
Evolving role of deubiquitinating enzymes in oral cancer (Review)
- Authors:
- Published online on: May 19, 2025 https://doi.org/10.3892/ol.2025.15100
- Article Number: 354
-
Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Oral diseases rank highly amongst widespread health conditions worldwide, and impose notable health and economic burdens, which substantially diminish the quality of life of patients, impacting overall health and well-being (1). Oral cancer is a generic term used to refer to malignant tumors in oral organs. Notably, oral cancer encompasses cancer types of the lip, and all subsites of the oral cavity and oropharynx (2). According to the Global Cancer Statistics 2022, oral cancer was the 16th most common malignancy and the 15th leading cause of mortality worldwide (3), which was similar to the rankings observed in 2018 (4). The incidence of oral cancer is 2.72 cases per 100,000 individuals in China (men, 3.87; women, 1.60). Notably, the aforementioned rate is lower compared with the global incidence rate (men, 5.8; women, 2.3) (3,5). Oral squamous cell carcinoma (OSCC) is the most prevalent and extensively researched type of oral cancer, and the predominant malignancy within the head and neck region (6,7). In China, a total of 65,400 new cases of oral and pharyngeal cancer occurred in 2022 (5). The prevalence of oral cancer is projected to increase by ~40% by 2040 and this may lead to an increase in mortality rates in the future (5). Thus, numerous studies have focused on the molecular mechanisms underlying tumor growth, invasion, migration and distant metastasis, with the aim of identifying novel therapeutic targets and key tumor markers in OSCC. Previous studies have demonstrated that deubiquitinating enzymes (DUBs) serve an important role in the development of OSCC (8,9). The present review aims to investigate the functional mechanisms, tumorigenic regulation and therapeutic targets of DUBs in OSCC, which may provide a novel theoretical basis for the diagnosis and treatment of oral cancer in the future.
Oral carcinogenesis
In total, ~90% of oral cancer types originate in the stratified non-keratinized epithelium of the oral mucosa (10). Notably, oral cancer may be caused by genetic, epigenetic and environmental factors, including tobacco, alcohol and poor nutrition, which lead to changes in oral keratinocytessize and shape (10). Early genetic and molecular alterations of oral keratinocytes occur in all tissue areas exposed to carcinogens, followed by varying degrees of damage to the epithelium, which may lead to oral epithelial carcinoma and metastasis (Fig. 1) (11). A previous study has demonstrated that the development of OSCC may also be caused by additional factors, such as autoimmune diseases, infectious diseases, immunosuppressive disorders and familial cancer syndromes that modulate the immune system (12).
The key characteristics of oral cancer include sustained cellular proliferation, resistance to apoptosis, invasion and metastasis, dysregulation of energy homeostasis, evasion of growth inhibitory signals, and the ability to circumvent immunotherapeutic interventions (13). Oral carcinogenesis is complex and multifactorial, involving genetic mutations, epigenetic modifications and imbalances in the tumor microenvironment (TME). Genetic alterations may lead to the abnormal activation of oncogenic signaling pathways, including PI3K/AKT/mTOR (14,15), EGFR (16), Wnt/β-catenin (17), Notch (18) and JAK/STAT (19) pathways, and simultaneously disrupt tumor suppressor pathways, such as the tumour protein 53/retinoblastoma pathway (20). Notably, the aforementioned alterations serve a key role in the progression of OSCC. Furthermore, epigenetic modifications, such as DNA methylation (21), histone covalent modifications (22) and chromatin remodeling (23), are also implicated in the initiation and progression of OSCC. Additional factors such as immune suppression (24), hypoxia (25) and imbalances in the oral microbiome (26) may also contribute to the dysregulated TME, thus facilitating OSCC progression.
Diagnosis and treatment of oral cancer
In clinical practice, patients with OSCC may present with early-stage lesions that are painless. However, as OSCC progresses, lesions may cause ulceration, nodules and tissue adherence (27). In total, ~50% of OSCC cases arise in the posterior lateral border of the tongue, with the remaining cases affecting the floor of the mouth, soft palate, gingiva, buccal mucosa and hard palate (28). Oral cancer is detected in clinical examinations; however, >50% of patients with OSCC are diagnosed during the advanced stages of the disease (stages III and IV) and >40% of patients with OSCC present with regional metastases at the time of diagnosis (28). Furthermore, OSCC may invade the ipsilateral cervical lymph nodes through lymphatic outflow or invade the contralateral or bilateral lymph nodes. Notably, the lungs, bones and liver are the main sites of OSCC metastasis (29).
At present, surgery is the primary treatment option for OSCC; however, adequate resection margins are difficult to achieve due to the complex anatomy of the affected area (13). Ionizing radiation (IR), immunotherapy and chemotherapy may be used to prevent or treat OSCC (13). Thus, the identification of novel biomarkers and therapeutic targets in OSCC is necessary. A recent systematic review has summarized the hallmarks of oral cancer and highlighted the importance of further studies focused on OSCC (30). In addition, numerous mono-antibodies or small molecular compounds that inhibit tumorigenesis have been developed. PRI-724, a specific inhibitor of the Wnt/β-catenin signaling pathway, works synergistically with vismodegib, erlotinib and HS-173 to effectively decrease cell viability, promote apoptosis and decrease cell migration in OSCC (31). Cetuximab, an EGFR-targeting antibody, may be used to enhance the antitumor function of PI3K/AKT inhibitors (32). Current research on oral cancer focuses on the role of DUBs, which exhibit potential as molecular targets in the treatment of oral cancer (8,9).
