
  30. Groeger, S.; Jarzina, F.; Domann, E.; Meyle, J. Porphyromonas Gingivalis Activates Nfκb and MAPK Pathways in Human Oral Epithelial Cells. BMC Immunol. 2017, 18, 1. [Google Scholar] [CrossRef] [Green Version]
  31. Yang, S.F.; Huang, H.D.; Fan, W.L.; Jong, Y.J.; Chen, M.K.; Huang, C.N.; Chuang, C.Y.; Kuo, Y.L.; Chung, W.H.; Su, S.C. Compositional and functional variations of oral microbiota associated with the mutational changes in oral cancer. Oral Oncol. 2018, 77, 1–8. [Google Scholar] [CrossRef] [PubMed]
  32. Hoppe, T.; Kraus, D.; Novak, N.; Probstmeier, R.; Frentzen, M.; Wenghoefer, M.; Jepsen, S.; Winter, J. Oral Pathogens Change Proliferation Properties of Oral Tumor Cells by Affecting Gene Expression of Human Defensins. Tumour Biol. 2016, 37, 13789–13798. [Google Scholar] [CrossRef] [PubMed]
  33. Cheng, W.C.; van Asten, S.D.; Burns, L.A.; Evans, H.G.; Walter, G.J.; Hashim, A.; Hughes, F.J.; Taams, L.S. Periodontitis-associated pathogens P. gingivalis and A. actinomycetemcomitans activate human CD14+ monocytes leading to enhanced Th17/IL-17 responses. Eur. J. Immunol. 2016, 46, 2211–2221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Husein-ElAhmed, H.; Steinhoff, M. Potential role of INTERLEUKIN-17 in the pathogenesis of oral lichen planus: A systematic review with META-analysis. J. Eur. Acad. Dermatol. Venereol. 2022, 36, 1735–1744. [Google Scholar] [CrossRef]
  35. Brunotto, M.; Zarate, A.M.; Bono, A.; Barra, J.L.; Berra, S. Risk Genes in Head and Neck Cancer: A Systematic Review and Meta-Analysis of Last 5 Years. Oral Oncol. 2014, 50, 178–188. [Google Scholar] [CrossRef]
  36. Li, N.; Zhang, C.; Chen, Z.; Bai, L.; Nie, M.; Zhou, B.; Xu, H. Interleukin 17A and interleukin 17F polymorphisms are associated with oral squamous cell carcinoma susceptibility in a Chinese population. J. Oral Maxillofac. Surg. 2015, 73, 267–273. [Google Scholar] [CrossRef]
  37. Mirza, A.H.; Thomas, G.; Ottensmeier, C.H.; King, E.V. Importance of the Immune System in Head and Neck Cancer. Head Neck 2019, 41, 2789–27800. [Google Scholar] [CrossRef]
  38. Milovanovic, J.; Arsenijevic, A.; Stojanovic, B.; Kanjevac, T.; Arsenijevic, D.; Radosavljevic, G.; Milovanovic, M.; Arsenijevic, N. Interleukin-17 in Chronic Inflammatory Neurological Diseases. Front. Immunol. 2020, 11, 947. [Google Scholar] [CrossRef]
  39. Razi Razi, S.; Baradaran Noveiry, B.; Keshavarz-Fathi, M.; Rezaei, N. IL-17 and colorectal cancer: From carcinogenesis to treatment. Cytokine 2019, 116, 7–12. [Google Scholar] [CrossRef]
  40. Bastid, J.; Dejou, C.; Docquier, A.; Bonnefoy, N. The Emerging Role of the IL-17B/IL-17RB Pathway in Cancer. Front. Immunol. 2020, 21, 718. [Google Scholar] [CrossRef] [Green Version]
  41. Hu, F.; Guo, F.; Zhu, Y.; Zhou, Q.; Li, T.; Xiang, H.; Shang, D. IL-17 in pancreatic disease: Pathogenesis and pharmacotherapy. Am. J. Cancer Res. 2020, 10, 3551–3564. [Google Scholar] [PubMed]
  42. Liu, H.; Chew, V. IFNγ-IL-17+ CD8 T cells contribute to immunosuppression and tumor progression in human hepatocellular carcinoma. Cancer Lett. 2023, 552, 215977. [Google Scholar]
  43. Li, J.; Zeng, M.; Yan, K.; Yang, Y.; Li, H.; Xu, X. IL-17 promotes hepatocellular carcinoma through inhibiting apoptosis induced by IFN-γ. Biochem. Biophys. Res. Commun. 2020, 522, 525–531. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, L.; Yi, T.; Zhang, W.; Pardoll, D.M.; Yu, H. IL-17 enhances tumor development in carcinogen-induced skin cancer. Cancer Res. 2010, 70, 10112–10120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Jin, C.; Lagoudas, G.K.; Zhao, C.; Bullman, S.; Bhutkar, A.; Hu, B.; Ameh, S.; Sandel, D.; Liang, X.S.; Mazzilli, S.; et al. Commensal Microbiota Promote Lung Cancer Development via gammadelta T Cells. Cell 2019, 176, 998–1013. [Google Scholar] [CrossRef] [Green Version]
  46. Calcinotto, A.; Brevi, A.; Chesi, M.; Ferrarese, R.; Garcia Perez, L.; Grioni, M.; Kumar, S.; Garbitt, V.M.; Sharik, M.E.; Henderson, K.J.; et al. Microbiota-driven interleukin-17-producing cells and eosinophils synergize to accelerate multiple myeloma progression. Nat. Commun. 2018, 9, 4832. [Google Scholar] [CrossRef] [Green Version]
  47. Mills, K.H.G. IL-17 and IL-17-producing cells in protection versus pathology. Nat. Rev. Immunol. 2023, 23, 38–54. [Google Scholar] [CrossRef]
  48. Papotto, P.H.; Ribot, J.C.; Silva-Santos, B. IL-17+ γδ T cells as kickstarters of inflammation. Nat. Immunol. 2017, 18, 604–611. [Google Scholar] [CrossRef]
  49. Ciric, B.; El-behi, M.; Cabrera, R.; Zhang, G.X.; Rostami, A. IL-23 drives pathogenic IL-17-producing CD8+ T cells. J. Immunol. 2009, 182, 5296–5305. [Google Scholar] [CrossRef] [Green Version]
  50. Cupedo, T.; Crellin, N.K.; Papazian, N.; Rombouts, E.J.; Weijer, K.; Grogan, J.L.; Fibbe, W.E.; Cornelissen, J.J.; Spits, H. Human fetal lymphoid tissue-inducer cells are interleukin 17-producing precursors to RORC+ CD127+ natural killer-like cel. Nat. Immunol. 2009, 10, 66–74. [Google Scholar] [CrossRef]
