Open Access

Long non‑coding RNAs are novel players in oral inflammatory disorders, potentially premalignant oral epithelial lesions and oral squamous cell carcinoma (Review)

  • Authors:
    • Kaiying Zhang
    • Wei Qiu
    • Buling Wu
    • Fuchun Fang
  • View Affiliations

  • Published online on: June 3, 2020     https://doi.org/10.3892/ijmm.2020.4628
  • Pages: 535-545
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In recent years, a large number of studies have shown that the abnormal expression of long non‑coding (lnc)RNAs can lead to a variety of different diseases, including inflammatory disorders, cardiovascular disease, nervous system diseases, and cancers. Recent research has demonstrated the biological characteristics of lncRNAs and the important functions of lncRNAs in oral inflammation, precancerous lesions and cancers. The present review aims to explore and discuss the potential roles of candidate lncRNAs in oral diseases by summarizing multiple lncRNA profiles in diseased and healthy oral tissues to determine the altered lncRNA signatures. In addition, to highlight the exact regulatory mechanism of lncRNAs in oral inflammatory disorders, potentially premalignant oral epithelial lesions and oral squamous cell carcinoma. The detection of lncRNAs in oral samples has the potential to be used as a diagnostic and an early detection tool for oral diseases. Furthermore, lncRNAs are promising future therapeutic targets in oral diseases, and research in this field may expand in the future.

1. Introduction

Oral inflammatory disorder is a series of processes associated with microbial infections (such as periodontal, endodontic and periapical diseases) and immune-mediated damage [for example Sjögren's syndrome (SS)] (1). Numerous factors, including non-coding (nc)RNAs, leukocytes, cytokines and complement components, are involved in this process (2,3). Long non-coding (lnc)RNAs affect oral inflammation by sponging microRNAs (miRNA/miR) (4) or activating downstream target miRNAs (5). Research on competing endogenous (ce)RNA mechanisms, in which lncRNAs sponge specific miRNAs to suppress their target genes, has increased in numbers in over the last 2 years (6). For example, lncRNA metastasis associated lung adenocarcinoma transcript 1 (MALAT1) acts as a sponge of miR-20a to induce Toll-like receptor 4 (TLR4) signaling and results in an inflammatory reaction from human gingival fibroblasts (HGFs) (7).

Several potentially premalignant oral epithelial lesions (PPOELs) are associated with the disease process of chronic inflammatory disorders (8,9). PPOEL is a broad term to define both histological and clinical oral lesions that have malignant potential, including oral lichen planus (OLP), oral submucous fibrosis (OSF), and oral dysplasia (10). In addition, other vital epigenetic and subcellular regulatory non-coding transcripts, such as lncRNAs and miRNAs are also known to regulate the mRNA expression of inflammation-related cytokines, and disturbance of the miRNA-mRNA-cytokine regulatory network is one of the common pathological features of PPOELs (11,12). For example, the significant upregulation of miR-31 and downregulation of its target gene, C-X-C motif chemokine ligand 12 (CXCL12) contributed to progression of PPOELs (13).

As the 7th hallmark of cancer, chronic inflammation has been linked to various stages of tumorigenesis (14). Several studies have reported that numerous regulators, including inflammatory cytokines and ncRNAs, facilitate tumor development (15,16). LncRNAs participate in the transformation of chronic inflammation into cancer by altering the expression of various inflammatory signaling pathways such as NF-κB and STAT3 and proinflammatory cytokines [such as tumor necrosis factor (TNF) family]. Interleukin (IL)-6-dependent STAT3 signaling activation contributes to the occurrence of colorectal cancer (17). The major histological type of oral cancer is oral squamous cell carcinoma (OSCC) (18). Jia et al (19), identified that differentially expressed (DE) lncRNAs and genes between OSCC, oral dysplasia, and normal oral tissues may control the initiation and development of OSCC through phosphatidylinositol-3-kinases (PI3K)/Akt signaling and mast cell NF-κB functional pathways.

LncRNAs play an essential role in the occurrence and development of inflammation and cancer (20,21). The oral diseases associated with the function of lncRNAs are shown in Fig. 1. According to previous reports, the DE lncRNAs between healthy and pathological oral tissues may affect the occurrence and process of oral diseases (Table I). The present review outlines the current understanding of the established functions and underlying mechanisms of lncRNAs in various oral inflammatory disorders, PPOELs and OSCC.

Table I

Expression profile of lncRNAs in diseased oral tissues.

Table I

Expression profile of lncRNAs in diseased oral tissues.

DiseaseNo. of sample/subjectsMethodConfirmationFindingsComments(Refs.)
Pulpitis7 inflamed pulp tissues and 5 healthy pulp tissuesMicroarrayRT-qPCR; 10 normal human pulp tissue and 10 inflamed samples338 upregulated and 414 downregulated lncRNAs; FC>2; P<0.05Inflamed pulp samples were collected from patients with pulpitis, to reflect the real pathological state(31)
PeriodontitisInflamed and healthy adjacent gingival tissue from two patients with chronic periodontitisMicroarrayRT-qPCR; 15 normal tissue samples and 30 chronic periodontitis tissue samples4313 upregulated and 4612 downregulated lncRNAs; FC≥2.0; P≤0.05First time to analyze DE lncRNAs between chronic periodontitis tissues samples and adjacent normal tissues(36)
SSPBMCs from 8 patients with pSS and 8 healthy subjectsMicroarrayqRT-PCR; No sample size reported199 dysregulated lncRNAs; FC≥1.5; P≤0.01aNo validation; first time to analyze DE lncRNAs in PBMCs of pSS(43)
SSLSGs from 4 pSS patients and 4 healthy individualsMicroarrayRT-qPCR; 30 pSS patients and 16 controls890 upregulated lncRNAs and 353 downregulated lncRNAs; FC>2; P<0.05First time to analyze DE lncRNAs in LSGs of pSS(44)
OSF20 OSCC samples, 10 OSF samples and 13 normal mucous samplesRNA-sequencingRT-qPCR; 20 OSCC samples, 10 OSF samples and 13 normal mucous samples231 upregulated lncRNAs and 456 downregulated lncRNAs; FC>2; P<0.05LncRNAs involved in the malignant transformation process were firstly analyzed and assessed(56)
OLP1 papillomavirus-related OSCC tissue, one OLP tissue and 1 normal oral mucosa tissue samplesRNA-sequencing-76.2% intergenic lncRNAs and 16.5% sense lncRNAs; A coding potential calculator score <0, and CPAT probability ≤0.3643 types of tissues samples but sample numbers were too small; lncRNAs involved in the malignant transformation process were firstly analyzed and assessed(70)
OSCC72 OSCC tissues and adjacent normal tissuesMicroarrayRT-qPCR; 72 OSCC tissues and adjacent normal tissues933 upregulated and 1361 downregulated lncRNAs; FC>2.0; P<0.053 types of OSCC tissues (tongue cancer, gingival carcinoma and carcinoma of the buccal mucosa) were analyzed and compared, and 4 critical lncRNA nodes were identified(62)

a Bonferroni corrected P-value. lnc, long non-coding; SS, Sjögren's syndrome; pSS, primary SS; OSCC, oral squamous cell carcinoma; OLP, oral lichen planus; OSF, oral submucous fibrosis; PBMCs, peripheral blood mononuclear cells; LSGs, labial salivary glands; RT-qPCR, reverse transcription-quantitative PCR; FC, fold change; CPAT, Coding Potential Assessment Tool; DE, differentially expressed.