Ubiquitination and deubiquitination
The sequential enzymatic processes that covalently attach ubiquitin, a 76-residue polypeptide with a molecular mass of ~8.5 kDa, to target proteins, are known as ubiquitylation. Ubiquitylation is achieved through a mechanism that involves several factors, including ubiquitin-activating enzyme (E1), ubiquitin-binding enzyme (E2) and ubiquitin ligase (E3). In humans, there are two variants of E1 enzymes, namely, ubiquitin-like modifier activating enzyme 1 and ubiquitin-like modifier activating enzyme 6, alongside ~50 distinct E2 enzymes and ~600 different E3 enzymes. Notably, E3 enzymes are pivotal in the selective identification of target proteins for ubiquitination and operate in a manner that is both spatially and temporally specific (33). Ubiquitin contains seven lysine residues and an N-terminal region that function as a site for ubiquitination, specifically at positions K6, K11, K27, K29, K33, K48, K63 and M1. Ubiquitin chains bind to substrates by linking the glycine residue of ubiquitin to a lysine molecule of ubiquitin (34). Different linkages exhibit different roles for the target substrate. Notably, K48-linked chains represent the most prevalent type of ubiquitin linkage within cellular environments, accounting for >50% of all ubiquitin linkages (33). The primary function of K48-linked chains is to facilitate the targeting of proteins to the proteasome for degradation. By contrast, K63-linked chains, which are the second most abundant type of ubiquitin linkages, exhibit a range of non-degradative functions (33). Ubiquitination serves a key role in numerous pathological conditions, such as neurodegenerative diseases, various cancers, aging and metabolic disorders (35). Alternate atypical ubiquitin modifications, linked through M1, K6, K11, K27, K29 or K33, also exhibit unique functions in substrate modification (36). Variations in the use of ubiquitin lysine residues may lead to the formation of homotypic chains, which are linked exclusively through a single type of residue, or heterotypic and branched chains. The aforementioned processes are exemplified by K63-linear and K48-K11 hybrid polymers, respectively (37).
DUBs are a class of proteases that facilitate the reversal of protein ubiquitination, a critical process for maintaining healthy cellular homeostasis. DUBs are responsible for the removal of ubiquitin from target proteins, which enables the recycling of ubiquitin, mediated by ~100 distinct DUBs (38). Ubiquitin molecules may be conjugated to the N-terminal amino group or lysine residues on other ubiquitin molecules, which results in the formation of ubiquitin chains (39). DUBs possess the ability to dismantle ubiquitin conjugations by cleaving the linkages between ubiquitin molecules or processing ubiquitin precursors to produce free pools of ubiquitin (Fig. 2A) (39). In total, there are ~100 DUBs that are classified into eight different families, namely, ubiquitin specific protease (USP), ubiquitin carboxy-terminal hydrolase, JAB1/MPN/MOV34 metalloenzyme, ovarian tumor protease (OTU), motif interacting with ubiquitin-containing novel DUB, monocyte chemotactic protein-induced proteins zinc finger-containing ubiquitin peptidase 1 and Machado-Joseph disease (Fig. 2B) (40,41). Furthermore, DUBs exhibit four distinct mechanisms of action, namely, processing of ubiquitin precursors, recycling of ubiquitin molecules during ubiquitination, cleavage of poly-ubiquitin chains and reversal of ubiquitin conjugation (42). The aforementioned mechanisms are used to regulate several cellular functions, including cell cycle progression, vesicle transport, signal transduction and chromosome segregation (43). DUBs also serve key roles in various developmental processes of eukaryotic cells, including apoptosis (44), DNA damage repair (45), maintenance of cell stemness (46) and tumorigenesis (47). Thus, the association between ubiquitination and DUBs is essential for cellular homeostasis.
Deubiquitination in oral cancer
Numerous DUBs may be associated with either tumor-suppressive or oncogenic activities and exhibit potential as candidates for therapeutic intervention. Table I highlights key studies that focus on the regulatory mechanisms of DUBs in oral cancer under reference summarized.
Association between DUBs and the Wnt/β-catenin pathway in oral cancer
Under healthy conditions, the Wnt family of proteins bind to Frizzled receptor and related ligands such as LDL receptor-related protein 5/6 (LRP5/6) on the cell surface to form a complex that recruits the protein framing protein, Dishevelled, which leads to the phosphorylation of LRP5/6 and the recruitment and activation of the Axin protein complex. In turn, the activation of the Axin protein complex inhibits the phosphorylation and degradation of β-catenin proteins and leads to stabilization. Accumulation of β-catenin in the cytoplasm will lead to entry into the nucleus spontaneously, where β-catenin binds to T cell factor/lymphoid enhancer factor family proteins, which promotes the transcription and expression of Wnt target genes, including Axin2, c-Myc and Cyclin D1. The expression levels of Wnt target genes serve a key role in cell proliferation, cycle regulation and differentiation. In the absence of Wnt activation, β-catenin is phosphorylated by the Axin protein complex, where β-catenin binds to ubiquitin E3 ligase (β-Trcp) (48). β-Trcp is subsequently presented to the proteasome for ubiquitination (48). To date, numerous studies have focused on the role of Wnt in OSCC and demonstrated that components of the Wnt/β-catenin signaling pathway, including Wnt ligands, Wnt inhibitors, membrane receptors and intracellular mediators, serve a key role in the inhibition of OSCC (11,49,50).
A previous study has demonstrated that USP14 activates the Wnt downstream pathway by regulating the deubiquitination and subsequent phosphorylation of Dishevelled (51). In OSCC tissues, USP14 expression levels are markedly upregulated (52). Furthermore, in vitro cellular experiments and investigations using mice transplantation tumor models have demonstrated that the proliferation, invasion and migration of OSCC were inhibited following USP14 knockdown (52).
Association between DUBs and the NF-κB pathway in oral cancer
NF-κB is a transcription factor that is often located in the cytoplasm and NF-κB regulates the expression of various genes, impacting cellular physiology and pathology. Activation of the NF-κB signaling pathway is often achieved through IκB protein degradation and nuclear translocation of NF-κB proteins, which serve key roles in inflammatory responses, immune responses and cell survival. In the inactive state, the IκB protein forms a complex with NF-κB, which leads to the prevention of NF-κB nuclear translocation (53). When inflammatory factors or cytokines stimulate the cell, the IκB protein is ubiquitinated and degraded, which allows the release of NF-κB protein into the nucleus to regulate the transcription of target genes (IL-6, inducible nitric oxide synthase) (53). Activated NF-κB promotes OSCC migration, invasion and resistance to radiotherapy (54).