  51. Michel, M.L.; Keller, A.C.; Paget, C.; Fujio, M.; Trottein, F.; Savage, P.M.; Wong, C.H.; Schneider, E.; Dy, M.; Leite-de-Moraes, M.C. Identification of an IL-17-producing NK1.1(neg) iNKT cell population involved in airway neutrophilia. J. Exp. Med. 2007, 204, 995–1001. [Google Scholar] [CrossRef] [PubMed]
  52. Buonocore, S.; Ahern, P.P.; Uhlig, H.H.; Ivanov, I.I.; Littman, D.R.; Maloy, K.J.; Powrie, F. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 2010, 464, 1371–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Lin, A.M.; Rubin, C.J.; Khandpur, R.; Wang, J.Y.; Riblett, M.; Yalavarthi, S.; Villanueva, E.C.; Shah, P.; Kaplan, M.J.; Bruce, A.T. Mast cells and neutrophils release IL-17 through extracellular trap formation in psoriasis. J. Immunol. 2011, 187, 490–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Yadav, B.; Specht, C.A.; Lee, C.K.; Pokrovskii, M.; Huh, J.R.; Littman, D.R.; Levitz, S.M. Lung eosinophils elicited during allergic and acute aspergillosis express RORγt and IL-23R but do not require IL-23 for IL-17 production. PLoS Pathog. 2021, 17, e1009891. [Google Scholar] [CrossRef]
  55. Kostareva, O.S.; Gabdulkhakov, A.G.; Kolyadenko, I.A.; Garber, M.B.; Tishchenko, S.V. Interleukin-17: Functional and Structural Features, Application as a Therapeutic Target. Biochemistry 2019, 84 (Suppl. 1), S193–S205. [Google Scholar] [CrossRef]
  56. Chung, Y.; Chang, S.H.; Martinez, G.J.; Yang, X.O.; Nurieva, R.; Kang, H.S.; Ma, L.; Watowich, S.S.; Jetten, A.M.; Tian, Q.; et al. Critical regulation of early Th17 cell differentiation by interleukin-1 signaling. Immunity 2009, 30, 576–587. [Google Scholar] [CrossRef] [Green Version]
  57. Revu, S.; Wu, J.; Henkel, M.; Rittenhouse, N.; Menk, A.; Delgoffe, G.M.; Poholek, A.C.; McGeachy, M.J. IL-23 and IL-1β Drive Human Th17 Cell Differentiation and Metabolic Reprogramming in Absence of CD28 Costimulation. Cell. Rep. 2018, 22, 2642–2653. [Google Scholar] [CrossRef] [Green Version]
  58. Langrish, C.L.; Chen, Y.; Blumenschein, W.M.; Mattson, J.; Basham, B.; Sedgwick, J.D.; McClanahan, T.; Kastelein, R.A.; Cua, D.J. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J. Exp. Med. 2005, 201, 233–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Sutton, C.E.; Lalor, S.J.; Sweeney, C.M.; Brereton, C.F.; Lavelle, E.C.; Mills, K.H. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity 2009, 31, 331–341. [Google Scholar] [CrossRef] [Green Version]
  60. Lee, S.Y.; Lee, A.R.; Choi, J.W.; Lee, C.R.; Cho, K.H.; Lee, J.H.; Cho, M.L. IL-17 Induces Autophagy Dysfunction to Promote Inflammatory Cell Death and Fibrosis in Keloid Fibroblasts via the STAT3 and HIF-1α Dependent Signaling Pathways. Front. Immunol. 2022, 10, 888719. [Google Scholar] [CrossRef]
  61. Ivanov, I.I.; McKenzie, B.S.; Zhou, L.; Tadokoro, C.E.; Lepelley, A.; Lafaille, J.J.; Cua, D.J.; Littman, D.R. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006, 126, 1121–1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Zuberbuehler, M.K.; Parker, M.E.; Wheaton, J.D.; Espinosa, J.R.; Salzler, H.R.; Park, E.; Ciofan, M. The transcription factor c-Maf is essential for the commitment of IL-17-producing γδ T cells. Nat. Immunol. 2019, 20, 73–85. [Google Scholar] [CrossRef] [PubMed]
  63. McGeachy, M.J.; Cua, D.J.; Gaffen, S.L. The IL-17 Family of Cytokines in Health and Disease. Immunity 2019, 50, 892–906. [Google Scholar] [CrossRef] [PubMed]
  64. Moseley, T.A.; Haudenschild, D.R.; Rose, L.; Reddi, A.H. Interleukin-17 family and IL-17 receptors. Cytokine Growth Factor Rev. 2003, 14, 155–174. [Google Scholar] [CrossRef]
  65. Amatya, N.; Garg, A.V.; Gaffen, S.L. IL-17 Signaling: The Yin and the Yang. Trends Immunol. 2017, 38, 310–322. [Google Scholar] [CrossRef] [Green Version]
  66. Novatchkova, M.; Leibbrandt, A.; Werzowa, J.; Neubüser, A.; Eisenhaber, F. The STIR-domain superfamily in signal transduction, development and immunity. Trends Biochem. Sci. 2003, 28, 226–229. [Google Scholar] [CrossRef]
  67. Gu, C.; Wu, L.; Li, X. IL-17 family: Cytokines, receptors and signaling. Cytokine 2013, 64, 477–485. [Google Scholar] [CrossRef] [Green Version]
  68. Chang, S.H.; Park, H.; Dong, C. Act1 adaptor protein is an immediate and essential signaling component of interleukin-17 receptor. J. Biol. Chem. 2006, 281, 35603–35607. [Google Scholar] [CrossRef] [Green Version]
  69. Maitra, A.; Shen, F.; Hanel, W.; Mossman, K.; Tocker, J.; Swart, D.; Gaffen, S.L. Distinct functional motifs within the IL-17 receptor regulate signal transduction and target gene expression. Proc. Natl. Acad. Sci. USA 2007, 104, 7506–7511. [Google Scholar] [CrossRef] [Green Version]