2. LncRNA biogenesis

LncRNAs are a class of RNA molecules whose transcript length is >200 nucleotides (22). Different lncRNA classifications have been established based on different criteria. The first criteria was presented by Jarroux et al (23), and lncRNAs which are >10 kb belong to the groups of very long intergenic RNAs and macro lncRNAs. Examples of macro lncRNAs include antisense of IGF2R non-protein coding RNA, KCNQ1 opposite strand/antisense transcript 1 and GNAS antisense RNA1, which act as cis-silencers in mouse genomic imprinting (24). The second criterion, location with respect to protein coding genes, is commonly used. In this classification, 5 types (sense, antisense, bidirectional, intronic and intergenic) of lncRNAs are included (23). An example of nature antisense transcripts, which is one type of antisense lncRNAs, is anti-sense non-coding RNA in the INK4 locus, which is encoded by the NK4b-ARF-INK4a locus on chromosome 9p21 (25). According to the classification based on association with other DNA regulatory elements and loci, lncRNAs are divided into pseudogenes, enhancer lncRNAs and promoter-associated lncRNAs and 3′-untranslated region-associated RNAs (23). Long intergenic non-coding (LINC)RNA-p21 is an enhancer RNA, which originates from a p53 binding site associated with regulation of cyclin-dependent kinase inhibitor 1A (26). The associated biochemical pathways or stability of lncRNAs serve as characteristics for their classification, as demonstrated by stable unannotated transcripts (27), Xrn1 sensitive unstable transcripts (XUTs) (such as 5′-long terminal repeat antisense TY1 RNA and XUT1678) (28), Nrd1-unterminated transcripts (29) and cryptic unstable transcripts (for example promoter upstream transcript) (30). Furthermore, several subgroups of lncRNAs with a precise subcellular localization have been defined. Long non-coding mitochondrial RNAs (ncmtRNAs) are cytoplasmic lncRNAs while GAA repeat-containing RNAs and chromatin-enriched RNAs (cheRNAs) locate in the nucleus (23). For example, antisense mitochondrial non-coding RNA-2 and hemin-induced chromatin-enriched RNA down-stream of fetal hemoglobin are ncmtRNAs and cheRNAs, respectively (31,32). Lastly, hypoxia-induced non-coding ultra-conserved transcripts (HINCUTs), stress-induced lncRNA (si-lncRNA), senescence-associated lncRNA (SAL), non-annotated stem transcript, prostate cancer-associated transcripts serve as subgroups of another attribute used for lncRNA classification: Association with specific biological processes (23). Hypoxia-inducible factor (HIF) induces HINCUTs to elevate transcription of nearby genes, which are involved in cellular signaling pathways and processes, such as glucose metabolism (33). Oxidative stress causes rapid and transient dynamics of si-lncRNAs in the nucleus and the cytosol, leading to their accumulation at polysomes, which subsequently induces transcription (34). Antisense very long intergenic ncRNA (VAD), one type of SAL, modulates chromatin structure in cis and increases gene expression in trans at the INK4 locus, which encodes cell cycle inhibitors, that are important to senescence-associated cell proliferation arrest (35). The schematic diagram illustrating various classes of lncRNAs are presented in Fig. 2.

LncRNA can also be classified according to their function. LncRNAs exert or execute their functions in four main ways: Signal, decoy, guide, and scaffold. They have been shown to impact cell macromolecular (protein, RNA and DNA) stability (36). The regulatory mechanism of lncRNAs vary based on their locations within the cells. They participate in chromatin modification and transcription in the nucleus, while they interact with RNA-binding proteins or modulate mRNA translation in the cytoplasm (37,38). There are a variety of mutual regulatory mechanisms for lncRNAs and miRNAs. LncRNAs can not only be mediated by miRNAs but also act as miRNA precursors. It is noteworthy that the ceRNA network is one of the common sites of posttranscriptional regulation (12). In the last 5 years, it has been demonstrated that lncRNAs are important in the regulation of a healthy immune system, which in turn is important for healthy oral tissue (39,40,41). An increasing number of lncRNAs have been reported to regulate the differentiation and activation of immune cells. The differentiation of granulocytes is partly mediated by HOX antisense intergenic RNA myeloid 1 (HOTAIRM1), an antisense lncRNA within the HOXA gene locus. Hallmark myeloid maturation-associated genes such as HOXA1/A2 would be inhibited by silencing HOTAIRM1 (42). The potential importance of lncRNAs in the immune response, inflammation and even cancers is emerging (43-45).

3. Role of lncRNAs in oral inflammatory disorders

Pulpitis

Pulpitis is a state of inflammation of the dental pulp. Most cases are due to penetration of a carious lesion into the pulp chamber (46). Pulpitis is classified as either reversible or irreversible (47). Huang and Chen (48) conducted a microarray analysis to establish lncRNA profiles of inflamed (n=7) and normal (n=5) pulp tissues. A total of 752 lncRNAs (338 upregulated and 414 downregulated) were significantly expressed. A total of 460 significantly upregulated genes were enriched in biological processes, such as immune system processes, immune and defense response, the response to stress, and cell activation. The results indicated that most lncRNAs might play roles in the immune system and inflammatory responses of dental pulp. Lei et al (49), performed a comprehensive analysis of a lncRNA-miRNA-mRNA ceRNA network by integrating the lncRNA profile from Huang and Chen (48), the miRNA profile from Zhong et al (50) and the gene expression profile from Galicia et al (51). A ceRNA regulatory network was created, which was composed of the lncRNA plasmacytoma variant translocation 1, miR-455-5p, and the mRNAs, suppressor of cytokine signaling 3 and Plexin C1. To the best of our knowledge, only one lncRNA microarray has been performed with a comprehensive analysis (49). There has been no mechanistic study focusing on specific lncRNAs in pulpitis. Thus, more research is required to explore the regulatory role of specific lncRNAs in pulpitis.

Periodontitis

Periodontitis is an inflammatory disease, which is primarily caused by bacterial infection (52). It can cause inflammation of the gingivae, loss of alveolar bone and loss of attachment (53). Tooth loss, which occurs in adults is largely due to periodontitis (54). In 2015, Zou et al (55) revealed for the first time that lncRNAs have critical roles in the pathogenesis of periodontitis. A total of 2 pairs of chronic periodontitis gingival samples and adjacent healthy samples were collected for lncRNA analysis and 8,925 DE lncRNAs were detected, of which 4,313 were upregulated and 4,612 were downregulated. Functional analysis of the nearby protein-coding genes revealed that different lncRNAs can regulate a common gene, and a single lncRNA can be regulated by different genes. Thus, lncRNAs might play crucial and dual roles in periodontitis.

Chen et al (4) found that lncRNA FGD5 antisense RNA1 (FGD5-AS1) was downregulated in the gingival samples from patients with chronic periodontal compared with that in healthy samples. FGD5-AS1 inhibited NF-κB signaling via the FGD5-AS1/miR-142-3p/suppressor of cytokine signaling 6 (SOCS6) ceRNA network and subsequently reduced the secretion of TNF-α, IL-6, IL-1β and IL-8. Thus, the axis may provide a promising strategy for the treatment of periodontitis.

