Next Article in Journal
Assessing Skin Cancer Risk Factors, Sun Safety Behaviors and Melanoma Concern in Atlantic Canada: A Comprehensive Survey Study
Previous Article in Journal
PET/CT in Prostate Cancer
Previous Article in Special Issue
Integration of p16/HPV DNA Status with a 24-miRNA-Defined Molecular Phenotype Improves Clinically Relevant Stratification of Head and Neck Cancer Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-Coding RNAs in Oral Cancer: Emerging Roles and Clinical Applications

Cancer and Translational Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth (DPU), Pimpri, Pune 411033, India
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(15), 3752; https://doi.org/10.3390/cancers15153752
Submission received: 7 June 2023 / Revised: 29 June 2023 / Accepted: 12 July 2023 / Published: 25 July 2023
(This article belongs to the Collection The Role of Non-coding RNA in Cancer)

Abstract

:

Simple Summary

Oral cancer (OC) is one of the most prevalent cancers in the world. Despite improvements in therapies, OC still has a poor survival rate of about 50%, with metastasis being the worst-case scenario. Thus, there is an urgent need to understand the disease process and to develop diagnostic and therapeutic strategies for OC. Advancement of high throughput genome sequencing shows that more than 90% of the human genome encodes non-coding transcripts that do not code for any protein. In this review, we discuss the role of various types of these non-coding RNAs (ncRNAs) in OC and their promising clinical implications. Dysregulated expressions of ncRNAs are associated with OC initiation and progression, as well as therapy resistance. Differential expressions of these ncRNAs in blood or saliva have indicated their potential diagnostic and prognostic importance. In this review, we have summarized all the promising aspects of ncRNAs in the management of OC.

Abstract

Oral cancer (OC) is among the most prevalent cancers in the world. Certain geographical areas are disproportionately affected by OC cases due to the regional differences in dietary habits, tobacco and alcohol consumption. However, conventional therapeutic methods do not yield satisfying treatment outcomes. Thus, there is an urgent need to understand the disease process and to develop diagnostic and therapeutic strategies for OC. In this review, we discuss the role of various types of ncRNAs in OC, and their promising clinical implications as prognostic or diagnostic markers and therapeutic targets. MicroRNA (miRNA), long ncRNA (lncRNA), circular RNA (circRNA), PIWI-interacting RNA (piRNA), and small nucleolar RNA (snoRNA) are the major ncRNA types whose involvement in OC are emerging. Dysregulated expression of ncRNAs, particularly miRNAs, lncRNAs, and circRNAs, are linked with the initiation, progression, as well as therapy resistance of OC via modulation in a series of cellular pathways through epigenetic, transcriptional, post-transcriptional, and translational modifications. Differential expressions of miRNAs and lncRNAs in blood, saliva or extracellular vesicles have indicated potential diagnostic and prognostic importance. In this review, we have summarized all the promising aspects of ncRNAs in the management of OC.

1. Introduction

Oral squamous cell carcinoma (OSCC) or oral cancer (OC) is the most prevalent type of head and neck cancer that arises in the tongue, lips, and floor of the mouth. In the year 2020, GLOBOCAN estimated around 377,713 total cases and 177,757 deaths worldwide from lip and oral cavity cancer, in which India alone showed a high burden of the disease [1]. The American Cancer Society has recently estimated the incidence of cancers in the oral cavity and pharynx, with around 54,000 new cases and about 11,580 deaths in the year 2023 alone [2]. Some of the most common factors with which OC progression is often linked include high tobacco and alcohol consumption, as well as an infection caused by human papillomavirus (HPV) [3]. Despite the advancement of conventional therapeutic strategies, the overall survival rate of OC is barely 50%, and even worse in the case of metastasis [4]. Since 1991, the USA has shown a continuous decline in overall mortality by 33%. However, the mortality rate of OC has shown a continued increase by 2% in men and 1% in women per year [2]. The aggressiveness and heterogeneity of the disease with delayed diagnosis, lack of early detection markers, lack of effective chemotherapeutic drugs, therapy resistance, and side effects often make the management of the disease complicated [5]. Although the U.S. Food and Drug Administration (FDA) approved EGFR targeted therapy and PD-1/PD-L1 immune therapy show some promising results, but these have achieved limited success [6,7]. Thus, there is an urgent need to understand the disease process and to develop better diagnostic and therapeutic strategies.
Recent developments in high throughput genome sequencing have revealed that more than 90% of the human genome encodes non-coding transcripts that do not code for any protein [8]. For a long while, non-coding RNAs (ncRNAs) were considered junk materials for cells. Recent knowledge on comprehensive molecular evaluations has led to the noncoding genes acquiring considerable attention nowadays in the advancement of several diseases, including cancers. Based on structural and functional characteristics, types of ncRNAs present in a cell are tRNA, rRNA, long non-coding RNA (lncRNA), circular RNA (circRNA), microRNA (miRNA), PIWI-interacting RNA (piRNA), and small nucleolar RNA (snoRNA) (Figure 1). The ncRNAs are sometimes tissue-specific and cellular-compartment-specific in their distribution and expression, and sometimes ubiquitous. NcRNAs are found to interact with nucleic acids or proteins to alter their conformation, activities, and stabilities. The differential expressions of ncRNAs are reported in different cancers. For example, miR-15/16, miR-29, miR-34, miR-200 family, let-7, miR-21, miR-155, miR-17–92 cluster, miR-221/222, miR-195 and miR-26b were significantly modulated in different cancers, including leukemia, prostate cancer, colorectal cancer, pancreatic cancer, liver cancer, lung cancer, breast cancer, glioblastoma, ovarian cancer, renal cancer, and thyroid cancer, and involved in cancer progression, metastasis, and drug resistance [9,10]. The high expression of miR-195 and miR-26b and down-regulation of their common target gene Semaphorin 6D (SEMA6D) were found to be associated with therapy resistance in breast cancer and, thus, this signaling axis is suggested as a predictive marker for chemotherapy response [10]. Likewise, up-regulation of lncRNA HOTAIR and MALAT1, down-regulation of lncRNA Meg3 and dual function of lnRNA H19 were seen in different cancer types, including lung cancer, ovarian cancer, prostate cancer, breast cancer, colorectal cancer, gastric cancer, and liver cancer [9]. Among the circRNAs, circPRKCI and circHIPK3 were found to be up-regulated in glioma, lung cancer, breast cancer, colorectal cancer, gallbladder cancer, gastric cancer, and ovarian cancer [9]. Differential expressions of piR-651, piR-823, piR-932 were reported in cancers of lung, breast, colorectal, esophageal, and gastric cancer [9]. Apart from the primary tissues, differential expressions of the ncRNAs in body fluid, including blood and saliva, suggest their importance as diagnostic and therapeutic biomarkers.
To date, RNAseq studies have revealed more than 200 ncRNAs (including miRNA, lncRNA, circRNA, snoRNA, and piRNA) to have an association with OC progression. In this review, we have described the types of ncRNAs, their functions, and their possible role as diagnostic and prognostic markers in OC. The correlations between the expressions of these ncRNAs and OC have been elaborated further in this review. Results from different studies obtained so far suggest a better understanding of the relationship between non-coding RNA transcriptomic alterations and disease development, since these modifications have the prospective to be used as diagnostic and therapeutic biomarkers. Thus, the review has practical implications for the treatment of OC and may offer fresh perspectives and ideas for future mechanistic research in various preclinical and clinical settings.

2. Oral Cancer: Current Diagnostic and Prognostic Markers

Tissue biopsy and histological evaluation are the gold standard for diagnosing oral cancer (OC). However, this technique is painful for patients and causes a delayed diagnosis [11]. As non-invasive methods, metachromasia using iodine staining, or toluidine blue, which stains cancerous lesions, chemiluminescence-based lumenoscopy, auto-fluorescence based techniques like laser-induced auto-fluorescence (LIAF) or visually enhanced lesion scope (Velscope), and optical coherence tomography (OCT) provide assistance in early diagnosis and identification of oral pathophysiologic lesions [11]. The advancement of complete human genome insight, and the numerous potentials of cellular and epigenetic research, can be utilized as prognostic and diagnostic techniques for conducting rapid evaluation and treatment of oral lesions. The molecular diagnostic measures are divided into two types: nucleic acid-associated markers and protein-associated markers. In recent years, several biomarkers have been identified that could be utilized for the prognosis, diagnosis, differential diagnosis, prediction of recurrence, distant metastasis, and chemotherapy or radiotherapy resistance of OC. The biomarkers in OC include up-regulated expression of epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), matrix metalloproteinases (MMPs), proliferating cell nuclear antigen (PCNA), Ki-67, Cyclin D1, Cathepsin-d, CD44, cytokeratins, p53 and p16 mutation or expression status, and the prevalence of human papilloma virus (HPV) and its oncogenes [12]. These biomarkers can aid in screening and early detection and prognosis of OC, which can improve treatment outcomes by allowing early intervention. However, these biomarkers are not enough as a primary diagnostic tool, and can only be useful in combination with other diagnostic methods to confirm malignancy and its stage. In addition, these markers are often not organ-specific. Therapeutic intervention by targeting these biomarkers shows limited efficacy and side effects.

3. Diverse ncRNAs in OC

Multiple ncRNAs have been identified in association with OC growth, invasion, migration, and therapy resistance. The importance of the ncRNAs in OC is summarized below.

3.1. Role of miRNAs in Oral Cancer

The effect of microRNAs (miRNAs) has gained considerable attention in the past few years for regulating the biological processes of multiple malignancies. The miRNAs are 19–25 nucleotide small RNA molecules that mainly interact with the 3′UTR of the target mRNA, and thus regulate gene expression. However, interactions with 5′UTR, promoter, and coding region have also been reported [13]. The miRNA can function as an oncogene or tumor suppressor gene, and the deregulation of miRNAs is seen in many cancers, including OC. Defects in miRNA biogenesis machinery, alterations in miRNA genes or transcriptional regulation, or epigenetic regulations are often associated with the miRNA deregulation mechanism.
With the aim of improving patient survival, numerous miRNAs have been discovered to have roles in the etiology of OC throughout time. The miRNAs associated with tissue, or bio-fluids like blood, and saliva, are found to regulate cancer growth, survival, invasion, metastasis, angiogenesis, chemotherapy, and radiotherapy resistance [14]. Simultaneous suppression of oncomiRs and replacement of tumor suppressor miRNAs are suggested to be an effective approach in designing an OC treatment strategy. OncomiRs like miR-21, miR-155, miR-196, miR-1237, miR-31, miR-455, miR-181, miR-184, miR-134, miR-146, miR-93, miR-372, miR-373, miR-103a-3p, miR-454, miR-654-5p, miR-188-5p, miR-626, miR-4513, miR-944, and miR-650 are found to be up-regulated in OC cell lines and patient samples (Table 1). As stated in Table 1, many of the oncomiRs are found to be associated with OC diagnosis or prognosis, and some are suggested as biomarkers. The functions of the oncomiRs are associated with tumor progression, cell migration, drug resistance, inhibition of apoptosis, and induction of metastasis in OC. On the other hand, miRNAs such as miR-204, miR125b, miR-9, miR-26a/b, miR-491-5p, miR-375, miR-320, miR-218, miR-205, miR-181a, miR-138, miR-124, miR-99a, miR-34, miR-29a, miR-17, etc., function like tumor suppressors and are found to be down-regulated in OC cell lines, and patient samples (Table 1). The miRNAs generally regulate the expression of oncogenes and tumor suppressors to modulate OC progression. For example, the miR-21 is one of the well-studied oncomiRs, and targets many tumor suppressor genes. In OC, the miR-21 interacts and regulates PTEN, RECK, PDCD4, TPM1, DKK2, and CADM1, resulting in induction of proliferation, invasion, and chemoresistance [15,16,17]. The transcription factor AP-1 (activating protein-1) activates miR-21 in various cancers [18]. Exposure of tobacco-smoke-associated nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) induces miR-21 and miR-155 expressions in lung and OC cell lines [19]. The miR-155 is involved in development of many cancers, and can act as both oncogene and tumor suppressor gene. In OC, the miR-155 is up-regulated as oncomiR, and associated with OC progression, metastasis, and drug resistance [20,21,22]. The transforming growth factor β (TGF-β)/Smad4 signaling activates miR-155 expression and promoter activity [23]. The TGF-β, a pleiotropic cytokine, regulates tumor suppression or promoting activity in OC [24]. Overexpression of TGF-β induces metastasis and immune modulation in OC [24]. Another well-studied miRNA is Let-7, which is responsible for cell differentiation in normal cells, and is found to become suppressed in OC [25]. The RNA binding protein Lin28A and B inhibit Let-7 biogenesis by binding at pre-miRNA gene [26]. In addition, lncRNA ROR, lncRNA H19, lncRNA PVT-1, and circ_CPA4, circ_HMCU act as miRNA sponges, and negatively regulate Let-7 expression in cancers [26]. Down-regulation of this miRNA increases expression of Twist and Snail which, in turn, promotes epithelial-to-mesenchymal transition (EMT) and plays a significant role in chemoresistance-to-cisplatin transition, as well as 5-fluorouracil (5-FU) [27]. The miR-124 is found to be deregulated in many cancers, including OC, due to promoter methylation. Overexpression of miR-124 inhibited integrin beta-1 (ITGβ1) and, thus, reduced OC migration [28]. Thus, the difference in the expression profiles gives both oncoMiRs and tumor suppressive miRNAs a role of potent biomarkers or therapeutic targets that might be beneficial to diagnose and treat OC.