  70. Onishi, R.; Park, S.J.; Hanel, W.; Ho, A.W.; Maitra, A.; Gaffen, S.L. The SEFIR is not enough: An extended region downstream of the Interleukin-17RA SEFIR domain is required for IL-17-dependent signal transduction. J. Biol. Chem. 2010, 285, 32751–32759. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, B.; Liu, C.; Qian, W.; Han, Y.; Li, X.; Deng, J. Structure of the unique SEFIR domain from human interleukin 17 receptor A reveals a composite ligand-binding site containing a conserved alpha-helix for Act1 binding and IL-17 signaling. Acta Crystallogr. D. Biol. Crystallogr. 2014, 70, 1476–1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Qian, Y.; Liu, C.; Hartupee, J.; Altuntas, C.Z.; Gulen, M.F.; Jane-Wit, D.; Xiao, J.; Lu, Y.; Giltiay, N.; Liu, J. The adaptor Act1 is required for interleukin 17-dependent signaling associated with autoimmune and inflammatory disease. Nat. Immunol. 2007, 8, 247–256. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, C.; Qian, W.; Qian, Y.; Giltiay, N.V.; Lu, Y.; Swaidani, S.; Misra, S.; Deng, L.; Chen, Z.J.; Li, X. Act1, a U-box E3 ubiquitin ligase for IL-17 signaling. Sci. Signal. 2009, 2, ra63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Schwandner, R.; Schwandner, R.; Yamaguchi, K.; Cao, Z. Requirement of tumor necrosis factor-associated factor (TRAF)6 in interleukin 17 signal transduction. J. Exp. Med. 2000, 191, 1233–1239. [Google Scholar] [CrossRef] [Green Version]
  75. Napetschnig, J.; Wu, H. Molecular basis of NF-κB signaling. Annu. Rev. Biophys. 2013, 42, 443–468. [Google Scholar] [CrossRef] [Green Version]
  76. Wu, N.L.; Huang, D.Y.; Tsou, H.N.; Lin, Y.C.; Lin, W.W. Syk mediates IL-17-induced CCL20 expression by targeting Act1-dependent K63-linked ubiquitination of TRAF6. J. Investig. Dermatol. 2015, 135, 490–498. [Google Scholar] [CrossRef] [Green Version]
  77. Ruddy, M.J.; Wong, G.C.; Liu, X.K.; Yamamoto, H.; Kasayama, S.; Kirkwood, K.L.; Gaffen, S.L. Functional cooperation between interleukin-17 and tumor necrosis factor-α is mediated by CCAAT/enhancer binding protein family members. J. Biol. Chem. 2004, 279, 2559–2567. [Google Scholar] [CrossRef] [Green Version]
  78. Shen, F.; Gaffen, S.L. Structure-function relationships in the IL-17 receptor: Implications for signal transduction and therapy. Cytokine 2008, 41, 92–104. [Google Scholar] [CrossRef] [Green Version]
  79. Xiao, Y.; Jin, J.; Chang, M.; Nakaya, M.; Hu, H.; Zou, Q.; Zhou, X.; Brittain, G.C.; Cheng, X.; Sun, S.C. TPL2 mediates autoimmune inflammation through activation of the TAK1 axis of IL-17 signaling. J. Exp. Med. 2014, 211, 1689–1702. [Google Scholar] [CrossRef] [Green Version]
  80. Wu, L.; Chen, X.; Zhao, J.; Martin, B.; Zepp, J.A.; Ko, J.S.; Gu, C.; Cai, G.; Ouyang, W.; Sen, G. A novel IL-17 signaling pathway controlling keratinocyte proliferation and tumorigenesis via the TRAF4-ERK5 axis. J. Exp. Med. 2015, 212, 1571–1587. [Google Scholar] [CrossRef]
  81. Akira, S.; Maeda, K. Control of RNA stability in immunity. Annu. Rev. Immunol. 2021, 39, 481–509. [Google Scholar] [CrossRef] [PubMed]
  82. Herjan, T.; Hong, L.; Bubenik, J.; Bulek, K.; Qian, W.; Liu, C.; Li, X.; Chen, X.; Yang, H.; Ouyang, S.; et al. IL-17-receptor-associated adaptor Act1 directly stabilizes mRNAs to mediate IL-17 inflammatory signaling. Nat. Immunol. 2018, 19, 354–365. [Google Scholar] [CrossRef] [PubMed]
  83. Tanaka, H.; Arima, Y.; Kamimura, D.; Tanaka, Y.; Takahashi, N.; Uehata, T.; Maeda, K.; Satoh, T.; Murakami, M.; Akira, S. Phosphorylation-dependent Regnase-1 release from endoplasmic reticulum is critical in IL-17 response. J. Exp. Med. 2019, 216, 1431–1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Herjan, T.; Yao, P.; Qian, W.; Li, X.; Liu, C.; Bulek, K.; Sun, D.; Yang, W.P.; Zhu, J.; He, A.; et al. HuR is required for IL-17-induced Act1-mediated CXCL1 and CXCL5 mRNA stabilization. J. Immunol. 2013, 191, 640–649. [Google Scholar] [CrossRef] [Green Version]
  85. Amatya, N.; Childs, E.E.; Cruz, J.A.; Aggor, F.E.Y.; Garg, A.V.; Berman, A.J.; Gudjonsson, J.E.; Atasoy, U.; Gaffen, S.L. IL-17 integrates multiple self-reinforcing, feed-forward mechanisms through the RNA binding protein Arid5a. Sci. Signal. 2018, 11, eaat4617. [Google Scholar] [CrossRef] [Green Version]
  86. Gaffen, S.L.; Jain, R.; Garg, A.V.; Cua, D.J. The IL-23-IL-17 immune axis: From mechanisms to therapeutic testing. Nat. Rev. Immunol. 2014, 14, 585–600. [Google Scholar] [CrossRef] [Green Version]
  87. Onishi, R.M.; Gaffen, S.L. Interleukin-17 and its target genes: Mechanisms of interleukin-17 function in disease. Immunology 2010, 129, 311–321. [Google Scholar] [CrossRef]
  88. Chiricozzi, A.; Guttman-Yassky, E.; Suárez-Fariñas, M.; Nograles, K.E.; Tian, S.; Cardinale, I.; Chimenti, S.; Krueger, J.G. Integrative responses to IL-17 and TNF-α in human keratinocytes account for key inflammatory pathogenic circuits in psoriasis. J. Investig. Dermatol. 2011, 131, 677–687. [Google Scholar] [CrossRef]
  89. Fabre, T.; Kared, H.; Friedman, S.L.; Shoukry, N.H. IL-17A enhances the expression of profibrotic genes through upregulation of the TGF-β receptor on hepatic stellate cells in a JNK-dependent manner. J. Immunol. 2014, 193, 3925–3933. [Google Scholar] [CrossRef] [Green Version]
  90. Kaiko, G.E.; Chen, F.; Lai, C.W.; Chiang, I.L.; Perrigoue, J.; Stojmirovic, A.; Li, K.; Muegge, B.D.; Jain, U.; VanDussen, K.L.; et al. PAI-1 augments mucosal damage in colitis. Sci. Transl. Med. 2019, 11, eaat0852. [Google Scholar] [CrossRef]