Several lncRNAs inhibit periodontitis; however, results from recent research have revealed that lncRNAs could promote the inflammatory process of periodontal-derived cells, including MALAT1 (7) and papillary thyroid carcinoma susceptibility candidate 3 (56). Li et al (7), explored the role of MALAT1 in inflammatory cytokine production in HGFs. The study indicated that MALAT1 bound to miR-20a, as a ceRNA and consequently led to increased mRNA levels of TLR4, which contributed to the activation of inflammation. Therefore, the effect and mechanism of MALAT1 in periodontal inflammation have been characterized. SS. SS is a chronic systemic autoimmune disease characterized by reduced secretions of the salivary and lacrimal glands and associated neuroendocrine disturbances (57). The disturbances of neuroendocrine include release of hormones (i.e., glucocorticoids) via the hypothalamic-pituitary-adrenal axis stimulation, production of mediators within the sympathetic innervation of immune organs (i.e., thymus) and production of proinflammatory cytokines (i.e., IL-2 and TNF-α) during the inflammatory response (58). The primary hallmark of SS is the infiltration of inflammatory mediators and cells, particularly T and B cells into the salivary and lacrimal glands (59). In addition, gland tissues have damaged acinar cells, fibrosis and increased adiposity with severe inflammatory lesions (60,61). Therefore, exploration into the relevant molecular mechanisms underlying SS is required.

In 2019, Dolcino et al (62) performed a high-throughput gene and lncRNA expression profiling in peripheral blood mononuclear cell samples from 8 patients with primary SS (pSS) and 8 healthy subjects. Among the 199 lncRNAs that were identified, CTD-2020K17.1, LINC00511 and LINC00657 and their target genes were found to be involved in apoptosis, immune response, cell proliferation, and several proinflammatory pathways. Shi et al (63), compared the expression profiles of lncRNAs from labial salivary glands between patients with pSS and healthy individuals. The gene ontology and pathway analysis results found 28 DE mRNAs associated with 8 DE lncRNAs were involved in chemokine signaling pathways, the NF-κB signaling pathway, and the TNF signaling pathway. Taken together, the results suggest that samples from multiple tissues could be utilized for investigating the same oral autoimmune disease. A comparison of DE lncRNA sets from different sample types of the same disease could provide valuable clues to the discovery of novel therapeutic targets to treat oral autoimmune diseases.

Subsequently, Xin et al (5) demonstrated that lncRNA myocardial infarction associated transcript 2 (Mirt2) reduces apoptosis and inflammatory levels in interferon (IFN)-γ-induced inflammation in salivary gland epithelial cells. It is hypothesized that Mirt2 might block NF-κB, and Janus kinase (JAK)/STAT3 signaling by increasing miR-377 expression levels.

Research investigating lncRNAs typically involves the fields of medicine and biology; however, research regarding lncRNAs in oral inflammatory disorders is still in the early stages. To date, the collection of clinical oral tissue samples and subsequent microarray or sequencing analysis has been the primary method of investigation in this field. In periodontitis, the majority of lncRNAs serve as ceRNAs (4,6,7). There are a high number of research models investigating oral tissue-derived cells treated with related inflammatory stimuli (i.e., lipopolysaccharide and IFN-γ) (5,7). Periodontitis animal models and lncRNAs have been well-established (64-66), while animal models involving lncRNAs in other oral inflammatory disorders requires further investigation. Thus, additional research is required to determine the mechanism of lncRNAs in oral inflammatory disorders.

4. Role of lncRNAs in PPOELs

OSF

OSF is a chronic, occult oral mucosal disease associated with chewing betel nut, characterized by a juxta-epithelial inflammatory response followed by generalized submucosal fibrosis (67,68). As a result, OSF typically leads to difficulty in opening the mouth and an increased malignant transformation rate (69,70). Therefore, the identification of molecules associated with OSF pathological progression is urgent. In the course of OSF, endothelial dysfunction may be accompanied by the dysregulation of multiple lncRNAs (71). For example, lncRNA growth arrest specific 5 antisense 1 (GAS5-AS1) was inhibited, while lncRNA hypoxia-inducible factor 1-α antisense RNA 1 (HIF1A-AS1) was upregulated during the development of OSF (72,73). Research into lncRNAs has increased in the last 5 years; however, the function of numerous lncRNAs in OSF remains unclear. To date, only four articles have illustrated the relative issues (72-75).

The lncRNA sequencing conducted in 2019 by Zhou et al (75) included 13 normal mucous samples, 10 OSF samples, and 20 OSCC samples. A total of 5 DE candidate lncRNAs were found to participate in the inflammatory signaling pathway and contributed to inflammatory and fibro-elastic pathogenetic changes by deregulating their cis-target and trans-target genes in OSF malignant development. Further functional analysis of these lncRNAs is required to provide conclusive evidence supporting an underlying regulatory mechanism during OSF.

Fang et al (74), determined that arecoline-induced myofibroblast trans-differentiation occurred via LINC00974-mediated activation of the transforming growth factor-β (TGF-β)/Smad signaling pathway. According to their study, collagen gel contractility and myofibroblast migration ability was increased in fibrotic buccal mucosal fibroblasts (fBMFs) overexpressing LINC00974. Increased expression of another lncRNAHIF1A-AS1 also positively modulates the TGF-β/Smad signaling pathway, similar to LINC00974 (72). However, the lncRNA GAS5-AS1 presented contrary results in arecoline-treated BMFs and fBMFs (73).

OLP

OLP is a chronic inflammatory disease affecting the oral mucosa with characteristic relapses and remissions (76-78). Emerging evidence shows that OLP may be premalignant (79). Unstable molecular changes can induce the production of several inflammatory cytokines and subsequently contribute to the course of OLP (77,80). For example, pathogen associated molecular patterns and adaptor molecules (i.e., myeloid differentiation factor 88) activation leads to nuclear translocation of NF-κB and augments the transcription of inflammatory genes (i.e., IL-6 and IL-8) (81). As with OSF, there have only been two studies on the role of lncRNAs in OLP to date.

Yang et al (82), examined DE genes and lncRNA targets in human papillomavirus-related OSCC (n=1), normal (n=1), and OLP (n=1) samples. Of the identified lncRNAs, most (697; 76.2%) were intergenic lncRNAs, followed by 151 sense lncRNAs (16.5%). Keratinization and MHC class I antigen processing and presentation were significantly enriched by OSCC-associated DE genes and lncRNA targets, and the olfactory transduction pathway was enriched by OLP- and OSCC-related DE genes. To the best of our knowledge, this has been the only study investigating the lncRNA profile of OLP so far; however, the number of tissue samples was too small to guarantee the validity of these findings. In addition, there is evidence suggesting that lncRNA DQ786243 significantly enhances the expression levels of miR-146a by inducing Forkhead box P3 (Foxp3), which subsequently blocks NF-κB signaling during OLP. Moreover, Foxp3+ regulatory T cells significantly suppressed the function of other CD4+ T cells, such as CD4+IL-1+ helper T cells and CD4+IL-17+ helper T cells, by inhibiting the mRNA expression levels of IFN-γ and IL-17 (83).