DUBs regulate the NF-κB signaling pathway, which leads to oncogenic and anti-oncogenic activity. Receptor-interacting protein 1 (RIP1) may be modified by K63-linked polyubiquitination, which leads to TNF-α-induced NF-κB activation, increased expression levels of anti-apoptotic proteins [cellular inhibitor of apoptosis protein-1/2, (cIAP1/2), Bcl-2] and the promotion of cell survival. The DUB USP4 exerts a regulatory effect on RIP1 and a previous study has demonstrated that USP4 was upregulated in OSCC (55). USP4 inhibits NF-κB activation and promotes apoptosis via cleavage of the K63 ubiquitin chain of RIP1, which leads to oncogenic activity (56). In addition, cylindromatosis lysine 63 deubiquitinase (CYLD) is a key negative regulator of NF-κB. CYLD specifically removes the K63 ubiquitin chain and the M1 linear ubiquitin chain, and inhibits NF-κB signaling within different pathways. Mutations or low expression levels of CYLD in OSCC result in abnormal activation of NF-κB and inhibition of TGF-β (57). Previous studies have also demonstrated that CYLD upregulation inhibited the invasion and metastasis of the SCC15 OSCC cell line (58,59).
Association between DUBs and the TGF-β pathway in oral cancer
Members of the TGF-β family exert cellular effects through the formation of heterotetrameric complexes, comprising type I and type II serine/threonine kinase transmembrane receptors. To date, five type II receptors and seven type I receptors, referred to as activin receptor-like kinases (ALKs), have been characterized. TGF-β and bone morphogenetic protein dimers induce the formation of a heterotetrameric complex between a specific type II receptor and a type I receptor, which leads to the transphosphorylation and subsequent activation of the type I receptor. Furthermore, type I receptors propagate signals into the cell through the phosphorylation of receptor-regulated SMADs, which form heteromeric complexes with SMAD4 (Co-SMAD) (60,61). Co-SMAD translocates to the nucleus and interacts with other transcription factors (p300/CBP, Snail), which leads to the regulation of gene transcription responses (60,61). TGF-β signal transduction pathways may elicit a variety of cellular responses, which serve a key role in embryonic development, maintenance of tissue homeostasis and the process of tumorigenesis (62,63).
Notably, CYLD knockdown induced stabilization of TGF-β receptor I (ALK5), which promoted TGF-β signaling in OSCC. Low CYLD expression levels may lead to increased phosphorylation of SMAD3, which is a key indicator for the activation of the TGF-β signaling pathway. Low CYLD expression was associated with poor overall survival of patients with invasive OSCC (64). In addition, results from a previous study have demonstrated that cell survival was markedly increased in cisplatin-resistant OSCC cells with CYLD knockdown, which was associated with activation of the TGF-β signaling pathway. EGFR tyrosine kinase inhibitors, such as gefitinib, may be used to decrease cell survival via inhibition of TGF-β (65).
Association between DUBs and the tumorigenesis of oral cancer
P53 is one of the most commonly mutated proteins in various cancer types and exhibits oncogenic activity in tumors. Notably, P53 is activated following cellular stress, which leads to the inhibition of cell cycle progression and induction of pro-apoptotic signaling (66). Murine double minute 2 (MDM2) is an E3 ubiquitin ligase that specifically binds to P53, which leads to ubiquitination and degradation of P53 proteins. Under healthy conditions, MDM2 regulates the stability of P53, which limits P53 activity and maintains low levels of protein expression. Following DNA damage, MDM2 is inhibited by DUBs, which induces the release of accumulated P53 and promotes P53 activity. In turn, P53 induces MDM2 gene expression, which forms a negative feedback loop known as the P53-MDM2 signaling pathway (67). In OSCC, P53 gene mutations result in a loss of the oncogenic function of P53 and the upregulation of P53 and MDM2, which are associated with poor prognosis in patients (68).
DUBs serve a key role in stabilizing P53. Notably, CYLD inhibits tumor growth by cleaving the K63 ubiquitination chain on P53, which indirectly removes the K48 chain and inhibits the ubiquitination degradation of P53 (69). Findings from previous studies have demonstrated that USP28 effectively removed the MDM2-catalyzed K48 ubiquitin chain from P53, which led to the stabilization of P53. However, transcription of USP28 was notably upregulated in OSCC. Another study has demonstrated that OSCC is often associated with mutations or genetic variations in P53 (70,71). Thus, USP28-mediated stabilization of P53 may be detrimental to patients with OSCC (70,71).
Notably, alternative mechanisms may also serve a role in DUB-regulated tumorigenesis. Programmed cell death-ligand 1 (PD-L1) is upregulated in OSCC and acts as an oncogene (8). Results from a previous study have demonstrated that ubiquitin-specific peptidase 9 X-linked (USP9X) interacted with PD-L1, which facilitated deubiquitination of PD-L1 and thereby enhanced the stability of protein expression, which may promote OSCC tumorigenesis (8). Furthermore, myeloid cell leukemia-1 (MCL1), an anti-apoptosis protein, is markedly upregulated in OSCC. MCL1 is also deubiquitinated by USP9X. Notably, pharmacological inhibition of USP9X may decrease MCL1 expression and induce cell death in OSCC (72). Another study also demonstrated that OTU deubiquitinase, ubiquitin aldehyde binding 1 (OTUB1) was positively associated with OSCC tumor stage. OTUB1 knockdown leads to the suppression of OSCC cell proliferation, invasion and migration, and promotes tumor-associated macrophage M1 polarization. However, OTUB1 knockdown leads to the suppression of M2 polarization, which, in turn, inhibits the survival of OSCC cells (73). Furthermore, USP14 knockdown suppresses OSCC cell proliferation in vitro and tumor growth in vivo, due to impaired Sox2 stability mediated by polyubiquitination. Additionally, USP14 interacts with phosphofructokinase-1 liver type (PFKL), a key rate-limiting enzyme in the glycolytic pathway, which enhances PFKL-mediated glycolytic metabolism, and ultimately promotes cellular proliferation, migration and tumorigenesis (74,75).