  91. Verma, A.H.; Richardson, J.P.; Zhou, C.; Coleman, B.M.; Moyes, D.L.; Ho, J.; Huppler, A.R.; Ramani, K.; McGeachy, M.J.; Mufazalov, I.A.; et al. Oral epithelial cells orchestrate innate type 17 responses to Candida albicans through the virulence factor candid. Sci. Immunol. 2017, 2, eaam8834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Wang, S.; Yu, X.; Li, F.; Fan, H.; Zhao, E.; Hu, Z. Targeting IL-17alpha to promote anti-PD-1 therapy effect by screening the tumor immune microenvironment in a mouse oral carcinogenesis model. Cancer Biomark. 2021, 31, 339–350. [Google Scholar] [CrossRef] [PubMed]
  93. Gaur, P.; Singh, A.K.; Shukla, N.K.; Das, S.N. Inter-relation of Th1, Th2, Th17 and Treg cytokines in oral cancer patients and their clinical significance. Hum. Immunol. 2014, 75, 330–337. [Google Scholar] [CrossRef] [PubMed]
  94. Xiaonan, H. Expression levels of BDNF, VEGF, IL-17 and IL-17F in oral and maxillofacial squamous cell carcinoma and their clinicopathological features. Acta Med. Mediterr. 2019, 35, 1225–1231. [Google Scholar]
  95. Ding, L.; Hu, E.L.; Xu, Y.J.; Huang, X.F.; Zhang, D.Y.; Li, B.; Hu, Q.G.; Ni, Y.H.; Hou, Y.Y. Serum IL-17F combined with VEGF as potential diagnostic biomarkers for oral squamous cell carcinoma. Tumour Biol. 2015, 36, 2523–2529. [Google Scholar] [CrossRef]
  96. Zielińska, K.; Karczmarek-Borowska, B.; Kwaśniak, K.; Czarnik-Kwaśniak, J.; Ludwin, A.; Lewandowski, B.; Tabarkiewicz, J. Salivary IL-17A, IL-17F, and TNF-α are associated with disease advancement in patients with oral and oropharyngeal cancer. J. Immunol. Res. 2020, 2020, 3928504. [Google Scholar] [CrossRef]
  97. Zhao, Y.; Huang, J.; Chen, J. The integration of differentially expressed genes based on multiple microarray datasets for prediction of the prognosis in oral squamous cell carcinoma. Bioengineered 2021, 12, 3309–3321. [Google Scholar] [CrossRef]
  98. Gaur, P.; Shukla, N.K.; Das, S.N. Phenotypic and Functional Characteristics of Th17 (CD4+IL17A+) Cells in Human Oral Squamous Cell Carcinoma and Its Clinical Relevance. Immunol. Investig. 2017, 46, 689–702. [Google Scholar] [CrossRef]
  99. Gaur, P.; Qadir, G.A.; Upadhyay, S.; Singh, A.K.; Shukla, N.K.; Das, S.N. Skewed immunological balance between Th17 (CD4+IL17A+) and Treg (CD4+CD25+FOXP3+) cells in human oral squamous cell carcinoma. Cell Oncol. 2012, 35, 335–343. [Google Scholar] [CrossRef]
  100. Lee, M.H.; Tung-Chieh Chang, J.; Liao, C.T.; Chen, Y.S.; Kuo, M.L.; Shen, C.R. Interleukin 17 and peripheral IL-17-expressing T cells are negatively correlated with the overall survival of head and neck cancer patients. Oncotarget 2018, 9, 9825–9837. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, S.; Wang, X.; Gupta, A.; Fang, X.; Wang, L.; Zhang, C. Expression of IL-17 with tumor budding as a prognostic marker in oral squamous cell carcinoma. Am. J. Transl. Res. 2019, 11, 1876–1883. [Google Scholar] [PubMed]
  102. Dawson, H.; Koelzer, V.H.; Karamitopoulou, E.; Economou, M.; Hammer, C.; Muller, D.E.; Lugli, A.; Zlobec, I. The apoptotic and proliferation rate of tumour budding cells in colorectal cancer outlines a heterogeneous population of cells with various impacts on clinical outcome. Histopathology 2014, 64, 577–584. [Google Scholar] [CrossRef]
  103. Almangush, A.; Pirinen, M.; Heikkinen, I.; Makitie, A.A.; Salo, T.; Leivo, I. Tumour budding in oral squamous cell carcinoma: A meta-analysis. Br. J. Cancer. 2018, 118, 577–586. [Google Scholar] [CrossRef] [Green Version]
  104. Avadhani, A.V.; Parachuru, V.P.; Milne, T.; Seymour, G.J.; Rich, A.M. Multiple cells express interleukin 17 in oral squamous cell carcinoma. J. Oral. Pathol. Med. 2017, 46, 39–45. [Google Scholar] [CrossRef] [PubMed]
  105. Quan, H.; Shan, Z.; Liu, Z.; Liu, S.; Yang, L.; Fang, X.; Li, K.; Wang, B.; Deng, Z.; Hu, Y.; et al. The repertoire of tumor-infiltrating lymphocytes within the microenvironment of oral squamous cell carcinoma reveals immune dysfunction. Cancer Immunol. Immunother. 2020, 69, 465–476. [Google Scholar] [CrossRef] [PubMed]
  106. Lee, J.J.; Chang, Y.L.; Lai, W.L.; Ko, J.Y.; Kuo, M.Y.; Chiang, C.P.; Azuma, M.; Chen, C.W.; Chia, J.S. Increased prevalence of interleukin-17-producing CD4(+) tumor infiltrating lymphocytes in human oral squamous cell carcinoma. Head Neck 2011, 33, 1301–1308. [Google Scholar] [CrossRef]
  107. Guery, L.; Hugues, S. Th17 cell plasticity and functions in cancer immunity. BioMed Res. Int. 2015, 2015, 314620. [Google Scholar] [CrossRef] [Green Version]
  108. Chao, J.L.; Korzinkin, M.; Zhavoronkov, A.; Ozerov, I.V.; Walker, M.T.; Higgins, K.; Lingen, M.W.; Izumchenko, E.; Savage, P.A. Effector T cell responses unleashed by regulatory T cell ablation exacerbate oral squamous cell carcinoma. Cell. Rep. Med. 2021, 2, 100399. [Google Scholar] [CrossRef]
  109. Miossec, P.; Kolls, J.K. Targeting IL-17 and TH17 cells in chronic inflammation. Nat. Rev. Drug. Discov. 2012, 11, 763–776. [Google Scholar] [CrossRef]