LncRNA microarray analysis is considered a reasonable option for comparing and determining DE lncRNAs in normal, PPOEL and OSCC tissues, which are difficult to simultaneously acquire in one patient. Animal models which investigate both lncRNAs and PPOELs are expected to be explored extensively, and to primarily include fBMFs treated with related stimuli (i.e., arecoline). Mechanistically, the classical signaling pathways in PPOELs focus on the TGF-β/Smad and NF-κB signaling pathways. Therefore, it is worth exploring more regulatory mechanisms [i.e., p38/MAPK and JAK/STAT3 signaling pathways] in subsequent studies on PPOELs.

5. Role of lncRNAs in OSCC

Oral cancers are cancers that exist in the oral cavity, such as the mucosal surfaces of the lips, floor of the mouth, tongue, buccal mucosa, lower and upper gingival surfaces, hard palate, and retromolar trigone (84). The histology of oral cancers varies widely; the majority of them are OSCCs (84). An increasing number of reports have revealed that lncRNAs play a broad role in the oncogenesis and progression of OSCC through transcriptional regulation, posttranscriptional modulation and epigenetic modifications (85). LncRNAs, functioning in oral cancer migration, epithelial-mesenchymal transition (EMT), metastasis, progression and invasion (86), could serve as biomarkers or therapeutic targets for OSCC diagnosis, prognosis and treatment (87).

With the development of whole transcriptome analyses, including serial analyses of gene expression, RNA sequencing and microarray data, several oral cancer-associated lncRNAs have been identified. In 2019, Qiu et al (88) screened 2,294 DE lncRNAs (933 upregulated and 1,361 downregulated) in OSCC tissues (n=72) compared with paired adjacent normal tissues. A total of four lncRNA-mRNA coexpression networks were constructed, and low expression levels of the four lncRNA nodes contributed to poor median progression-free survival and overall survival. This study provided novel insights into the role of lncRNAs in OSCC.

LncRNAs could transcriptionally regulate the progression of OSCC via interactions with proteins or interactions with RNA and DNA molecules (85). Zhu et al (89), verified that hypoxia induces the overexpression of hyaluronan synthase 2 antisense 1 (HAS2-AS1) in a HIF-1α- and NF-κB-dependent manner. HAS2-AS1 mediates hypoxia-induced EMT and invasiveness of OSCC cells by binding and stabilizing the HAS2 gene. Additional research found that LINC01133 inhibited OSCC cell migration and invasion by inhibiting growth and differentiation factor 15 (GDF15) protein expression and formed a feedback regulatory loop with GDF15 (90). Kong et al (91), found that expression of lncRNA FOXC1 upstream transcript, which is the adjacent promoter upstream of the Fork head box C1 (FOXC1) gene, was positively correlated with FOXC1 mRNA expression and promoted OSCC proliferation and migration. Thus, a vast amount of research has elucidated transcriptional regulatory mechanisms to address the role of lncRNAs in OSCC.

Posttranscriptional regulation through pre-mRNA alternative splicing, mRNA decay acceleration, mRNA protection, or translational activation or repression is also one of the primary mechanisms of lncRNAs (92). Chang and Hu (93), demonstrated that MALAT1 could function as a ceRNA to mediate STAT3 expression by sponging miR-125b in OSCC. OSCC cell viability and growth were enhanced by increasing expression levels of MALAT1, and a role of MALAT1/miR-125b/STAT3 axis was confirmed in vivo using a nude mouse xenograft model with OSCC Tca8113 cells. Fang et al (94), found that lncRNA urothelial cancer associated 1 (UCA1) facilitated proliferation, enhanced cisplatin chemoresistance, and suppressed apoptosis in OSCC cells by suppressing miR-184 expression to increase the mRNA expression levels of splicing factor 1. Thus, ceRNA appears to be a promising posttranscriptional regulatory mechanism in OSCC.

In addition, lncRNAs could affect the characteristics of OSCC through epigenetic modifications, including DNA methylation, chromatin modification and imprinting (95). LncRNA HOX transcript antisense RNA (HOTAIR) repressed the expression of E-cadherin by binding to enhancer of zeste homolog 2, the enzymatic component of polycomb repressive complex 2 (PRC2) and H3K27me3 at the E-cadherin promoter (96). HOTAIR silenced transcription factors by interacting with PRC2, lysine-specific histone demethylase-1 and RE-1 elements, leading to chromatin remodelling, thus trans-inhibiting the expression of the homeobox D cluster gene and promoting the occurrence, invasion and metastasis of tumors (96). Additional lncRNAs, participating in the progression of OSCC through epigenetic modifications, are expected to be detected, as there is an increasing number of the research being performed (95,97).

Accumulating evidence has shown that lncRNAs modulate the metastasis, proliferation, invasiveness and migration of oral cancer, especially OSCC, in cellular physiological processes (86,98,99). As aforementioned, lncRNAs interact with different cellular macromolecules, including chromatin, protein and RNA (36). According to the aforementioned studies, unstimulated OSCC cell lines are typically used for external models. Animal models are also well-established to provide more convincing evidence to confirm the role of lncRNAs in OSCC (89,93,94). This suggests that lncRNAs have the potential to be prognostic and therapeutic markers, providing valid approaches for clinical treatment.

6. Conclusion

With the advent of genomic technologies, including micro-arrays and RNA sequencing, investigations of lncRNA genomic profiles have been widely performed in the last 5 years (6,100,101). A large number of studies include micro-array analyses of clinical tissue samples, which enhances the clinical value and significance of the findings (48,55,88). The present review not only summarized the lncRNA profiles but also elucidated the potential underlying mechanisms of lncRNAs in oral diseases (Table II and Fig. 3). These mechanisms may be significant to the clinical diagnosis and therapy of oral diseases. Moreover, the present review focused on the identification and associations of oral inflammatory disorders, PPOELs and OSCC and discussed the functions of lncRNAs in the pathological process. By summarizing the studies involving the mechanisms of lncRNAs in oral diseases, ceRNA regulation was found to be the most common, while lncRNAs interacting with proteins is relatively rare. Thus, additional research is required to expand the early findings and characterize the mechanisms of DE lncRNAs. Overall, lncRNAs, the novel candidates in oral inflammatory disorders, PPOELs and oral cancer, should be investigated further, and their diagnostic and therapeutic functions may have significant value.

Table II

Regulatory effect of lncRNAs in oral inflammatory disorders, PPOELs and OSCC.