3.2. Role of Long Non-Coding RNAs in Oral Cancer

Long non coding RNAs (LncRNAs) are more than 200 nucleotides long and they have similar biogenesis to messenger RNAs (mRNAs). They are transcribed by RNA-Polymerase II (Pol II), and undergo polyadenylation, splicing, and 5′-capping [115]. LncRNAs are localized either in the nucleus, cytoplasm, or both. Depending on the localization, they perform a variety of cellular processes by interacting with DNAs, RNAs, and proteins such as epigenetic modification, transcriptional regulation, RNA splicing, mRNA stabilization, translational regulation, sequestration of proteins, protein stabilization, and miRNA sponges [115,116,117,118]. Due to these regulatory functions, lncRNAs are associated with numerous disorders such as diabetes, inflammatory bowel diseases, AIDS, neurodegenerative diseases, different blood-related disorders, as well as cancers [119,120].
The lncRNAs exhibit aberrant expression patterns in OC, and play a significant role in the advancement of the disease [121]. In OC, the lncRNAs may either act as oncogenes or as tumor suppressor genes. The functions of some reported lncRNAs are summarized in Table 2. Oncogenic lncRNAs, such as lncRNAs MALAT1, NEAT1, PVT1, ELDR, DLEU1, HOTAIR, HIFCAR, PCAT1, DANCR, etc., are associated with tumor proliferation, invasion, metastasis, angiogenesis, and drug resistance. For example, high MALAT1 expression is correlated with OC recurrence and metastasis [122]. It has also been demonstrated that MALAT1 promotes cellular proliferation and metastasis via controlling multiple signaling events, including Wnt/β-catenin signaling and PI3K/AKT/mTOR signaling pathways [122]. MALAT-1 is present in circulation, and shows importance as a serum biomarker. The MALAT1 is associated with resistance in multiple drugs, such as cisplatin, 5-fluorouracil, and paclitaxel [122]. Currently, the lncRNA MALAT1 is in OC clinical trial. The MALAT1 promoter contains transcription factors SP1 and SP3 binding sites [123]. Cooperative function of SP1 and SP3 activates MALAT1 expression [123]. In addition, STAT3/ TGF-β signaling axis, hypoxia-inducible factor (HIF)-2α, Yes-associated oncoprotein (YAP)-1, Jumonji C-domain–containing protein (JMJD)-1A, Octamer-binding transcription factor (OCT)-4, chemokine (C-C motif) ligand (CCL)-5, β-catenin, and Lysine-specific demethylase (KDM)-5B are involved in up-regulation of MALAT1 in OC and other cancers [123]. Another lncRNA ELDR is found to be up-regulated in OC cell lines and patient samples [124]. The ELDR inhibits miRNA-7, resulting in stabilization of EGFR. In addition, the ELDR interacts with RNA binding protein Interleukin Enhancer Binding Factor 3 (ILF3), and stabilizes cell cycle gene Cyclin E1. Interestingly, targeted inhibition of ELDR could inhibit in vivo tumor growth in a mouse model [124]. Overexpression of ELDR in normal oral keratinocytes (NOKs) induces cell proliferation and G2/M cell cycle progression through activation of CTCF/FOXM1/AURKA axis showing importance of the lncRNA as one of the OC driver genes [125]. It is not clear why the ELDR is up-regulated in OC; however, around 11% of samples contain the gene amplification in TCGA database (www.cbioportal.org, accessed on 28 June 2023).
On the other hand, the lncRNAs MEG3, GAS5, FENDRR, and PTCSC3 are down-regulated in OC. For example, the down-regulation of the lncRNA GAS5 increases miR-21 expression and helps in proliferation, invasion, EMT, and migration [126]. MEG3, being another tumor-suppressive lncRNA, suppresses miR-421 and the Wnt/β-catenin pathway [127,128]. Down-regulation of the lncRNA helps in the induction of the cell cycle, cell proliferation, metastasis, and suppression of cell apoptosis. Promoter methylation of MEG3 and GAS5 is one of the mechanisms of down-regulation in cancers [129,130]. As stated in Table 2, many lncRNAs are associated with OC diagnosis and prognosis, and are thus suggested to be OC biomarkers and therapeutic targets.
Table 2. List of lncRNAs regulating oral cancer.
Table 2. List of lncRNAs regulating oral cancer.
LncRNAsFunction on Up or Down RegulationTargets/Associated PathwaysStudy ModelBiomarker/TherapyReferences
List of up-regulated lncRNAs
lncRNA DANCR ↑Promotes proliferation, invasion, and migration, and suppresses apoptosismiR-135a-5p/KLF8 axis, DANCR/miR-4707-3p/FOXC2 pathwayPatient samples and cell linesBiomarker (prognostic and diagnostic), therapeutic target [131]
lncRNA PCAT1 ↑Regulation of proliferation, and inhibits apoptosisc-Myc-AKT1-p38 MAPK signaling pathwaysCell lines, patient tissue, and xenograft tumor modelTherapeutic target[132]
lncRNA PVT1 ↑Increases metastasis, proliferation, and invasion, enhanced EMT and cancer cell stemnessWnt/β-catenin signalingCell lines and xenograft tumor modelTherapeutic target[133]
lncRNA MALAT1 ↑Associated with differentiation and clinical staging in TSCC. Correlated with tumor occurrence, development, and prognosis in HNSCC.
Chemoresistance in LSCC and OSCC cells
Wnt/β-catenin in TSCC.
G2/M in HNSCC.
PI3K/AKT/mTORsignaling pathway in OSCC.
Cancer cell lines and tissue and plasma samplesDiagnostic biomarker[122]
lncRNA-ROR ↑Regulates cellular differentiation, represses p53 miR-145Clinical specimensPrognostic biomarker[134]
lncRNA NORAD ↑Causes cell proliferation, migration, decreasing apoptosis, sponges miR-577 to enhance TPM4.miR-577/TPM4 axisOC tissues and cell linesTherapeutic target [135]
lncRNA ELDR ↑Induces cell proliferation, and inhibits miR-7 to regulate EGFR. Regulates Cyclin E1 signaling through ILF3 ILF3-cyclin E1 signaling, Tissue
samples, cell
lines, and
xenograft mouse model
Therapeutic target[124]
lncRNA HOTAIR ↑Promotes tumor cell invasion and metastasis, and represses E-cadherin in OSCCEZH2Tissue, saliva
samples and cell lines
Biomarker and therapeutic target[136]
lncRNA HIFCAR ↑Modulates the hypoxia signal pathway and contributes to OSCC progressionhypoxia-inducible factor (HIF-1α)OC cell lines and xenograft mice modelPrognostic biomarker[137]
lncRNA UCA1 ↑Promotes proliferation and cisplatin resistance, as well as suppressed apoptosis in OSCC cellsmiR-184Tissue, saliva samples, cell lines, and xenograft mice modelTherapeutic target[138]
lncRNA XIST ↑Regulates miR-29b expression, which induces cell apoptosis through the p53 pathway and promotes tumor growth in in vivo modelmiR-29bXenograft model and OC cell linesTherapeutic target[139]
lncRNA ARNIL ↑Promotes proliferation, invasion, and migrationmiR-125aTissues and serum, cell lines, and xenograft mouse model Biomarker (prognostic)[140]
lncRNA NEAT1 ↑Promotes proliferation, migration, and invasionmiR-365/RGS20OC cell lines, tissue, saliva samples, and mice modelBiomarkers and therapeutic target[141,142]
lncRNA DLEU1 ↑Promotes proliferation, invasion, and migrationmiR-149/CDK6 axisOC cell linesTherapeutic target[143,144]
lncRNA AC007271.3 ↑Promotes proliferation, migration, and invasion, inhibits apoptosis, and induces tumor growth in vivoWnt/β-catenin signaling pathway, miR-125b-2-3p/Slug/E-cadherin axisOC tissues, saliva, plasma, cell lines, and mice modelTherapeutic target[145,146]
lncRNA LHFPL3-AS1 ↑Promotes OSCC growth and cisplatin resistanceLHFPL3-AS1/miR-362-5p/CHSY1 PathwayOC tissues and cell lines-[147]
lncRNA 01296 ↑Promotes proliferation, invasion, and migrationSRSF1 proteinTissue
samples, cell lines, and xenograft mice model
Therapeutic target[148]
lncRNA JPX ↑Promotes proliferation, invasion, and migrationmiR-944/CDH2 axisOC cell linesTherapeutic target[149]
lncRNA LINC00974 ↑Promotes invasion and migrationmiR-122, RhoATissue samples and cell lines-[150]
lncRNA PRNCR1 ↑Promotes proliferation, invasion, and migrationmiR-326/FSCN1 axisCell lines, saliva, and plasma-[151]
lncRNA LOLA1 ↑Promotes migration, invasion, and EMTAKT/GSK-3β pathwayTissue samples and cell linesTherapeutic target [152]
lncRNA MCM3AP-AS1 ↑Promotes proliferation, migration, and invasionmiR-204-5p/FOXC1Tissues and cells-[153]
lncRNA LINC00662 ↑Increased TNM stage and lymph node metastasis of the patients. Promotes cell growth and metastasismiR-144-3p/EZH2 AxisTissues and cell linesTherapeutic target[154]
lncRNA HOTTIP ↑Causes lymph node metastasis. Regulates proliferation, migration, and invasionHMGA2-Mediated Wnt/β-Catenin PathwayXenograft model, patient tissue and saliva, and cell linesBiomarker (diagnosis) and therapeutic target[155]
lncRNA MIR4435-2HG ↑Regulates cancer cell behavior. Involved in the promotion of cancer cell proliferation, migration, and invasionTGF-β1Plasma samples and cell linesTherapeutic target[156]
lncRNA GACAT1 ↑Promotes tumor growth and migrationmiR-149Tissue samples and cell linesTherapeutic target[157]
lncRNA TSPEAR-AS2 ↑Promotes tumor cell progression and is associated with advanced TNM stagingTSPEAR-AS2/miR-487a-3p/PPM1A axisTissues and cell linesBiomarker and therapeutic strategy[158]
lncRNA FTH1P3 ↑Induces cancer cell proliferation, migration, and invasionPI3K/Akt/GSK3b/ Wnt/β-cateninTissues and cell linesBiomarker [159]
lncRNA PLAC2 ↑Promotes proliferation and invasion in OSCC cells as well as tumor growth and metastasis in vivo Downstream Wnt/β-catenin signaling pathwayTissues cell lines, and xenograft mice modelBiomarker (prognosis and therapy) [160]
lncRNA SNHG20 ↑Promotes proliferation, migration, and invasionmiRNA-19b-3p/RAB14 axis, miR-29a/DIXDC1/Wntsignaling pathwayTissue samples and cell linesBiomarker (diagnosis), and therapeutic target [161,162]
lncRNA LINC01137 ↑Promotes proliferation, invasion, and migrationmiR-22-3pCell lines Therapeutic target[163]
lncRNA PSMA3-AS1 ↑Promotes proliferation, invasion, and migrationmiR-136-5p/FN1 axisPatient samplesPrognostic marker[164]
lncRNA DCST1-AS1 ↑Causes M2 polarization of tumor-associated macrophages, which thereby promotes tumor malignancy and metastasisNF-κB pathwayOC cell lines and tumor xenograft model Prognostic indicator[165]
lncRNA TUG1 ↑Promotes proliferation, invasion, and migration, prevents apoptosisTUG1/miR-593-3p/MAPK axisOC cells, tissues, saliva and nude mice model Therapeutic target and biomarker (diagnosis)[166]
lncRNA FOXD2-AS1 ↑Associated with poor pathological grading and prognosis in patients. Promotes proliferation and colony formation as well as regulates the cell cycle signaling pathwaysCDK2, CDK4, and P21OC cell linesTherapeutic target and biomarker (prognosis)[167]
lncRNA LINC01234 ↑Promotes growth, invasiveness, and metastasismiR-637/NUPR1 axis, miR-433/PAK4 axisTissue samples, cell lines, and nude mice modelTherapeutic target[168,169]
lncRNA LINC01207 ↑Promotes proliferation and migration, reduces apoptosis and autophagy of cellsmiR-1301-3p/LDHA axisTissue samples and cell lines Novel diagnostic and therapeutic target[170]
lncRNA TTN-AS1 ↑Promotes cell growth, and migration and restricts apoptosismiR-411-3p/NFAT5 axisTissue samples and cells, as well as mice modelTherapeutic target[171]
lncRNA ZEB1-AS1 ↑Promotes EMT, cell invasion, and migration. Act as a tumor promotermiR-23aPatient samples, cell lines, and xenograft mice modelTherapeutic target[172]
lncRNA H19↑Promotes proliferation, associated with the TNM staging and nodal invasionH19/miR-138/EZH2 axisCell lines, tissue samples, and mice model Therapeutic target[173]
lncRNA HCP5 ↑Facilitates Cell Invasion And EMTmiR-140-5p/SOX4 axisPatient samples and cell lines Therapeutic target[174]
lncRNA PTTG3P ↑Promotes proliferation and migrationPTTG3P/miR-142-5p/JAG1 axisOC cell lines-[175]
lncRNA IGF2BP2-AS1 ↑Promotes cell growth, and migration and restricts apoptosisWnt/β-catenin pathwayTissue samples, plasma, and cell linesTherapeutic target[176]
lncRNA ADAMTS9-AS2 ↑Promotes migration and invasion and facilitated metastasis in salivary adenoid cystic carcinoma (SACC)Binds with miR-143-3p and activates PI3K/Akt and MEK/ErksignalingTissue samples, cell lines, and xenograft mouse modelTherapeutic target[177]
lncRNA LTSCCAT ↑Promotes EMT and promotes invasion and metastasis in both in vivo and in vitromiR-103a-2-5p/SMYD3/TWIST1 axisTissue samples, cell lines and xenograft tumor modelTherapeutic target[178]
lncRNA RP11-284F21.9 ↑Promotes proliferation, invasion, and migrationmiR-383-5p/MAL2 axisTissue samples and cancer cell lines Therapeutic target[179]
lncRNA WWTR1-AS1 ↑Associated with larger tumor size, cervical node metastasis, and poor prognosisWWTR1-AS1/WWTR1 axisCell linesBiomarkers with
prognostic significance
[180]
LINC00668 ↑Facilitate VEGFA expression, and promotes tumor growthmiR-297/VEGFA axisOC cell lines and tissues Biomarker (diagnosis) and therapeutic target[181]
lncRNA HNF1A-AS1 ↑Promotes OSCC progressionNotch signaling pathwayTissue samples and cell linesTherapeutic target [182]
lncRNA MINCR ↑Causes proliferation and migrationWnt/β-catenin pathwayTissue samples and cancer cell lines Prognostic biomarker and therapeutic target[183]
lncRNA LACAT1 ↑Promotes malignant progressionmicroRNA-4301Tissue samples and cancer cell lines-[184]
lncRNA CASC9 ↑Enhances tumor progression via suppression of autophagy-mediated cell apoptosisAKT/mTOR pathwayTissue samples, cell lines, and mice modelBiomarker (diagnosis and prognosis) [185]
lncRNA SNHG26 ↑Promotes TSCC growth, metastasis, and cisplatin resistancePGK1/Akt/mTORTissue samples, cell lies, and xenograft mice modelTherapeutic target and biomarker (diagnosis)[186]
lncRNA PART1 ↑Promotes proliferation and inhibits apoptosisEZH2Tissue samples, cell lines, and mice modelDiagnosis biomarker and a novel therapeutic
target
[187]
lncRNA LINC00152 ↑Induces tumor progression, and is associated with tumor size, invasion of muscles of the tongue, lymph node metastasis, and recurrence as well-Tissue samplesBiomarker (diagnosis and prognosis)[188]
lncRNA HOXA-AS2 ↑Causes OC cell proliferation and promotes tumor growth in vivomiR-567/CDK8Tissue and plasma samples, cell lines, and xenograft modelBiomarker (prognostic) and therapeutic target[189]
lncRNA DNM3OS ↑Modulates cell viability and migrationDNM3OS/miR-204-5p/HIP1 axisClinical samples and OC cell lines Therapeutic target[190]
lncRNA SNHG1 ↑Leads to the proliferation of cancer cellsmiR-421/HMGB2 axisCancer cell linesTherapeutic target[191]
lncRNA HOXA10-AS ↑Promotes OC growth, and metastasis.TP63 mRNAXenograft model and cancer cell linesTherapeutic target[192]
lncRNA BBOX1 ↑Encourages proliferation and migration, and suppresses apoptosismiR-3940-3p/laminin subunit gamma 2 axisTissue, saliva and cell lines Therapeutic target[193]
lncRNA IFITM4P ↑Induces cell proliferation and enhanced immune escapePD-L1Tissue samples, cell lines, and xenografted tumorsTherapeutic target[194]
lncRNA CACS15 ↑Promotes proliferation and reduces expression of lncRNA MEG3lncRNA MEG3Tissue and plasma samplesDiagnostic biomarker [195]
lncRNA LINC00963 ↑Promotes cancer stemness, increases cancer aggressiveness, and reduces chemosensitivityABCB5Tissue samples, cancer cell lines, and xenograft nude mice modelTherapeutic target[196]
lncRNA OIP5-AS1 ↑Enhances cancer stemness, and is associated with poor clinical outcome -Clinical specimens -[197]
lncRNA KCNQ1OT1 ↑Increases cisplatin resistance, regulates proliferation and metastasis of cisplatin-resistant TSCC KCNQ1OT1/miR-124-3p/TRIM14 axisCisplatin-resistant TSCC samples and TSCC cell lines-[198]
lncRNA BLACAT1 ↑Regulates viability, and causes migration and invasion of cellsmiR-142-5pOC cell lines Therapeutic target[199]
lncRNA AFAP1-AS1 ↑Encourages tumor proliferation and indicates a poor prognosis CCNA2OC cell lines, xenograft tumor modelUnfavorable biomarker, therapeutic target [200]
lncRNA FAL1 ↑Causes proliferation and develops OSCCmicroRNA-761/CRKL pathwayTissues and cell linesTherapeutic target[201]
lncRNA HOXA11-AS ↑Promotes proliferation, and facilitates CDDP-resistancemiR-214-3p/PIM1Clinical tissue specimens, cell lines, and xenograft mice model Therapeutic target[202]
lncRNA FEZF1-AS1 ↑Promote the malignant progression of OSCCmiR-196aPatient samples and OSCC cell lines -[203]
lncRNA SNHG6 ↑Improves cell viability, proliferation, and EMT. Inhibits apoptosisβ-catenin and E-cadherinTca1183 cells-[204]
lncRNA HOXC13-AS ↑Induces proliferation, migration, and EMTmiR-378g/HOXC13 axisPatient samples and cell lines Therapeutic target[205]
lncRNA ORAOV1-B ↑Induces invasion, migration, and metastasis Binds to Hsp90 and activates the NF-κB-TNFα loop.OC cell linesTherapeutic target[206]
lncRNA LINC00319 ↑Induces proliferation, metastasis, EMT, invasion, and angiogenesismiR-199a-5p/FZD4 axisCancer cells and tissues Therapeutic target[207]
lncRNA SLC16A1-AS1 ↑Promotes proliferation and accelerates cell cycleSLC16A1-AS1/CCND1 (requires further elucidation)OC cell lines and patient tissue and plasma samplesTherapeutic target and diagnostic indicator[208]
lncRNA RP5-916L7.2 ↑Induces proliferation and represses apoptosismiR-328 and miR-939Patient samples and OC cell lines-[209]
lncRNA LINC00284 ↑Causes cell proliferation and migrationmiR-211-3p/MAFG axisPatient samples and cell linesBiomarker [210]
lncRNA LINC00958 ↑Promotes proliferation, migration, EMT and retards apoptosismiR-627-5p/YBX2 axisTissue, saliva, and cell lines Therapeutic target and biomarker (prognostic)[211]
lncRNA LEF1-AS1 ↑Increases cell survival, proliferation, and migration. Retards cell apoptosisLATS1Plasma samples and cell linesTherapeutic target and biomarker[212]
lncRNA FOXC2-AS1 ↑Improves proliferation, invasion, migration, and EMT and regulates the cell cyclemiR-6868-5p/E2F3 axisPatient samples and cell linesTherapeutic target[213]
lncRNA LINC01116 ↑Causes migration and invasionLINC01116/miR-9/MMP1 axisPatient samples and cell linesTherapeutic target[214]
List of down regulated lncRNAs
lncRNA MORT ↓Low expression is associated with increased proliferation and poor survivalROCK1Cell lines and patient samplesTherapeutic target[215]
lncRNA AC012456.4 ↓Significantly associated with tumor staging and survival rates for patientsJAK-STAT and MAPK signaling pathwaysCell lines and patient samplesDiagnostic, therapeutic and prognostic biomarker[216]
lncRNA MEG3 ↓Down-regulation alleviates the aggressiveness of cancer, and is associated with poor prognosis. Induces tumor growth by promotion of cell proliferation and metastasis, induced cell cycle and suppressed cell apoptosismiR-421, Wnt/β-catenin pathwayOC cell lines and patient samples-[127]
lncRNA HCG11 ↓Enhances OSCC proliferation, increases G1/S transition and Ki67 levelsmiR-455-5pOC cell linesTherapeutic target[217]
lncRNA SCIRT ↓Inhibits cancer cell apoptosismiR-221Patient samplesBiomarker [218]
lncRNA C5orf66-AS1 ↓Induces proliferation, invasion, and migration and inhibits apoptosisCYC1Clinical specimens and cell cultureTherapeutic target[219]
lncRNA PTCSC3 ↓Promotes cancer cell proliferation and invasion-Tissues and cell linesTherapeutic target[220]
lncRNA GAS5 ↓Promotes proliferation, invasion, EMT, and migrationmiR-21/PTEN axisTissue, serum, and cancer cell linesTherapeutic target[126]
lncRNA FENDRR ↓Fails to inhibit angiogenesis of OSCC PI3K/AKT pathwayCell lines and patient samplesTherapeutic target[221]
Up arrows ↑ indicate upregulation and down arrows ↓ indicate downregulation.

3.3. Role of Circular RNAs (circRNA) in Oral Cancer

CircRNAs are closed-loop, extremely stable ncRNA molecules with no 3′ or 5′ ends and a poly (A) tail. They belong to the lncRNA kingdom, and have a higher half-life compared to linear RNAs [222,223]. Back-splicing and exon skipping of pre-mRNAs are the two processes by which circRNAs are produced. The transcription process is carried out via RNA pol II [224]. These RNA structures have the ability to withstand exonucleolytic breakdown by RNase R. It has been seen that 80% of circRNAs are present in the cytoplasm. However, the presence of circRNAs in the nucleus makes the control of gene expression possible [225].
Due to their closed-loop orientation, tissue specificity, high stability, and conservation, they serve as significant biomarkers for several diseases. These RNA molecules perform several regulatory functions, such as miRNA sponging, direct protein binding, and certain circRNAs are even translated into proteins [226,227]. Numerous studies have revealed that the majority of aberrantly expressed circRNAs play a significant role in controlling the progression of cancer by influencing a number of cancer hallmarks. Proliferative signaling, encouraging tumor and antitumor immunity, triggering angiogenesis, promoting invasion, metastasis, and deregulating cellular energetics are some functions that are associated with an aberrant circRNA profile [228].
Research on circRNA in OC has increased in recent years, and it has been discovered that these RNA molecules have a significant influence on the development, management, and prognosis of OC [229]. Like miRNA and lncRNA, these RNA molecules also play a role as both oncogenes and tumor suppressors in OC. For example, circ_0002185, circ_PVT1, circ_100290, circ_0001742, circ_HIPK3, circ_0001971, circ_DOCK1, circ_FLNA, circ_GOLPH3, circ_CLK3, circ_CDR1, circ_0014359, circ_LPAR3, circ_SEPT9, etc., are up-regulated, whereas circ_0000140, circ-PKD2, circ_0005379, circ_0004491, circ_SPATA6, circ_0086414, circ_0008309, circGDI2, circ_0007059, etc., are down-regulated in OC (Table 3). The mechanism of differential expression of the circRNA in OC is not clear. The oncogenic circular RNA Circ_100290 acts as competing endogenous RNA (ceRNA), and inhibits miR-378a mediated suppression of glucose transporter GLUT1, resulting in the induction of glycolysis and cell growth [230]. The circ_PVT1 is derived from exon 3 of the oncogene of lncRNA PVT1 [231]. Recently, it was discovered that the mutant p53/ YAP/ TEAD transcription-competent complex is responsible for the up-regulation of circ_PVT1 in head and neck squamous cancer [231]. Through sponging miR-125b, Circ_PVT1 worked as a competitive endogenous RNA (ceRNA) to induce STAT3 signaling and cell proliferation [231]. Another circular RNA, called Circ_CDR1, has been stated to encourage autophagy under the hypoxic condition to enhance cell survival in OC, via control of the AKT/ERK-1/2/mTOR signaling pathway [232]. On the contrary, a study showed the association between suppressed expression of circ_0007059 and OC, via regulation of the AKT/mTOR pathway [233]. A high throughput sequencing study of OC samples identified significant down-regulation of circ_0005379 as compared to the adjacent normal tissues [234]. Up-regulation of circ_0005379 enhances cetuximab sensitivity, efficiently reduces OC proliferation, migration, invasion, and angiogenesis in vitro, and slows tumor growth in nude mice via inhibiting EGFR signaling [234]. All these studies suggest that these RNA molecules regulate OC via control over the major signaling pathways like MAPK, WNT/β-catenin, Notch, VEGF, and PI3K/AKT in OC [235]. The aggressive trait of OC was found to be linked to circ_PKD2 down-regulation. Overexpression of circ_PKD2 induces cell cycle arrest and apoptosis, and inhibited proliferation, migration, and invasion of OC through inhibiting miR-204-3p [236]. All these studies indicate the potential role of circRNA in OC. Table 3 enlists several other circRNAs whose aberrant expression level regulates OC.

3.4. Role of Small Nucleolar RNA (SnoRNA) in Oral Cancer

SnoRNAs are one of the many classes of non-coding RNA molecules present in the body. There are around 300 snoRNA sequences identified in the human genome. Although, snoRNAs are small in size, they are present in large quantities within the nucleus of cells [269]. Most snoRNAs are expressed in the intron of both coding and non-coding genes, while the remaining gets transcribed by RNA polymerase II (RNA pol II). Splicing, debranching, and co-transcription are the stages involved in their biogenesis. They play diverse roles in the development of ribosomal, small nuclear, and other pre-mRNA molecules via endonucleolytic disintegration and post-transcriptional regulation [269]. They also have the ability to control gene expressions by modifying and splicing mRNA. The snoRNAs form small nucleolar ribonucleoprotein complexes (snoRNP complexes) by binding to protein molecules, which then leads to the modification of rRNA bases [269].
SnoRNAs have roles in a variety of pathological and physiological processes. Studies have shown that snoRNAs control tumor growth, invasion, and metastasis, as well as cell death during the carcinogenesis process. More importantly, snoRNAs play a significant role in the development of OC tumors. Today, the differential expression of snoRNAs in OCs leads to the possibility of them being used as diagnostic and prognostic biomarkers [270]. However, the mechanism of differential expression of snoRNA in OC is not known clearly. Alteration in snoRNA biogenesis and post-transcriptional regulation may be involved in differential expression of different snoRNAs in OC. An in silico investigation using RNA seq data of 567 samples from the TCGA head and neck cancer cohort could identify 113 snoRNAs using p  < 0 .05 as the cut-off [271]. The top significantly modulated snoRNAs were associated with DNA template regulation, RNA editing, regulation of cell proliferation, adhesion, invasion, metastasis, PI3K-AKT signaling, EMT, and angiogenesis pathways. Further analysis with the top five snoRNAs (SNORD114-17: ENSG00000201569, SNORA36B: ENSG00000222370, SNORD78: ENSG00000212378, U3: ENSG00000212182, and U3: ENSG00000212195) showed association with patient survival, indicating the importance of snoRNAs in disease progression and as biomarkers in OC [271]. A microarray analysis from eight OC samples identified 16 significantly modulated snoRNAs as compared to control samples; among them, 15 were significantly down-regulated and associated with patient survival [270]. The SNHG3, a snoRNA that gets up-regulated in OC patients, induces migration and cell proliferation of oral squamous cells. It targets the nuclear transcription factor-Y subunit gamma (NFYC) via the SNHG3/ miR-2682-5p axis and functions as a biomarker [272,273]. SnoRNA SNHG15 also gets overexpressed in OC cell lines, and facilitates the malignant behaviors of OC via miR-188-5p/ DAAM1 as a target [274]. Thus, snoRNAs play a role in assisting tumor growth in OC. Further studies can increase their significance in cancer therapy in the future. Table 4 enlists expressions and functions of some snoRNAs in OC.

3.5. Role of piRNAs in Oral Cancer

PIWI-interacting RNAs (piRNAs) are a subclass of ncRNAs that can be divided into three primary categories: transposon-derived piRNAs, mRNA-derived piRNAs, and lncRNA-derived piRNAs. They are 24–31 nucleotides long, have 5′-end uridine or 10th position adenosine bias, and lack proper secondary structural features [278]. The piRNAs, composed of an array of different nucleotide sequences, are single-stranded ncRNAs that interact with P-element-induced wimpy testis (PIWI) proteins [279]. They are the largest group of ncRNAs, and are multi-functional. PiRNAs are instrumental in genome rearrangement, spermiogenesis, protein regulation, transposon silencing, epigenetic regulation, and germ stem-cell maintenance by binding to PIWI proteins to make a piRNA/PIWI complex [280].
The piRNAs are mostly known to be expressed in germ cells; however, their existence is also observed in cancer cells. Therefore, the question of employing these RNA molecules as a prognostic marker or therapeutic target arises. Current research has given evidence of the piRNAs/PIWI complex being used for the occurrence, development, metastasis, and recurrence of breast cancer [281] and lung cancer [282]. Some piRNAs have been found to have a role in the development of OC, and can be a potential biomarker or therapeutic target for OC in the future [283]. In the OC mouse model, piR354, piR415, piR832, and piR1584, have been found to interact with mRNA molecules [284]. Longer survival of patients with head and neck cancer is associated with low levels of piR-58510 and piR-35373 [285]. It is observed that genes like GALNT6, SPEDF, and MYBL2 that are paired with piRNAs are responsible for the suppression or progression of several OC tumors [284]. An in silico analysis using the RNA sequencing data of 455 head and neck cancer samples, and 43 matched non-tumors from The Cancer Genome Atlas (TCGA), showed a total of 305 piRNAs in both tumor and non-tumor tissues [286]. Among a total of 247 significantly altered genes, 25 piRNAs were exclusively expressed in non-tumor samples, and 87 were only expressed in tumors. The significantly up-regulated piRNAs, including the topmost gene FR140858, were associated with poor patient survival. This indicates the importance of piRNAs in OC as diagnostic and prognostic biomarkers [286]. Another study identified a panel of 30 piRNAs in 77 HPV positive head and neck cancer samples from the TCGA RNA seq data [287]. Simultaneous validation in cell lines further reported key piRNAs NONHSAT077364, NONHSAT102574, and NONHSAT128479 in HPV associated head and neck cancer development. Based on analysis of the tongue cancer GEO database (GSE196674 and GSE196688), 406 differentially expressed piRNAs were identified [288]. Further investigation identified a down-regulated piRNA: piR-33422 and its association with mevalonate/ cholesterol-pathway-related gene FDFT1 in tongue cancer. Using TCGA RNA seq data of 256 smoking-related head and neck cancer samples, a panel of 13 piRNAs were identified [289]. Among them, NONHSAT123636 and NONHSAT113708 were found to be associated with tumor stage, NONHSAT067200 with patient survival, and 6 other piRNAs with TP53 mutation and 3q26, 8q24, and 11q13 amplification. Further studies are needed to know the regulation of their expression and functional mechanism of piRNAs in OC.

4. NcRNAs in Oral Cancer Progression

OSCC or OC is a multistep process originating from epithelial cells by progressive accumulation of genetic and epigenetic alterations. The histological changes that occur during the carcinogenesis begin with atypical squamous cell hyperplasia to carcinoma in situ (CIS) through stages of dysplasia [290]. The molecular events associated with alterations in different protein coding genes during the development of OC are extensively studied [290]. However, the role of ncRNAs is not well-studied in this regard. Few studies have reported the potential modulation of miRNAs, lncRNAs, and circRNAs in premalignant lesions, including oral leukoplakia (LK), oral lichen planus (OLP), oral submucous fibrosis (OSF), and oral dysplasia, with respect to OC (Figure 2).
Up-regulation of miR-7, miR-31, miR-1293, and down-regulation of miR-133a, miR-204 and miR-206 were reported in OC samples. Among these miRNAs, significantly high expressions of miR-31 and down-regulation of its target gene C-X-C motif chemokine ligand 12 (CXCL12) were seen in LK and OLP tissues, suggesting their importance in OC progression from pre-cancerous stages [291]. The miR-21, miR-181b, miR-345, miR-549 and miR-205, were found to be overexpressed both in progressive dysplasia and OC [292]. Another study reported up-regulation of miR-145, lncRNA RoR, and SNHG1 and down-regulation of miR-34a from low-grade to high-grade dysplasia and, finally, to OC during carcinogenesis [293]. The lncRNA FGD5-AS1 inhibits NF-kB signaling, and gets down-regulated in chronic periodontal samples as compared to the healthy tissues [294]. On the other hand, lncRNA MALAT1 was found to be up-regulated in periodontal samples, and induced inflammation through TLR4 by targeting miR-20a [294]. In OSF, up-regulation of lncRNA LINC00974, HIF1A-AS1, and down-regulation of GAS5-AS1 were seen during the development of OSF [294]. An in silico study examined microarray data of 167 OC, 17 dysplasia, and 45 normal oral tissues from the GEO database for expression analysis of lncRNAs [295]. Among these groups, 200 lncRNAs were found to be common in three groups, and 1206 genes are common in OC vs. dysplasia groups. The differentially expressed genes (DEGs) identified among the three groups were found to regulate OC development through PI3K–Akt signaling and NF-kB signaling. Among the DEGs, lncRNA DUXAP10 is relatively new, and associated with the progression of OC development [295]. A high throughput sequencing study identified 366 significantly modulated circRNAs, including 65 up-regulated and 301 down-regulated in LK tissues as compared to the normal mucosa indicating their importance in OC development [296]. Some of the top significantly up-regulated circRNAs were Circ_HLA-C, Circ_PLIN4, Circ_MTX2, Circ_RNF13, and the down-regulated ones were Circ_SENP2, Circ_PLEKHM2, Circ_ERICH1, Circ_EMB, Circ_ALDH3A2, and Circ_ZNF720. The Circ_HLA-C showed stage-wise up-regulation from mild to severe dysplasia [296]. All these studies indicate the importance of ncRNAs in OC development from precancerous lesions to the most aggressive form; however, more mechanistic investigation is needed in this regard.