  110. Murugaiyan, G.; Saha, B. Protumor vs antitumor functions of IL-17. J. Immunol. 2009, 183, 4169–4175. [Google Scholar] [CrossRef] [Green Version]
  111. Wei, W.; Li, J.; Shen, X.; Lyu, J.; Yan, C.; Tang, B.; Ma, W.; Xie, H.; Zhao, L.; Cheng, L.; et al. Oral Microbiota from Periodontitis Promote Oral Squamous Cell Carcinoma Development via γδ T Cell Activation. mSystems 2022, 7, e0046922. [Google Scholar] [CrossRef] [PubMed]
  112. Roy, N.K.; Monisha, J.; Padmavathi, G.; Lalhruaitluanga, H.; Kumar, N.S.; Singh, A.K.; Bordoloi, D.; Baruah, M.N.; Ahmed, G.N.; Longkumar, I.; et al. Isoform-Specific Role of Akt in Oral Squamous Cell Carcinoma. Biomolecules 2019, 9, 253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Caughron, B.; Yang, Y.; Young, M.R.I. Role of IL-23 signaling in the progression of premalignant oral lesions to cancer. PLoS ONE 2018, 13, e0196034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Young, M.R.; Levingston, C.A.; Johnson, S.D. Treatment to sustain a Th17-type phenotype to prevent skewing toward Treg and to limit premalignant lesion progression to cancer. Int. J. Cancer 2016, 138, 2487–2498. [Google Scholar] [CrossRef] [Green Version]
  115. Vitiello, G.A.; Miller, G. Targeting the interleukin-17 immune axis for cancer immunotherapy. J. Exp. Med. 2020, 217, e20190456. [Google Scholar] [CrossRef] [PubMed]
  116. Dan, H.; Liu, S.; Liu, J.; Liu, D.; Yin, F.; Wei, Z.; Wang, J.; Zhou, Y.; Jiang, L.; Ji, N.; et al. RACK1 promotes cancer progression by increasing the M2/M1 macrophage ratio via the NF-κB pathway in oral squamous cell carcinoma. Mol. Oncol. 2020, 14, 795–807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Feng, X.; Deng, P.; Zeng, X.; Zhou, M.; Zhou, Y.; Dan, H.; Jiang, L.; Chen, Q. ORAOV1-B Promotes OSCC Metastasis via the NF-κB-TNFα Loop. J. Dent. Res. 2021, 100, 858–867. [Google Scholar]
  118. Xu, Q.; Chen, X.; Yu, T.; Tang, Q.; Zhou, Z.; Wang, H.; Huang, W.; Huang, T.; Liang, F. Downregulation of VAP-1 in OSCC suppresses tumor growth and metastasis via NF-κB/IL-8 signaling and reduces neutrophil infiltration. J. Oral Pathol. Med. 2022, 51, 332–341. [Google Scholar] [CrossRef]
  119. Shan, F.; Shen, S.; Wang, X.; Chen, G. BST2 regulated by the transcription factor STAT1 can promote metastasis, invasion and proliferation of oral squamous cell carcinoma via the AKT/ERK1/2 signaling pathway. Int. J. Oncol. 2023, 62, 54. [Google Scholar] [CrossRef]
  120. Cui, B.; Chen, J.; Luo, M.; Liu, Y.; Chen, H.; Lü, D.; Wang, L.; Kang, Y.; Feng, Y.; Huang, L.; et al. PKD3 promotes metastasis and growth of oral squamous cell carcinoma through positive feedback regulation with PD-L1 and activation of ERK-STAT1/3-EMT signalling. Int. J. Oral Sci. 2021, 13, 8. [Google Scholar] [CrossRef]
  121. Yang, W.; Zhang, S.; Li, T.; Zhou, Z.; Pan, J. Single-cell analysis reveals that cancer-associated fibroblasts stimulate oral squamous cell carcinoma invasion via the TGF-β/Smad pathway. Acta Biochim. Biophys. Sin. 2022, 55, 262–273. [Google Scholar] [CrossRef] [PubMed]
  122. Lv, S.; Luo, T.; Yangm, Y.; Li, Y.; Yang, J.; Xu, J.; Zheng, J.; Zeng, Y. Naa10p and IKKα interaction regulates EMT in oral squamous cell carcinoma via TGF-β1/Smad pathway. J. Cell Mol. Med. 2021, 25, 6760–6772. [Google Scholar] [CrossRef] [PubMed]
  123. Yap, T.; Pruthi, N.; Seers, C.; Belobrov, S.; McCullough, M.; Celentano, A. Extracellular Vesicles in Oral Squamous Cell Carcinoma and Oral Potentially Malignant Disorders: A Systematic Review. Int. J. Mol. Sci. 2020, 21, 1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Simón, L.; Sanhueza, S.; Gaete-Ramírez, B.; Varas-Godoy, M.; Quest, A.F.G. Role of the Pro-Inflammatory Tumor Microenvironment in Extracellular Vesicle-Mediated Transfer of Therapy Resistance. Front. Oncol. 2022, 12, 897205. [Google Scholar] [CrossRef] [PubMed]
  125. Huang, Y.; Kanada, M.; Ye, J.; Deng, Y.; He, Q.; Lei, Z.; Chen, Y.; Li, Y.; Qin, P.; Zhang, J.; et al. Exosome-mediated remodeling of the tumor microenvironment: From local to distant intercellular communication. Cancer Lett. 2022, 543, 215796. [Google Scholar] [CrossRef] [PubMed]
  126. Li, R.; Zhou, Y.; Zhang, M.; Xie, R.; Duan, N.; Liu, H.; Qin, Y.; Ma, J.; Li, Z.; Ye, P.; et al. Oral squamous cell carcinoma-derived EVs promote tumor progression by regulating inflammatory cytokines and the IL-17A-induced signaling pathway. Int. Immunopharmacol. 2023, 118, 110094. [Google Scholar] [CrossRef]
  127. Ye, P.; Rodriguez, F.H.; Kanaly, S.; Stocking, K.L.; Schurr, J.; Schwarzenberger, P.; Oliver, P.; Huang, W.; Zhang, P.; Zhang, J.; et al. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J. Exp. Med. 2001, 194, 519–527. [Google Scholar] [CrossRef]
  128. Veldhoen, M. Interleukin 17 is a chief orchestrator of immunity. Nat. Immunol. 2017, 18, 612–621. [Google Scholar] [CrossRef]
  129. Coffelt, S.B.; Kersten, K.; Doornebal, C.W.; Weiden, J.; Vrijland, K.; Hau, C.S.; Verstegen, N.J.M.; Ciampricotti, M.; Hawinkels, L.J.A.C.; Jonkers, J.; et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastas. Nature 2015, 522, 345–348. [Google Scholar] [CrossRef] [Green Version]