Table II

Regulatory effect of lncRNAs in oral inflammatory disorders, PPOELs and OSCC.

lncRNACell categoryCell stimulation (bacteria)DiseaseEffectsModes of actionAssociated targets or pathways(Refs.)
FGD5-AS1PDLCsLPS (P. g)PeriodontitisInhibit apoptosis and reduce inflammatory cytokine productionSponging miR-142-3pSOCS6 and NF-κB(37)
MALAT1HGFsLPS (P. g or E. coli)Periodontitis inflammatoryEnhance cytokine productionSponging miR-20aTLR4(7)
Mirt2SGECsIFN-γSSRepress apoptosis inflammatory cytokine productionFacilitate miR-377 and enhanceJAK/STAT3 and NF-κB(5)
LINC00974fBMFs-OSFIncrease myofibroblasts activation-TGF-β/Smad(55)
HIF1A-AS1fBMFs-OSFIncrease myofibroblasts activation--(53)
GAS5-AS1fBMFsArecolineOSFInhibit myofibroblasts activities in OSFInhibiting p-Smad and α-SMATGF-β/Smad(54)
DQ786243CD4+ Treg cells-OLPSuppress the function of CD4+ T cells such as Th1 and Th17Elevating Foxp3miR-146a/NF-κB(63)
HAS2-AS1Human OSCC cell linesHypoxiaOSCCMediate hypoxia-induced EMT and invasiveness of OSCCStabilizing HAS2HF-1α and NF-κB(69)
LINC01133Human OSCC cell lines-OSCCInhibit OSCC cell migration and invasion-GDF15(70)
FOXCUTHuman OSCC cell lines-OSCCPromote OSCC proliferation and migration ability-FOXC1(71)
MALAT1Human OSCC cell lines-OSCCAccelerates EMT and development of OSCCSponging miR-125bSTAT3(73)
UCA1Human OSCC cell lines-OSCCFacilitate proliferation, enhance CDDP chemoresistance, and suppresses apoptosisSponging miR-184SF1(74)
HOTAIRHuman OSCC cell lines-OSCCPromote occurrence, invasion and metastasisEZH2 and H3K27me3E-cadherin(76)

[i] lnc, long non-coding; miR, microRNA; PDLCs, periodontal ligament cells; HGFs, human gingival fibroblasts; SGECs, salivary gland epithelial cells; fBMFs, fibrotic buccal mucosal fibroblasts; OSCC, oral squamous cell carcinoma; OLP, oral lichen planus; OSF, oral submucous fibrosis; SS, Sjögren's syndrome; LPS, lipopolysaccharide; IFN, interferon; HIF1-α, hypoxia-inducible factor 1-α; Mirt2, myocardial infarction associated transcript 2; FGD5-AS1, FGD5 antisense RNA 1; MALAT1, metastasis associated lung adenocarcinoma transcript 1; HOTAIR, HOX transcript antisense RNA; TLR4, Toll-like receptor 4; HAS2, hyaluronan synthase 2; FOXC1, Fork head box C1; SOCS6, suppressor of cytokine signaling 6; NF-κB, nuclear factor-κ B; TGF-β, transforming growth factor-β; EMT, epithelial-mesenchymal transition; CDDP, cis-diamine dichloroplatinum; p, phosphorylated.

Funding

This study was supported by the National Natural Science Foundation of China (grant nos. 81600882 and 81870755), a China Postdoctoral Science Foundation funded project (grant no. 2019M663009) and the President Foundation of Nanfang Hospital, Southern Medical University (grant no. 2019B002).

Availability of data and materials

Not applicable.

Authors' contributions

FF and BW contributed to the conception and design of the review. KZ contributed to the writing and drafting of the manuscript. FF and WQ contributed to the critical revision of the manuscript for important intellectual content. All the authors have given approval of the final version to be published and agree to be accountable for all aspects of the work.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Brogden KA, Johnson GK, Vincent SD, Abbasi T and Vali S: Oral inflammation, a role for antimicrobial peptide modulation of cytokine and chemokine responses. Expert Rev Anti Infect Ther. 11:1097–1113. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Grivennikov SI, Greten FR and Karin M: Immunity, inflammation, and cancer. Cell. 140:883–899. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Kulkarni V, Uttamani JR, Naqvi AR and Nares S: microRNAs: Emerging players in oral cancers and inflammatory disorders. Tumour Biol. 39:10104283176983792017. View Article : Google Scholar : PubMed/NCBI

4 

Chen H, Lan Z, Li Q and Li Y: Abnormal expression of long noncoding RNA FGD5-AS1 affects the development of periodontitis through regulating miR-142-3p/SOCS6/NF-kB pathway. Artif Cells Nanomed Biotechnol. 47:2098–2106. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Xin M, Liang H, Wang H, Wen D, Wang L, Zhao L, Sun M and Wang J: Mirt2 functions in synergy with miR-377 to participate in inflammatory pathophysiology of sjogren's syndrome. Artif Cells Nanomed Biotechnol. 47:2473–2480. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Li S, Liu X, Li H, Pan H, Acharya A, Deng Y, Yu Y, Haak R, Schmidt J, Schmalz G and Ziebolz D: Integrated analysis of long noncoding RNA-associated competing endogenous RNA network in periodontitis. J Periodontal Res. 53:495–505. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Li J, Wang M, Song L, Wang X, Lai W and Jiang S: LncRNA MALAT1 regulates inflammatory cytokine production in lipopolysaccharide-stimulated human gingival fibroblasts through sponging miR-20a and activating TLR4 pathway. J Periodontal Res. 55:182–190. 2020. View Article : Google Scholar

8 

Lee PH, Chu PM, Hsieh PL, Yang HW, Chueh PJ, Huang YF, Liao YW and Yu CC: Glabridin inhibits the activation of myofi-broblasts in human fibrotic buccal mucosal fibroblasts through TGF-β/smad signaling. Environ Toxicol. 33:248–255. 2018. View Article : Google Scholar

9 

Ganesh D, Sreenivasan P, Ohman J, Wallström M, Braz-Silva PH, Giglio D, Kjeller G and Hasséus B: Potentially malignant oral disorders and cancer transformation. Anticancer Res. 38:3223–3229. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Awadallah M, Idle M, Patel K and Kademani D: Management update of potentially premalignant oral epithelial lesions. Oral Surg Oral Med Oral Pathol Oral Radiol. 125:628–636. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Han X, Wei YB, Tian G, Tang Z, Gao JY and Xu XG: Screening of crucial long non-coding RNAs in oral epithelial dysplasia by serial analysis of gene expression. Genet Mol Res. 14:11729–11738. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Chen L, Zhou Y and Li H: LncRNA, miRNA and lncRNA-miRNA interaction in viral infection. Virus Res. 257:25–32. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Chattopadhyay E, Singh R, Ray A, Roy R, Sarkar ND, Paul RR, Pal M, Aich R and 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. 6:327352016. View Article : Google Scholar : PubMed/NCBI

14 

Fiaschi T and Chiarugi P: Oxidative stress, tumor microenvironment, and metabolic reprogramming: A diabolic liaison. Expression deregulation of mir31 and CXCL12 in two types of oral precancers and cancer: Importance in progression of precancer and cancer. Int J Cell Biol. 2012:7628252012. View Article : Google Scholar

15 

Naylor MS, Stamp GW, Foulkes WD, Eccles D and Balkwill FR: Tumor necrosis factor and its receptors in human ovarian cancer. Potential role in disease progression. J Clin Invest. 91:2194–2206. 1993. View Article : Google Scholar : PubMed/NCBI

16 

Yang G, Lu X and Yuan L: LncRNA: A link between RNA and cancer. Biochim Biophys Acta. 1839:1097–1109. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Yang ZH, Dang YQ and Ji G: Role of epigenetics in transformation of inflammation into colorectal cancer. World J Gastroenterol. 25:2863–2877. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Katsanos KH, Roda G, Brygo A, Delaporte E and Colombel JF: Oral cancer and oral precancerous lesions in inflammatory bowel diseases: A systematic review. J Crohns Colitis. 9:1043–1052. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Jia H, Wang X and Sun Z: Exploring the long noncoding RNAs-based biomarkers and pathogenesis of malignant trans-formation from dysplasia to oral squamous cell carcinoma by bioinformatics method. Eur J Cancer Prev. 29:174–181. 2020. View Article : Google Scholar