Association between DUBs and the treatment of oral cancer
Treatment of OSCC requires a multidisciplinary approach, which often consists of surgical resection of the primary lesion, followed by post-operative radiotherapy (76). Molecular targeted therapy is a novel therapeutic strategy, and at present, two types of drugs are approved by the Food and Drug Administration for the treatment of OSCC, namely, cetuximab and nabulizumab (77). A key determinant of mortality in patients with OSCC is the elevated incidence of recurrence following treatment. Numerous studies have indicated that the overall recurrence rate ranges from 28 to 44.5% (26,78,79). Cisplatin resistance is a major obstacle in the treatment of middle- and late-stage OSCC, which leads to recurrence, metastasis and a poor prognosis. Cisplatin resistance mediated by decreased CYLD expression is associated with the diminished accumulation of intracellular cisplatin and the inhibition of cisplatin-induced apoptosis, which occurs as a result of hyperactivation of the NF-κB signaling pathway (80). The tolerance of OSCC to radiotherapy also affects patient prognosis and IR may induce the apoptosis of tumor cells. In a previous study, USP14 was knocked down in nude mice bearing OSCC tumors. USP14 knockdown facilitated IR-induced autophagy via upregulation of LC3BII and γH2AX expression levels in OSCC cells subjected to IR (81).
Conclusions
Aberrant activation and expression of signaling pathway components are commonly observed in OSCC, which may promote tumor cell proliferation, invasion and metastasis, and inhibit apoptosis (82). Regulation of DUBs in OSCC is considered to be an important factor in the abnormal activation of signaling pathways (83–89). Notably, DUBs operate through four distinct mechanisms: i) The processing of ubiquitin protein precursors; ii) the retrieval of ubiquitin molecules during the ubiquitination process; iii) the cleavage of ubiquitin protein chains; and iv) the disassociation of ubiquitin proteins from substrate targets. According to the aforementioned functions, DUBs may reverse the ubiquitination of target proteins, thereby contributing to the equilibrium between ubiquitination and deubiquitination of substrate proteins (90). While DUBs are known to be involved in the initiation and progression of OSCC, the specific mechanisms and downstream effects of DUBs remain poorly understood. DUBs have a dual role in oral cancer. The upregulation of some DUBs, such as USP14 and USP9X, promotes tumor development, while the downregulation of others like CYLD is linked to tumor invasion and drug resistance. By regulating key pathways (Wnt/β-catenin, NF-κB, TGF-β and P53), DUBs influence tumor progression. Their expression levels correlate with patient prognosis, suggesting a potential as therapeutic targets. Clinically, DUBs can indicate the prognosis of invasive OSCC patients. Targeting DUBs may overcome treatment resistance, and some DUBs inhibitors might enhance therapeutic effects when combined with other treatments. Given their potential as therapeutic targets in the treatment of OSCC, further research is warranted to elucidate the regulatory mechanisms associated with DUBs and to assess the potential side effects of targeted therapies.
Acknowledgements
Not applicable.
Funding
This work was supported by Tianjin Beichen Hospital (Beichen District Health System Technology Project; grant no. SHGY-2023005).
Availability of data and materials
Not applicable.
Authors' contributions
WY and FL conceived and organized the manuscript. ZW, SC and JW wrote the manuscript. JH revised the manuscript for important intellectual content. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Peres MA, Macpherson LMD, Weyant RJ, Daly B, Venturelli R, Mathur MR, Listl S, Celeste RK, Guarnizo-Herreño CC, Kearns C, et al: Oral diseases: A global public health challenge. Lancet. 394:249–260. 2019. View Article : Google Scholar : PubMed/NCBI | |
Inchingolo F, Santacroce L, Ballini A, Topi S, Dipalma G, Haxhirexha K, Bottalico L and Charitos I: Oral cancer: A historical review. Int J Environ Res Public Health. 17:31682020. View Article : Google Scholar : PubMed/NCBI | |
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI | |
Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A and Bray F: Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 144:1941–1953. 2019. View Article : Google Scholar : PubMed/NCBI | |
Han B, Zheng R, Zeng H, Wang S, Sun K, Chen R, Li L, Wei W and He J: Cancer incidence and mortality in China, 2022. J Natl Cancer Cent. 4:47–53. 2024. View Article : Google Scholar : PubMed/NCBI | |
Irfan M, Delgado RZR and Frias-Lopez J: The oral microbiome and cancer. Front Immunol. 11:5910882020. View Article : Google Scholar : PubMed/NCBI | |
Mody MD, Rocco JW, Yom SS, Haddad RI and Saba NF: Head and neck cancer. Lancet. 398:2289–2299. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wu J, Guo W, Wen D, Hou G, Zhou A and Wu W: Deubiquitination and stabilization of programmed cell death ligand 1 by ubiquitin-specific peptidase 9, X-linked in oral squamous cell carcinoma. Cancer Med. 7:4004–4011. 2018. View Article : Google Scholar : PubMed/NCBI | |