  130. Veglia, F.; Perego, M.; Gabrilovich, D. Myeloid-derived suppressor cells coming of age. Nat. Immunol. 2018, 19, 108–119. [Google Scholar] [CrossRef]
  131. Weber, R.; Groth, C.; Lasser, S.; Arkhypov, I.; Petrova, V.; Altevogt, P.; Utikal, J.; Umansky, V. IL-6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. Cell Immunol. 2021, 359, 104254. [Google Scholar] [CrossRef] [PubMed]
  132. Zhong, L.M.; Liu, Z.G.; Zhou, X.; Song, S.H.; Weng, G.Y.; Wen, Y.; Liu, F.B.; Cao, D.L.; Liu, Y.F. Expansion of PMN-myeloid derived suppressor cells and their clinical relevance in patients with oral squamous cell carcinoma. Oral Oncol. 2019, 95, 157–163. [Google Scholar] [CrossRef] [PubMed]
  133. Lo, Y.W.; Lee, A.Y.; Liu, Y.C.; Ko, H.H.; Peng, H.H.; Lee, H.C.; Pan, P.Y.; Chiang, C.P.; Cheng, S.J. β-glucan therapy converts the inhibition of myeloid-derived suppressor cells in oral cancer patients. Oral Dis. 2022, 28, 1484–1495. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, W.C.; Lai, C.H.; Chuang, H.C.; Lin, P.Y.; Chen, M.F. Inflammation-induced myeloid-derived suppressor cells associated with squamous cell carcinoma of the head and neck. Head Neck 2017, 39, 347–355. [Google Scholar] [CrossRef] [PubMed]
  135. Dar, A.A.; Patil, R.S.; Pradhan, T.N.; Chaukar, D.A.; D’Cruz, A.K.; Chiplunkar, S.V. Myeloid-derived suppressor cells impede T cell functionality and promote Th17 differentiation in oral squamous cell carcinoma. Cancer Immunol. Immunother. 2020, 69, 1071–1086. [Google Scholar] [CrossRef]
  136. Qiu, C.; She, P.; Yao, C.; Zhou, H.; Su, Z.; Kong, F. The number of myeloid suppressor cells, Th17 cells of peripheral blood and the serum IL-17 level increase in patients with oral squamous cell carcinoma. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2016, 32, 1382. [Google Scholar]
  137. Yu, T.; Tang, Q.; Chen, X.; Fan, W.; Zhou, Z.; Huang, W.; Liang, F. TGF-β1 and IL-17A comediate the protumor phenotype of neutrophils to regulate the epithelial-mesenchymal transition in oral squamous cell carcinoma. J. Oral Pathol. Med. 2021, 50, 353–361. [Google Scholar] [CrossRef]
  138. Silva, R.N.F.; Dallarmi, L.B.; Araujo, A.K.C.; Alencar, R.C.G.; Mendonça, E.F.; Silva, T.A.; Batista, A.C.; Costa, N.L. Immunohistochemical analysis of neutrophils, interleukin-17, matrix metalloproteinase-9, and neoformed vessels in oral squamous cell carcinoma. J. Oral Pathol. Med. 2018, 47, 856–863. [Google Scholar] [CrossRef]
  139. Wu, J.S.; Li, L.; Wang, S.S.; Pang, X.; Wu, J.B.; Sheng, S.R.; Tang, Y.J.; Tang, Y.L.; Zheng, M.; Liang, X.H. Autophagy is positively associated with the accumulation of myeloid-derived suppressor cells in 4-nitroquinoline-1-oxide-induced oral cancer. Oncol. Rep. 2018, 40, 338. [Google Scholar] [CrossRef]
  140. Chu, M.; Su, Y.X.; Wang, L.; Zhang, T.H.; Liang, Y.J.; Liang, L.Z.; Liao, G.Q. Myeloid-derived suppressor cells contribute to oral cancer progression in 4NQO-treated mice. Oral Dis. 2012, 18, 67–73. [Google Scholar] [CrossRef]
  141. Peng, J.; Hu, Q.; Chen, X.; Wang, C.; Zhang, J.; Ren, X.; Wang, Y.; Tao, X.; Li, H.; Song, M.; et al. Diet-induced obesity accelerates oral carcinogenesis by recruitment and functional enhancement of myeloid-derived suppressor cells. Cell Death Dis. 2021, 12, 946. [Google Scholar] [CrossRef] [PubMed]
  142. Fugle, C.W.; Zhang, Y.; Hong, F.; Sun, S.; Westwater, C.; Rachidi, S.; Yu, H.; Garret-Mayer, E.; Kirkwood, K.; Liu, B.; et al. CD24 blunts oral squamous cancer development and dampens the functional expansion of myeloid-derived suppressor cells. Oncoimmunology 2016, 5, e1226719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Wen, L.; Mu, W.; Lu, H.; Wang, X.; Fang, J.; Jia, Y.; Li, Q.; Wang, D.; Wen, S.; Guo, J.; et al. Porphyromonas gingivalis Promotes Oral Squamous Cell Carcinoma Progression in an Immune Microenvironment. J. Dent. Res. 2020, 99, 666–675. [Google Scholar] [CrossRef] [PubMed]
  144. Wang, X.; Wu, S.; Wu, W.; Zhang, W.; Li, L.; Liu, Q.; Yan, Z. Candida albicans Promotes Oral Cancer via IL-17A/IL-17RA-Macrophage Axis. mBio 2023, e0044723. [Google Scholar] [CrossRef] [PubMed]
  145. Chen, Y.; Li, Q.; Li, X.; Ma, D.; Fang, J.; Luo, L.; Liu, X.; Wang, X.; Lui, V.W.Y.; Xia, J.; et al. Blockade of PD-1 effectively inhibits in vivo malignant transformation of oral mucosa. Oncoimmunology 2017, 7, e1388484. [Google Scholar] [CrossRef]
  146. Lan, Z.; Zou, K.L.; Cui, H.; Chen, H.; Zhao, Y.Y.; Yu, G.T. PFC@O2 Targets HIF-1α to Reverse the Immunosuppressive TME in OSCC. J. Clin. Med. 2023, 12, 560. [Google Scholar] [CrossRef]
  147. Chen, X.; Zhao, J.; Herjan, T.; Hong, L.; Liao, Y.; Liu, C.; Vasu, K.; Wang, H.; Thompson, A.; Fox, P.L.; et al. IL-17-induced HIF1α drives resistance to anti-PD-L1 via fibroblast-mediated immune exclusion. J. Exp. Med. 2022, 219, e20210693. [Google Scholar] [CrossRef]
  148. Majumder, S.; McGeachy, M.J. IL-17 in the Pathogenesis of Disease: Good Intentions Gone Awry. Annu. Rev. Immunol. 2021, 39, 537–556. [Google Scholar] [CrossRef]