5. NcRNAs in Body Fluid and Exosomes of Oral Cancer as Diagnostic Markers

Even though there has been a lot of improvement in the treatment of OC in the last few years, the prognosis for OC remains poor. Involvement of extracellular vesicles or exosomes in biofluids, like blood and saliva, is seen in disease progression, cellular communication, and metastasis of many cancer types, including OC [297,298,299]. Moreover, studies have revealed that biomarkers in the blood are more intriguing, due to their low invasiveness and increased stability. In a recent study, aberrant expressions of 18 different circulating miRNAs have been discovered that possess a direct association with a poor prognosis for head and neck cancer [300]. Salivary exosomal miRNA-1307-5p was seen as a potent prognostic indicator for oral malignancies, as it has shown the ability to indicate poor prognosis as well as poor patient outcomes [301]. Increased levels of lncRNA TIRY derived from exosomes have been found to reduce miR-14 expression levels which, in turn, enhances OC progression and metastasis [302]. In another study, Li et al., have mentioned two lncRNAs, namely MAGI2-AS3 and CCDC144NL-AS1, derived from serum exosomes that encourage cellular proliferation, migration, and invasion in OC, via regulating the PI3K-AKT-mTOR pathway [303]. Several circRNAs have also been found to have their role as potent biomarkers in OC. A high level of circ_0000199 was seen in circulating exosomes of OC patients, which was found to be associated with poor survival outcomes, proving itself as a potent biomarker for OC [247]. The circ_0001874 and circ_0001971 derived from the saliva of patients were up-regulated in OC and identified as a potential diagnostic biomarker [250]. Table 5 enlists the expression of some of the ncRNAs, which are derived from exosomes, blood, serum/ plasma, and saliva samples, and have significance as biomarkers.

6. Non-Coding RNA in Oral Cancer Clinical Trials

The utilization of cancer-specific ncRNAs as biomarkers and therapeutic targets could aid in tailoring treatment options to specific patients or patient subgroups. In contrast to the typical tumor markers being used in latest medical applications, such as protein biomarkers and metabolic products, growing research indicates that ncRNAs could be ideal agents in cancer diagnosis and therapy. This is because the ncRNAs target multiple druggable and non-druggable targets and signaling events at a time. Further, they have tissue specificity, distinct RNA attributes of rapid detection, extra tissue-associated activity, and a far more stable structure [325]. Several clinical trials have been conducted using ncRNAs, especially using miRNAs as diagnostic and therapeutic biomarkers [326]. Through head-on targeting of gene sequences using antisense oligonucleotides (ASOs) and siRNA-associated therapeutic applications, the most sophisticated and concise therapeutic efforts at RNA screening have been achieved to date [326]. The lncRNA H19 promoter sequence has been introduced with the coding sequence of diphtheria toxin in BC-819 plasmid in clinical trials of the bladder, pancreatic and ovarian cancer [115,327]. The lncRNA HOTAIR and CCAT1 (ClinicalTrials.gov) are in clinical trials for thyroid and colorectal cancer diagnostic biomarker studies, whereas inhibitors of LINC01212 and lncMyoD are used as therapeutic markers for melanoma (US2016271163) and sarcoma therapy (WO2015020960) [115,327]. All these studies indicate possible clinical implications of ncRNAs in cancer diagnosis and therapy. A clinical study has been initiated to evaluate the sensitivity and specificity of miRNA-412 and miR-512 in extracellular vesicles from saliva in the malignant progression of OC (ClinicalTrials.gov Identifier: NCT04913545). Another trial has recently been initiated to identify diagnostic and prognostic miRNA biomarkers from blood, saliva, and tissue samples of head and neck cancer (ClinicalTrials.gov Identifier: NCT04305366). The diagnostic and therapeutic importance of lncRNA MALAT1 and its target miR-124 has been studied in saliva samples from OC patients (ClinicalTrials.gov Identifier: NCT05708209). A randomized phase II study has been initiated to identify salivary and plasma miRNAs of head and neck cancer patients, and monitor their change during the dietary intervention (ClinicalTrials.gov Identifier: NCT02869399). Two studies have been performed to assess the correlation of head–neck cancer immunotherapy with blood and plasma miRNAs profiles (ClinicalTrials.gov Identifier: NCT03843515 and NCT04453046). Low EGFR-AS1 lncRNA expression is determined as companion diagnostic biomarker of OC, and a phase II clinical study is recently ongoing to evaluate therapeutic efficacy of a certain drug of EGFR-associated advanced OC with low-EGFR-AS1 OC patients (ClinicalTrials.gov Identifier: NCT04946968). Outcomes of the study may indicate the diagnostic and therapeutic importance of EGFR-AS1 in OC treatment.

7. Future Perspectives

Specific ncRNA expression changes have been linked to disease progression and poor outcomes in OC patients. Moreover, the ncRNAs show a promising role in regulating multiple signaling pathways in the modulation of OC progression, invasion, and metastasis. Nevertheless, such conclusions still have to be transformed into medical settings for OC patient populations. This could be attributable to a lack of scientific and technological evaluation research in pre-clinical systems, a dearth of prospective cohort clinical studies, inadequate standardization of the work process to extract and enhance ncRNA, and an absence of standardization targets. Since OC is linked to both behavioral risk factors and genetic predisposition, investigations must account for population-level variability; thus, substantial research regarding patient cohort study from various global locations are required to confirm these benchmarks. In short, the field of ncRNA research in OC is rapidly advancing, and holds great promise for improving our understanding of the disease and developing new strategies for diagnosis, treatment, and prevention. However, there are still many unknowns, and much more research is needed to fully understand the complex roles that ncRNAs play in OC. There are many ncRNAs that are yet to be explored. Many modified ncRNAs are designed to improve their stability and target specific efficacy. Thus, studies in pre-clinical and clinical systems, modifications to increase in-vivo stability and organ-specific targeting, extensive validation, and follow-up studies are needed to fully realize the potential of ncRNAs in OC. A rapid detection kit to identify ncRNAs in blood or saliva samples would greatly improve the diagnostic accuracy of OC. Thus, the acquired information in these areas will pave the way for more important healthcare, prognostic, and treatment options for the management of OC in the near future.

8. Conclusions

In summary, the review describes the substantial role of both quantitative and qualitative alterations of different ncRNAs (such as miRNA, lncRNA, circRNA, snoRNA, and piRNA) in the development of OC. The ncRNAs target multiple signaling molecules at a time, and regulate cell proliferation, survival, angiogenesis, metastasis and drug resistance. Studies have revealed that the high death rate and morbidity in OC are correlated with the complexity of conducting a quick diagnosis and appropriate management. Therefore, a timely diagnosis can prove to be crucial for controlling potential invasion and metastasis of oral premalignant conditions, and can also increase the overall life expectancies of patients. Keeping this in mind, a lot of emphasis is being given to the regulative function of different ncRNA profiles, as they demonstrate great promise in identifying OC lesions. Large numbers of ncRNAs like miRNAs, lncRNAs, and circRNAs obtained from exosomes or blood, serum, and saliva unveil their implicit role as non-invasive diagnostic and prognostic biomarkers for OC. Some clinical studies have been initiated to identify blood or saliva miRNA biomarkers in OC patients. miR-412, miR-512, miR-124, lncRNAs MALAT-1, and EGFR-AS1 are currently in OC clinical trials as diagnostic and therapeutic biomarkers, and the outcomes are yet to be received. Recent research on the role of other ncRNA molecules, like snoRNAs and piRNAs, behind the cause of OC development also makes them potential contenders for early diagnosis tools. However, in order to properly integrate liquid biopsy tests into the clinical practice in OC diagnosis, more in-depth pre-clinical research, as well as experimental studies with large cohorts, would indeed be required to confirm these findings and the effectiveness of these biomarkers in OC.

Author Contributions

Conceptualization: S.S., S.B., J.K.P. and S.D.; writing—original draft preparation: S.D., B.B., A.M.A., J.S., S.B. and S.S.; writing—review and editing: J.K.P., S.B., S.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by the Ramalingaswami Re-entry fellowship, Department of Biotechnology, Govt. of India to S. Sur [BT/RLF/Re-entry/47/2021] and Intramural Grants, Dr. D. Y. Patil Vidyapeeth (DPU), Pimpri, Pune, India to S. Basu [DPU/644-43/2021].

Acknowledgments

The authors would like to thank Ratna B. Ray, Saint Louis University, USA for the critical review of our manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