Apoorva CC, Ananthaneni A, Kumar AJ, Guduru VS and Puneeth HK: Evaluation of USP22 and Ki-67 expression in oral squamous cell carcinoma: An immunohistochemical study. J Oral Maxillofac Pathol. 27:679–684. 2023. View Article : Google Scholar : PubMed/NCBI | |
Barsouk A, Aluru JS, Rawla P, Saginala K and Barsouk A: Epidemiology, risk factors, and prevention of head and neck squamous cell carcinoma. Med Sci (Basel). 11:422023.PubMed/NCBI | |
Reyes M, Flores T, Betancur D, Pena-Oyarzun D and Torres VA: Wnt/β-catenin signaling in oral carcinogenesis. Int J Mol Sci. 21:46822020. View Article : Google Scholar : PubMed/NCBI | |
Tarle M and Luksic I: Pathogenesis and therapy of oral carcinogenesis. Int J Mol Sci. 25:63432024. View Article : Google Scholar : PubMed/NCBI | |
Tan Y, Wang Z, Xu M, Li B, Huang Z, Qin S, Nice EC, Tang J and Huang C: Oral squamous cell carcinomas: State of the field and emerging directions. Int J Oral Sci. 15:442023. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Jiang C, Li N, Wang F, Xu Y, Shen Z, Yang L, Li Z and He C: The circEPSTI1/mir-942-5p/LTBP2 axis regulates the progression of OSCC in the background of OSF via EMT and the PI3K/Akt/mTOR pathway. Cell Death Dis. 11:6822020. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Dong Y, Zhao M, Ding L, Yang X, Jing Y, Song Y, Chen S, Hu Q and Ni Y: ITGB2-mediated metabolic switch in CAFs promotes OSCC proliferation by oxidation of NADH in mitochondrial oxidative phosphorylation system. Theranostics. 10:12044–12059. 2020. View Article : Google Scholar : PubMed/NCBI | |
Huang Z, Rui X, Yi C, Chen Y, Chen R, Liang Y, Wang Y, Yao W, Xu X and Huang Z: Silencing LCN2 suppresses oral squamous cell carcinoma progression by reducing EGFR signal activation and recycling. J Exp Clin Cancer Res. 42:602023. View Article : Google Scholar : PubMed/NCBI | |
Pena-Oyarzun D, Flores T, Torres VA, Quest AFG, Lobos-González L, Kretschmar C, Contreras P, Maturana-Ramírez A, Criollo A and Reyes M: Inhibition of PORCN blocks wnt signaling to attenuate progression of oral carcinogenesis. Clin Cancer Res. 30:209–223. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Chen Y and Liu W: Chaperonin containing TCP1 subunit 6A may activate Notch and Wnt pathways to facilitate the malignant behaviors and cancer stemness in oral squamous cell carcinoma. Cancer Biol Ther. 25:22871222024. View Article : Google Scholar : PubMed/NCBI | |
Wang S, Wang X, Sun J, Yang J, Wu D, Wu F and Zhou H: Down-regulation of DNA key protein-FEN1 inhibits OSCC growth by affecting immunosuppressive phenotypes via IFN-gamma/JAK/STAT-1. Int J Oral Sci. 15:172023. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Chen T, Yang X, Cheng H, Späth SS, Clavijo PE, Chen J, Silvin C, Issaeva N, Su X, et al: Attenuated TRAF3 fosters activation of alternative NF-ĸB and reduced expression of antiviral interferon, TP53, and RB to promote HPV-positive head and neck cancers. Cancer Res. 78:4613–4626. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Sun Y, Yang J, Wu D, Yu S, Liu J, Hu T, Luo J and Zhou H: DNMT1-targeting remodeling global DNA hypomethylation for enhanced tumor suppression and circumvented toxicity in oral squamous cell carcinoma. Mol Cancer. 23:1042024. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Li R, Wu L, Chen Y, Liu S, Zhao H, Wang Y, Wang L and Shao Z: Histone methyltransferase KMT2D cooperates with MEF2A to promote the stem-like properties of oral squamous cell carcinoma. Cell Biosci. 12:492022. View Article : Google Scholar : PubMed/NCBI | |
Oh SY, Kim J, Lee KY, Lee HJ, Kwon TG, Kim JW, Lee ST, Kim DG, Choi SY and Hong SH: Chromatin remodeling-driven autophagy activation induces cisplatin resistance in oral squamous cell carcinoma. Cell Death Dis. 15:5892024. View Article : Google Scholar : PubMed/NCBI | |
Hu S, Lu H, Xie W, Wang D, Shan Z, Xing X, Wang XM, Fang J, Dong W, Dai W, et al: TDO2+ myofibroblasts mediate immune suppression in malignant transformation of squamous cell carcinoma. J Clin Invest. 132:e1576492022. View Article : Google Scholar : PubMed/NCBI | |
Fang K, Sun M, Leng Z, Chu Y, Zhao Z, Li Z, Zhang Y, Xu A, Zhang Z, Zhang L, et al: Targeting IGF1R signaling enhances the sensitivity of cisplatin by inhibiting proline and arginine metabolism in oesophageal squamous cell carcinoma under hypoxia. J Exp Clin Cancer Res. 42:732023. View Article : Google Scholar : PubMed/NCBI | |
Lyu WN, Lin MC, Shen CY, Chen LH, Lee YH, Chen SK, Lai LC, Chuang EY, Lou PJ and Tsai MH: An oral microbial biomarker for early detection of recurrence of oral squamous cell carcinoma. ACS Infect Dis. 9:1783–1792. 2023. View Article : Google Scholar : PubMed/NCBI | |
Bagan J, Sarrion G and Jimenez Y: Oral cancer: Clinical features. Oral Oncol. 46:414–417. 2010. View Article : Google Scholar : PubMed/NCBI | |