  149. Sisto, M.; Lorusso, L.; Tamma, R.; Ingravallo, G.; Ribatti, D.; Lisi, S. Interleukin-17 and -22 synergy linking inflammation and EMT-dependent fibrosis in Sjögren’s syndrome. Clin. Exp. Immunol. 2019, 198, 261–272. [Google Scholar] [CrossRef]
  150. Zhang, X.; Dong, Y.; Zhao, M.; Ding, L.; Yang, X.; Jing, Y.; Song, Y.; Chen, S.; Hu, Q.; Ni, Y. ITGB2-mediated metabolic switch in CAFs promotes OSCC proliferation by oxidation of NADH in mitochondrial oxidative phosphorylation system. Theranostics 2020, 10, 12044. [Google Scholar] [CrossRef]
  151. Huang, T.X.; Tan, X.Y.; Huang, H.S.; Li, Y.T.; Liu, B.L.; Liu, K.S.; Chen, X.; Chen, Z.; Guan, X.Y.; Zou, C.; et al. Targeting cancer-associated fibroblast-secreted WNT2 restores dendritic cell-mediated antitumour immunity. Gut 2022, 71, 333–344. [Google Scholar] [CrossRef] [PubMed]
  152. Li, Y.Y.; Tao, Y.W.; Gao, S.; Li, P.; Zheng, J.M.; Zhang, S.E.; Liang, J.; Zhang, Y. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine 2018, 36, 209–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Sun, L.P.; Xu, K.; Cui, J.; Yuan, D.Y.; Zou, B.; Li, J.; Liu, J.L.; Li, K.Y.; Meng, Z.; Zhang, B. Cancer-associated fibroblast-derived exosomal miR-382-5p promotes the migration and invasion of oral squamous cell carcinoma. Oncol. Rep. 2019, 42, 1319–1328. [Google Scholar] [CrossRef] [PubMed]
  154. Zheng, Y.; Liu, Y. TRAP1 suppresses oral squamous cell carcinoma progression by reducing oxidative phosphorylation metabolism of Cancer-associated fibroblasts. BMC Cancer 2021, 21, 1329. [Google Scholar]
  155. Tong, Z.; Yang, X.O.; Yan, H.; Liu, W.; Niu, X.; Shi, Y.; Fang, W.; Xiong, B.; Wan, Y.; Dong, C. A Protective Role by Interleukin-17F in Colon Tumorigenesis. PLoS ONE 2012, 7, e34959. [Google Scholar] [CrossRef] [Green Version]
  156. Xie, Y.; Sheng, W.; Xiang, J.; Ye, Z.; Yang, J. Interleukin-17F suppresses hepatocarcinoma cell growth via inhibition of tumor angiogenesis. Cancer Investig. 2010, 28, 598–607. [Google Scholar] [CrossRef]
  157. Almahmoudi, R.; Salem, A.; Sieviläinen, M.; Sundquist, E.; Almangush, A.; Toppila-Salmi, S.; Paavonen, T.; Salo, T.; Al-Samadi, A. Extracellular interleukin-17F has a protective effect in oral tongue squamous cell carcinoma. Head Neck 2018, 40, 2155–2165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Almahmoudi, R.; Salem, A.; Murshid, S.; Dourado, M.R.; Apu, E.H.; Salo, T.; Al-Samadi, A. Interleukin-17F Has Anti-Tumor Effects in Oral Tongue Cancer. Cancers 2019, 11, 650. [Google Scholar] [CrossRef] [Green Version]
  159. Almahmoudi, R.; Salem, A.; Hadler-Olsen, E.; Svineng, G.; Salo, T.; Al-Samadi, A. The effect of interleukin-17F on vasculogenic mimicry in oral tongue squamous cell carcinoma. Cancer Sci. 2021, 112, 2223–2232. [Google Scholar] [CrossRef]
  160. Ren, Y.; Ma, J.; Wang, T.; Bu, R.; Kong, X.; Shi, Y.; Zhang, L. Interleukin-17 inhibits the growth of oral squamous cell carcinoma by promoting the differentiation of T helper 17 cells. Transl. Cancer Res. 2018, 7, 839–848. [Google Scholar] [CrossRef]
  161. Zepp, J.A.; Zhao, J.; Liu, C.; Bulek, K.; Wu, L.; Chen, X.; Hao, Y.; Wang, Z.; Wang, X.; Ouyang, W.; et al. IL-17A-Induced PLET1 Expression Contributes to Tissue Repair and Colon Tumorigenesis. J. Immunol. 2017, 199, 3849–3857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Song, X.; Dai, D.; He, X.; Zhu, S.; Yao, Y.; Gao, H.; Wang, J.; Qu, F.; Qiu, J.; Wang, H.; et al. Growth Factor FGF2 Cooerates with Interleukin-17 to Repair Intestinal Epithelial Damage. Immunity 2015, 43, 488–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Hovav, A.H.; Wilharm, A.; Barel, O.; Prinz, I. Development and Function of γδT Cells in the Oral Mucosa. J. Dent. Res. 2020, 99, 498–505. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, X.; Cai, G.; Liu, C.; Zhao, J.; Gu, C.; Wu, L.; Hamilton, T.A.; Zhang, C.J.; Ko, J.; Zhu, L.; et al. IL-17R-EGFR axis links wound healing to tumorigenesis in Lrig1+ stem cells. J. Exp. Med. 2019, 216, 195–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Gur, G.; Rubin, C.; Katz, M.; Amit, I.; Citri, A.; Nilsson, J.; Amarigli, N.; Henriksson, R.; Rechavi, G.; Hedman, H.; et al. LRIG1 restricts growth factor signaling by enhancing receptor ubiquitylation and degradation. EMBO J. 2004, 23, 3270–3281. [Google Scholar] [CrossRef] [Green Version]
  166. Byrd, K.M.; Piehl, N.C.; Patel, J.H.; Huh, W.J.; Sequeira, I.; Lough, K.J.; Wagner, B.L.; Marangoni, P.; Watt, F.M.; Klein, O.D.; et al. Heterogeneity within Stratified Epithelial Stem Cell Populations Maintains the Oral Mucosa in Response to Physiological Stress. Cell Stem Cell 2019, 25, 814–829.e6. [Google Scholar] [CrossRef]
  167. Wei, T.; Cong, X.; Wang, X.T.; Xu, X.J.; Min, S.N.; Ye, P.; Peng, X.; Wu, L.L.; Yu, G.Y. Interleukin-17A promotes tongue squamous cell carcinoma metastasis through activating miR-23b/versican pathway. Oncotarget 2017, 8, 6663–6680. [Google Scholar] [CrossRef] [Green Version]