OC: Oral Cancer, OSCC: oral squamous cell carcinoma, HNSCC: head and neck squamous cell carcinomas, TSCC: tongue squamous cell carcinoma, LSCC: laryngeal squamous cell cancer, SACC: salivary adenoid cystic carcinoma, ncRNA: non-coding RNA, miRNA: microRNA, lncRNA: long non-coding RNA, CircRNA: circular RNA, piRNA: PIWI-interacting RNA, snoRNA: small nucleolar RNA, siRNA: small interacting RNA, TSG: tumor suppressive gene, mRNA: messenger RNA, Pri-miRNA: primary microRNA, Pre-miRNA: precursor microRNA, EMT: epithelial–mesenchymal transition, snoRNP: small nucleolar protein, MALAT1: metastasis-associated lung adenocarcinoma transcript 1, NEAT1: nuclear paraspeckle assembly transcript 1, CCAT1: colon cancer associated transcript 1, EIciRNA: exon–intron circRNA, tricRNAs: tRNAintronic circular RNA, ecircRNAs: exonic circular RNAs, RBP: RNA-binding protein, LK: leukoplakia, OLP: oral lichen planus, OSF: oral submucous fibrosis.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef]
  3. Kumar, M.; Nanavati, R.; Modi, T.G.; Dobariya, C. Oral cancer: Etiology and risk factors: A review. J. Cancer Res. Ther. 2016, 12, 458–463. [Google Scholar] [CrossRef] [PubMed]
  4. Wong, T.; Wiesenfeld, D. Oral Cancer. Aust. Dent. J. 2018, 63 (Suppl. S1), S91–S99. [Google Scholar] [CrossRef]
  5. Sujir, N.; Ahmed, J.; Pai, K.; Denny, C.; Shenoy, N. Challenges in Early Diagnosis of Oral Cancer: Cases Series. Acta Stomatol. Croat. 2019, 53, 174–180. [Google Scholar] [CrossRef] [PubMed]
  6. Ai, L.; Chen, J.; Yan, H.; He, Q.; Luo, P.; Xu, Z.; Yang, X. Research Status and Outlook of PD-1/PD-L1 Inhibitors for Cancer Therapy. Drug Des. Dev. Ther. 2020, 14, 3625–3649. [Google Scholar] [CrossRef]
  7. Gharat, S.A.; Momin, M.; Bhavsar, C. Oral Squamous Cell Carcinoma: Current Treatment Strategies and Nanotechnology-Based Approaches for Prevention and Therapy. Crit. Rev. Ther. Drug Carr. Syst. 2016, 33, 363–400. [Google Scholar] [CrossRef]
  8. Dhamija, S.; Menon, M.B. Non-coding transcript variants of protein-coding genes—What are they good for? RNA Biol. 2018, 15, 1025–1031. [Google Scholar] [CrossRef]
  9. Le, P.; Romano, G.; Nana-Sinkam, P.; Acunzo, M. Non-Coding RNAs in Cancer Diagnosis and Therapy: Focus on Lung Cancer. Cancers 2021, 13, 1372. [Google Scholar] [CrossRef]
  10. Baxter, D.E.; Allinson, L.M.; Al Amri, W.S.; Poulter, J.A.; Pramanik, A.; Thorne, J.L.; Verghese, E.T.; Hughes, T.A. MiR-195 and Its Target SEMA6D Regulate Chemoresponse in Breast Cancer. Cancers 2021, 13, 5979. [Google Scholar] [CrossRef]
  11. Chaurasia, A.; Alam, S.I.; Singh, N. Oral cancer diagnostics: An overview. Natl. J. Maxillofac. Surg. 2021, 12, 324–332. [Google Scholar] [CrossRef]
  12. Basheeth, N.; Patil, N. Biomarkers in Head and Neck Cancer an Update. Indian J. Otolaryngol. Head Neck Surg. 2019, 71, 1002–1011. [Google Scholar] [CrossRef] [PubMed]
  13. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  14. Fang, C.; Li, Y. Prospective applications of microRNAs in oral cancer. Oncol. Lett. 2019, 18, 3974–3984. [Google Scholar] [CrossRef] [PubMed]
  15. Jung, H.M.; Phillips, B.L.; Patel, R.S.; Cohen, D.M.; Jakymiw, A.; Kong, W.W.; Cheng, J.Q.; Chan, E.K. Keratinization-associated miR-7 and miR-21 regulate tumor suppressor reversion-inducing cysteine-rich protein with kazal motifs (RECK) in oral cancer. J. Biol. Chem. 2012, 287, 29261–29272. [Google Scholar] [CrossRef] [PubMed]
  16. Kawakita, A.; Yanamoto, S.; Yamada, S.; Naruse, T.; Takahashi, H.; Kawasaki, G.; Umeda, M. MicroRNA-21 promotes oral cancer invasion via the Wnt/beta-catenin pathway by targeting DKK2. Pathol. Oncol. Res. 2014, 20, 253–261. [Google Scholar] [CrossRef] [PubMed]
  17. Zheng, G.; Li, N.; Jia, X.; Peng, C.; Luo, L.; Deng, Y.; Yin, J.; Song, Y.; Liu, H.; Lu, M.; et al. MYCN-mediated miR-21 overexpression enhances chemo-resistance via targeting CADM1 in tongue cancer. J. Mol. Med. 2016, 94, 1129–1141. [Google Scholar] [CrossRef]
  18. Feng, Y.H.; Tsao, C.J. Emerging role of microRNA-21 in cancer. Biomed. Rep. 2016, 5, 395–402. [Google Scholar] [CrossRef]
  19. Doukas, S.G.; Vageli, D.P.; Lazopoulos, G.; Spandidos, D.A.; Sasaki, C.T.; Tsatsakis, A. The Effect of NNK, A Tobacco Smoke Carcinogen, on the miRNA and Mismatch DNA Repair Expression Profiles in Lung and Head and Neck Squamous Cancer Cells. Cells 2020, 9, 1031. [Google Scholar] [CrossRef]
  20. Kirave, P.; Gondaliya, P.; Kulkarni, B.; Rawal, R.; Garg, R.; Jain, A.; Kalia, K. Exosome mediated miR-155 delivery confers cisplatin chemoresistance in oral cancer cells via epithelial-mesenchymal transition. Oncotarget 2020, 11, 1157–1171. [Google Scholar] [CrossRef]
  21. Ni, Y.H.; Huang, X.F.; Wang, Z.Y.; Han, W.; Deng, R.Z.; Mou, Y.B.; Ding, L.; Hou, Y.Y.; Hu, Q.G. Upregulation of a potential prognostic biomarker, miR-155, enhances cell proliferation in patients with oral squamous cell carcinoma. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2014, 117, 227–233. [Google Scholar] [CrossRef] [PubMed]
  22. Yete, S.; Saranath, D. MicroRNAs in oral cancer: Biomarkers with clinical potential. Oral Oncol. 2020, 110, 105002. [Google Scholar] [CrossRef] [PubMed]
  23. Kong, W.; Yang, H.; He, L.; Zhao, J.J.; Coppola, D.; Dalton, W.S.; Cheng, J.Q. MicroRNA-155 is regulated by the transforming growth factor beta/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA. Mol. Cell. Biol. 2008, 28, 6773–6784. [Google Scholar] [CrossRef] [PubMed]
  24. Pang, X.; Tang, Y.L.; Liang, X.H. Transforming growth factor-beta signaling in head and neck squamous cell carcinoma: Insights into cellular responses. Oncol. Lett. 2018, 16, 4799–4806. [Google Scholar] [CrossRef]
  25. Su, J.L.; Chen, P.S.; Johansson, G.; Kuo, M.L. Function and regulation of let-7 family microRNAs. MicroRNA 2012, 1, 34–39. [Google Scholar] [CrossRef]
  26. Ma, Y.; Shen, N.; Wicha, M.S.; Luo, M. The Roles of the Let-7 Family of MicroRNAs in the Regulation of Cancer Stemness. Cells 2021, 10, 2415. [Google Scholar] [CrossRef]
  27. Chang, C.J.; Hsu, C.C.; Chang, C.H.; Tsai, L.L.; Chang, Y.C.; Lu, S.W.; Yu, C.H.; Huang, H.S.; Wang, J.J.; Tsai, C.H.; et al. Let-7d functions as novel regulator of epithelial-mesenchymal transition and chemoresistant property in oral cancer. Oncol. Rep. 2011, 26, 1003–1010. [Google Scholar] [CrossRef]
  28. Hunt, S.; Jones, A.V.; Hinsley, E.E.; Whawell, S.A.; Lambert, D.W. MicroRNA-124 suppresses oral squamous cell carcinoma motility by targeting ITGB1. FEBS Lett. 2011, 585, 187–192. [Google Scholar] [CrossRef]
  29. Li, J.; Huang, H.; Sun, L.; Yang, M.; Pan, C.; Chen, W.; Wu, D.; Lin, Z.; Zeng, C.; Yao, Y.; et al. MiR-21 indicates poor prognosis in tongue squamous cell carcinomas as an apoptosis inhibitor. Clin. Cancer Res. 2009, 15, 3998–4008. [Google Scholar] [CrossRef]
  30. Rajan, C.; Roshan, V.G.D.; Khan, I.; Manasa, V.G.; Himal, I.; Kattoor, J.; Thomas, S.; Kondaiah, P.; Kannan, S. MiRNA expression profiling and emergence of new prognostic signature for oral squamous cell carcinoma. Sci. Rep. 2021, 11, 7298. [Google Scholar] [CrossRef]
  31. Darda, L.; Hakami, F.; Morgan, R.; Murdoch, C.; Lambert, D.W.; Hunter, K.D. The role of HOXB9 and miR-196a in head and neck squamous cell carcinoma. PLoS ONE 2015, 10, e0122285. [Google Scholar] [CrossRef] [PubMed]
  32. Lu, Y.C.; Chang, J.T.; Liao, C.T.; Kang, C.J.; Huang, S.F.; Chen, I.H.; Huang, C.C.; Huang, Y.C.; Chen, W.H.; Tsai, C.Y.; et al. OncomiR-196 promotes an invasive phenotype in oral cancer through the NME4-JNK-TIMP1-MMP signaling pathway. Mol. Cancer 2014, 13, 218. [Google Scholar] [CrossRef]
  33. Kao, Y.Y.; Chou, C.H.; Yeh, L.Y.; Chen, Y.F.; Chang, K.W.; Liu, C.J.; Fan Chiang, C.Y.; Lin, S.C. MicroRNA miR-31 targets SIRT3 to disrupt mitochondrial activity and increase oxidative stress in oral carcinoma. Cancer Lett. 2019, 456, 40–48. [Google Scholar] [CrossRef]
  34. Lai, Y.H.; Liu, H.; Chiang, W.F.; Chen, T.W.; Chu, L.J.; Yu, J.S.; Chen, S.J.; Chen, H.C.; Tan, B.C. MiR-31-5p-ACOX1 Axis Enhances Tumorigenic Fitness in Oral Squamous Cell Carcinoma Via the Promigratory Prostaglandin E2. Theranostics 2018, 8, 486–504. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, C.J.; Tsai, M.M.; Hung, P.S.; Kao, S.Y.; Liu, T.Y.; Wu, K.J.; Chiou, S.H.; Lin, S.C.; Chang, K.W. miR-31 ablates expression of the HIF regulatory factor FIH to activate the HIF pathway in head and neck carcinoma. Cancer Res. 2010, 70, 1635–1644. [Google Scholar] [CrossRef] [PubMed]
  36. Lu, W.C.; Liu, C.J.; Tu, H.F.; Chung, Y.T.; Yang, C.C.; Kao, S.Y.; Chang, K.W.; Lin, S.C. miR-31 targets ARID1A and enhances the oncogenicity and stemness of head and neck squamous cell carcinoma. Oncotarget 2016, 7, 57254–57267. [Google Scholar] [CrossRef] [PubMed]
  37. Tseng, S.H.; Yang, C.C.; Yu, E.H.; Chang, C.; Lee, Y.S.; Liu, C.J.; Chang, K.W.; Lin, S.C. K14-EGFP-miR-31 transgenic mice have high susceptibility to chemical-induced squamous cell tumorigenesis that is associating with Ku80 repression. Int. J. Cancer 2015, 136, 1263–1275. [Google Scholar] [CrossRef]
  38. Cheng, C.M.; Shiah, S.G.; Huang, C.C.; Hsiao, J.R.; Chang, J.Y. Up-regulation of miR-455-5p by the TGF-beta-SMAD signalling axis promotes the proliferation of oral squamous cancer cells by targeting UBE2B. J. Pathol. 2016, 240, 38–49. [Google Scholar] [CrossRef]
  39. Yang, C.C.; Hung, P.S.; Wang, P.W.; Liu, C.J.; Chu, T.H.; Cheng, H.W.; Lin, S.C. miR-181 as a putative biomarker for lymph-node metastasis of oral squamous cell carcinoma. J. Oral Pathol. Med. 2011, 40, 397–404. [Google Scholar] [CrossRef]
  40. Wong, T.S.; Liu, X.B.; Wong, B.Y.; Ng, R.W.; Yuen, A.P.; Wei, W.I. Mature miR-184 as Potential Oncogenic microRNA of Squamous Cell Carcinoma of Tongue. Clin. Cancer Res. 2008, 14, 2588–2592. [Google Scholar] [CrossRef]
  41. Liu, C.J.; Shen, W.G.; Peng, S.Y.; Cheng, H.W.; Kao, S.Y.; Lin, S.C.; Chang, K.W. miR-134 induces oncogenicity and metastasis in head and neck carcinoma through targeting WWOX gene. Int. J. Cancer 2014, 134, 811–821. [Google Scholar] [CrossRef] [PubMed]
  42. Peng, S.Y.; Tu, H.F.; Yang, C.C.; Wu, C.H.; Liu, C.J.; Chang, K.W.; Lin, S.C. miR-134 targets PDCD7 to reduce E-cadherin expression and enhance oral cancer progression. Int. J. Cancer 2018, 143, 2892–2904. [Google Scholar] [CrossRef] [PubMed]
  43. Hung, P.S.; Liu, C.J.; Chou, C.S.; Kao, S.Y.; Yang, C.C.; Chang, K.W.; Chiu, T.H.; Lin, S.C. miR-146a enhances the oncogenicity of oral carcinoma by concomitant targeting of the IRAK1, TRAF6 and NUMB genes. PLoS ONE 2013, 8, e79926. [Google Scholar] [CrossRef] [PubMed]
  44. Li, G.; Ren, S.; Su, Z.; Liu, C.; Deng, T.; Huang, D.; Tian, Y.; Qiu, Y.; Liu, Y. Increased expression of miR-93 is associated with poor prognosis in head and neck squamous cell carcinoma. Tumour Biol. 2015, 36, 3949–3956. [Google Scholar] [CrossRef] [PubMed]
  45. Tu, H.F.; Chang, K.W.; Lin, S.C.; Hung, W.W.; Ji, S.H.; Wu, H.L.; Liu, C.J. Aberrant miR-10b, miR-372, and miR-375 expression in the cytobrushed samples from oral potentially malignant disorders. J. Dent. Sci. 2022, 17, 688–695. [Google Scholar] [CrossRef] [PubMed]
  46. Yeh, L.Y.; Liu, C.J.; Wong, Y.K.; Chang, C.; Lin, S.C.; Chang, K.W. miR-372 inhibits p62 in head and neck squamous cell carcinoma in vitro and in vivo. Oncotarget 2015, 6, 6062–6075. [Google Scholar] [CrossRef]
  47. Yeh, L.Y.; Yang, C.C.; Wu, H.L.; Kao, S.Y.; Liu, C.J.; Chen, Y.F.; Lin, S.C.; Chang, K.W. The miR-372-ZBTB7A Oncogenic Axis Suppresses TRAIL-R2 Associated Drug Sensitivity in Oral Carcinoma. Front. Oncol. 2020, 10, 47. [Google Scholar] [CrossRef]
  48. Zhang, X.J.; Jin, Y.; Song, J.L.; Deng, F. MiR-373 promotes proliferation and metastasis of oral squamous cell carcinoma by targeting SPOP. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5270–5276. [Google Scholar] [CrossRef]
  49. Zhang, G.; Chen, Z.; Zhang, Y.; Li, T.; Bao, Y.; Zhang, S. Inhibition of miR-103a-3p suppresses the proliferation in oral squamous cell carcinoma cells via targeting RCAN1. Neoplasma 2020, 67, 461–472. [Google Scholar] [CrossRef]
  50. Guo, J.Y.; Wang, Y.K.; Lv, B.; Jin, H. miR-454 performs tumor-promoting effects in oral squamous cell carcinoma via reducing NR3C2. J. Oral Pathol. Med. 2020, 49, 286–293. [Google Scholar] [CrossRef]
  51. Lu, M.; Wang, C.; Chen, W.; Mao, C.; Wang, J. miR-654-5p Targets GRAP to Promote Proliferation, Metastasis, and Chemoresistance of Oral Squamous Cell Carcinoma Through Ras/MAPK Signaling. DNA Cell Biol. 2018, 37, 381–388. [Google Scholar] [CrossRef] [PubMed]
  52. Jeong, S.; Kim, S.A.; Ahn, S.G. HOXC6-Mediated miR-188-5p Expression Induces Cell Migration through the Inhibition of the Tumor Suppressor FOXN2. Int. J. Mol. Sci. 2021, 23, 9. [Google Scholar] [CrossRef] [PubMed]
  53. Lou, C.; Shi, J.; Xu, Q. Exosomal miR-626 promotes the malignant behavior of oral cancer cells by targeting NFIB. Mol. Biol. Rep. 2022, 49, 4829–4840. [Google Scholar] [CrossRef]
  54. Xu, Y.X.; Sun, J.; Xiao, W.L.; Liu, Y.S.; Yue, J.; Xue, L.F.; Deng, J.; Zhi, K.Q.; Wang, Y.L. MiR-4513 mediates the proliferation and apoptosis of oral squamous cell carcinoma cells via targeting CXCL17. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 3821–3828. [Google Scholar] [CrossRef]
  55. Peng, H.Y.; Hsiao, J.R.; Chou, S.T.; Hsu, Y.M.; Wu, G.H.; Shieh, Y.S.; Shiah, S.G. MiR-944/CISH mediated inflammation via STAT3 is involved in oral cancer malignance by cigarette smoking. Neoplasia 2020, 22, 554–565. [Google Scholar] [CrossRef]
  56. Chang, K.W.; Liu, C.J.; Chu, T.H.; Cheng, H.W.; Hung, P.S.; Hu, W.Y.; Lin, S.C. Association between high miR-211 microRNA expression and the poor prognosis of oral carcinoma. J. Dent. Res. 2008, 87, 1063–1068. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, Y.F.; Yang, C.C.; Kao, S.Y.; Liu, C.J.; Lin, S.C.; Chang, K.W. MicroRNA-211 Enhances the Oncogenicity of Carcinogen-Induced Oral Carcinoma by Repressing TCF12 and Increasing Antioxidant Activity. Cancer Res. 2016, 76, 4872–4886. [Google Scholar] [CrossRef]
  58. Chu, T.H.; Yang, C.C.; Liu, C.J.; Lui, M.T.; Lin, S.C.; Chang, K.W. miR-211 promotes the progression of head and neck carcinomas by targeting TGFbetaRII. Cancer Lett. 2013, 337, 115–124. [Google Scholar] [CrossRef]
  59. Zheng, J.; Wang, J.; Jia, Y.; Liu, T.; Duan, Y.; Liang, X.; Liu, L. microRNA-211 promotes proliferation, migration, and invasion ability of oral squamous cell carcinoma cells via targeting the bridging integrator 1 protein. J. Cell. Biochem. 2019, 120, 4644–4653. [Google Scholar] [CrossRef]
  60. Jiang, L.; Lv, L.; Liu, X.; Jiang, X.; Yin, Q.; Hao, Y.; Xiao, L. MiR-223 promotes oral squamous cell carcinoma proliferation and migration by regulating FBXW7. Cancer Biomark. 2019, 24, 325–334. [Google Scholar] [CrossRef]
  61. Yang, C.J.; Shen, W.G.; Liu, C.J.; Chen, Y.W.; Lu, H.H.; Tsai, M.M.; Lin, S.C. miR-221 and miR-222 expression increased the growth and tumorigenesis of oral carcinoma cells. J. Oral Pathol. Med. 2011, 40, 560–566. [Google Scholar] [CrossRef] [PubMed]
  62. Zhou, L.; Jiang, F.; Chen, X.; Liu, Z.; Ouyang, Y.; Zhao, W.; Yu, D. Downregulation of miR-221/222 by a microRNA sponge promotes apoptosis in oral squamous cell carcinoma cells through upregulation of PTEN. Oncol. Lett. 2016, 12, 4419–4426. [Google Scholar] [CrossRef] [PubMed]
  63. Jiang, F.; Zhao, W.; Zhou, L.; Zhang, L.; Liu, Z.; Yu, D. miR-222 regulates the cell biological behavior of oral squamous cell carcinoma by targeting PUMA. Oncol. Rep. 2014, 31, 1255–1262. [Google Scholar] [CrossRef] [PubMed]
  64. Zhao, L.; Ren, Y.; Tang, H.; Wang, W.; He, Q.; Sun, J.; Zhou, X.; Wang, A. Deregulation of the miR-222-ABCG2 regulatory module in tongue squamous cell carcinoma contributes to chemoresistance and enhanced migratory/invasive potential. Oncotarget 2015, 6, 44538–44550. [Google Scholar] [CrossRef] [PubMed]
  65. Wei, Z.; Lyu, B.; Hou, D.; Liu, X. Mir-5100 Mediates Proliferation, Migration and Invasion of Oral Squamous Cell Carcinoma Cells Via Targeting SCAI. J. Investig. Surg. 2021, 34, 834–841. [Google Scholar] [CrossRef]
  66. 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]
  67. Li, T.; Wu, Q.; Liu, D.; Wang, X. miR-27b Suppresses Tongue Squamous Cell Carcinoma Epithelial-Mesenchymal Transition by Targeting ITGA5. OncoTargets Ther. 2020, 13, 11855–11867. [Google Scholar] [CrossRef]
  68. Wang, X.; Li, F.; Zhou, X. miR-204-5p regulates cell proliferation and metastasis through inhibiting CXCR4 expression in OSCC. Biomed. Pharmacother. 2016, 82, 202–207. [Google Scholar] [CrossRef]
  69. Yu, C.C.; Chen, P.N.; Peng, C.Y.; Yu, C.H.; Chou, M.Y. Suppression of miR-204 enables oral squamous cell carcinomas to promote cancer stemness, EMT traits, and lymph node metastasis. Oncotarget 2016, 7, 20180–20192. [Google Scholar] [CrossRef]
  70. Henson, B.J.; Bhattacharjee, S.; O’Dee, D.M.; Feingold, E.; Gollin, S.M. Decreased expression of miR-125b and miR-100 in oral cancer cells contributes to malignancy. Genes Chromosom. Cancer 2009, 48, 569–582. [Google Scholar] [CrossRef]
  71. Lu, L.; Xue, X.; Lan, J.; Gao, Y.; Xiong, Z.; Zhang, H.; Jiang, W.; Song, W.; Zhi, Q. MicroRNA-29a upregulates MMP2 in oral squamous cell carcinoma to promote cancer invasion and anti-apoptosis. Biomed. Pharmacother. 2014, 68, 13–19. [Google Scholar] [CrossRef] [PubMed]
  72. Kim, J.S.; Choi, D.W.; Kim, C.S.; Yu, S.K.; Kim, H.J.; Go, D.S.; Lee, S.A.; Moon, S.M.; Kim, S.G.; Chun, H.S.; et al. MicroRNA-203 Induces Apoptosis by Targeting Bmi-1 in YD-38 Oral Cancer Cells. Anticancer Res. 2018, 38, 3477–3485. [Google Scholar] [CrossRef] [PubMed]
  73. Lee, S.A.; Kim, J.S.; Park, S.Y.; Kim, H.J.; Yu, S.K.; Kim, C.S.; Chun, H.S.; Kim, J.; Park, J.T.; Go, D.; et al. miR-203 downregulates Yes-1 and suppresses oncogenic activity in human oral cancer cells. J. Biosci. Bioeng. 2015, 120, 351–358. [Google Scholar] [CrossRef]
  74. Lim, H.S.; Kim, C.S.; Kim, J.S.; Yu, S.K.; Go, D.S.; Lee, S.A.; Moon, S.M.; Chun, H.S.; Kim, S.G.; Kim, D.K. Suppression of Oral Carcinoma Oncogenic Activity by microRNA-203 via Down-regulation of SEMA6A. Anticancer Res. 2017, 37, 5425–5433. [Google Scholar] [CrossRef] [PubMed]
  75. Lin, J.; Lin, Y.; Fan, L.; Kuang, W.; Zheng, L.; Wu, J.; Shang, P.; Wang, Q.; Tan, J. miR-203 inhibits cell proliferation and promotes cisplatin induced cell death in tongue squamous cancer. Biochem. Biophys. Res. Commun. 2016, 473, 382–387. [Google Scholar] [CrossRef]
  76. Arunkumar, G.; Deva Magendhra Rao, A.K.; Manikandan, M.; Prasanna Srinivasa Rao, H.; Subbiah, S.; Ilangovan, R.; Murugan, A.K.; Munirajan, A.K. Dysregulation of miR-200 family microRNAs and epithelial-mesenchymal transition markers in oral squamous cell carcinoma. Oncol. Lett. 2018, 15, 649–657. [Google Scholar] [CrossRef]
  77. Wong, T.S.; Liu, X.B.; Chung-Wai Ho, A.; Po-Wing Yuen, A.; Wai-Man Ng, R.; Ignace Wei, W. Identification of pyruvate kinase type M2 as potential oncoprotein in squamous cell carcinoma of tongue through microRNA profiling. Int. J. Cancer 2008, 123, 251–257. [Google Scholar] [CrossRef]
  78. Mutallip, M.; Nohata, N.; Hanazawa, T.; Kikkawa, N.; Horiguchi, S.; Fujimura, L.; Kawakami, K.; Chiyomaru, T.; Enokida, H.; Nakagawa, M.; et al. Glutathione S-transferase P1 (GSTP1) suppresses cell apoptosis and its regulation by miR-133alpha in head and neck squamous cell carcinoma (HNSCC). Int. J. Mol. Med. 2011, 27, 345–352. [Google Scholar] [CrossRef]
  79. Jiang, L.; Dai, Y.; Liu, X.; Wang, C.; Wang, A.; Chen, Z.; Heidbreder, C.E.; Kolokythas, A.; Zhou, X. Identification and experimental validation of G protein alpha inhibiting activity polypeptide 2 (GNAI2) as a microRNA-138 target in tongue squamous cell carcinoma. Hum. Genet. 2011, 129, 189–197. [Google Scholar] [CrossRef]
  80. Shiiba, M.; Shinozuka, K.; Saito, K.; Fushimi, K.; Kasamatsu, A.; Ogawara, K.; Uzawa, K.; Ito, H.; Takiguchi, Y.; Tanzawa, H. MicroRNA-125b regulates proliferation and radioresistance of oral squamous cell carcinoma. Br. J. Cancer 2013, 108, 1817–1821. [Google Scholar] [CrossRef]
  81. Yu, T.; Liu, K.; Wu, Y.; Fan, J.; Chen, J.; Li, C.; Yang, Q.; Wang, Z. MicroRNA-9 inhibits the proliferation of oral squamous cell carcinoma cells by suppressing expression of CXCR4 via the Wnt/beta-catenin signaling pathway. Oncogene 2014, 33, 5017–5027. [Google Scholar] [CrossRef] [PubMed]
  82. Fukumoto, I.; Hanazawa, T.; Kinoshita, T.; Kikkawa, N.; Koshizuka, K.; Goto, Y.; Nishikawa, R.; Chiyomaru, T.; Enokida, H.; Nakagawa, M.; et al. MicroRNA expression signature of oral squamous cell carcinoma: Functional role of microRNA-26a/b in the modulation of novel cancer pathways. Br. J. Cancer 2015, 112, 891–900. [Google Scholar] [CrossRef] [PubMed]
  83. Huang, W.C.; Chan, S.H.; Jang, T.H.; Chang, J.W.; Ko, Y.C.; Yen, T.C.; Chiang, S.L.; Chiang, W.F.; Shieh, T.Y.; Liao, C.T.; et al. miRNA-491-5p and GIT1 serve as modulators and biomarkers for oral squamous cell carcinoma invasion and metastasis. Cancer Res. 2014, 74, 751–764. [Google Scholar] [CrossRef] [PubMed]
