Open Access

Analysis of transcriptome expression profiling data in oral leukoplakia and early and late‑stage oral squamous cell carcinoma

  • Authors:
    • Lihui Yao
    • Bin Guo
    • Jiannan Wang
    • Jiale Wu
  • View Affiliations

  • Published online on: March 3, 2023     https://doi.org/10.3892/ol.2023.13742
  • Article Number: 156
  • Copyright: © Yao 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

The present study screened, potential prognostic biomarkers for oral carcinogenesis. The GSE85195 dataset, which consisted of oral leukoplakia (OL) and early and late‑stage oral squamous cell carcinoma (OSCC) samples, was used. The differentially expressed genes (DEGs) in early OSCC vs. OL, late OSCC vs. OL and late OSCC vs. early OSCC groups were screened using the limma package in R. The Short Time‑series Expression Miner software package was used to cluster DEGs with similar expression patterns in the course of disease progression (from OL to early and then late‑stage OSCC). Moreover, the Database for Annotation, Visualization and Integrated Discovery online analysis tool was used to perform Gene Ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. A protein‑protein interaction (PPI) network was also constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins database. Reverse transcription‑quantitative PCR was performed to assess the mRNA expression levels of hub node genes in clinical samples, and receiver operating characteristic curve analysis was performed to assess the prognostic value of the hub genes. A total of 4,595, 6,042 and 2,738 DEGs were screened in the early OSCC vs. OL, late OSCC vs. OL and late OSCC vs. early OSCC groups, respectively. A total of 665 overlapping genes were identified when the screened DEGs were compared. Cluster 1 and cluster 7 were identified as the significant clusters, which contained 496 and 341 DEGs, respectively. A PPI network was constructed with 440 interaction pairs. There were five differentially expressed hub nodes identified in different stages from OL to OSCC. The results of the present study indicated that fibronectin 1, signal transducer and activator of transcription 1, collagen type II α1 chain, collagen type X α1 chain and collagen type IV α6 chain might serve as independent diagnostic factors for OL and OSCC, and as prognostic biomarkers for OL carcinogenesis.

Introduction

Oral leukoplakia (OL) is a common oral mucosal disease characterized by white plaques or patches (1). As reported by the World Health Organization (WHO), OL is a manifestation of precancerous lesions; in other words, leukoplakia is likely to progress into cancer (2). According to the statistics reported in previous studies, the cancerous rate of leukoplakia is 3–5% (3). Oral squamous cell carcinoma (OSCC) is a frequently diagnosed head and neck cancer, accounting for >90% of oral and maxillofacial cancer cases (4). OSCC demonstrates a high incidence, strong invasiveness and a poor prognosis (5). Currently, the treatment of OSCC is mainly performed using surgical resection, supplemented by radiotherapy and chemotherapy (6). Therefore, it is important to prevent, treat and predict the prognosis of OSCC at the early stage and more studies have focused on the early prognosis of OSCC.

Previous studies have reported that biomarkers, such as keratin 8 whose high levels independently predict a poor prognosis for patients with lung adenocarcinoma, serve critical roles in the prognosis of numerous cancer types, (7). The autophagy-related gene, P4HB, has been reported as a novel prognostic biomarker for renal clear cell carcinoma (8). Rivera et al (9) reported that radiotherapy was normally used in OSCC treatment but that it was mostly applied without stratification by molecular diagnostics, and that it was urgent to provide clinically useful biomarkers for oral cancer. Biomarkers are expected to become the key to OSCC prognosis prediction in the future (10) and could also be used to guide the selection of appropriate treatment options. Notably, 35% of Asian patients with oral cancer present with H-ras mutations and patients who have the habit of chewing tobacco exhibit a significantly higher frequency of H-ras mutations (11). As reported by Sathyan et al (12), H-ras mutation reduced the expression of cyclin D1 and cyclin-dependent kinase 4, and upregulated that of RB transcriptional corepressor 1 and cyclin-dependent kinase inhibitor 2A, which might be used as favorable prognostic criterion. Moreover, the upregulation of human telomerase reverse transcriptase (hTRT) protein is an early event in oral cancer (13). Quantitative detection of hTRT expression in the cytoplasm and nucleus is of use in the assessment of the progression, recurrence and prognosis of OSCC (14). Patients with oral cancer with high expression of epidermal growth factor receptor (EGFR) are often more sensitive to gene therapy; therefore, antibodies targeting EGFR may be an effective tool to treat OSCC and precancerous lesions (15). Moreover, patients with higher expression of tumor protein P53 (TP53) and lower expression of Ki-67 are more likely to develop disease relapse after initial treatment (16). Therefore, these patients should be given more active combination therapy.