  168. Baram, T.; Rubinstein-Achiasaf, L.; Ben-Yaakov, H.; Ben-Baruch, A. Inflammation-Driven Breast Tumor Cell Plasticity: Stemness/EMT, Therapy Resistance and Dormancy. Front. Oncol. 2021, 10, 614468. [Google Scholar] [CrossRef]
  169. Lee, E.J.; Park, H.J.; Lee, I.J.; Kim, W.W.; Ha, S.J.; Suh, Y.G.; Seong, J. Inhibition of IL-17A suppresses enhanced-tumor growth in low dose pre-irradiated tumor beds. PLoS ONE 2014, 9, e106423. [Google Scholar] [CrossRef] [Green Version]
  170. Saul-McBeth, J.; Dillon, J.; Lee, A.; Launder, D.; Kratch, J.M.; Abutaha, E.; Williamson, A.A.; Schroering, A.G.; Michalski, G.; Biswas, P.; et al. Tissue Damage in Radiation-Induced Oral Mucositis Is Mitigated by IL-17 Receptor Signaling. Front. Immunol. 2021, 12, 687627. [Google Scholar] [CrossRef]
  171. Ling, J.; Zhang, L.; Wang, Y.; Chang, A.; Huang, Y.; Zhao, H.; Zhuo, X. Fisetin, a dietary flavonoid, increases the sensitivity of chemoresistant head and neck carcinoma cells to cisplatin possibly through HSP90AA1/IL-17 pathway. Phytother. Res. PTR 2023, 37, 1997–2011. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Interleukin-17 signaling. The IL-17R mediated transcriptional and posttranscriptional changes in target cells.
Figure 1. Interleukin-17 signaling. The IL-17R mediated transcriptional and posttranscriptional changes in target cells.
Ijms 24 09874 g001
Figure 2. The roles of IL-17 induced mediators in OSCC pathogenesis. Interleukin-17 from γδ T cells Th17 cells could stimulate proliferation of oral epithelial stem cells and their transformation which could be the initiating factor in OSCC pathogenesis. These could also contribute to progression of already established tumors and to therapy resistance. Interleukin-17 directly stimulates proliferation of tumor, prevents apoptosis leading to chemotherapy insensitivity. Moreover, IL-17 stimulates the production of G-CSF and GM-CSF, cytokines that expand, and chemokines that recruit myeloid cells, and neutrophils or granulocytic myeloid derived suppressor cells (MDSCs). These myeloid cells produce various angiogenic factors, MMPs, and promote tumor progression by stimulation of proliferation, survival, invasiveness, and metastasis and by suppressing antitumor immune activity of Tc17 and NK cells. Interleukin-17 stimulates cancer associated fibroblasts (CAFs) that produce mediators which promotes epithelial to mesenchymal transition and deposition of collagen resulting in escape from immune response. In addition, IL-17–induced protumoral cytokines, such as IL-6, function in a paracrine manner to enhance tumor growth and survival.
Figure 2. The roles of IL-17 induced mediators in OSCC pathogenesis. Interleukin-17 from γδ T cells Th17 cells could stimulate proliferation of oral epithelial stem cells and their transformation which could be the initiating factor in OSCC pathogenesis. These could also contribute to progression of already established tumors and to therapy resistance. Interleukin-17 directly stimulates proliferation of tumor, prevents apoptosis leading to chemotherapy insensitivity. Moreover, IL-17 stimulates the production of G-CSF and GM-CSF, cytokines that expand, and chemokines that recruit myeloid cells, and neutrophils or granulocytic myeloid derived suppressor cells (MDSCs). These myeloid cells produce various angiogenic factors, MMPs, and promote tumor progression by stimulation of proliferation, survival, invasiveness, and metastasis and by suppressing antitumor immune activity of Tc17 and NK cells. Interleukin-17 stimulates cancer associated fibroblasts (CAFs) that produce mediators which promotes epithelial to mesenchymal transition and deposition of collagen resulting in escape from immune response. In addition, IL-17–induced protumoral cytokines, such as IL-6, function in a paracrine manner to enhance tumor growth and survival.
Ijms 24 09874 g002
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

Ladjevac, N.; Milovanovic, M.; Jevtovic, A.; Arsenijevic, D.; Stojanovic, B.; Dimitrijevic Stojanovic, M.; Stojanovic, B.; Arsenijevic, N.; Arsenijevic, A.; Milovanovic, J. The Role of IL-17 in the Pathogenesis of Oral Squamous Cell Carcinoma. Int. J. Mol. Sci. 2023, 24, 9874. https://doi.org/10.3390/ijms24129874

AMA Style

Ladjevac N, Milovanovic M, Jevtovic A, Arsenijevic D, Stojanovic B, Dimitrijevic Stojanovic M, Stojanovic B, Arsenijevic N, Arsenijevic A, Milovanovic J. The Role of IL-17 in the Pathogenesis of Oral Squamous Cell Carcinoma. International Journal of Molecular Sciences. 2023; 24(12):9874. https://doi.org/10.3390/ijms24129874

Chicago/Turabian Style

Ladjevac, Nevena, Marija Milovanovic, Andra Jevtovic, Dragana Arsenijevic, Bojana Stojanovic, Milica Dimitrijevic Stojanovic, Bojan Stojanovic, Nebojsa Arsenijevic, Aleksandar Arsenijevic, and Jelena Milovanovic. 2023. "The Role of IL-17 in the Pathogenesis of Oral Squamous Cell Carcinoma" International Journal of Molecular Sciences 24, no. 12: 9874. https://doi.org/10.3390/ijms24129874

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