  84. Cao, Z.H.; Cheng, J.L.; Zhang, Y.; Bo, C.X.; Li, Y.L. MicroRNA-375 inhibits oral squamous cell carcinoma cell migration and invasion by targeting platelet-derived growth factor-A. Mol. Med. Rep. 2017, 15, 922–928. [Google Scholar] [CrossRef]
  85. Jung, H.M.; Patel, R.S.; Phillips, B.L.; Wang, H.; Cohen, D.M.; Reinhold, W.C.; Chang, L.J.; Yang, L.J.; Chan, E.K. Tumor suppressor miR-375 regulates MYC expression via repression of CIP2A coding sequence through multiple miRNA-mRNA interactions. Mol. Biol. Cell 2013, 24, 1638–1648. [Google Scholar] [CrossRef]
  86. Wu, Y.; Sun, X.; Song, B.; Qiu, X.; Zhao, J. MiR-375/SLC7A11 axis regulates oral squamous cell carcinoma proliferation and invasion. Cancer Med. 2017, 6, 1686–1697. [Google Scholar] [CrossRef]
  87. Wu, Y.Y.; Chen, Y.L.; Jao, Y.C.; Hsieh, I.S.; Chang, K.C.; Hong, T.M. miR-320 regulates tumor angiogenesis driven by vascular endothelial cells in oral cancer by silencing neuropilin 1. Angiogenesis 2014, 17, 247–260. [Google Scholar] [CrossRef]
  88. Uesugi, A.; Kozaki, K.; Tsuruta, T.; Furuta, M.; Morita, K.; Imoto, I.; Omura, K.; Inazawa, J. The tumor suppressive microRNA miR-218 targets the mTOR component Rictor and inhibits AKT phosphorylation in oral cancer. Cancer Res. 2011, 71, 5765–5778. [Google Scholar] [CrossRef]
  89. Kim, J.S.; Park, S.Y.; Lee, S.A.; Park, M.G.; Yu, S.K.; Lee, M.H.; Park, M.R.; Kim, S.G.; Oh, J.S.; Lee, S.Y.; et al. MicroRNA-205 suppresses the oral carcinoma oncogenic activity via down-regulation of Axin-2 in KB human oral cancer cell. Mol. Cell. Biochem. 2014, 387, 71–79. [Google Scholar] [CrossRef]
  90. Nagai, H.; Hasegawa, S.; Uchida, F.; Terabe, T.; Ishibashi Kanno, N.; Kato, K.; Yamagata, K.; Sakai, S.; Kawashiri, S.; Sato, H.; et al. MicroRNA-205-5p suppresses the invasiveness of oral squamous cell carcinoma by inhibiting TIMP-2 expression. Int. J. Oncol. 2018, 52, 841–850. [Google Scholar] [CrossRef]
  91. Liu, M.; Wang, J.; Huang, H.; Hou, J.; Zhang, B.; Wang, A. miR-181a-Twist1 pathway in the chemoresistance of tongue squamous cell carcinoma. Biochem. Biophys. Res. Commun. 2013, 441, 364–370. [Google Scholar] [CrossRef] [PubMed]
  92. Shin, K.H.; Bae, S.D.; Hong, H.S.; Kim, R.H.; Kang, M.K.; Park, N.H. miR-181a shows tumor suppressive effect against oral squamous cell carcinoma cells by downregulating K-ras. Biochem. Biophys. Res. Commun. 2011, 404, 896–902. [Google Scholar] [CrossRef] [PubMed]
  93. Shao, Y.; Qu, Y.; Dang, S.; Yao, B.; Ji, M. MiR-145 inhibits oral squamous cell carcinoma (OSCC) cell growth by targeting c-Myc and Cdk6. Cancer Cell Int. 2013, 13, 51. [Google Scholar] [CrossRef] [PubMed]
  94. Kai, Y.; Peng, W.; Ling, W.; Jiebing, H.; Zhuan, B. Reciprocal effects between microRNA-140-5p and ADAM10 suppress migration and invasion of human tongue cancer cells. Biochem. Biophys. Res. Commun. 2014, 448, 308–314. [Google Scholar] [CrossRef]
  95. Yen, Y.C.; Shiah, S.G.; Chu, H.C.; Hsu, Y.M.; Hsiao, J.R.; Chang, J.Y.; Hung, W.C.; Liao, C.T.; Cheng, A.J.; Lu, Y.C.; et al. Reciprocal regulation of microRNA-99a and insulin-like growth factor I receptor signaling in oral squamous cell carcinoma cells. Mol. Cancer 2014, 13, 6. [Google Scholar] [CrossRef]
  96. Kumar, B.; Yadav, A.; Lang, J.; Teknos, T.N.; Kumar, P. Dysregulation of microRNA-34a expression in head and neck squamous cell carcinoma promotes tumor growth and tumor angiogenesis. PLoS ONE 2012, 7, e37601. [Google Scholar] [CrossRef]
  97. Chang, C.C.; Yang, Y.J.; Li, Y.J.; Chen, S.T.; Lin, B.R.; Wu, T.S.; Lin, S.K.; Kuo, M.Y.; Tan, C.T. MicroRNA-17/20a functions to inhibit cell migration and can be used a prognostic marker in oral squamous cell carcinoma. Oral Oncol. 2013, 49, 923–931. [Google Scholar] [CrossRef]
  98. Sun, C.; Li, J. Expression of MiRNA-137 in oral squamous cell carcinoma and its clinical significance. J. BUON 2018, 23, 167–172. [Google Scholar]
  99. Fang, R.; Lu, Q.; Xu, B. hsa-miR-5580-3p inhibits oral cancer cell viability, proliferation and migration by suppressing LAMC2. Mol. Med. Rep. 2021, 23, 453. [Google Scholar] [CrossRef]
  100. Du, Y.; Li, Y.; Lv, H.; Zhou, S.; Sun, Z.; Wang, M. miR-98 suppresses tumor cell growth and metastasis by targeting IGF1R in oral squamous cell carcinoma. Int. J. Clin. Exp. Pathol. 2015, 8, 12252–12259. [Google Scholar]
  101. Peng, C.Y.; Liao, Y.W.; Lu, M.Y.; Yu, C.H.; Yu, C.C.; Chou, M.Y. Downregulation of miR-1 enhances tumorigenicity and invasiveness in oral squamous cell carcinomas. J. Formos. Med. Assoc. = Taiwan Yi Zhi 2017, 116, 782–789. [Google Scholar] [CrossRef] [PubMed]
  102. Rastogi, B.; Kumar, A.; Raut, S.K.; Panda, N.K.; Rattan, V.; Joshi, N.; Khullar, M. Downregulation of miR-377 Promotes Oral Squamous Cell Carcinoma Growth and Migration by Targeting HDAC9. Cancer Investig. 2017, 35, 152–162. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, F.; Qi, S.; Zhang, X.; Wu, J.; Yang, X.; Wang, R. miR-23a-3p suppresses cell proliferation in oral squamous cell carcinomas by targeting FGF2 and correlates with a better prognosis: miR-23a-3p inhibits OSCC growth by targeting FGF2. Pathol. Res. Pract. 2019, 215, 660–667. [Google Scholar] [CrossRef] [PubMed]
  104. Feng, X.; Luo, Q.; Wang, H.; Zhang, H.; Chen, F. MicroRNA-22 suppresses cell proliferation, migration and invasion in oral squamous cell carcinoma by targeting NLRP3. J. Cell. Physiol. 2018, 233, 6705–6713. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, K.; Jin, J.; Ma, T.; Zhai, H. MiR-139-5p inhibits the tumorigenesis and progression of oral squamous carcinoma cells by targeting HOXA9. J. Cell. Mol. Med. 2017, 21, 3730–3740. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, X.; Chang, K.; Gao, J.; Wei, J.; Xu, G.; Xiao, L.; Song, G. MicroRNA-504 functions as a tumor suppressor in oral squamous cell carcinoma through inhibiting cell proliferation, migration and invasion by targeting CDK6. Int. J. Biochem. Cell Biol. 2020, 119, 105663. [Google Scholar] [CrossRef]
  107. Shi, B.; Ma, C.; Liu, G.; Guo, Y. MiR-106a directly targets LIMK1 to inhibit proliferation and EMT of oral carcinoma cells. Cell. Mol. Biol. Lett. 2019, 24, 1. [Google Scholar] [CrossRef]
  108. Wang, X.; Li, G.H. MicroRNA-16 functions as a tumor-suppressor gene in oral squamous cell carcinoma by targeting AKT3 and BCL2L2. J. Cell. Physiol. 2018, 233, 9447–9457. [Google Scholar] [CrossRef]
  109. Lv, L.; Wang, Q.; Yang, Y.; Ji, H. MicroRNA-495 targets Notch1 to prohibit cell proliferation and invasion in oral squamous cell carcinoma. Mol. Med. Rep. 2019, 19, 693–702. [Google Scholar] [CrossRef]
  110. Shiah, S.G.; Hsiao, J.R.; Chang, W.M.; Chen, Y.W.; Jin, Y.T.; Wong, T.Y.; Huang, J.S.; Tsai, S.T.; Hsu, Y.M.; Chou, S.T.; et al. Downregulated miR329 and miR410 promote the proliferation and invasion of oral squamous cell carcinoma by targeting Wnt-7b. Cancer Res. 2014, 74, 7560–7572. [Google Scholar] [CrossRef]
  111. Chen, L.; Zhu, Q.; Lu, L.; Liu, Y. MiR-132 inhibits migration and invasion and increases chemosensitivity of cisplatin-resistant oral squamous cell carcinoma cells via targeting TGF-beta1. Bioengineered 2020, 11, 91–102. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, K.; Jin, J.; Ma, T.; Zhai, H. MiR-376c-3p regulates the proliferation, invasion, migration, cell cycle and apoptosis of human oral squamous cancer cells by suppressing HOXB7. Biomed. Pharmacother. 2017, 91, 517–525. [Google Scholar] [CrossRef] [PubMed]
  113. Zhou, Y.; Xu, X.M.; Feng, Y. MiR-769-5p inhibits cancer progression in oral squamous cell carcinoma by directly targeting JAK1/STAT3 pathway. Neoplasma 2020, 67, 528–536. [Google Scholar] [CrossRef] [PubMed]
  114. Chou, S.T.; Peng, H.Y.; Mo, K.C.; Hsu, Y.M.; Wu, G.H.; Hsiao, J.R.; Lin, S.F.; Wang, H.D.; Shiah, S.G. MicroRNA-486-3p functions as a tumor suppressor in oral cancer by targeting DDR1. J. Exp. Clin. Cancer Res. 2019, 38, 281. [Google Scholar] [CrossRef] [PubMed]
  115. Sur, S.; Ray, R.B. Emerging role of lncRNA ELDR in development and cancer. FEBS J. 2022, 289, 3011–3023. [Google Scholar] [CrossRef]
  116. Gao, N.; Li, Y.; Li, J.; Gao, Z.; Yang, Z.; Li, Y.; Liu, H.; Fan, T. Long Non-Coding RNAs: The Regulatory Mechanisms, Research Strategies, and Future Directions in Cancers. Front. Oncol. 2020, 10, 598817. [Google Scholar] [CrossRef]
  117. Huang, W.; Li, H.; Yu, Q.; Xiao, W.; Wang, D.O. LncRNA-mediated DNA methylation: An emerging mechanism in cancer and beyond. J. Exp. Clin. Cancer Res. CR 2022, 41, 100. [Google Scholar] [CrossRef]
  118. Ishteyaq Majeed, S.; Mashooq Ahmad, D.; Kaiser Ahmad, B.; Tashook Ahmad, D.; Fayaz, A.; Syed Mudasir, A. Long Non-Coding RNAs: Biogenesis, Mechanism of Action and Role in Different Biological and Pathological Processes. In Recent Advances in Noncoding RNAs; Lütfi, T., Ed.; IntechOpen: Rijeka, Croatia, 2022; p. 118. [Google Scholar]
  119. Aznaourova, M.; Schmerer, N.; Schmeck, B.; Schulte, L.N. Disease-Causing Mutations and Rearrangements in Long Non-coding RNA Gene Loci. Front. Genet. 2020, 11, 527484. [Google Scholar] [CrossRef]
  120. Dahariya, S.; Paddibhatla, I.; Kumar, S.; Raghuwanshi, S.; Pallepati, A.; Gutti, R.K. Long non-coding RNA: Classification, biogenesis and functions in blood cells. Mol. Immunol. 2019, 112, 82–92. [Google Scholar] [CrossRef]
  121. Xu, Y.; Jiang, E.; Shao, Z.; Shang, Z. Long Noncoding RNAs in the Metastasis of Oral Squamous Cell Carcinoma. Front. Oncol. 2020, 10, 616717. [Google Scholar] [CrossRef]
  122. Ye, D.; Deng, Y.; Shen, Z. The Role and Mechanism of MALAT1 Long Non-Coding RNA in the Diagnosis and Treatment of Head and Neck Squamous Cell Carcinoma. OncoTargets Ther. 2021, 14, 4127–4136. [Google Scholar] [CrossRef] [PubMed]
  123. Fu, S.; Wang, Y.; Li, H.; Chen, L.; Liu, Q. Regulatory Networks of LncRNA MALAT-1 in Cancer. Cancer Manag. Res. 2020, 12, 10181–10198. [Google Scholar] [CrossRef] [PubMed]
  124. Sur, S.; Nakanishi, H.; Steele, R.; Zhang, D.; Varvares, M.A.; Ray, R.B. Long non-coding RNA ELDR enhances oral cancer growth by promoting ILF3-cyclin E1 signaling. EMBO Rep. 2020, 21, e51042. [Google Scholar] [CrossRef] [PubMed]
  125. Sur, S.; Steele, R.; Ko, B.C.B.; Zhang, J.; Ray, R.B. Long noncoding RNA ELDR promotes cell cycle progression in normal oral keratinocytes through induction of a CTCF-FOXM1-AURKA signaling axis. J. Biol. Chem. 2022, 298, 101895. [Google Scholar] [CrossRef] [PubMed]
  126. Zeng, B.; Li, Y.; Jiang, F.; Wei, C.; Chen, G.; Zhang, W.; Zhao, W.; Yu, D. LncRNA GAS5 suppresses proliferation, migration, invasion, and epithelial-mesenchymal transition in oral squamous cell carcinoma by regulating the miR-21/PTEN axis. Exp. Cell Res. 2019, 374, 365–373. [Google Scholar] [CrossRef]
  127. Chen, P.Y.; Hsieh, P.L.; Peng, C.Y.; Liao, Y.W.; Yu, C.H.; Yu, C.C. LncRNA MEG3 inhibits self-renewal and invasion abilities of oral cancer stem cells by sponging miR-421. J. Formos. Med. Assoc. = Taiwan Yi Zhi 2021, 120, 1137–1142. [Google Scholar] [CrossRef]
  128. Zhang, L.; Meng, X.; Zhu, X.W.; Yang, D.C.; Chen, R.; Jiang, Y.; Xu, T. Long non-coding RNAs in Oral squamous cell carcinoma: Biologic function, mechanisms and clinical implications. Mol. Cancer 2019, 18, 102. [Google Scholar] [CrossRef]
  129. Zhou, Y.; Zhang, X.; Klibanski, A. MEG3 noncoding RNA: A tumor suppressor. J. Mol. Endocrinol. 2012, 48, R45–R53. [Google Scholar] [CrossRef]
  130. Andresini, O.; Ciotti, A.; Rossi, M.N.; Battistelli, C.; Carbone, M.; Maione, R. A cross-talk between DNA methylation and H3 lysine 9 dimethylation at the KvDMR1 region controls the induction of Cdkn1c in muscle cells. Epigenetics 2016, 11, 791–803. [Google Scholar] [CrossRef]
  131. Wang, M.; Gu, J.; Zhang, X.; Yang, J.; Zhang, X.; Fang, X. Long Non-coding RNA DANCR in Cancer: Roles, Mechanisms, and Implications. Front. Cell Dev. Biol. 2021, 9, 753706. [Google Scholar] [CrossRef]
  132. Sur, S.; Nakanishi, H.; Steele, R.; Ray, R.B. Depletion of PCAT-1 in head and neck cancer cells inhibits tumor growth and induces apoptosis by modulating c-Myc-AKT1-p38 MAPK signalling pathways. BMC Cancer 2019, 19, 354. [Google Scholar] [CrossRef] [PubMed]
  133. Yu, C.; Wang, Y.; Li, G.; She, L.; Zhang, D.; Chen, X.; Zhang, X.; Qin, Z.; Cao, H.; Liu, Y. LncRNA PVT1 promotes malignant progression in squamous cell carcinoma of the head and neck. J. Cancer 2018, 9, 3593–3602. [Google Scholar] [CrossRef] [PubMed]
  134. Arunkumar, G.; Deva Magendhra Rao, A.K.; Manikandan, M.; Arun, K.; Vinothkumar, V.; Revathidevi, S.; Rajkumar, K.S.; Rajaraman, R.; Munirajan, A.K. Expression profiling of long non-coding RNA identifies linc-RoR as a prognostic biomarker in oral cancer. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2017, 39, 1010428317698366. [Google Scholar] [CrossRef]
  135. Qi, C.; Liu, J.; Guo, P.; Xu, Y.; Hu, J.; Han, X. LncRNA NORAD facilitates oral squamous cell carcinoma progression by sponging miR-577 to enhance TPM4. Biol. Direct 2022, 17, 1. [Google Scholar] [CrossRef] [PubMed]
  136. Wu, Y.; Zhang, L.; Zhang, L.; Wang, Y.; Li, H.; Ren, X.; Wei, F.; Yu, W.; Liu, T.; Wang, X.; et al. Long non-coding RNA HOTAIR promotes tumor cell invasion and metastasis by recruiting EZH2 and repressing E-cadherin in oral squamous cell carcinoma. Int. J. Oncol. 2015, 46, 2586–2594. [Google Scholar] [CrossRef]
  137. Shih, J.W.; Chiang, W.F.; Wu, A.T.H.; Wu, M.H.; Wang, L.Y.; Yu, Y.L.; Hung, Y.W.; Wang, W.C.; Chu, C.Y.; Hung, C.L.; et al. Long noncoding RNA LncHIFCAR/MIR31HG is a HIF-1alpha co-activator driving oral cancer progression. Nat. Commun. 2017, 8, 15874. [Google Scholar] [CrossRef]
  138. Fang, Z.; Zhao, J.; Xie, W.; Sun, Q.; Wang, H.; Qiao, B. LncRNA UCA1 promotes proliferation and cisplatin resistance of oral squamous cell carcinoma by sunppressing miR-184 expression. Cancer Med. 2017, 6, 2897–2908. [Google Scholar] [CrossRef]
  139. Tao, B.; Wang, D.; Yang, S.; Liu, Y.; Wu, H.; Li, Z.; Chang, L.; Yang, Z.; Liu, W. Cucurbitacin B Inhibits Cell Proliferation by Regulating X-Inactive Specific Transcript Expression in Tongue Cancer. Front. Oncol. 2021, 11, 651648. [Google Scholar] [CrossRef]
  140. Chai, L.; Yuan, Y.; Chen, C.; Zhou, J.; Wu, Y. The role of long non-coding RNA ANRIL in the carcinogenesis of oral cancer by targeting miR-125a. Biomed. Pharmacother. 2018, 103, 38–45. [Google Scholar] [CrossRef]
  141. Huang, G.; He, X.; Wei, X.L. lncRNA NEAT1 promotes cell proliferation and invasion by regulating miR-365/RGS20 in oral squamous cell carcinoma. Oncol. Rep. 2018, 39, 1948–1956. [Google Scholar] [CrossRef]
  142. Liu, X.; Shang, W.; Zheng, F. Long non-coding RNA NEAT1 promotes migration and invasion of oral squamous cell carcinoma cells by sponging microRNA-365. Exp. Ther. Med. 2018, 16, 2243–2250. [Google Scholar] [CrossRef] [PubMed]
  143. Hatanaka, Y.; Niinuma, T.; Kitajima, H.; Nishiyama, K.; Maruyama, R.; Ishiguro, K.; Toyota, M.; Yamamoto, E.; Kai, M.; Yorozu, A.; et al. DLEU1 promotes oral squamous cell carcinoma progression by activating interferon-stimulated genes. Sci. Rep. 2021, 11, 20438. [Google Scholar] [CrossRef]
  144. Lv, T.; Liu, H.; Wu, Y.; Huang, W. Knockdown of lncRNA DLEU1 inhibits the tumorigenesis of oral squamous cell carcinoma via regulation of miR-149-5p/CDK6 axis. Mol. Med. Rep. 2021, 23, 447. [Google Scholar] [CrossRef] [PubMed]
  145. Shao, T.R.; Zheng, Z.N.; Chen, Y.C.; Wu, Q.Q.; Huang, G.Z.; Li, F.; Zeng, W.S.; Lv, X.Z. LncRNA AC007271.3 promotes cell proliferation, invasion, migration and inhibits cell apoptosis of OSCC via the Wnt/beta-catenin signaling pathway. Life Sci. 2019, 239, 117087. [Google Scholar] [CrossRef]
  146. Zheng, Z.N.; Huang, G.Z.; Wu, Q.Q.; Ye, H.Y.; Zeng, W.S.; Lv, X.Z. NF-kappaB-mediated lncRNA AC007271.3 promotes carcinogenesis of oral squamous cell carcinoma by regulating miR-125b-2-3p/Slug. Cell Death Dis. 2020, 11, 1055. [Google Scholar] [CrossRef] [PubMed]
  147. Li, J.; Xu, X.; Zhang, D.; Lv, H.; Lei, X. LncRNA LHFPL3-AS1 Promotes Oral Squamous Cell Carcinoma Growth and Cisplatin Resistance Through Targeting miR-362-5p/CHSY1 Pathway. OncoTargets Ther. 2021, 14, 2293–2300. [Google Scholar] [CrossRef]
  148. Zhang, Y.; Wang, A.; Zhang, X.; Wang, X.; Zhang, J.; Ma, J. lncRNA LINC01296 Promotes Oral Squamous Cell Carcinoma Development by Binding with SRSF1. BioMed Res. Int. 2021, 2021, 6661520. [Google Scholar] [CrossRef]
  149. Yao, Y.; Chen, S.; Lu, N.; Yin, Y.; Liu, Z. LncRNA JPX overexpressed in oral squamous cell carcinoma drives malignancy via miR-944/CDH2 axis. Oral Dis. 2021, 27, 924–933. [Google Scholar] [CrossRef]
  150. Tian, Y.; Zhong, L.; Gao, S.; Yu, Y.; Sun, D.; Liu, X.; Ji, J.; Yao, Y.; Liu, Y.; Jiang, Z. LncRNA LINC00974 Downregulates miR-122 to Upregulate RhoA in Oral Squamous Cell Carcinoma. Cancer Biother. Radiopharm. 2021, 36, 18–22. [Google Scholar] [CrossRef]
  151. Liu, D.K.; Li, Y.J.; Tian, B.; Sun, H.M.; Li, Q.Y.; Ren, B.F. LncRNA PRNCR1 aggravates the malignancy of oral squamous cell carcinoma by regulating miR-326/FSCN1 axis. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 3226–3234. [Google Scholar] [CrossRef]
  152. Liu, W.; Yao, Y.; Shi, L.; Tang, G.; Wu, L. A novel lncRNA LOLA1 may predict malignant progression and promote migration, invasion, and EMT of oral leukoplakia via the AKT/GSK-3beta pathway. J. Cell. Biochem. 2021, 122, 1302–1312. [Google Scholar] [CrossRef] [PubMed]
  153. Li, H.; Jiang, J. LncRNA MCM3AP-AS1 promotes proliferation, migration and invasion of oral squamous cell carcinoma cells via regulating miR-204-5p/FOXC1. J. Investig. Med. Off. Publ. Am. Fed. Clin. Res. 2020, 68, 1282–1288. [Google Scholar] [CrossRef] [PubMed]
  154. Yao, Y.; Liu, Y.; Jin, F.; Meng, Z. LINC00662 Promotes Oral Squamous Cell Carcinoma Cell Growth and Metastasis through miR-144-3p/EZH2 Axis. Yonsei Med. J. 2021, 62, 640–649. [Google Scholar] [CrossRef] [PubMed]
  155. Xiong, L.; Tang, Y.; Tang, J.; Liu, Z.; Wang, X. Downregulation of lncRNA HOTTIP Suppresses the Proliferation, Migration, and Invasion of Oral Tongue Squamous Cell Carcinoma by Regulation of HMGA2-Mediated Wnt/beta-Catenin Pathway. Cancer Biother. Radiopharm. 2020, 35, 720–730. [Google Scholar] [CrossRef]
  156. Shen, H.; Sun, B.; Yang, Y.; Cai, X.; Bi, L.; Deng, L.; Zhang, L. MIR4435-2HG regulates cancer cell behaviors in oral squamous cell carcinoma cell growth by upregulating TGF-beta1. Odontology 2020, 108, 553–559. [Google Scholar] [CrossRef]
  157. Wang, X.; Gong, Z.; Ma, L.; Wang, Q. LncRNA GACAT1 induces tongue squamous cell carcinoma migration and proliferation via miR-149. J. Cell. Mol. Med. 2021, 25, 8215–8221. [Google Scholar] [CrossRef] [PubMed]
  158. Xia, Y.C.; Cao, J.; Yang, J.; Zhang, Y.; Li, Y.S. lncRNA TSPEAR-AS2, a Novel Prognostic Biomarker, Promotes Oral Squamous Cell Carcinoma Progression by Upregulating PPM1A via Sponging miR-487a-3p. Dis. Mrk. 2021, 2021, 2217663. [Google Scholar] [CrossRef] [PubMed]
  159. Liu, M.; Gao, X.; Liu, C.L. Increased expression of lncRNA FTH1P3 promotes oral squamous cell carcinoma cells migration and invasion by enhancing PI3K/Akt/GSK3b/ Wnt/beta-catenin signaling. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 8306–8314. [Google Scholar] [CrossRef]
  160. Chen, F.; Qi, S.; Zhang, X.; Wu, J.; Yang, X.; Wang, R. lncRNA PLAC2 activated by H3K27 acetylation promotes cell proliferation and invasion via the activation of Wnt/beta-catenin pathway in oral squamous cell carcinoma. Int. J. Oncol. 2019, 54, 1183–1194. [Google Scholar] [CrossRef]
  161. Zhu, X.; Zhang, H.; Xu, J. Long noncoding RNA SNHG20 regulates cell migration, invasion, and proliferation via the microRNA-19b-3p/RAB14 axis in oral squamous cell carcinoma. Bioengineered 2021, 12, 3993–4003. [Google Scholar] [CrossRef]
  162. Chen, Z.F.; Wang, Y.; Sun, L.L.; Ding, S.Y.; Jinag, H. LncRNA SNHG20 enhances the progression of oral squamous cell carcinoma by regulating the miR-29a/DIXDC1/Wnt regulatory axis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 5436–5445. [Google Scholar] [CrossRef] [PubMed]
  163. Du, Y.; Yang, H.; Li, Y.; Guo, W.; Zhang, Y.; Shen, H.; Xing, L.; Li, Y.; Wu, W.; Zhang, X. Long non-coding RNA LINC01137 contributes to oral squamous cell carcinoma development and is negatively regulated by miR-22-3p. Cell. Oncol. 2021, 44, 595–609. [Google Scholar] [CrossRef] [PubMed]
  164. Cao, X.; Luan, K.; Yang, J.; Huang, Y. Targeting lncRNA PSMA3-AS1, a Prognostic Marker, Suppresses Malignant Progression of Oral Squamous Cell Carcinoma. Dis. Mrk. 2021, 2021, 3138046. [Google Scholar] [CrossRef] [PubMed]
  165. Ai, Y.; Liu, S.; Luo, H.; Wu, S.; Wei, H.; Tang, Z.; Li, X.; Zou, C. lncRNA DCST1-AS1 Facilitates Oral Squamous Cell Carcinoma by Promoting M2 Macrophage Polarization through Activating NF-kappaB Signaling. J. Immunol. Res. 2021, 2021, 5524231. [Google Scholar] [CrossRef]