20 

Camacho CV, Choudhari R and Gadad SS: Long noncoding RNAs and cancer, an overview. Steroids. 133:93–95. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Zhang J and Chu M: Targeting of IL-6-relevant long noncoding RNA profiles in inflammatory and tumorous disease. Inflammation. 42:1139–1146. 2019. View Article : Google Scholar : PubMed/NCBI

22 

St LG, Wahlestedt C and Kapranov P: The landscape of long noncoding RNA classification. Trends Genet. 31:239–251. 2015. View Article : Google Scholar

23 

Jarroux J, Morillon A and Pinskaya M: History, discovery, and classification of lncRNAs. Adv Exp Med Biol. 1008:1–46. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Guenzl PM and Barlow DP: Macro lncRNAs: A new layer of cis-regulatory information in the mammalian genome. RNA Biol. 9:731–741. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Beltrami C, Angelini TG and Emanueli C: Noncoding RNAs in diabetes vascular complications. J Mol Cell Cardiol. 89:42–50. 2015. View Article : Google Scholar

26 

Allen MA, Andrysik Z, Dengler VL, Mellert HS, Guarnieri A, Freeman JA, Sullivan KD, Galbraith MD, Luo X, Kraus WL, et al: Global analysis of p53-regulated transcription identifies its direct targets and unexpected regulatory mechanisms. Elife. 3:e22002014. View Article : Google Scholar

27 

Xu Z, Wei W, Gagneur J, Perocchi F, Clauder-Münster S, Camblong J, Guffanti E, Stutz F, Huber W and Steinmetz LM: Bidirectional promoters generate pervasive transcription in yeast. Nature. 457:1033–1037. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Neil H, Malabat C, D'Aubenton-Carafa Y, Xu Z, Steinmetz LM and Jacquier A: Widespread bidirectional promoters are the major source of cryptic transcripts in yeast. Nature. 457:1038–1042. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Fox MJ, Gao H, Smith-Kinnaman WR, Liu Y and Mosley AL: The exosome component Rrp6 is required for RNA polymerase II termination at specific targets of the Nrd1-Nab3 pathway. PLoS Genet. 11:e10049992015. View Article : Google Scholar : PubMed/NCBI

30 

Yu D, Ma X, Zuo Z, Wang H and Meng Y: Classification of tran-scription boundary-associated RNAs (TBARs) in animals and plants. Front Genet. 9:1682018. View Article : Google Scholar

31 

Bianchessi V, Badi I, Bertolotti M, Nigro P, D'Alessandra Y, Capogrossi MC, Zanobini M, Pompilio G, Raucci A and Laur A: The mitochondrial lncRNA ASncmtRNA-2 is induced in aging and replicative senescence in endothelial cells. J Mol Cell Cardiol. 81:62–70. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Werner MS, Sullivan MA, Shah RN, Nadadur RD, Grzybowski AT, Galat V, Moskowitz IP and Ruthenburg AJ: Chromatin-enriched lncRNAs can act as cell-type specific activators of proximal gene transcription. Nat Struct Mol Biol. 24:596–603. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Ferdin J, Nishida N, Wu X, Nicoloso MS, Shah MY, Devlin C, Ling H, Shimizu M, Kumar K, Cortez MA, et al: HINCUTs in cancer: Hypoxia-induced noncoding ultraconserved transcripts. Cell Death Differ. 20:1675–1687. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Giannakakis A, Zhang J, Jenjaroenpun P, Nama S, Zainolabidin N, Aau MY, Yarmishyn AA, Vaz C, Ivshina AV, Grinchuk OV, et al: Contrasting expression patterns of coding and noncoding parts of the human genome upon oxidative stress. Sci Rep. 5:97372015. View Article : Google Scholar : PubMed/NCBI

35 

Lazorthes S, Vallot C, Briois S, Aguirrebengoa M, Thuret JY, St Laurent G, Rougeulle C, Kapranov P, Mann C, Trouche D and Nicolas E: A vlincRNA participates in senescence maintenance by relieving H2AZ-mediated repression at the INK4 locus. Nat Commun. 6:59712015. View Article : Google Scholar : PubMed/NCBI

36 

Dahariya S, Paddibhatla I, Kumar S, Raghuwanshi S, Pallepati A and Gutti RK: Long non-coding RNA: Classification, biogenesis and functions in blood cells. Mol Immunol. 112:82–92. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Chen LL and Carmichael GG: Decoding the function of nuclear long non-coding RNAs. Curr Opin Cell Biol. 22:357–364. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Fang F, Zhang K, Chen Z and Wu B: Noncoding RNAs: New insights into the odontogenic differentiation of dental tissue-derived mesenchymal stem cells. Stem Cell Res Ther. 10:2972019. View Article : Google Scholar : PubMed/NCBI

39 

Geng F, Liu J, Guo Y, Li C, Wang H, Wang H, Zhao H and Pan Y: Persistent exposure to porphyromonas gingivalis promotes proliferative and invasion capabilities, and tumorigenic proper-ties of human immortalized oral epithelial cells. Front Cell Infect Microbiol. 7:572017. View Article : Google Scholar

40 

Song Y, Pan Y and Liu J: Functional analysis of lncRNAs based on competitive endogenous RNA in tongue squamous cell carcinoma. PeerJ. 7:e69912019. View Article : Google Scholar : PubMed/NCBI

41 

Ranzani V, Rossetti G, Panzeri I, Arrigoni A, Bonnal RJ, Curti S, Gruarin P, Provasi E, Sugliano E, Marconi M, et al: The long intergenic noncoding RNA landscape of human lymphocytes highlights the regulation of T cell differentiation by linc-MAF-4. Nat Immunol. 16:318–325. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Zhang X, Lian Z, Padden C, Gerstein MB, Rozowsky J, Snyder M, Gingeras TR, Kapranov P, Weissman SM and Newburger PE: A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster. Blood. 113:2526–2534. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Elling R, Chan J and Fitzgerald KA: Emerging role of long noncoding RNAs as regulators of innate immune cell development and inflammatory gene expression. Eur J Immunol. 46:504–512. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Heward JA and Lindsay MA: Long non-coding RNAs in the regulation of the immune response. Trends Immunol. 35:408–419. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Qu Q, Fang F, Wu B, Hu Y, Chen M, Deng Z, Ma D, Chen T, Hao Y and Ge Y: Potential role of long non-coding RNA in osteogenic differentiation of human periodontal ligament stem cells. J Periodontol. 8:e127–e137. 2016. View Article : Google Scholar

46 

Bjørndal L, Simon S, Tomson PL and Duncan HF: Management of deep caries and the exposed pulp. Int Endod J. 52:949–973. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Hui T, Wang C, Chen D, Zheng L, Huang D and Ye L: Epigenetic regulation in dental pulp inflammation. Oral Dis. 23:22–28. 2017. View Article : Google Scholar

48 

Huang X and Chen K: Differential expression of long noncoding RNAs in normal and inflamed human dental pulp. J Endod. 44:62–72. 2018. View Article : Google Scholar