Harada H, Kikuchi M, Asato R, Hamaguchi K, Tamaki H, Mizuta M, Hori R, Kojima T, Honda K, Tsujimura T, et al: Characteristics of oral squamous cell carcinoma focusing on cases unaffected by smoking and drinking: A multicenter retrospective study. Head Neck. 45:1812–1822. 2023. View Article : Google Scholar : PubMed/NCBI | |
Jerjes W, Upile T, Petrie A, Riskalla A, Hamdoon Z, Vourvachis M, Karavidas K, Jay A, Sandison A, Thomas GJ, et al: Clinicopathological parameters, recurrence, locoregional and distant metastasis in 115 T1-T2 oral squamous cell carcinoma patients. Head Neck Oncol. 2:92010. View Article : Google Scholar : PubMed/NCBI | |
Gonzalez-Moles MA, Warnakulasuriya S, Lopez-Ansio M and Ramos-Garcia P: Hallmarks of cancer applied to oral and oropharyngeal carcinogenesis: A scoping review of the evidence gaps found in published systematic reviews. Cancers (Basel). 14:38342022. View Article : Google Scholar : PubMed/NCBI | |
Kleszcz R, Frackowiak M, Dorna D and Paluszczak J: Combinations of PRI-724 Wnt/beta-catenin pathway inhibitor with vismodegib, erlotinib, or HS-173 synergistically inhibit head and neck squamous cancer cells. Int J Mol Sci. 24:104482023. View Article : Google Scholar : PubMed/NCBI | |
Tathineni P, Joshi N and Jelinek MJ: Current state and future directions of EGFR-Directed therapy in head and neck cancer. Curr Treat Options Oncol. 24:680–692. 2023. View Article : Google Scholar : PubMed/NCBI | |
Swatek KN and Komander D: Ubiquitin modifications. Cell Res. 26:399–422. 2016. View Article : Google Scholar : PubMed/NCBI | |
Harrigan JA, Jacq X, Martin NM and Jackson SP: Deubiquitylating enzymes and drug discovery: Emerging opportunities. Nat Rev Drug Discov. 17:57–78. 2018. View Article : Google Scholar : PubMed/NCBI | |
Song L and Luo ZQ: Post-translational regulation of ubiquitin signaling. J Cell Biol. 218:1776–1786. 2019. View Article : Google Scholar : PubMed/NCBI | |
van Wijk SJ, Fulda S, Dikic I and Heilemann M: Visualizing ubiquitination in mammalian cells. EMBO Rep. 20:e465202019. View Article : Google Scholar : PubMed/NCBI | |
Yau RG, Doerner K, Castellanos ER, Haakonsen DL, Werner A, Wang N, Yang XW, Martinez-Martin N, Matsumoto ML, Dixit VM and Rape M: Assembly and function of heterotypic ubiquitin chains in cell-cycle and protein quality control. Cell. 171:918–933.e20. 2017. View Article : Google Scholar : PubMed/NCBI | |
Loix M, Zelcer N, Bogie JFJ and Hendriks JJA: The ubiquitous role of ubiquitination in lipid metabolism. Trends Cell Biol. 34:416–429. 2024. View Article : Google Scholar : PubMed/NCBI | |
De Cesare V, Carbajo Lopez D, Mabbitt PD, Fletcher AJ, Soetens M, Antico O, Wood NT and Virdee S: Deubiquitinating enzyme amino acid profiling reveals a class of ubiquitin esterases. Proc Natl Acad Sci USA. 118:e20069471182021. View Article : Google Scholar : PubMed/NCBI | |
Abdul Rehman SA, Kristariyanto YA, Choi SY, Nkosi PJ, Weidlich S, Labib K, Hofmann K and Kulathu Y: MINDY-1 Is a member of an evolutionarily conserved and structurally distinct new family of deubiquitinating enzymes. Mol Cell. 63:146–155. 2016. View Article : Google Scholar : PubMed/NCBI | |
Kwasna D, Abdul Rehman SA, Natarajan J, Matthews S, Madden R, De Cesare V, Weidlich S, Virdee S, Ahel I, Gibbs-Seymour I and Kulathu Y: Discovery and characterization of ZUFSP/ZUP1, a distinct deubiquitinase class important for genome stability. Mol Cell. 70:150–164. e62018. View Article : Google Scholar : PubMed/NCBI | |
Tsuchida S and Nakayama T: Ubiquitination and deubiquitination in oral disease. Int J Mol Sci. 22:54882021. View Article : Google Scholar : PubMed/NCBI | |
Trulsson F, Akimov V, Robu M, van Overbeek N, Berrocal DAP, Shah RG, Cox J, Shah GM, Blagoev B and Vertegaal ACO: Deubiquitinating enzymes and the proteasome regulate preferential sets of ubiquitin substrates. Nat Commun. 13:27362022. View Article : Google Scholar : PubMed/NCBI | |
Schwickart M, Huang X, Lill JR, Liu J, Ferrando R, French DM, Maecker H, O'Rourke K, Bazan F, Eastham-Anderson J, et al: Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival. Nature. 463:103–107. 2010. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Zhao Y, Yang X, Ren X, Huang S, Gong S, Tan X, Li J, He S, Li Y, et al: USP44 regulates irradiation-induced DNA double-strand break repair and suppresses tumorigenesis in nasopharyngeal carcinoma. Nat Commun. 13:5012022. View Article : Google Scholar : PubMed/NCBI | |
Ling S, Shan Q, Zhan Q, Ye Q, Liu P, Xu S, He X, Ma J, Xiang J, Jiang G, et al: USP22 promotes hypoxia-induced hepatocellular carcinoma stemness by a HIF1α/USP22 positive feedback loop upon TP53 inactivation. Gut. 69:1322–1334. 2020. View Article : Google Scholar : PubMed/NCBI | |
Deng L, Meng T, Chen L, Wei W and Wang P: The role of ubiquitination in tumorigenesis and targeted drug discovery. Signal Transduct Target Ther. 5:112020. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, Zhou Z, Shu G and Yin G: Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther. 7:32022. View Article : Google Scholar : PubMed/NCBI | |
Xie J, Huang L, Lu YG and Zheng DL: Roles of the wnt signaling pathway in head and neck squamous cell carcinoma. Front Mol Biosci. 7:5909122020. View Article : Google Scholar : PubMed/NCBI | |