  166. Jiang, L.; Zhou, B.; Fu, D.; Cheng, B. lncRNA TUG1 promotes the development of oral squamous cell carcinoma by regulating the MAPK signaling pathway by sponging miR-593-3p. Cell Cycle 2022, 21, 1856–1866. [Google Scholar] [CrossRef]
  167. Liang, X.; Chen, Z.; Wu, G. FOXD2-AS1 Predicts Dismal Prognosis for Oral Squamous Cell Carcinoma and Regulates Cell Proliferation. Cell Transplant. 2020, 29, 963689720964411. [Google Scholar] [CrossRef]
  168. Huang, W.; Cao, J.; Peng, X. LINC01234 facilitates growth and invasiveness of oral squamous cell carcinoma through regulating the miR-637/NUPR1 axis. Biomed. Pharmacother. 2019, 120, 109507. [Google Scholar] [CrossRef]
  169. Liu, D.; Jian, X.; Xu, P.; Zhu, R.; Wang, Y. Linc01234 promotes cell proliferation and metastasis in oral squamous cell carcinoma via miR-433/PAK4 axis. BMC Cancer 2020, 20, 107. [Google Scholar] [CrossRef]
  170. Lu, X.; Chen, L.; Li, Y.; Huang, R.; Meng, X.; Sun, F. Long non-coding RNA LINC01207 promotes cell proliferation and migration but suppresses apoptosis and autophagy in oral squamous cell carcinoma by the microRNA-1301-3p/lactate dehydrogenase isoform A axis. Bioengineered 2021, 12, 7780–7793. [Google Scholar] [CrossRef]
  171. Fu, S.W.; Zhang, Y.; Li, S.; Shi, Z.Y.; Zhao, J.; He, Q.L. LncRNA TTN-AS1 promotes the progression of oral squamous cell carcinoma via miR-411-3p/NFAT5 axis. Cancer Cell Int. 2020, 20, 415. [Google Scholar] [CrossRef]
  172. Wang, X.; Guo, Y.; Wang, C.; Wang, Q.; Yan, G. Long Noncoding RNA ZEB1-AS1 Downregulates miR-23a, Promotes Tumor Progression, and Predicts the Survival of Oral Squamous Cell Carcinoma Patients. OncoTargets Ther. 2021, 14, 2699–2710. [Google Scholar] [CrossRef] [PubMed]
  173. Hong, Y.; He, H.; Sui, W.; Zhang, J.; Zhang, S.; Yang, D. Long non-coding RNA H1 promotes cell proliferation and invasion by acting as a ceRNA of miR-138 and releasing EZH2 in oral squamous cell carcinoma. Int. J. Oncol. 2018, 52, 901–912. [Google Scholar] [CrossRef]
  174. Zhao, J.; Bai, X.; Feng, C.; Shang, X.; Xi, Y. Long Non-Coding RNA HCP5 Facilitates Cell Invasion And Epithelial-Mesenchymal Transition In Oral Squamous Cell Carcinoma By miR-140-5p/SOX4 Axis. Cancer Manag. Res. 2019, 11, 10455–10462. [Google Scholar] [CrossRef] [PubMed]
  175. Zhang, T.; Liu, X.; Su, K.; Zheng, Q.; Liu, P.; Xu, Z.; Zhang, Y. A novel mechanism of the lncRNA PTTG3P/miR-142-5p/JAG1 axis modulating tongue cancer cell phenotypes through the Notch1 signaling. Cells Dev. 2022, 169, 203762. [Google Scholar] [CrossRef] [PubMed]
  176. Tong, S.; Wang, X.; Guo, X.; Lu, Z. Knockdown of lncRNA IGF2BP2-AS1 inhibits proliferation and migration of oral squamous cell carcinoma cells via the Wnt/beta-catenin pathway. J. Oral Pathol. Med. 2022, 51, 272–280. [Google Scholar] [CrossRef] [PubMed]
  177. Xie, S.; Yu, X.; Li, Y.; Ma, H.; Fan, S.; Chen, W.; Pan, G.; Wang, W.; Zhang, H.; Li, J.; et al. Upregulation of lncRNA ADAMTS9-AS2 Promotes Salivary Adenoid Cystic Carcinoma Metastasis via PI3K/Akt and MEK/Erk Signaling. Mol. Ther. 2018, 26, 2766–2778. [Google Scholar] [CrossRef]
  178. Liu, M.; Liu, Q.; Fan, S.; Su, F.; Jiang, C.; Cai, G.; Wang, Y.; Liao, G.; Lei, X.; Chen, W.; et al. LncRNA LTSCCAT promotes tongue squamous cell carcinoma metastasis via targeting the miR-103a-2-5p/SMYD3/TWIST1 axis. Cell Death Dis. 2021, 12, 144. [Google Scholar] [CrossRef]
  179. Shao, B.; Fu, X.; Li, X.; Li, Y.; Gan, N. RP11-284F21.9 promotes oral squamous cell carcinoma development via the miR-383-5p/MAL2 axis. J. Oral Pathol. Med. 2020, 49, 21–29. [Google Scholar] [CrossRef]
  180. Li, J.; Li, Z.; Wu, Y.; Diao, P.; Zhang, W.; Wang, Y.; Yang, J.; Cheng, J. Overexpression of lncRNA WWTR1-AS1 associates with tumor aggressiveness and unfavorable survival in head-neck squamous cell carcinoma. J. Cell. Biochem. 2019, 120, 18266–18277. [Google Scholar] [CrossRef]
  181. Zhang, C.Z. Long intergenic non-coding RNA 668 regulates VEGFA signaling through inhibition of miR-297 in oral squamous cell carcinoma. Biochem. Biophys. Res. Commun. 2017, 489, 404–412. [Google Scholar] [CrossRef]
  182. Liu, Z.; Li, H.; Fan, S.; Lin, H.; Lian, W. STAT3-induced upregulation of long noncoding RNA HNF1A-AS1 promotes the progression of oral squamous cell carcinoma via activating Notch signaling pathway. Cancer Biol. Ther. 2019, 20, 444–453. [Google Scholar] [CrossRef]
  183. Lyu, Q.; Jin, L.; Yang, X.; Zhang, F. LncRNA MINCR activates Wnt/beta-catenin signals to promote cell proliferation and migration in oral squamous cell carcinoma. Pathol. Res. Pract. 2019, 215, 924–930. [Google Scholar] [CrossRef] [PubMed]
  184. Wang, J.; Huo, F.; Wang, X.R.; Xu, Y.Y. LncRNA LACAT1 promotes proliferation of oral squamous cell carcinoma cells by inhibiting microRNA-4301. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 2427–2435. [Google Scholar] [CrossRef] [PubMed]
  185. Yang, Y.; Chen, D.; Liu, H.; Yang, K. Increased expression of lncRNA CASC9 promotes tumor progression by suppressing autophagy-mediated cell apoptosis via the AKT/mTOR pathway in oral squamous cell carcinoma. Cell Death Dis. 2019, 10, 41. [Google Scholar] [CrossRef] [PubMed]
  186. Jiang, Q.; Wang, Z.; Qi, Q.; Li, J.; Xin, Y.; Qiu, J. lncRNA SNHG26 promoted the growth, metastasis, and cisplatin resistance of tongue squamous cell carcinoma through PGK1/Akt/mTOR signal pathway. Mol. Ther. Oncolytics 2022, 24, 355–370. [Google Scholar] [CrossRef]
  187. Yu, Q.; Du, Y.; Wang, S.; Zheng, X. LncRNA PART1 promotes cell proliferation and inhibits apoptosis of oral squamous cell carcinoma by blocking EZH2 degradation. J. Biochem. 2021, 169, 721–730. [Google Scholar] [CrossRef]
  188. Yu, J.; Liu, Y.; Guo, C.; Zhang, S.; Gong, Z.; Tang, Y.; Yang, L.; He, Y.; Lian, Y.; Li, X.; et al. Upregulated long non-coding RNA LINC00152 expression is associated with progression and poor prognosis of tongue squamous cell carcinoma. J. Cancer 2017, 8, 523–530. [Google Scholar] [CrossRef]
  189. Chen, R.; Wang, X.; Zhou, S.; Zeng, Z. LncRNA HOXA-AS2 Promotes Tumor Progression by Suppressing miR-567 Expression in Oral Squamous Cell Carcinoma. Cancer Manag. Res. 2021, 13, 5443–5455. [Google Scholar] [CrossRef]
  190. Fang, X.; Tang, Z.; Zhang, H.; Quan, H. Long non-coding RNA DNM3OS/miR-204-5p/HIP1 axis modulates oral cancer cell viability and migration. J. Oral Pathol. Med. 2020, 49, 865–875. [Google Scholar] [CrossRef]
  191. Liu, J.; Zhao, C.; Yang, S.; Dong, C. LncRNA SNHG1 promotes the development of oral cavity cancer via regulating the miR-421/HMGB2 axis. Cell. Mol. Biol. 2020, 66, 14–19. [Google Scholar] [CrossRef]
  192. Chen, Y.T.; Kan, C.H.; Liu, H.; Liu, Y.H.; Wu, C.C.; Kuo, Y.P.; Chang, I.Y.; Chang, K.P.; Yu, J.S.; Tan, B.C. Modular scaffolding by lncRNA HOXA10-AS promotes oral cancer progression. Cell Death Dis. 2022, 13, 629. [Google Scholar] [CrossRef] [PubMed]
  193. Zhao, C.; Shi, W.; Chen, M. Long non-coding RNA BBOX1-antisense RNA 1 enhances cell proliferation and migration and suppresses apoptosis in oral squamous cell carcinoma via the miR-3940-3p/laminin subunit gamma 2 axis. Bioengineered 2022, 13, 11138–11153. [Google Scholar] [CrossRef] [PubMed]
  194. Shi, L.; Yang, Y.; Li, M.; Li, C.; Zhou, Z.; Tang, G.; Wu, L.; Yao, Y.; Shen, X.; Hou, Z.; et al. LncRNA IFITM4P promotes immune escape by up-regulating PD-L1 via dual mechanism in oral carcinogenesis. Mol. Ther. 2022, 30, 1564–1577. [Google Scholar] [CrossRef]
  195. Zhang, X.; Guo, B.; Zhu, Y.; Xu, W.; Ning, S.; Liu, L. Up-regulation of plasma lncRNA CACS15 distinguished early-stage oral squamous cell carcinoma patient. Oral Dis. 2020, 26, 1619–1624. [Google Scholar] [CrossRef] [PubMed]
  196. Lee, S.P.; Hsieh, P.L.; Fang, C.Y.; Chu, P.M.; Liao, Y.W.; Yu, C.H.; Yu, C.C.; Tsai, L.L. LINC00963 Promotes Cancer Stemness, Metastasis, and Drug Resistance in Head and Neck Carcinomas via ABCB5 Regulation. Cancers 2020, 12, 1073. [Google Scholar] [CrossRef]
  197. Arunkumar, G.; Anand, S.; Raksha, P.; Dhamodharan, S.; Prasanna Srinivasa Rao, H.; Subbiah, S.; Murugan, A.K.; Munirajan, A.K. LncRNA OIP5-AS1 is overexpressed in undifferentiated oral tumors and integrated analysis identifies as a downstream effector of stemness-associated transcription factors. Sci. Rep. 2018, 8, 7018. [Google Scholar] [CrossRef]
  198. Qiao, C.Y.; Qiao, T.Y.; Jin, H.; Liu, L.L.; Zheng, M.D.; Wang, Z.L. LncRNA KCNQ1OT1 contributes to the cisplatin resistance of tongue cancer through the KCNQ1OT1/miR-124-3p/TRIM14 axis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 200–212. [Google Scholar] [CrossRef]
  199. Dai, D.; Feng, X.D.; Zhu, W.Q.; Bao, Y.N. LncRNA BLACAT1 regulates the viability, migration and invasion of oral squamous cell carcinoma cells by targeting miR-142-5p. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10313–10323. [Google Scholar] [CrossRef]
  200. Li, T.; Liu, D.; Li, C.; Ru, L.; Wang, X. Silencing of LncRNA AFAP1-AS1 Inhibits Cell Proliferation in Oral Squamous Cancer by Suppressing CCNA2. Cancer Manag. Res. 2021, 13, 7897–7908. [Google Scholar] [CrossRef]
  201. Ye, J.; Jiao, Y. LncRNA FAL1 promotes the development of oral squamous cell carcinoma through regulating the microRNA-761/CRKL pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5779–5786. [Google Scholar] [CrossRef]
  202. Wang, X.; Li, H.; Shi, J. LncRNA HOXA11-AS Promotes Proliferation and Cisplatin Resistance of Oral Squamous Cell Carcinoma by Suppression of miR-214-3p Expression. BioMed Res. Int. 2019, 2019, 8645153. [Google Scholar] [CrossRef] [PubMed]
  203. Xu, L.; Hou, T.J.; Yang, P. Mechanism of lncRNA FEZF1-AS1 in promoting the occurrence and development of oral squamous cell carcinoma through targeting miR-196a. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6505–6515. [Google Scholar] [CrossRef] [PubMed]
  204. Zhao, Y.; Wang, J.; Ma, K. [Knockdown of lncRNA SNHG6 inhibites the proliferation and epithelial mesenchymal transition in tongue cancer cells]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi = Chin. J. Cell. Mol. Immunol. 2018, 34, 806–811. [Google Scholar]
  205. Li, W.; Zhu, Q.; Zhang, S.; Liu, L.; Zhang, H.; Zhu, D. HOXC13-AS accelerates cell proliferation and migration in oral squamous cell carcinoma via miR-378g/HOXC13 axis. Oral Oncol. 2020, 111, 104946. [Google Scholar] [CrossRef]
  206. Luo, X.; Jiang, Y.; Chen, F.; Wei, Z.; Qiu, Y.; Xu, H.; Tian, G.; Gong, W.; Yuan, Y.; Feng, H.; et al. ORAOV1-B Promotes OSCC Metastasis via the NF-kappaB-TNFalpha Loop. J. Dent. Res. 2021, 100, 858–867. [Google Scholar] [CrossRef]
  207. Jiang, X.; Liu, J.; Li, S.; Jia, B.; Huang, Z.; Shen, J.; Luo, H.; Zhao, J. CCL18-induced LINC00319 promotes proliferation and metastasis in oral squamous cell carcinoma via the miR-199a-5p/FZD4 axis. Cell Death Dis. 2020, 11, 777. [Google Scholar] [CrossRef]
  208. Feng, H.; Zhang, X.; Lai, W.; Wang, J. Long non-coding RNA SLC16A1-AS1: Its multiple tumorigenesis features and regulatory role in cell cycle in oral squamous cell carcinoma. Cell Cycle 2020, 19, 1641–1653. [Google Scholar] [CrossRef]
  209. Chen, Y.; Guo, Y.; Yan, W. lncRNA RP5-916L7.2 correlates with advanced tumor stage, and promotes cells proliferation while inhibits cells apoptosis through targeting miR-328 and miR-939 in tongue squamous cell carcinoma. Clin. Biochem. 2019, 67, 24–32. [Google Scholar] [CrossRef]
  210. Yan, D.; Wu, F.; Peng, C.; Wang, M. Silencing of LINC00284 inhibits cell proliferation and migration in oral squamous cell carcinoma by the miR-211-3p/MAFG axis and FUS/KAZN axis. Cancer Biol. Ther. 2021, 22, 149–163. [Google Scholar] [CrossRef]
  211. Chen, F.; Liu, M.; Yu, Y.; Sun, Y.; Li, J.; Hu, W.; Wang, X.; Tong, D. LINC00958 regulated miR-627-5p/YBX2 axis to facilitate cell proliferation and migration in oral squamous cell carcinoma. Cancer Biol. Ther. 2019, 20, 1270–1280. [Google Scholar] [CrossRef]
  212. Zhang, C.; Bao, C.; Zhang, X.; Lin, X.; Pan, D.; Chen, Y. Knockdown of lncRNA LEF1-AS1 inhibited the progression of oral squamous cell carcinoma (OSCC) via Hippo signaling pathway. Cancer Biol. Ther. 2019, 20, 1213–1222. [Google Scholar] [CrossRef] [PubMed]
  213. Chen, C.; Chen, Q.; Wu, J.; Zou, H. H3K27ac-induced FOXC2-AS1 accelerates tongue squamous cell carcinoma by upregulating E2F3. J. Oral Pathol. Med. 2021, 50, 1018–1030. [Google Scholar] [CrossRef] [PubMed]
  214. Ying, Y.; Liu, D.; Zhao, Y.; Zhong, Y.; Xu, X.; Luo, J.; Zhang, Z. LINC01116 Promotes Migration and Invasion of Oral Squamous Cell Carcinoma by Acting as a Competed Endogenous RNA in Regulation of MMP1 Expression. Comput. Math. Methods Med. 2022, 2022, 2857022. [Google Scholar] [CrossRef]
  215. Jin, Z.; Jiang, S.; Jian, S.; Shang, Z. Long noncoding RNA MORT overexpression inhibits cancer cell proliferation in oral squamous cell carcinoma by downregulating ROCK1. J. Cell. Biochem. 2019, 120, 11702–11707. [Google Scholar] [CrossRef]
  216. Hu, X.; Qiu, Z.; Zeng, J.; Xiao, T.; Ke, Z.; Lyu, H. A novel long non-coding RNA, AC012456.4, as a valuable and independent prognostic biomarker of survival in oral squamous cell carcinoma. PeerJ 2018, 6, e5307. [Google Scholar] [CrossRef]
  217. Wu, J.; Li, Y.; Liu, J.; Xu, Y. Down-regulation of lncRNA HCG11 promotes cell proliferation of oral squamous cell carcinoma through sponging miR-455-5p. J. Gene Med. 2021, 23, e3293. [Google Scholar] [CrossRef] [PubMed]
  218. Yang, L.; Yang, K.; Wang, H.; Chen, Y.; Zhao, J. LncRNA SCIRT absorbs miR-221 to advance the expression of lncRNA GAS5 in oral squamous cell carcinoma to inhibit cancer cell apoptosis. J. Oral Pathol. Med. 2022, 51, 256–262. [Google Scholar] [CrossRef]
  219. Lu, T.; Liu, H.; You, G. Long non-coding RNA C5orf66-AS1 prevents oral squamous cell carcinoma through inhibiting cell growth and metastasis. Int. J. Mol. Med. 2018, 42, 3291–3299. [Google Scholar] [CrossRef]
  220. Zhang, H.; Wang, J.; Xun, W.; Wang, J.; Song, W.; Wang, X. Long non-coding RNA PTCSC3 inhibits human oral cancer cell proliferation by inducing apoptosis and autophagy. Arch. Med. Sci. 2021, 17, 492–499. [Google Scholar] [CrossRef]
  221. Xu, Y.; Jiang, E.; Shao, Z.; Shang, Z. LncRNA FENDRR in Carcinoma-Associated Fibroblasts Regulates the Angiogenesis of Oral Squamous Cell Carcinoma Through the PI3K/AKT Pathway. Front. Oncol. 2021, 11, 616576. [Google Scholar] [CrossRef]
  222. Wang, M.; Yu, F.; Wu, W.; Zhang, Y.; Chang, W.; Ponnusamy, M.; Wang, K.; Li, P. Circular RNAs: A novel type of non-coding RNA and their potential implications in antiviral immunity. Int. J. Biol. Sci. 2017, 13, 1497–1506. [Google Scholar] [CrossRef] [PubMed]
  223. Jeck, W.R.; Sharpless, N.E. Detecting and characterizing circular RNAs. Nat. Biotechnol. 2014, 32, 453–461. [Google Scholar] [CrossRef] [PubMed]
  224. Chen, L.L.; Yang, L. Regulation of circRNA biogenesis. RNA Biol. 2015, 12, 381–388. [Google Scholar] [CrossRef] [PubMed]
  225. Geng, X.; Jia, Y.; Zhang, Y.; Shi, L.; Li, Q.; Zang, A.; Wang, H. Circular RNA: Biogenesis, degradation, functions and potential roles in mediating resistance to anticarcinogens. Epigenomics 2020, 12, 267–283. [Google Scholar] [CrossRef]
  226. Yu, T.; Wang, Y.; Fan, Y.; Fang, N.; Wang, T.; Xu, T.; Shu, Y. CircRNAs in cancer metabolism: A review. J. Hematol. Oncol. 2019, 12, 90. [Google Scholar] [CrossRef] [PubMed]
  227. Zeng, Y.; Zou, Y.; Gao, G.; Zheng, S.; Wu, S.; Xie, X.; Tang, H. The biogenesis, function and clinical significance of circular RNAs in breast cancer. Cancer Biol. Med. 2021, 19, 14–29. [Google Scholar] [CrossRef]
  228. Zhang, Q.; Wang, W.; Zhou, Q.; Chen, C.; Yuan, W.; Liu, J.; Li, X.; Sun, Z. Roles of circRNAs in the tumour microenvironment. Mol. Cancer 2020, 19, 14. [Google Scholar] [CrossRef]
  229. Fan, H.Y.; Jiang, J.; Tang, Y.J.; Liang, X.H.; Tang, Y.L. CircRNAs: A New Chapter in Oral Squamous Cell Carcinoma Biology. OncoTargets Ther. 2020, 13, 9071–9083. [Google Scholar] [CrossRef]
  230. Chen, X.; Yu, J.; Tian, H.; Shan, Z.; Liu, W.; Pan, Z.; Ren, J. Circle RNA hsa_circRNA_100290 serves as a ceRNA for miR-378a to regulate oral squamous cell carcinoma cells growth via Glucose transporter-1 (GLUT1) and glycolysis. J. Cell. Physiol. 2019, 234, 19130–19140. [Google Scholar] [CrossRef]
  231. He, T.; Li, X.; Xie, D.; Tian, L. Overexpressed circPVT1 in oral squamous cell carcinoma promotes proliferation by serving as a miRNA sponge. Mol. Med. Rep. 2019, 20, 3509–3518. [Google Scholar] [CrossRef]
  232. Gao, L.; Dou, Z.C.; Ren, W.H.; Li, S.M.; Liang, X.; Zhi, K.Q. CircCDR1as upregulates autophagy under hypoxia to promote tumor cell survival via AKT/ERK((1/2))/mTOR signaling pathways in oral squamous cell carcinomas. Cell Death Dis. 2019, 10, 745. [Google Scholar] [CrossRef] [PubMed]
  233. Su, W.; Wang, Y.; Wang, F.; Zhang, B.; Zhang, H.; Shen, Y.; Yang, H. Circular RNA hsa_circ_0007059 indicates prognosis and influences malignant behavior via AKT/mTOR in oral squamous cell carcinoma. J. Cell. Physiol. 2019, 234, 15156–15166. [Google Scholar] [CrossRef] [PubMed]
  234. Su, W.; Wang, Y.; Wang, F.; Sun, S.; Li, M.; Shen, Y.; Yang, H. Hsa_circ_0005379 regulates malignant behavior of oral squamous cell carcinoma through the EGFR pathway. BMC Cancer 2019, 19, 400. [Google Scholar] [CrossRef] [PubMed]
  235. Papatsirou, M.; Artemaki, P.I.; Karousi, P.; Scorilas, A.; Kontos, C.K. Circular RNAs: Emerging Regulators of the Major Signaling Pathways Involved in Cancer Progression. Cancers 2021, 13, 2744. [Google Scholar] [CrossRef] [PubMed]
  236. Gao, L.; Zhao, C.; Li, S.; Dou, Z.; Wang, Q.; Liu, J.; Ren, W.; Zhi, K. circ-PKD2 inhibits carcinogenesis via the miR-204-3p/APC2 axis in oral squamous cell carcinoma. Mol. Carcinog. 2019, 58, 1783–1794. [Google Scholar] [CrossRef]
  237. Zhao, W.; Cui, Y.; Liu, L.; Qi, X.; Liu, J.; Ma, S.; Hu, X.; Zhang, Z.; Wang, Y.; Li, H.; et al. Splicing factor derived circular RNA circUHRF1 accelerates oral squamous cell carcinoma tumorigenesis via feedback loop. Cell Death Differ. 2020, 27, 919–933. [Google Scholar] [CrossRef]
  238. Hu, Y.T.; Li, X.X.; Zeng, L.W. Circ_0001742 promotes tongue squamous cell carcinoma progression via miR-431-5p/ATF3 axis. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10300–10312. [Google Scholar] [CrossRef]
  239. Tan, X.; Zhou, C.; Liang, Y.; Lai, Y.F.; Liang, Y. Circ_0001971 regulates oral squamous cell carcinoma progression and chemosensitivity by targeting miR-194/miR-204 in vitro and in vivo. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 2470–2481. [Google Scholar] [CrossRef]
  240. Wang, L.; Wei, Y.; Yan, Y.; Wang, H.; Yang, J.; Zheng, Z.; Zha, J.; Bo, P.; Tang, Y.; Guo, X.; et al. CircDOCK1 suppresses cell apoptosis via inhibition of miR-196a-5p by targeting BIRC3 in OSCC. Oncol. Rep. 2018, 39, 951–966. [Google Scholar] [CrossRef]
  241. Wang, J.X.; Liu, Y.; Jia, X.J.; Liu, S.X.; Dong, J.H.; Ren, X.M.; Xu, O.; Zhang, H.Z.; Duan, H.J.; Shan, C.G. Upregulation of circFLNA contributes to laryngeal squamous cell carcinoma migration by circFLNA-miR-486-3p-FLNA axis. Cancer Cell Int. 2019, 19, 196. [Google Scholar] [CrossRef]
  242. Zou, C.; Li, X.; Wei, H.; Wu, S.; Song, J.; Tang, Z.; Luo, H.; Lv, X.; Ai, Y. Circular GOLPH3 RNA exerts oncogenic effects in vitro by regulating the miRNA-1299/LIF axis in oral squamous cell carcinoma. Bioengineered 2022, 13, 11012–11025. [Google Scholar] [CrossRef] [PubMed]
  243. Yu, H.; Yu, Z.; Wang, X.; Wang, D. Circular RNA circCLK3 promotes the progression of tongue squamous cell carcinoma via miR-455-5p/PARVA axis. Biotechnol. Appl. Biochem. 2022, 69, 431–441. [Google Scholar] [CrossRef] [PubMed]
  244. Chen, X.; Chen, J.; Fu, L.; Chen, J.; Chen, Y.; Liu, F. Circ_0014359 promotes oral squamous cell carcinoma progression by sponging miR-149. Acta biochimica Polonica 2022, 69, 315–320. [Google Scholar] [CrossRef] [PubMed]
  245. Li, L.; Yin, Y.; Nan, F.; Ma, Z. Circ_LPAR3 promotes the progression of oral squamous cell carcinoma (OSCC). Biochem. Biophys. Res. Commun. 2022, 589, 215–222. [Google Scholar] [CrossRef]
  246. Ai, Y.; Tang, Z.; Zou, C.; Wei, H.; Wu, S.; Huang, D. circ_SEPT9, a newly identified circular RNA, promotes oral squamous cell carcinoma progression through miR-1225/PKN2 axis. J. Cell. Mol. Med. 2020, 24, 13266–13277. [Google Scholar] [CrossRef]
  247. Luo, Y.; Liu, F.; Guo, J.; Gui, R. Upregulation of circ_0000199 in circulating exosomes is associated with survival outcome in OSCC. Sci. Rep. 2020, 10, 13739. [Google Scholar] [CrossRef]
  248. Chen, F.; Lao, Z.; Zhang, H.; Wang, J.; Wang, S. Knockdown of circ_0001883 may inhibit epithelial-mesenchymal transition in laryngeal squamous cell carcinoma via the miR-125-5p/PI3K/AKT axis. Exp. Ther. Med. 2021, 22, 1007. [Google Scholar] [CrossRef]
  249. Zheng, X.; Du, F.; Gong, X.; Xu, P. Circ_0005320 promotes oral squamous cell carcinoma tumorigenesis by sponging microRNA-486-3p and microRNA-637. Bioengineered 2022, 13, 440–454. [Google Scholar] [CrossRef]