49 

Lei F, Zhang H and Xie X: Comprehensive analysis of an lncRNA-miRNA-mRNA competing endogenous RNA network in pulpitis. PeerJ. 7:e71352019. View Article : Google Scholar : PubMed/NCBI

50 

Zhong S, Zhang S, Bair E, Nares S and Khan AA: Differential expression of microRNAs in normal and inflamed human pulps. J Endod. 38:746–752. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Galicia JC, Henson BR, Parker JS and Khan AA: Gene expression profile of pulpitis. Genes Immun. 17:239–243. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Mombelli A: Microbial colonization of the periodontal pocket and its significance for periodontal therapy. Periodontol 2000. 76:85–96. 2018. View Article : Google Scholar

53 

Singhrao SK, Harding A, Poole S, Kesavalu L and Crean S: Porphyromonas gingivalis periodontal infection and its putative links with Alzheimer's disease. Mediators Inflamm. 2015:1373572015. View Article : Google Scholar : PubMed/NCBI

54 

Michaud DS, Fu Z, Shi J and Chung M: Periodontal disease, tooth loss, and cancer risk. Epidemiol Rev. 39:49–58. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Zou Y, Li C, Shu F, Tian Z, Xu W, Xu H, Tian H, Shi R and Mao X: lncRNA expression signatures in periodontitis revealed by microarray: The potential role of lncRNAs in periodontitis pathogenesis. J Cell Biochem. 116:640–647. 2015. View Article : Google Scholar

56 

Liu W, Zheng Y, Chen B, Ke T and Shi Z: LncRNA papillary thyroid carcinoma susceptibility candidate 3 (PTCSC3) regulates the proliferation of human periodontal ligament stem cells and toll-like receptor 4 (TLR4) expression to improve periodontitis. BMC Oral Health. 19:1082019. View Article : Google Scholar : PubMed/NCBI

57 

Malathi N, Mythili S and Vasanthi HR: Salivary diagnostics: A brief review. ISRN Dent. 2014:1587862014.PubMed/NCBI

58 

Tzioufas AG, Tsonis J and Moutsopoulos HM: Neuroendocrine dysfunction in Sjogren's syndrome. Neuroimmunomodulation. 15:37–45. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Zoukhri D: Effect of inflammation on lacrimal gland function. Exp Eye Res. 82:885–898. 2006. View Article : Google Scholar :

60 

Gliozzi M, Greenwell-Wild T, Jin W, Moutsopoulos NM, Kapsogeorgou E, Moutsopoulos HM and Wahl SM: A link between interferon and augmented plasmin generation in exocrine gland damage in Sjögren's syndrome. J Autoimmun. 40:122–133. 2013. View Article : Google Scholar

61 

Reksten TR, Jonsson MV, Szyszko EA, Brun JG, Jonsson R and Brokstad KA: Cytokine and autoantibody profiling related to histopathological features in primary Sjogren's syndrome. Rheumatology (Oxford). 48:1102–1106. 2009. View Article : Google Scholar

62 

Dolcino M, Tinazzi E, Vitali C, Del PN, Puccetti A and Lunardi C: Long non-coding RNAs modulate Sjögren's syndrome associated gene expression and are involved in the pathogenesis of the disease. J Clin Med. 8:13492019. View Article : Google Scholar

63 

Shi H, Cao N, Pu Y, Xie L, Zheng L and Yu C: Long non-coding RNA expression profile in minor salivary gland of primary Sjögren's syndrome. Arthritis Res Ther. 18:1092016. View Article : Google Scholar

64 

Jia B, Qiu X, Chen J, Sun X, Zheng X, Zhao J, Li Q and Wang Z: A feed-forward regulatory network lncPCAT1/miR-106a-5p/E2F5 regulates the osteogenic differentiation of periodontal ligament stem cells. J Cell Physiol. 234:19523–19538. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Huang Y, Han Y, Guo R, Liu H, Li X, Jia L, Zheng Y and Li W: Long non-coding RNA FER1L4 promotes osteogenic differentiation of human periodontal ligament stromal cells via miR-874-3p and vascular endothelial growth factor A. Stem Cell Res Ther. 11:52020. View Article : Google Scholar : PubMed/NCBI

66 

Wang L, Wu F, Song Y, Li X, Wu Q, Duan Y and Jin Z: Long noncoding RNA related to periodontitis interacts with miR-182 to upregulate osteogenic differentiation in periodontal mesenchymal stem cells of periodontitis patients. Cell Death Dis. 7:e23272016. View Article : Google Scholar : PubMed/NCBI

67 

Canniff JP, Harvey W and Harris M: Oral submucous fibrosis: Its pathogenesis and management. Br Dent J. 160:429–434. 1986. View Article : Google Scholar : PubMed/NCBI

68 

Tilakaratne WM, Klinikowski MF, Saku T, Peters TJ and Warnakulasuriya S: Oral submucous fibrosis: Review on aetiology and pathogenesis. Oral Oncol. 42:561–568. 2006. View Article : Google Scholar

69 

Sharma M and Radhakrishnan R: Limited mouth opening in oral submucous fibrosis: Reasons, ramifications, and remedies. J Oral Pathol Med. 46:424–430. 2017. View Article : Google Scholar

70 

Arakeri G, Patil SG, Aljabab AS, Lin KC, Merkx MAW, Gao S and Brennan PA: Oral submucous fibrosis: An update on pathophysiology of malignant transformation. J Oral Pathol Med. 46:413–417. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Sharma M, Shetty SS and Radhakrishnan R: Oral submucous fibrosis as an overhealing wound: Implications in malignant transformation. Recent Pat Anticancer Drug Discov. 13:272–291. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Wang YK, Liu CM, Lin T, Fang CY, Yu CC and Yu CH: Inhibition of HIF1A-AS1 impedes the arecoline-induced migration activity of human oral mucosal fibroblasts. J Formos Med Assoc. 119:879–883. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Lin CY, Liao YW, Hsieh PL, Lu MY, Peng CY, Chu PM, Yang HW, Huang YF, Yu CC and Yu CH: LncRNA GAS5-AS1 inhibits myofibroblasts activities in oral submucous fibrosis. J Formos Med Assoc. 117:727–733. 2018. View Article : Google Scholar

74 

Fang CY, Yu CC, Liao YW, Hsieh PL, Lu MY, Lin KC, Wu CZ and Tsai LL: LncRNA LINC00974 activates TGF-β/Smad signaling to promote oral fibrogenesis. J Oral Pathol Med. 48:151–158. 2019.