Currently, gene detection for OSCC is limited due to its low sensitivity. Consequently, the combined detection of multiple tumor markers is required to evaluate and predict tumor prognosis. The present study performed transcriptome analysis in different stages from OL to OSCC. Based on the protein-protein interaction (PPI) network constructed, the top 10 hub differentially expressed genes (DEGs) were screened and evaluated using reverse transcription-quantitative PCR (RT-qPCR).

Materials and methods

Data sources

The GSE85195 dataset (17) was downloaded from the NCBI Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/). The dataset included 15 OL, 24 early (stage 1–2) OSCC and 10 late (stage 3–4) OSCC samples; these samples were later divided into OL, early and late groups, respectively. The dataset was generated using the Agilent-014850 Whole Human Genome Microarray 4×44K G4112F platform (Agilent Technologies, Inc.).

Screening of DEGs between two groups

Samples from the three groups were compared in pairs. DEGs between two groups were screened using the R (version, 3.4.1) limma package (http://bioconductor.org/packages/release/bioc/html/limma.html) (version, 3.32.5) (18) for Bioconductor using thresholds of false discovery rate (FDR)<0.05 and log2 fold-change (FC)>1. The R (version, 3.4.1) pheatmap package (version, 1.0.8) (https://cran.rproject.org/web/packages/pheatmap/index.html) was used to generate two-way hierarchical clustering of the screened DEGs (19). The DEG sets among the three groups were then compared (early OSCC vs. OL, late OSCC vs. OL and late OSCC vs. early OSCC) and the overlapping DEG set was selected and retained as the object set for subsequent experiments.

Short Time-series Expression Miner (STEM) and enrichment analysis

DEGs that demonstrated similar expression patterns during the OL-early-late OSCC development process were clustered using STEM (version, 1.3.11; http://www.cs.cmu.edu/~jernst/stem/), using a similarity threshold of 0.8 and a significance threshold of FDR<0.05 (20). The Database for Annotation, Visualization and Integrated Discovery (DAVID) online analysis tool (version, 6.8; http://david.ncifcrf.gov/) was used to perform Gene Ontology (GO; http://geneontology.org/) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.kegg.jp/) pathway enrichment analysis on the DEGs in each cluster (21). Based on the GO and KEGG results, the Fisher's exact test of hypergeometric distribution was used to calculate the significance level of each function and pathway. The significance level was set at P<0.05.

Construction of the PPI network and analysis of topological structure

The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; version: 10.0; http://string-db.org/) database was used to evaluate the interaction relationships between DEGs in the clusters, and an interaction network of these proteins was then constructed (22). Cytoscape (version, 3.6.1; http://www.cytoscape.org/) was used for the visual display of the network. The DAVID 6.8 online analysis tool (http://david.abcc.ncifcrf.gov/) (23) was used for GO biological process and KEGG pathway enrichment analyses of hub genes in the network using the P<0.05 threshold.