  250. Zhao, S.Y.; Wang, J.; Ouyang, S.B.; Huang, Z.K.; Liao, L. Salivary Circular RNAs Hsa_Circ_0001874 and Hsa_Circ_0001971 as Novel Biomarkers for the Diagnosis of Oral Squamous Cell Carcinoma. Cell. Physiol. Biochem. 2018, 47, 2511–2521. [Google Scholar] [CrossRef]
  251. Zhou, Y.; Zhang, S.; Min, Z.; Yu, Z.; Zhang, H.; Jiao, J. Knockdown of circ_0011946 targets miR-216a-5p/BCL2L2 axis to regulate proliferation, migration, invasion and apoptosis of oral squamous cell carcinoma cells. BMC Cancer 2021, 21, 1085. [Google Scholar] [CrossRef]
  252. Ai, Y.; Song, J.; Wei, H.; Tang, Z.; Li, X.; Lv, X.; Luo, H.; Wu, S.; Zou, C. circ_0001461 promotes oral squamous cell carcinoma progression through miR-145/TLR4/NF-kappaB axis. Biochem. Biophys. Res. Commun. 2021, 566, 108–114. [Google Scholar] [CrossRef] [PubMed]
  253. Wu, Z.; He, X.; Chen, S. Oncogenic circDHTKD1 promotes tumor growth and metastasis of oral squamous cell carcinoma in vitro and in vivo via upregulating miR-326-mediated GAB1. Braz. J. Med. Biol. Res. = Rev. Bras. De Pesqui. Med. E Biol. 2021, 54, e10837. [Google Scholar] [CrossRef] [PubMed]
  254. Liu, J.; Jiang, X.; Zou, A.; Mai, Z.; Huang, Z.; Sun, L.; Zhao, J. circIGHG-Induced Epithelial-to-Mesenchymal Transition Promotes Oral Squamous Cell Carcinoma Progression via miR-142-5p/IGF2BP3 Signaling. Cancer Res. 2021, 81, 344–355. [Google Scholar] [CrossRef] [PubMed]
  255. Yang, Y.; Ci, H.S.; Mao, Y.L.; Li, J.W.; Zuo, J.H. CircRNA_002178 promotes the proliferation and migration of oral squamous cell carcinoma cells by activating the Akt/mTOR pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 6122–6130. [Google Scholar] [CrossRef] [PubMed]
  256. Chen, H.; Zhang, Y.; Wu, K.; Qiu, X. circVAPA promotes the proliferation, migration and invasion of oral cancer cells through the miR-132/HOXA7 axis. J. Int. Med. Res. 2021, 49, 3000605211013207. [Google Scholar] [CrossRef] [PubMed]
  257. Zhang, Q.; Jiang, C.; Ren, W.; Li, S.; Zheng, J.; Gao, Y.; Zhi, K.; Gao, L. Circ-LRP6 mediates epithelial-mesenchymal transition and autophagy in oral squamous cell carcinomas. J. Oral Pathol. Med. 2021, 50, 660–667. [Google Scholar] [CrossRef]
  258. Peng, Q.S.; Cheng, Y.N.; Zhang, W.B.; Fan, H.; Mao, Q.H.; Xu, P. circRNA_0000140 suppresses oral squamous cell carcinoma growth and metastasis by targeting miR-31 to inhibit Hippo signaling pathway. Cell Death Dis. 2020, 11, 112. [Google Scholar] [CrossRef]
  259. Li, X.; Zhang, H.; Wang, Y.; Sun, S.; Shen, Y.; Yang, H. Silencing circular RNA hsa_circ_0004491 promotes metastasis of oral squamous cell carcinoma. Life Sci. 2019, 239, 116883. [Google Scholar] [CrossRef]
  260. Fan, X.; Wang, Y. Circular RNA circSPATA6 Inhibits the Progression of Oral Squamous Cell Carcinoma Cells by Regulating TRAF6 via miR-182. Cancer Manag. Res. 2021, 13, 1817–1829. [Google Scholar] [CrossRef]
  261. Li, L.; Zhang, Z.T. Hsa_circ_0086414 Might Be a Diagnostic Biomarker of Oral Squamous Cell Carcinoma. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2020, 26, e919383. [Google Scholar] [CrossRef]
  262. Li, B.; Wang, F.; Li, X.; Sun, S.; Shen, Y.; Yang, H. Hsa_circ_0008309 May Be a Potential Biomarker for Oral Squamous Cell Carcinoma. Dis. Mrk. 2018, 2018, 7496890. [Google Scholar] [CrossRef] [PubMed]
  263. Shi, D.; Li, H.; Zhang, J.; Li, Y. CircGDI2 Regulates the Proliferation, Migration, Invasion and Apoptosis of OSCC via miR-454-3p/FOXF2 Axis. Cancer Manag. Res. 2021, 13, 1371–1382. [Google Scholar] [CrossRef] [PubMed]
  264. Dong, W.; Zhao, L.; Zhang, S.; Zhang, S.; Si, H. Circ-KIAA0907 inhibits the progression of oral squamous cell carcinoma by regulating the miR-96-5p/UNC13C axis. World J. Surg. Oncol. 2021, 19, 75. [Google Scholar] [CrossRef] [PubMed]
  265. Dai, Y.; Zhu, Y.; Xu, H. circ_0004872 inhibits proliferation, invasion, and glycolysis of oral squamous cell carcinoma by sponged miR-424-5p. J. Clin. Lab. Anal. 2022, 36, e24486. [Google Scholar] [CrossRef] [PubMed]
  266. Gao, L.; Wang, Q.B.; Zhi, Y.; Ren, W.H.; Li, S.M.; Zhao, C.Y.; Xing, X.M.; Dou, Z.C.; Liu, J.C.; Jiang, C.M.; et al. Down-regulation of hsa_circ_0092125 is related to the occurrence and development of oral squamous cell carcinoma. Int. J. Oral Maxillofac. Surg. 2020, 49, 292–297. [Google Scholar] [CrossRef]
  267. Ando, T.; Kasamatsu, A.; Kawasaki, K.; Hiroshima, K.; Fukushima, R.; Iyoda, M.; Nakashima, D.; Endo-Sakamoto, Y.; Uzawa, K. Tumor Suppressive Circular RNA-102450: Development of a Novel Diagnostic Procedure for Lymph Node Metastasis from Oral Cancer. Cancers 2021, 13, 5708. [Google Scholar] [CrossRef]
  268. Han, L.; Cheng, J.; Li, A. hsa_circ_0072387 Suppresses Proliferation, Metastasis, and Glycolysis of Oral Squamous Cell Carcinoma Cells by Downregulating miR-503-5p. Cancer Biother. Radiopharm. 2021, 36, 84–94. [Google Scholar] [CrossRef]
  269. Dragon, F.; Lemay, V.; Trahan, C. snoRNAs: Biogenesis, Structure and Function. In Encyclopedia of Life Sciences; Wiley: Hoboken, NJ, USA, 2006. [Google Scholar]
  270. Chamorro-Petronacci, C.; Perez-Sayans, M.; Padin-Iruegas, M.E.; Marichalar-Mendia, X.; Gallas-Torreira, M.; Garcia Garcia, A. Differential expression of snoRNAs in oral squamous cell carcinomas: New potential diagnostic markers. J. Enzym. Inhib. Med. Chem. 2018, 33, 424–427. [Google Scholar] [CrossRef]
  271. Xing, L.; Zhang, X.; Zhang, X.; Tong, D. Expression scoring of a small-nucleolar-RNA signature identified by machine learning serves as a prognostic predictor for head and neck cancer. J. Cell. Physiol. 2020, 235, 8071–8084. [Google Scholar] [CrossRef]
  272. Liu, Z.; Tao, H. Small nucleolar RNA host gene 3 facilitates cell proliferation and migration in oral squamous cell carcinoma via targeting nuclear transcription factor Y subunit gamma. J. Cell. Biochem. 2020, 121, 2150–2158. [Google Scholar] [CrossRef]
  273. Lu, N.; Yin, Y.; Yao, Y.; Zhang, P. SNHG3/miR-2682-5p/HOXB8 promotes cell proliferation and migration in oral squamous cell carcinoma. Oral Dis. 2021, 27, 1161–1170. [Google Scholar] [CrossRef] [PubMed]
  274. Wang, T.; Liang, D.; Yang, H. SNHG15 facilitated malignant behaviors of oral squamous cell carcinoma through targeting miR-188-5p/DAAM1. J. Oral Pathol. Med. 2021, 50, 681–691. [Google Scholar] [CrossRef] [PubMed]
  275. Jia, Y.; Chen, X.; Zhao, D.; Ma, S. SNHG1/miR-194-5p/MTFR1 Axis Promotes TGFbeta1-Induced EMT, Migration and Invasion of Tongue Squamous Cell Carcinoma Cells. Mol. Biotechnol. 2022, 64, 780–790. [Google Scholar] [CrossRef]
  276. Li, S.; Zhang, S.; Chen, J. c-Myc induced upregulation of long non-coding RNA SNHG16 enhances progression and carcinogenesis in oral squamous cell carcinoma. Cancer Gene Ther. 2019, 26, 400–410. [Google Scholar] [CrossRef] [PubMed]
  277. Tong, F.; Guo, J.; Miao, Z.; Li, Z. LncRNA SNHG17 promotes the progression of oral squamous cell carcinoma by modulating miR-375/PAX6 axis. Cancer Biomark. 2021, 30, 1–12. [Google Scholar] [CrossRef]
  278. Liu, P.; Dong, Y.; Gu, J.; Puthiyakunnon, S.; Wu, Y.; Chen, X.G. Developmental piRNA profiles of the invasive vector mosquito Aedes albopictus. Parasites Vectors 2016, 9, 524. [Google Scholar] [CrossRef]
  279. Girard, A.; Sachidanandam, R.; Hannon, G.J.; Carmell, M.A. A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature 2006, 442, 199–202. [Google Scholar] [CrossRef]
  280. Han, Y.N.; Li, Y.; Xia, S.Q.; Zhang, Y.Y.; Zheng, J.H.; Li, W. PIWI Proteins and PIWI-Interacting RNA: Emerging Roles in Cancer. Cell. Physiol. Biochem. 2017, 44, 1–20. [Google Scholar] [CrossRef]
  281. Tan, L.; Mai, D.; Zhang, B.; Jiang, X.; Zhang, J.; Bai, R.; Ye, Y.; Li, M.; Pan, L.; Su, J.; et al. PIWI-interacting RNA-36712 restrains breast cancer progression and chemoresistance by interaction with SEPW1 pseudogene SEPW1P RNA. Mol. Cancer 2019, 18, 9. [Google Scholar] [CrossRef]
  282. Reeves, M.E.; Firek, M.; Jliedi, A.; Amaar, Y.G. Identification and characterization of RASSF1C piRNA target genes in lung cancer cells. Oncotarget 2017, 8, 34268–34282. [Google Scholar] [CrossRef]
  283. Liu, Y.; Dou, M.; Song, X.; Dong, Y.; Liu, S.; Liu, H.; Tao, J.; Li, W.; Yin, X.; Xu, W. The emerging role of the piRNA/piwi complex in cancer. Mol. Cancer 2019, 18, 123. [Google Scholar] [CrossRef] [PubMed]
  284. Wu, L.; Jiang, Y.; Zheng, Z.; Li, H.; Cai, M.; Pathak, J.L.; Li, Z.; Huang, L.; Zeng, M.; Zheng, H.; et al. mRNA and P-element-induced wimpy testis-interacting RNA profile in chemical-induced oral squamous cell carcinoma mice model. Exp. Anim. 2020, 69, 168–177. [Google Scholar] [CrossRef] [PubMed]
  285. Saad, M.A.; Ku, J.; Kuo, S.Z.; Li, P.X.; Zheng, H.; Yu, M.A.; Wang-Rodriguez, J.; Ongkeko, W.M. Identification and characterization of dysregulated P-element induced wimpy testis-interacting RNAs in head and neck squamous cell carcinoma. Oncol. Lett. 2019, 17, 2615–2622. [Google Scholar] [CrossRef]
  286. Firmino, N.; Martinez, V.D.; Rowbotham, D.A.; Enfield, K.S.S.; Bennewith, K.L.; Lam, W.L. HPV status is associated with altered PIWI-interacting RNA expression pattern in head and neck cancer. Oral Oncol. 2016, 55, 43–48. [Google Scholar] [CrossRef]
  287. Krishnan, A.R.; Qu, Y.; Li, P.X.; Zou, A.E.; Califano, J.A.; Wang-Rodriguez, J.; Ongkeko, W.M. Computational methods reveal novel functionalities of PIWI-interacting RNAs in human papillomavirus-induced head and neck squamous cell carcinoma. Oncotarget 2018, 9, 4614–4624. [Google Scholar] [CrossRef]
  288. Chattopadhyay, T.; Gupta, P.; Nayak, R.; Mallick, B. Genome-wide profiling of dysregulated piRNAs and their target genes implicated in oncogenicity of tongue squamous cell carcinoma. Gene 2023, 849, 146919. [Google Scholar] [CrossRef] [PubMed]
  289. Krishnan, A.R.; Korrapati, A.; Zou, A.E.; Qu, Y.; Wang, X.Q.; Califano, J.A.; Wang-Rodriguez, J.; Lippman, S.M.; Hovell, M.F.; Ongkeko, W.M. Smoking status regulates a novel panel of PIWI-interacting RNAs in head and neck squamous cell carcinoma. Oral Oncol. 2017, 65, 68–75. [Google Scholar] [CrossRef] [PubMed]
  290. Celetti, A.; Merolla, F.; Luise, C.; Siano, M.; Staibano, S. Novel Markers for Diagnosis and Prognosis of Oral Intraepithelial Neoplasia; IntechOpen: Rijeka, Croatia, 2012. [Google Scholar]
  291. Chattopadhyay, E.; Singh, R.; Ray, A.; Roy, R.; De Sarkar, N.; Paul, R.R.; Pal, M.; Aich, R.; Roy, B. Expression deregulation of mir31 and CXCL12 in two types of oral precancers and cancer: Importance in progression of precancer and cancer. Sci. Rep. 2016, 6, 32735. [Google Scholar] [CrossRef]
  292. Osan, C.; Chira, S.; Nutu, A.M.; Braicu, C.; Baciut, M.; Korban, S.S.; Berindan-Neagoe, I. The Connection between MicroRNAs and Oral Cancer Pathogenesis: Emerging Biomarkers in Oral Cancer Management. Genes 2021, 12, 1989. [Google Scholar] [CrossRef]
  293. Grubelnik, G.; Bostjancic, E.; Anicin, A.; Dovsak, T.; Zidar, N. MicroRNAs and Long Non-Coding RNAs as Regulators of NANOG Expression in the Development of Oral Squamous Cell Carcinoma. Front. Oncol. 2020, 10, 579053. [Google Scholar] [CrossRef]
  294. Zhang, K.; Qiu, W.; Wu, B.; Fang, F. Long non-coding RNAs are novel players in oral inflammatory disorders, potentially premalignant oral epithelial lesions and oral squamous cell carcinoma (Review). Int. J. Mol. Med. 2020, 46, 535–545. [Google Scholar] [CrossRef] [PubMed]
  295. Jia, H.; Wang, X.; Sun, Z. Exploring the long noncoding RNAs-based biomarkers and pathogenesis of malignant transformation from dysplasia to oral squamous cell carcinoma by bioinformatics method. Eur. J. Cancer Prev. 2020, 29, 174–181. [Google Scholar] [CrossRef] [PubMed]
  296. Xu, S.; Song, Y.; Shao, Y.; Zhou, H. Comprehensive analysis of circular RNA in oral leukoplakia: Upregulated circHLA-C as a potential biomarker for diagnosis and prognosis. Ann. Transl. Med. 2020, 8, 1375. [Google Scholar] [CrossRef] [PubMed]
  297. Chen, C.M.; Chu, T.H.; Chou, C.C.; Chien, C.Y.; Wang, J.S.; Huang, C.C. Exosome-derived microRNAs in oral squamous cell carcinomas impact disease prognosis. Oral Oncol. 2021, 120, 105402. [Google Scholar] [CrossRef]
  298. Momen-Heravi, F.; Bala, S. Extracellular vesicles in oral squamous carcinoma carry oncogenic miRNA profile and reprogram monocytes via NF-kappaB pathway. Oncotarget 2018, 9, 34838–34854. [Google Scholar] [CrossRef]
  299. 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]
  300. Patil, S.; Warnakulasuriya, S. Blood-based circulating microRNAs as potential biomarkers for predicting the prognosis of head and neck cancer-a systematic review. Clin. Oral Investig. 2020, 24, 3833–3841. [Google Scholar] [CrossRef]
  301. Patel, A.; Patel, S.; Patel, P.; Mandlik, D.; Patel, K.; Tanavde, V. Salivary Exosomal miRNA-1307-5p Predicts Disease Aggressiveness and Poor Prognosis in Oral Squamous Cell Carcinoma Patients. Int. J. Mol. Sci. 2022, 23, 10639. [Google Scholar] [CrossRef]
  302. Jin, N.; Jin, N.; Bu, W.; Li, X.; Liu, L.; Wang, Z.; Tong, J.; Li, D. Long non-coding RNA TIRY promotes tumor metastasis by enhancing epithelial-to-mesenchymal transition in oral cancer. Exp. Biol. Med. 2020, 245, 585–596. [Google Scholar] [CrossRef]
  303. Li, C.; Guo, H.; Xiong, J.; Feng, B.; Zhu, P.; Jiang, W.; Jiang, P.; Su, X.; Huang, X. Exosomal long noncoding RNAs MAGI2-AS3 and CCDC144NL-AS1 in oral squamous cell carcinoma development via the PI3K-AKT-mTOR signaling pathway. Pathol. Res. Pract. 2022, 240, 154219. [Google Scholar] [CrossRef]
  304. Li, W.; Han, Y.; Zhao, Z.; Ji, X.; Wang, X.; Jin, J.; Wang, Q.; Guo, X.; Cheng, Z.; Lu, M.; et al. Oral mucosal mesenchymal stem cell-derived exosomes: A potential therapeutic target in oral premalignant lesions. Int. J. Oncol. 2019, 54, 1567–1578. [Google Scholar] [CrossRef] [PubMed]
  305. Cui, J.; Wang, H.; Zhang, X.; Sun, X.; Zhang, J.; Ma, J. Exosomal miR-200c suppresses chemoresistance of docetaxel in tongue squamous cell carcinoma by suppressing TUBB3 and PPP2R1B. Aging 2020, 12, 6756–6773. [Google Scholar] [CrossRef] [PubMed]
  306. Kawakubo-Yasukochi, T.; Morioka, M.; Hazekawa, M.; Yasukochi, A.; Nishinakagawa, T.; Ono, K.; Kawano, S.; Nakamura, S.; Nakashima, M. miR-200c-3p spreads invasive capacity in human oral squamous cell carcinoma microenvironment. Mol. Carcinog. 2018, 57, 295–302. [Google Scholar] [CrossRef] [PubMed]
  307. He, L.; Liao, L.; Du, L. miR-144-3p inhibits tumor cell growth and invasion in oral squamous cell carcinoma through the downregulation of the oncogenic gene, EZH2. Int. J. Mol. Med. 2020, 46, 828–838. [Google Scholar] [CrossRef]
  308. Husna, A.A.; Rahman, M.M.; Lai, Y.C.; Chen, H.W.; Hasan, M.N.; Nakagawa, T.; Miura, N. Identification of melanoma-specific exosomal miRNAs as the potential biomarker for canine oral melanoma. Pigment Cell Melanoma Res. 2021, 34, 1062–1073. [Google Scholar] [CrossRef]
  309. Liu, T.; Chen, G.; Sun, D.; Lei, M.; Li, Y.; Zhou, C.; Li, X.; Xue, W.; Wang, H.; Liu, C.; et al. Exosomes containing miR-21 transfer the characteristic of cisplatin resistance by targeting PTEN and PDCD4 in oral squamous cell carcinoma. Acta Biochim. Et Biophys. Sin. 2017, 49, 808–816. [Google Scholar] [CrossRef]
  310. Kavitha, M.; Jayachandran, D.; Aishwarya, S.Y.; Md Younus, P.; Venugopal, A.; Suresh Babu, H.W.; Ajay, E.; Sanjana, M.; Arul, N.; Balachandar, V. A new insight into the diverse facets of microRNA-31 in oral squamous cell carcinoma. Egypt. J. Med. Hum. Genet. 2022, 23, 149. [Google Scholar] [CrossRef]
  311. He, L.; Ping, F.; Fan, Z.; Zhang, C.; Deng, M.; Cheng, B.; Xia, J. Salivary exosomal miR-24-3p serves as a potential detective biomarker for oral squamous cell carcinoma screening. Biomed. Pharmacother. 2020, 121, 109553. [Google Scholar] [CrossRef]
  312. Lu, Y.C.; Chen, Y.J.; Wang, H.M.; Tsai, C.Y.; Chen, W.H.; Huang, Y.C.; Fan, K.H.; Tsai, C.N.; Huang, S.F.; Kang, C.J.; et al. Oncogenic function and early detection potential of miRNA-10b in oral cancer as identified by microRNA profiling. Cancer Prev. Res. 2012, 5, 665–674. [Google Scholar] [CrossRef]
  313. Karimi, A.; Bahrami, N.; Sayedyahossein, A.; Derakhshan, S. Evaluation of circulating serum 3 types of microRNA as biomarkers of oral squamous cell carcinoma; A pilot study. J. Oral Pathol. Med. 2020, 49, 43–48. [Google Scholar] [CrossRef]
  314. Faur, C.I.; Roman, R.C.; Jurj, A.; Raduly, L.; Almasan, O.; Rotaru, H.; Chirila, M.; Moldovan, M.A.; Hedesiu, M.; Dinu, C. Salivary Exosomal MicroRNA-486-5p and MicroRNA-10b-5p in Oral and Oropharyngeal Squamous Cell Carcinoma. Medicina 2022, 58, 1478. [Google Scholar] [CrossRef] [PubMed]
  315. Sayyed, A.A.; Gondaliya, P.; Mali, M.; Pawar, A.; Bhat, P.; Khairnar, A.; Arya, N.; Kalia, K. MiR-155 Inhibitor-Laden Exosomes Reverse Resistance to Cisplatin in a 3D Tumor Spheroid and Xenograft Model of Oral Cancer. Mol. Pharm. 2021, 18, 3010–3025. [Google Scholar] [CrossRef] [PubMed]
  316. Dickman, C.T.; Lawson, J.; Jabalee, J.; MacLellan, S.A.; LePard, N.E.; Bennewith, K.L.; Garnis, C. Selective extracellular vesicle exclusion of miR-142-3p by oral cancer cells promotes both internal and extracellular malignant phenotypes. Oncotarget 2017, 8, 15252–15266. [Google Scholar] [CrossRef] [PubMed]
  317. Baber, S.; Bayat, M.; Mohamadnia, A.; Shamshiri, A.; Amini Shakib, P.; Bahrami, N. Role of miR153 and miR455-5p Expression in Oral Squamous Cell Carcinoma Isolated from Plasma. Asian Pac. J. Cancer Prev. 2021, 22, 157–161. [Google Scholar] [CrossRef]
  318. Ries, J.; Vairaktaris, E.; Agaimy, A.; Kintopp, R.; Baran, C.; Neukam, F.W.; Nkenke, E. miR-186, miR-3651 and miR-494: Potential biomarkers for oral squamous cell carcinoma extracted from whole blood. Oncol. Rep. 2014, 31, 1429–1436. [Google Scholar] [CrossRef]
  319. Maclellan, S.A.; Lawson, J.; Baik, J.; Guillaud, M.; Poh, C.F.; Garnis, C. Differential expression of miRNAs in the serum of patients with high-risk oral lesions. Cancer Med. 2012, 1, 268–274. [Google Scholar] [CrossRef]
  320. 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]
  321. Ries, J.; Baran, C.; Wehrhan, F.; Weber, M.; Motel, C.; Kesting, M.; Nkenke, E. The altered expression levels of miR-186, miR-494 and miR-3651 in OSCC tissue vary from those of the whole blood of OSCC patients. Cancer Biomark. 2019, 24, 19–30. [Google Scholar] [CrossRef]
  322. Zhou, S.; Zhu, Y.; Li, Z.; Zhu, Y.; He, Z.; Zhang, C. Exosome-derived long non-coding RNA ADAMTS9-AS2 suppresses progression of oral submucous fibrosis via AKT signalling pathway. J. Cell. Mol. Med. 2021, 25, 2262–2273. [Google Scholar] [CrossRef]
  323. Chen, Y.; Li, Z.; Liang, J.; Liu, J.; Hao, J.; Wan, Q.; Liu, J.; Luo, C.; Lu, Z. CircRNA has_circ_0069313 induced OSCC immunity escape by miR-325-3p-Foxp3 axes in both OSCC cells and Treg cells. Aging 2022, 14, 4376–4389. [Google Scholar] [CrossRef]
  324. Ye, D.; Gong, M.; Deng, Y.; Fang, S.; Cao, Y.; Xiang, Y.; Shen, Z. Roles and clinical application of exosomal circRNAs in the diagnosis and treatment of malignant tumors. J. Transl. Med. 2022, 20, 161. [Google Scholar] [CrossRef] [PubMed]
  325. Mazumder, S.; Datta, S.; Ray, J.G.; Chaudhuri, K.; Chatterjee, R. Liquid biopsy: miRNA as a potential biomarker in oral cancer. Cancer Epidemiol. 2019, 58, 137–145. [Google Scholar] [CrossRef] [PubMed]
  326. Winkle, M.; El-Daly, S.M.; Fabbri, M.; Calin, G.A. Noncoding RNA therapeutics—Challenges and potential solutions. Nature reviews. Drug Discov. 2021, 20, 629–651. [Google Scholar] [CrossRef] [PubMed]
  327. Qian, Y.; Shi, L.; Luo, Z. Long Non-coding RNAs in Cancer: Implications for Diagnosis, Prognosis, and Therapy. Front. Med. 2020, 7, 612393. [Google Scholar] [CrossRef]
Figure 1. Classification of non-coding RNAs based on structures and functions. Abbreviations: non-coding RNA: ncRNA, rRNA: ribosomal RNA, tRNA: transfer RNA, miRNA: microRNA, piRNA: PIWI-interacting RNA, siRNA: small interfering RNA, snoRNA: small nucleolar RNA, tiRNA: transcription initiation RNA, snRNA: small nuclear RNA, scRNA: small cytoplasmic RNA, ceRNA: competing endogenous RNA, PROMPT: promoter upstream transcript.
Figure 1. Classification of non-coding RNAs based on structures and functions. Abbreviations: non-coding RNA: ncRNA, rRNA: ribosomal RNA, tRNA: transfer RNA, miRNA: microRNA, piRNA: PIWI-interacting RNA, siRNA: small interfering RNA, snoRNA: small nucleolar RNA, tiRNA: transcription initiation RNA, snRNA: small nuclear RNA, scRNA: small cytoplasmic RNA, ceRNA: competing endogenous RNA, PROMPT: promoter upstream transcript.
Cancers 15 03752 g001
Figure 2. Differential expression of non-coding RNAs in oral pre-cancerous lesions. Up ↑ and down ↓ arrows indicate their up- or down-regulation status.
Figure 2. Differential expression of non-coding RNAs in oral pre-cancerous lesions. Up ↑ and down ↓ arrows indicate their up- or down-regulation status.
Cancers 15 03752 g002
Table 1. List of miRNAs regulating oral cancer.
Table 1. List of miRNAs regulating oral cancer.