Moon JH, Lee SH and Lim YC: Wnt/β-catenin/Slug pathway contributes to tumor invasion and lymph node metastasis in head and neck squamous cell carcinoma. Clin Exp Metastasis. 38:163–174. 2021. View Article : Google Scholar : PubMed/NCBI | |
Jung H, Kim BG, Han WH, Lee JH, Cho JY, Park WS, Maurice MM, Han JK, Lee MJ, Finley D and Jho EH: Deubiquitination of dishevelled by Usp14 is required for Wnt signaling. Oncogenesis. 2:e642013. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Wu J, Chen Y, Ye D, Lei H, Xu H, Yang L, Wu Y and Gu W: Ubiquitin-specific protease 14 regulates cell proliferation and apoptosis in oral squamous cell carcinoma. Int J Biochem Cell Biol. 79:350–359. 2016. View Article : Google Scholar : PubMed/NCBI | |
Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T and Zhang J: NF-kappaB in biology and targeted therapy: New insights and translational implications. Signal Transduct Target Ther. 9:532024. View Article : Google Scholar : PubMed/NCBI | |
Chiu HW, Lee HL, Lee HH, Lu HW, Lin KY, Lin YF and Lin CH: AIM2 promotes irradiation resistance, migration ability and PD-L1 expression through STAT1/NF-ĸB activation in oral squamous cell carcinoma. J Transl Med. 22:132024. View Article : Google Scholar : PubMed/NCBI | |
Weinlich R and Green DR: The two faces of receptor interacting protein kinase-1. Mol Cell. 56:469–480. 2014. View Article : Google Scholar : PubMed/NCBI | |
Hou X, Wang L, Zhang L, Pan X and Zhao W: Ubiquitin-specific protease 4 promotes TNF-α-induced apoptosis by deubiquitination of RIP1 in head and neck squamous cell carcinoma. FEBS Lett. 587:311–316. 2013. View Article : Google Scholar : PubMed/NCBI | |
Zhao Y, Thornton AM, Kinney MC, Ma CA, Spinner JJ, Fuss IJ, Shevach EM and Jain A: The deubiquitinase CYLD targets Smad7 protein to regulate transforming growth factor β (TGF-β) signaling and the development of regulatory T cells. J Biol Chem. 286:40520–40530. 2011. View Article : Google Scholar : PubMed/NCBI | |
Morgan EL, Chen Z and Van Waes C: Regulation of NFĸB signalling by ubiquitination: A potential therapeutic target in head and neck squamous cell carcinoma? Cancers (Basel). 12:28772020. View Article : Google Scholar : PubMed/NCBI | |
Ge WL, Xu JF and Hu J: Regulation of oral squamous cell carcinoma proliferation through crosstalk between SMAD7 and CYLD. Cell Physiol Biochem. 38:1209–1217. 2016. View Article : Google Scholar : PubMed/NCBI | |
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C and He J: TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther. 9:612024. View Article : Google Scholar : PubMed/NCBI | |
Derynck R, Turley SJ and Akhurst RJ: TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol. 18:9–34. 2021. View Article : Google Scholar : PubMed/NCBI | |
Tauriello DVF, Sancho E and Batlle E: Overcoming TGFβ-mediated immune evasion in cancer. Nat Rev Cancer. 22:25–44. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wen B, Liao H, Lin W, Li Z, Ma X, Xu Q and Yu F: The Role of TGF-β during pregnancy and pregnancy complications. Int J Mol Sci. 24:168822023. View Article : Google Scholar : PubMed/NCBI | |
Shinriki S, Jono H, Maeshiro M, Nakamura T, Guo J, Li JD, Ueda M, Yoshida R, Shinohara M, Nakayama H, et al: Loss of CYLD promotes cell invasion via ALK5 stabilization in oral squamous cell carcinoma. J Pathol. 244:367–379. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kanemaru A, Shinriki S, Kai M, Tsurekawa K, Ozeki K, Uchino S, Suenaga N, Yonemaru K, Miyake S, Masuda T, et al: Potential use of EGFR-targeted molecular therapies for tumor suppressor CYLD-negative and poor prognosis oral squamous cell carcinoma with chemoresistance. Cancer Cell Int. 22:3582022. View Article : Google Scholar : PubMed/NCBI | |
Hassin O and Oren M: Drugging p53 in cancer: One protein, many targets. Nat Rev Drug Discov. 22:127–144. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhu H, Gao H, Ji Y, Zhou Q, Du Z, Tian L, Jiang Y, Yao K and Zhou Z: Targeting p53-MDM2 interaction by small-molecule inhibitors: Learning from MDM2 inhibitors in clinical trials. J Hematol Oncol. 15:912022. View Article : Google Scholar : PubMed/NCBI | |
Hong A, Zhang X, Jones D, Veillard AS, Zhang M, Martin A, Lyons JG, Lee CS and Rose B: Relationships between p53 mutation, HPV status and outcome in oropharyngeal squamous cell carcinoma. Radiother Oncol. 118:342–349. 2016. View Article : Google Scholar : PubMed/NCBI | |
Fernandez-Majada V, Welz PS, Ermolaeva MA, Schell M, Adam A, Dietlein F, Komander D, Büttner R, Thomas RK, Schumacher B and Pasparakis M: The tumour suppressor CYLD regulates the p53 DNA damage response. Nat Commun. 7:125082016. View Article : Google Scholar : PubMed/NCBI | |
Muller I, Strozyk E, Schindler S, Beissert S, Oo HZ, Sauter T, Lucarelli P, Raeth S, Hausser A, Al Nakouzi N, et al: Cancer cells employ nuclear caspase-8 to overcome the p53-Dependent G2/M checkpoint through cleavage of USP28. Mol Cell. 77:970–984. e72020. View Article : Google Scholar : PubMed/NCBI | |