75 

Zhou S, Zhu Y, He Z, Zhang D, Guo F, Jian X and Zhang C: Long non-coding RNA expression profile associated with malignant progression of oral submucous fibrosis. J Oncol. 2019:68351762019. View Article : Google Scholar : PubMed/NCBI

76 

Lodi G, Scully C, Carrozzo M, Griffiths M, Sugerman PB and Thongprasom K: Current controversies in oral lichen planus: Report of an international consensus meeting. Part 1. Viral infections and etiopathogenesis. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 100:40–51. 2005. View Article : Google Scholar : PubMed/NCBI

77 

Eisen D, Carrozzo M, Bagan SJ and Thongprasom K: Number V oral lichen planus: Clinical features and management. Oral Dis. 11:338–349. 2005. View Article : Google Scholar : PubMed/NCBI

78 

Scully C, Beyli M, Ferreiro MC, Ficarra G, Gill Y, Griffiths M, Holmstrup P, Mutlu S, Porter S and Wray D: Update on oral lichen planus: Etiopathogenesis and management. Crit Rev Oral Biol Med. 9:86–122. 1998. View Article : Google Scholar : PubMed/NCBI

79 

Lončar-Brzak B, Klobučar M, Veliki-Dalic I, Sabol I, Pavelić SK, Krušlin B and Mravak-Stipetić M: Expression of small leucine-rich extracellular matrix proteoglycans biglycan and lumican reveals oral lichen planus malignant potential. Clin Oral Investig. 22:1071–1082. 2018. View Article : Google Scholar

80 

Santoro A, Majorana A, Bardellini E, Festa S, Sapelli P and Facchetti F: NF-kappaB expression in oral and cutaneous lichen planus. J Pathol. 201:466–472. 2003. View Article : Google Scholar : PubMed/NCBI

81 

Groeger S and Meyle J: Oral mucosal epithelial cells. Feonr Immunol. 10:2082019.

82 

Yang Q, Xu B, Sun H, Wang X, Zhang J, Yu X and Ma X: A genome-wide association scan of biological processes involved in oral lichen planus and oral squamous cell carcinoma. Medicine (Baltimore). 96:e70122017. View Article : Google Scholar

83 

Wang J, Zhai X, Guo J, Li Y, Yang Y, Wang L, Yang L and Liu F: Long non-coding RNA DQ786243 modulates the induction and function of CD4(+) Treg cells through Foxp3-miR-146a-NF-kB axis: Implications for alleviating oral lichen planus. Int Immunopharmacol. 75:1057612019. View Article : Google Scholar

84 

Huang SH and O'Sullivan B: Oral cancer: Current role of radio-therapy and chemotherapy. Med Oral Patol Oral Cir Bucal. 18:e233–e240. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Gomes CC, de Sousa SF, Calin GA and Gomez RS: The emerging role of long noncoding RNAs in oral cancer. Oral Surg Oral Med Oral Pathol Oral Radiol. 123:235–241. 2017. View Article : Google Scholar

86 

Luo X, Qiu Y, Jiang Y, Chen F, Jiang L, Zhou Y, Dan H, Zeng X, Lei YL and Chen Q: Long non-coding RNA implicated in the invasion and metastasis of head and neck cancer: Possible function and mechanisms. Mol Cancer. 17:142018. View Article : Google Scholar : PubMed/NCBI

87 

Zhang L, Meng X, Zhu XW, Yang DC, Chen R, Jiang Y and Xu T: Long non-coding RNAs in Oral squamous cell carcinoma: Biologic function, mechanisms and clinical implications. Mol Cancer. 18:1022019. View Article : Google Scholar : PubMed/NCBI

88 

Qiu YL, Liu YH, Ban JD, Wang WJ, Han M, Kong P and Li BH: Pathway analysis of a genomewide association study on a long noncoding RNA expression profile in oral squamous cell carcinoma. Oncol Rep. 41:895–907. 2019.

89 

Zhu G, Wang S, Chen J, Wang Z, Liang X, Wang X, Jiang J, Lang J and Li L: Long noncoding RNA HAS2-AS1 mediates hypoxia-induced invasiveness of oral squamous cell carcinoma. Mol Carcinog. 56:2210–2222. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Kong J, Sun W, Zhu W, Liu C, Zhang H and Wang H: Long noncoding RNA LINC01133 inhibits oral squamous cell carcinoma metastasis through a feedback regulation loop with GDF15. J Surg Oncol. 118:1326–1334. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Kong XP, Yao J, Luo W, Feng FK, Ma JT, Ren YP, Wang DL and Bu RF: The expression and functional role of a FOXC1 related mRNA-lncRNA pair in oral squamous cell carcinoma. Mol Cell Biochem. 394:177–186. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Li Y, Zhang J, Pan J, Feng X, Duan P, Yin X, Xu Y, Wang X and Zou S: Insights into the roles of lncRNAs in skeletal and dental diseases. Cell Biosci. 8:82018. View Article : Google Scholar : PubMed/NCBI

93 

Chang SM and Hu WW: Long non-coding RNA MALAT1 promotes oral squamous cell carcinoma development via microRNA-125b/STAT3 axis. J Cell Physiol. 233:3384–3396. 2018. View Article : Google Scholar

94 

Fang Z, Zhao J, Xie W, Sun Q, Wang H and Qiao B: LncRNA UCA1 promotes proliferation and cisplatin resistance of oral squamous cell carcinoma by sunppressing miR-184 expression. Cancer Med. 6:2897–2908. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Gonzalez-Ramirez I, Soto-Reyes E, Sanchez-Perez Y, Herrera LA and Garcia-Cuellar C: Histones and long non-coding RNAs: The new insights of epigenetic deregulation involved in oral cancer. Oral Oncol. 50:691–695. 2014. View Article : Google Scholar : PubMed/NCBI

96 

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. 46:2586–2594. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Yang CM, Wang TH, Chen HC, Li SC, Lee MC, Liou HH, Liu PF, Tseng YK, Shiue YL, Ger LP and Tsai KW: Aberrant DNA hypermethylation-silenced SOX21-AS1 gene expression and its clinical importance in oral cancer. Clin Epigenetics. 8:1292016. View Article : Google Scholar : PubMed/NCBI

98 

Shao TR, Zheng ZN, Chen YC, Wu QQ, Huang GZ, Li F, Zeng WS and Lv XZ: LncRNA AC007271.3 promotes cell proliferation, invasion, migration and inhibits cell apoptosis of OSCC via the Wnt/β-catenin signaling pathway. Life Sci. 239:1170872019. View Article : Google Scholar

99 

Chen F, Qi S, Zhang X, Wu J, Yang X and Wang R: lncRNA PLAC2 activated by H3K27 acetylation promotes cell proliferation and invasion via the activation of Wnt/β-catenin pathway in oral squamous cell carcinoma. Int J Oncol. 54:1183–1194. 2019.PubMed/NCBI

100 

Meseure D, Drak AK, Nicolas A, Bieche I and Morillon A: Long noncoding RNAs as new architects in cancer epigenetics, prognostic biomarkers, and potential therapeutic targets. Biomed Res Int. 2015:3202142015. View Article : Google Scholar : PubMed/NCBI

101 

Ju H, Zhang L, Mao L, Wu Y, Liu S, Ruan M, Hu J and Ren G: A comprehensive genome-wide analysis of the long noncoding RNA expression profile in metastatic lymph nodes of oral mucosal melanoma. Gene. 675:44–53. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 46 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang K, Qiu W, Wu B and 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 46: 535-545, 2020
APA
Zhang, K., Qiu, W., Wu, B., & Fang, F. (2020). Long non‑coding RNAs are novel players in oral inflammatory disorders, potentially premalignant oral epithelial lesions and oral squamous cell carcinoma (Review). International Journal of Molecular Medicine, 46, 535-545. https://doi.org/10.3892/ijmm.2020.4628
MLA
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)". International Journal of Molecular Medicine 46.2 (2020): 535-545.
Chicago
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)". International Journal of Molecular Medicine 46, no. 2 (2020): 535-545. https://doi.org/10.3892/ijmm.2020.4628