miRNA Name Function on Up or Down RegulationTarget Genes/Associated PathwaysStudy ModelBio-Marker/TherapyReferences
List of Up-regulated miRNAs
miR-21 ↑Associated with cell growth, cell proliferation, and cell cycle progression, as well as inhibits apoptosisDKK2, PTEN, PDCD4, CADM1, RECKPatient tissue, saliva, and plasma samples, cell linesBiomarker (diagnosis and prognosis) and therapeutic target[15,22,29,30]
miR-155 ↑Enhances cell proliferation in patients and confers cisplatin chemoresistance via EMTFOXO3aPatient tissue samples and blood exosome Prognostic biomarker[20,21,22]
miR-196a ↑Promotes cell migration, invasion, and lymph node metastasisSPRR2C, ANXA1, S100A9, NME4-JNK-TIMP1-MMP signaling pathway, MAMDC2, Patient tissue, saliva, plasma samples, and OC cell linesBiomarker[30,31,32]
miR-1237 ↑Causes cellular progression FAM69C, PACS2, SPOPPatient tissue and plasma samples Biomarker (diagnosis and prognosis)[30]
miR-31 ↑Increases oxidative stress in OSCC and enriches the oncogenicity and stemness of HNSCC. Regulates the reprogramming of lipid metabolism and enhances cell migration in OSCCKu80, FIH, ARID1A, SIRT3, ACOX1 OC cell lines, tissue, plasma and saliva samples, and transgenic miceBiomarker [33,34,35,36,37]
miR-455 ↑Increases proliferation and anchorage-independent growth of OSCC cellUBE2BOC Cells, tissue and plasma samples, and xenograft mice model Biomarker (prognosis)[38]
miR-181 ↑Increases cell migration and invasion and may enhance lymph-node metastasis-Patient tissue sample and plasmaBiomarker[39]
miR-184 ↑Increased cell proliferation rate and suppressed apoptosisc-MYCTumor tissue, cell lines, and plasma Biomarker (diagnosis)[40]
miR-134 ↑Increases migration and reduces E-cadherin expression in OSCC cell lines.
Enhances metastatic potential of OSCC xenografts
WWOX, PDCD7 Tissue and plasma samples and OSCC cell linesBiomarker[41,42]
miR-146a ↑Increases the tumorigenicity and metastasis IRAK1, TRAF6 and NUMBTissue, plasma and saliva samples, cell lines, and xenograft tumor modelBiomarker (diagnosis) and therapeutic target[43]
miR-93 ↑Promotes metastasis, correlated with poor prognosis--OC tissue, saliva samples and cell linesPrognostic marker and therapeutic target[44]
miR-10b ↑Role in cancer stemness and facilitates metastatic colonizationDIAPH2Patient tissue and saliva samples and cell linesDiagnostic marker and therapeutic target[45]
miR-372 ↑ Enhanced drug resistance and migration. Also associated with lymph node metastasis and poor prognosisp62, ZBTB7ACell lines, patient tissue and saliva samples, and xenograft tumor modelDiagnostic and prognostic marker[45,46,47]
miR-373 ↑Enhances proliferation and metastasisSPOPOC samples and cell linesTherapeutic target[48]
miR-103a-3p ↑Induces proliferation and restricts cell apoptosisRCAN1OC cell lines, tumor tissues, saliva, plasma, and xenograft tumor modelDiagnostic marker and therapeutic target[49]
miR-454 ↑Involved in proliferation and colony formation, as well as inducing invasion and migration NR3C2OC cell lines, tissues and saliva samplesPotential therapeutic direction for diagnosis and prognosis[50]
miR-654-5p ↑Facilitates EMT, promotes cellular proliferation, tumor metastasis, and chemoresistanceGRAP, Ras/MAPK SignalingOC cell lines, patient tissues, plasma, and xenograft tumor modelBiomarker for the clinical diagnosis and prognosis[51]
miR-188-5p ↑Causes cell migrationFOXN2Cell lines Potential prognostic marker and therapeutic target[52]
miR-626 ↑Promotes cancer cell proliferation, colony formation, migration, and G0/G1-to-S phase transitionNFIBPatient tissue, plasma, cell lines, and xenograft tumor modelTherapeutic target[53]
miR-4513 ↑Causes proliferation and apoptosisCXCL17Saliva, plasma, and OC cell lines Therapeutic target[54]
miR-944 ↑Induces pro-inflammation cytokines secretion, migration, and invasion CISH/STAT regulationOC cell lines, tissue and saliva samples--[55]
miR-211 ↑Causes proliferation, migration, invasion, and metastasisBIN1, TCF12, TGFBRII, c-MYC Tissue samples, cell lines, and xenograft tumor modelTherapeutic target and prognostic marker[56,57,58,59]
miR-223 ↑Promotes OSCC proliferation and migrationFBXW7Tissue, saliva samples and cell lines Therapeutic target[60]
miR-221 ↑Regulates proliferation and invasion and inhibits apoptosisPTENOC cell linesBiomarker (diagnosis)[61,62]
miR-222 ↑Causes proliferation, metastasis, and invasion and decreases apoptosisPTEN, ABCG2, PUMATumor tissue, saliva samples, and cell linesBiomarker (diagnosis) and therapeutic target[61,62,63,64]
Mir-5100 ↑Causes proliferation, migration, and invasionSCAIOC cell linesTherapeutic target[65]
miR-382-5p ↑Induced cell migration and invasion-Patients tissue and saliva samples, and TSCC cell linesTherapeutic target[66]
List of Down regulated miRNAs
miR-27b ↓ Causes proliferation, migration, and invasion. Induces tumor growth in vivo. Activates the EMT process and metastatic pathwaysITGA5TSCC cell lines, saliva, and xenograft tumor modelTherapeutic target[67]
miR-204 ↓Regulates cancer stemness, cell proliferation, and metastasisCXCR4, CDC42, RAB22A, EZR, Slug and Sox4OSCC cell lines, patient saliva, plasma samples, and nude mice modelBiomarker[30,68,69]
miR-100 ↓Plays role in the development or progression of the disease, and may contribute to the loss of sensitivity to ionizing radiationMMP13, ID1, FGFR3, EGR2OC cell lines Therapeutic target[70]
miR-29a ↓Promoted OSCC cell invasion and induced drug-resistance in vitroMMP2OC cell lines and patient tissue and saliva samplesTherapeutic target[71]
Let-7d ↓Induces chemoresistance to cisplatin and 5-FUSnail and TwistOC cell lines and saliva samplesTherapeutic target[27]
miR-203 ↓Causes tumor proliferation and shows resistance to drugs. Also regulates apoptotic signalingBMI-1, PI3KCA, YES-1, SEMA6A, Plasma samples and cell linesBiomarker (diagnosis and prognosis), anti-cancer therapeutics[72,73,74,75]
miR-200 ↓Regulate EMT leading to OSCC progressionZEB1, ZEB2Clinical specimens-[76]
miR-133a ↓Regulates tumor proliferation and differentiation, as well as showing antiapoptotic characteristicsPKM2, GSTP1Patient samples and cell linesTherapeutic target[77,78]
miR-133b ↓Regulates the proliferation rate of cancer cells as well as the number of apoptotic cellsPKM2Patient samples and cell linesTherapeutic target[77]
miR-138 ↓Causes cell proliferation and inhibits apoptosisGNAI2TSCC cell lines and plasma-[79]
miR-125b ↓Regulates tumor proliferation and radioresistance mechanismsICAM2 signalingCell lines and tissue specimensPrognostic markers[80]
miR-9 ↓Cause proliferation of OSCC cellsCXCR4-Wnt/β-cateninPatient plasma sample and cell linesTherapeutic target[81]
miR-26a/b ↓Promotes invasion and migrationTMEM184BOC clinical specimens and cell linesTherapeutic target [82]
miR-491-5p ↓Responsible for poor overall survival of patients, invasion, and metastasisGIT1OC tissues, plasma, cell lines, and mice modelBiomarker (prognosis)[83]
miR-375 ↓Promotes tumor proliferation, migration, and invasionCIP2A, SLC7A11, PDGF-APatient tissue and saliva samples and cell linesBiomarker (diagnosis), therapeutic target[84,85,86]
miR-320 ↓Regulates tumor angiogenesisHIF-1α-NRP1-VEGFTissue, saliva, plasma samples, cell lines, and mice modelTherapeutic target[87]
miR-218 ↓Contributes to oral carcinogenesis.mTOR-Rictor-AktOC cell linesTherapeutic target[88]
miR-205 ↓Regulates cancer development by promoting migration, invasion, and inhibiting apoptosisIL-24, caspase-3/-7, Axin-2, TIMP-2OC cell linesTherapeutic target[89,90]
miR-181a ↓Cause tumorigenesis via stimulating EMT, and enhances metastatic potentialK-ras, Twist1Cell lines Therapeutic target[91,92]
miR-145 ↓Causes cell proliferation and colony formationc-Myc, Cdk6Cell lines and tissue samplesPotential biomarker for diagnosis and therapeutic target[93]
miR-140-5p ↓Promotes invasion and migration ADAM10, ERBB4, PAX6, and LAMC1 TSCC cell lines--[94]
miR-124 ↓Causes OSCC progressionITGB1Cell lines--[28]
miR-99a ↓Causes migration, invasion, and lung colonization in OSCC cellsIGF1RTissue samplesTherapeutic target[95]
miR-34a ↓Promotes tumor growth and angiogenesisE2F3Cell lines, plasma, and SCID mouse xenograft modelTherapeutic target[96]
miR-17/20a ↓Causes tumor progression and migration. Expression is negatively correlated with TNM staging and lymphatic metastasisITGβ8Cell lines and tissue and saliva samplesPrognostic marker[97]
miR-137 ↓Induces tumor differentiation--OC tissues, plasma and cells Potential marker for early diagnosis[98]
miR-5580-3p ↓Possibly associated with cell viability, proliferation, and migration LAMC2Tissue and cell linesTherapeutic target and potential biomarker[99]
miR-98 ↓Causes cell growth and metastasisIGF1ROC tissues and cell linesPotential therapeutic target[100]
miR-1 ↓Promotes migration and invasiveness in OSCC cells. SlugOC cell lines and tumor tissuesTherapeutic target[101]
miR-377 ↓Promotes OSCC growth and migrationHDAC9Tissue samples and cell linesTherapeutic target[102]
miR-23a-3p ↓It is correlated with more advanced cancerous stage and poorer differentiation of OSCC cellFGF2OC cell lines and tissue Prognostic biomarker and therapeutic target[103]
miR-22 ↓Increased cell viability, migration, and invasionNLRP3OC tissues and plasma, cell lines, and nude mice xenograft modelsPrognostic biomarker and therapeutic target [104]
miR-139-5p ↓Causes tumorigenesis and progressionHOXA9Patients tissue and saliva samples, and cell lines Therapeutic strategy for the treatment[105]
miRNA-504 ↓Facilitates proliferation, causes migration and invasionCDK6OC animal model and cell linesTherapeutic target[106]
miR-106a ↓Induces proliferation and EMT LIMK1OC tissues and cell linesPrognostic factor[107]
miR-16 ↓Promotes proliferation and inhibited apoptosisAKT3, BCL2L2OC patients and cancer cell linesTherapeutic target[108]
miR-495 ↓Induces cell proliferation and invasionNotch1OC tissues and plasma and OC cellsTherapeutic target[109]
miRNA-329 ↓,
miRNA-410 ↓
Promotes proliferation and invasionWnt-7bOC cell lines and tissues --[110]
miR-132 ↓Promotes proliferation, invasion, and migration, and confers OSCC cell resistance to CDDP-induced apoptosis in vitroTGF-β1/Smad2/3Tissues and cell lines Therapeutic target[111]
miR-376c-3p ↓Associated with tumor progression, poorer differentiation, lymphoid metastasis, and lymphovascular invasion.HOXB7Tissue specimens and OC cell linesBiomarker (early diagnosis)[112]
miR-769-5p ↓Causes cancer progressionJAK1/STAT3 pathwayOC tissues and cellsTherapeutic target[113]
miR-486-3p ↓Promotes proliferation and inhibits apoptosisDDR1Tissue, saliva, and plasma samplesTherapeutic target[114]
Up arrows ↑ indicate upregulation and down arrows ↓ indicate downregulation.
Table 3. List of circRNAs involved in oral cancer.
Table 3. List of circRNAs involved in oral cancer.
circRNAsFunction on Up- or Down-RegulationTargets/Associated PathwayStudy ModelBiomarker/TherapyReferences
List of up-regulated circRNAs
circ_0002185 ↑Promotes the proliferation, invasion, migration, EMT in vitro, and tumor growth in vivocircUHRF1/miR-526-5p/c-Myc/TGFB1/ESRP1 feedback loopOC tissues and cellsTherapeutic target[237]
circPVT1 ↑Induces proliferation by serving as a miRNA spongecircPVT1/miR-125b axisTissue samples and cell lines Biomarker and therapeutic target[231]
circ_100290 ↑Promotes glycolysis and cell proliferation in OSCCcirc_100290/miR-378a/GLUT1OC cell lines and tissues--[230]
circ_0001742 ↑Encourages proliferation, migration, invasion, and EMT, and resists apoptosis in TSCC cellscirc_0001742/miR-431-5p/ATF3 axisTSCC tissues and cellsTherapeutic target[238]
circ_0001971 ↑Regulates cell proliferation, migration, invasion, apoptosis, and chemosensitivity of OSCCcirc_0001971/miR-194/miR-204Tissues, saliva and cell lines--[239]
circDOCK1 ↑Suppresses cell apoptosiscircDOCK1/miR-196-5p/BIRC3OC cell lines, tissue and saliva samplesBiomarker and therapeutic target[240]
circFLNA ↑Contributes to laryngeal squamous cell carcinoma (LSCC) migrationcircFLNA/miR-486-3pLSCC tissues and cell lines Therapeutic target[241]
circGOLPH3 ↑Promotes the growth of OSCC in vitro and in vivo as well as up-regulating its cell migration and invasioncircGOLPH3/miR-1299/LIF axisOC cell lines and plasmaTherapeutic target[242]
circCLK3 ↑Promotes cell proliferation,
migration, invasion, and cell cycle in TSCC cells
miR-455-5p/PARVA axisTSCC tissues and cell lines--[243]
circCDR1 ↑Enhances OSCC cell viability, endoplasmic reticulum (ER) stress, and inhibits cell apoptosis under a hypoxic microenvironmentAKT/ERK½/mTORsignaling pathwayOSCC cell lines and mice model Treatment strategy[232]
circ_0014359 ↑Promotes cancer progressioncirc_0014359/miR-149 pathwayTissues, cell lines, and xenograft mouse modelDiagnosis and treatment[244]
circ_LPAR3 ↑Causes tumor growth and angiogenesisCirc_LPAR3/miR-513b-5p/VEGFC/AKT1OC cell lines and mice model--[245]
circ_SEPT9 ↑Induced OSCC proliferation, migration, and invasioncirc_SEPT9/miR-1225/PKN2OC cell lines, tissues, and xenograft modelTherapeutic target[246]
circ_0000199 ↑Associated with tumor size, lymphatic metastasis, and TNM staging in patients with OSCCmiR-145-5p and miR-29b-3p (in silico study)Patient plasma samples and cell linesBiomarker and potential therapeutic target[247]
circ_0001883 ↑May be responsible for cell migration, invasion, and EMT of LSCCmiR-125-5p/PI3K/AKT axisPatient samples and cell linesTherapeutic target for LSCC treatment[248]
circ_0005320 ↑Promotes tumorigenesiscirc_0005320-miR-486-3p/miR-637 axisOC tissues, cells, and nude mice modelBiomarker and therapeutic target[249]
circ_0001874 ↑, circ_0001971 ↑Associated with tumorigenesis (development and progression of OSCC)miR-661, miR-662, miR-593-5p, miR-107, and miR-103a-3p (targets of circ_0001874), miR-152-5p, miR-103a-3p, miR-107, miR-505-3p, and miR-9-5p (targets of circ_0001971)Patient sample (saliva)Diagnostic, prognostic, biomarker and therapeutic target[250]
circ_0011946 ↑Causes proliferation, migration, and invasion, as well as restricted apoptosismiR-216a-5p/BCL2L2 axisOC tissues and cell lines--[251]
circ_0001461 ↑Promotes cell proliferation, migration, and invasion. Promotes resistance to TNF-α-induced apoptosismiR-145/TLR4/NF-κB axisOC cells and tissues--[252]
Circ_DHTKD1 ↑Promotes tumor growth and metastasis of OSCCmiR-326/GAB1 axisOC cell lines, tissues, and xenograft modelTherapeutic target for clinical diagnosis[253]
Circ_IGHG ↑ Induces EMT and promotes cancer progressionmiR-142-5p/IGF2BP3 SignalingOC cell linesDiagnosis and treatment[254]
circ_002178 ↑ Promotes the proliferation and migrationAkt/mTOR pathwayOC tissues and cell lines--[255]
Circ_VAPA ↑ Promotes the proliferation, migration, and invasion miR-132/HOXA7 axisOC tissues and cellsPrognosis[256]
circ-LRP6 ↑ Mediates EMT and autophagy-OC cells and tissues--[257]
List of down regulated circRNAs
circ_0000140 ↓Correlated negatively with poor prognostic outcomes in OSCC patientsmiR-31/LATS2 axis of Hippo signaling pathwayTissue, cell lines, and xenograft mouse modelTherapeutic target[258]
circ-PKD2 ↓Reduced expression in OSCC patients is significantly correlated with aggressive tumor characteristicscircPDK2/miR-204-3p/APC2OC tissue samples and cell linesTherapeutic target[236]
circ_0005379 ↓Its expression is inversely correlated with tumor size and differentiationEGFR pathwayOC patient samples, cell line, and xenograft mouse modelsTherapeutic target[234]
circ_0004491 ↓Reduced expression is associated with lymph node metastasis, and may facilitate OSCC cell invasion and migrationcirc_0004491/miR-155-5p/SIRT1OC tissues and cellsPotential biomarker [259]
circSPATA6 ↓Lower expression in OSCC cells fails to impede migration and invasion and facilitate cell cycle arrest and apoptosismiR-182/TRAF6 axisCell lines and xenograft tumor model Targeted therapy[260]
circ_0086414 ↓Its lower expression promotes the processes of growth and metastasis in OSCCAMPK and cAMPsignaling pathwayOC tissues and cellsDiagnostic biomarker and OSCC therapy[261]
circ_0008309 ↓Associated with pathological differentiation of OSCC patientshsa_circ_0008309-miR-136-5P/hsa-miR-382-5P-ATXN1OC cell linesTherapeutic biomarker [262]
circGDI2 ↓Fails to serve as a repressor to restrain OSCC malignancy and, thus, regulate OSCC progressionmiR-454-3p/FOXF2 AxisOC tissues, cells, and mice modelBiomarker for targeted OSCC therapy[263]
circ-KIAA0907 ↓Fails to inhibit migration, invasion, glycolysis, and promote apoptosis, thereby leading to OSCC progressionmiR-96-5p/UNC13C axisOC tissues Potential target for OSCC treatment[264]
circ_0007059 ↓Determined to alter cell growthAKT/mTORsignalingSCC15 and CAL27 cellsPrognostic/therapeutic target [233]
circ_0004872 ↓Reverse the promoting effect of miR-424-5p overexpression on the process of OSCC cellssponges miR-424-5pOC cell linesEarly diagnosis and targeted therapy of OSCC[265]
circ_0092125 ↓Associated with clinicopathological factors in OSCC patients, including tumor size, TNM stage, and lymph node metastasis--OC cells and tissuesBiomarker of the OSCC prognosis[266]
circRNA-102450 ↓Low expression is associated with the tumor metastatic properties and act as a tumor suppressorcircRNA-102450/miR-1178 axisPatient plasma samplesBiomarker[267]
circ_0072387 ↓Associated with cell proliferation, migration, invasion, EMT, and glycolysis in OSCCmiR-503-5pPatient sample, OSCC cells, and tissuesTherapeutic target[268]
Up arrows ↑ indicate upregulation and down arrows ↓ indicate downregulation.
Table 4. List of snoRNAs involved in oral cancer.
Table 4. List of snoRNAs involved in oral cancer.
snoRNAsFunctionTargetsStudy ModelBiomarker/TherapyReferences
SNHG1 ↑Promotes TGFβ1-Induced EMT, migration, and invasion of TSCCsSNHG1/miR-194-5p/MTFR1 AxisTSCC cell linesTherapeutic target[275]
SNHG3 ↑Facilitates cell proliferation and migration in OSCC, and regulates HOXB8transcription factor Y subunit gamma, SNHG3/miR-2682-5p axisOC cell lines Biomarker[272,273]
SNHG15 ↑Increases growth, and facilitates malignant behaviors, of OSCC cellsmiR-188-5p/DAAM1OC cell linesTreatment purpose[274]
SNHG16 ↑Enhances progression and carcinogenesis-CAL-27 and TSCCA cells--[276]
SNHG17 ↑Accelerates proliferation and metastasis of OSCC cells, while reducing apoptosismiR-375/PAX6 axisCAL-27 and Tca8113 cells--[277]
SNHG20 ↑Enhances the progression of OSCCmiR-29a/DIXDC1/Wnt regulatory axisSCC9 and SCC15 cellsA theoretical basis for the treatment[162]
Up arrows ↑ indicate upregulation and down arrows ↓ indicate downregulation.
Table 5. ncRNAs derived from exosomes, blood, serum, and saliva.
Table 5. ncRNAs derived from exosomes, blood, serum, and saliva.
ncRNA NameSampleExpression Level in OC vs. Normal CellsReferences
miR-8485Chondrocyte-derived exosomesHigher[304]
miRNA-1307-5pSalivary exosomesHigher[301]
miR-200c-3pSerum exosomesHigher[305,306]
miR-143 and miR-221Plasma exosomesLower and higher, respectively[307,308]
miR-21Plasma/seminal exosomesHigher[309]
miR-31-5pMacrophage-derived exosomesHigher[310]
miR-24-3pSalivary exosomesHigher[311]
miR-382-5pFibroblast-associated exosomesHigher[66]
miR-10bPlasmaHigher[312]
miR-29a-3pSerumHigher[313]
miR-486-5pPlasma, salivary exosomesHigher (in Stage II)[314]
miR-155ExosomesHigher[315]
miR-142-3pExosomesHigher[316]
miR 455-5p and miR153Blood plasmaHigher and lower, respectively[317]
miR-200b-3p, miR-483-5p, miR-425-5p, miR-374b-5p, miR-191-5p, miR-let-7c, miR-29a, miR-103, miR-1234, miR-638, miR-572, miR-22, miR-29b, miR-24-3p, miR-223, miR-20a, miR-29c, miR-17, miR-196aBloodHigher[300]
miR-187, miR-9,
miR-223, and miR-29c
Lower
miR-494Whole bloodHigher[318]
miR-16 and let-7bSerum Higher[319]
miR-34a-5pCancer-associated fibroblast-derived exosomesLower[320]
miR-3651Whole bloodLower[321]
lncRNAs MAGI2-AS3 and CCDC144NL-AS1Serum exosomesHigher[303]
lncRNA TIRYCancer-associated fibroblast-derived exosomesHigher[302]
lncRNA ADAMTS9-AS2Saliva, exosomesLower[322]
circ_0069313ExosomesHigher[323]
circ_0000199Serum exosomesHigher[247]
circ_0026611Serum exosomesHigher[324]
circ_0001874Salivary exosomesHigher[250]
circ_0001971Salivary exosomesHigher[250]
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

Dey, S.; Biswas, B.; Manoj Appadan, A.; Shah, J.; Pal, J.K.; Basu, S.; Sur, S. Non-Coding RNAs in Oral Cancer: Emerging Roles and Clinical Applications. Cancers 2023, 15, 3752. https://doi.org/10.3390/cancers15153752

AMA Style

Dey S, Biswas B, Manoj Appadan A, Shah J, Pal JK, Basu S, Sur S. Non-Coding RNAs in Oral Cancer: Emerging Roles and Clinical Applications. Cancers. 2023; 15(15):3752. https://doi.org/10.3390/cancers15153752

Chicago/Turabian Style

Dey, Saurabh, Bini Biswas, Angela Manoj Appadan, Jaladhi Shah, Jayanta K. Pal, Soumya Basu, and Subhayan Sur. 2023. "Non-Coding RNAs in Oral Cancer: Emerging Roles and Clinical Applications" Cancers 15, no. 15: 3752. https://doi.org/10.3390/cancers15153752

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