Prieto-Garcia C, Tomaskovic I, Shah VJ, Dikic I and Diefenbacher M: USP28: Oncogene or tumor suppressor? A unifying paradigm for squamous cell carcinoma. Cells. 10:26522021. View Article : Google Scholar : PubMed/NCBI | |
Sulkshane P, Pawar SN, Waghole R, Pawar SS, Rajput P, Uthale A, Oak S, Kalkar P, Wani H, Patil R, et al: Elevated USP9X drives early-to-late-stage oral tumorigenesis via stabilisation of anti-apoptotic MCL-1 protein and impacts outcome in oral cancers. Br J Cancer. 125:547–560. 2021. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Li R, Qin H, He H and Li S: OTUB1′s role in promoting OSCC development by stabilizing RACK1 involves cell proliferation, migration, invasion, and tumor-associated macrophage M1 polarization. Cell Signal. 110:1108352023. View Article : Google Scholar : PubMed/NCBI | |
Liu C, Zhou S and Tang W: USP14 promotes the cancer stem-like cell properties of OSCC via promoting SOX2 deubiquitination. Oral Dis. 30:4255–4265. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Geng L, Tang Y, Wang Y, Zhang Y, Zhu C, Lei H, Xu H, Zhu Q, Wu Y and Gu W: Ubiquitin-specific protease 14 targets PFKL-mediated glycolysis to promote the proliferation and migration of oral squamous cell carcinoma. J Transl Med. 22:1932024. View Article : Google Scholar : PubMed/NCBI | |
Millen R, De Kort WWB, Koomen M, van Son GJF, Gobits R, Penning de Vries B, Begthel H, Zandvliet M, Doornaert P, Raaijmakers CPJ, et al: Patient-derived head and neck cancer organoids allow treatment stratification and serve as a tool for biomarker validation and identification. Med. 4:290–310. e122023. View Article : Google Scholar : PubMed/NCBI | |
Li H, Zhang Y, Xu M and Yang D: Current trends of targeted therapy for oral squamous cell carcinoma. J Cancer Res Clin Oncol. 148:2169–2186. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang W, Adeoye J, Thomson P and Choi SW: Multiple tumour recurrence in oral, head and neck cancer: Characterising the patient journey. J Oral Pathol Med. 50:979–984. 2021. View Article : Google Scholar : PubMed/NCBI | |
Blatt S, Kruger M and Sagheb K, Barth M, Kämmerer PW, Al-Nawas B and Sagheb K: Tumor recurrence and follow-up intervals in oral squamous cell carcinoma. J Clin Med. 11:70612022. View Article : Google Scholar : PubMed/NCBI | |
Suenaga N, Kuramitsu M, Komure K, Kanemaru A, Takano K, Ozeki K, Nishimura Y, Yoshida R, Nakayama H, Shinriki S, et al: Loss of tumor suppressor CYLD expression triggers cisplatin resistance in oral squamous cell carcinoma. Int J Mol Sci. 20:51942019. View Article : Google Scholar : PubMed/NCBI | |
Xie W and Xu L: Ubiquitin-specific protease 14 promotes radio-resistance and suppresses autophagy in oral squamous cell carcinoma. Exp Cell Res. 398:1123852021. View Article : Google Scholar : PubMed/NCBI | |
Patni AP, Harishankar MK, Joseph JP, Sreeshma B, Jayaraj R and Devi A: Comprehending the crosstalk between Notch, Wnt and Hedgehog signaling pathways in oral squamous cell carcinoma-clinical implications. Cell Oncol (Dordr). 44:473–494. 2021. View Article : Google Scholar : PubMed/NCBI | |
Sun T, Liu Z and Yang Q: The role of ubiquitination and deubiquitination in cancer metabolism. Mol Cancer. 19:1462020. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Bai Q, Pang N and Xue J: TCF12 induces ferroptosis by suppressing OTUB1-mediated SLC7A11 deubiquitination to promote cisplatin sensitivity in oral squamous cell carcinoma. Cell Biol Int. 48:1649–1663. 2024. View Article : Google Scholar : PubMed/NCBI | |
Feng L, Zhang J, Sun M, Qiu F, Chen W and Qiu W: Tumor Suppressor LINC02487 inhibits oral squamous cell carcinoma cell migration and invasion through the USP17-SNAI1 Axis. Front Oncol. 10:5598082020. View Article : Google Scholar : PubMed/NCBI | |
Lu R, Wu G, Chen M, Ji D, Liu Y, Zhou GG and Fu W: USP18 and USP20 restrict oHSV-1 replication in resistant human oral squamous carcinoma cell line SCC9 and affect the viability of SCC9 cells. Mol Ther Oncolytics. 23:477–487. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kobayashi E, Hwang D, Bheda-Malge A, Whitehurst CB, Kabanov AV, Kondo S, Aga M, Yoshizaki T, Pagano JS, Sokolsky M and Shakelford J: Inhibition of UCH-L1 deubiquitinating activity with two forms of LDN-57444 has anti-invasive effects in metastatic carcinoma cells. Int J Mol Sci. 20:37332019. View Article : Google Scholar : PubMed/NCBI | |
Chen S, Wu K, Zong Y, Hou Z, Deng Z and Xia Z: USP44 regulates HEXIM1 stability to inhibit tumorigenesis and metastasis of oral squamous cell carcinoma. Biol Direct. 19:1432024. View Article : Google Scholar : PubMed/NCBI | |
Chang W, Luo Q, Wu X, Nan Y, Zhao P, Zhang L, Luo A, Jiao W, Zhu Q, Fu Y and Liu Z: OTUB2 exerts tumor-suppressive roles via STAT1-mediated CALML3 activation and increased phosphatidylserine synthesis. Cell Rep. 41:1115612022. View Article : Google Scholar : PubMed/NCBI | |
Dewson G, Eichhorn PJA and Komander D: Deubiquitinases in cancer. Nat Rev Cancer. 23:842–862. 2023. View Article : Google Scholar : PubMed/NCBI |