The majority of biological networks obeyed the properties of scale-free networks; typically, nodes with the most connections in the network were identified as the hub nodes (24). The topological structure of the constructed interaction network was analyzed and the four important network topological parameters [degree, betweenness centrality (BC), closeness centrality (CC) and path length] were calculated. The degree distribution represented the probability distribution function P(k) of the degree of a node. The BC algorithm reflected the degree of the pivotal position of a node in the topological structure of the interaction network. The BC values of hub nodes were calculated as follows: CB(v)=∑t≠v≠u∈Vst(v)/σst). Where σst was the number of the shortest paths from s to t and σst(v) was the number of nodes (v) in the shortest path from s to t. The BC value was 0–1 and a value closer to 1 indicated a higher degree for the central hub. The CC algorithm assessed the connection between hub nodes and other nodes in the interactive network topology. The CC value of significantly related genes was calculated as follows: CC=1/(∑t∈V/tdG(v,t), where V represented the node set, t represented a node in the node set and dG(v,t) indicated the sum of the path distances from node t to the rest nodes. The CC value ranged from 0 to 1 and a value closer to 1 indicated stronger node centrality. Furthermore, the average path length described the degree of separation between nodes in the network.

Patient samples

Between June 2019 and January 2020, patients with OL and OSCC were enrolled from the Department of Stomatology in The First Affiliated Hospital of Zhengzhou University (Zhengzhou, China). All the enrolled samples were graded according to the WHO standards (25), with pathological diagnosis by two pathologists. There were 35 normal oral mucosal samples (including those from 21 males and 14 females), 41 OL samples (including those from 23 males and 18 females) and 37 OSCC samples (including those from 22 males and 15 females; 13 early OSCC and 24 late OSCC). The collected tissue samples were immediately frozen in liquid nitrogen for subsequent total RNA extraction. Differences in the age, sex ratio and underlying diseases of patients among the three groups were not statistically significant. All patients had not received radiotherapy, chemotherapy or other intervention before biopsy and they provided written informed consent for use of their tissues in this study. The study protocol was approved by the Ethics Committee of The First Affiliated Hospital of Zhengzhou University (approval number: 2018KJ06).

RT-qPCR for DEG verification in clinical samples

The mRNA sequence of hub genes was searched using Genebank (https://www.ncbi.nlm.nih.gov/genbank/), and RT-PCR primers were designed using Primer 5 software (https://www.bioprocessonline.com/doc/primer-premier-5-design-program-0001). Primer sequences (Table I) were synthesized by Sangon Bioengineering (Shangahi) Co., Ltd. The aforementioned patient tissue samples were weighed and total RNA was extracted from 100 mg by RNA extraction kit (cat. no. 9767) according to the manufacturer's protocol (Takara Bio, Inc.). Afterwards, complementary DNA was synthesized from the extracted RNA using a RevertAid RT Reverse Transcription kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions and used as the template for qPCR amplification on the Rotor Gene 3000 (GE Healthcare). For qPCR, SYBR Premix Ex Taq II (cat. no. RR420A; Takara Bio, Inc.) was used. The PCR conditions were as follows: 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec and 60°C for 45 sec. β-actin was used as the internal reference. The expression levels of DEGs were calculated using the 2−ΔΔCq method (26) and compared with that of β-actin.

Table I.

Sequences of primers used for reverse transcription-quantitative PCR.

Table I.

Sequences of primers used for reverse transcription-quantitative PCR.

GeneSequence (5′-3′)
FN1F: CCAGCAGAGGCATAAGGT
R: GTAGGGGTCAAAGCACGA
APPF: GATTCCCTACCGCTGCTT
R: CACTGCATGTCTCTTTGGC
STAT1F: TGCTCCCTCTCTGGAATG
R: CTCCTTGCTGATGAAGCC
SDC4F: GAAGGGGATGGTGGGAT
R: CAGGAACAGGGCAAGAGA
COL2A1F: TCCCACCCTCTCACAGTTC
R: TGCCCAGTTCAGGTCTCTT
COL10A1F: GGATCAGGCTTCAGGGAGTG
R: GGCCATTTGACTCGGCATTG
COL4A6F: GGATTGCCAGCATTATCAGGT
R: GTCTCAAATTCTGGACTAGGTGG
β-actinF: CAAAGACCTGTACGCCAACAC
R: CATACTCCTGCTTGCTGATCC

[i] FN1, fibronectin 1; STAT1, signal transducer and activator of transcription 1; COL2A1, collagen type II α1 chain; COL10A1, collagen type X α1 chain; COL4A6, collagen type IV α6 chain; SDC4, syndecan 4; APP, amyloid β precursor protein.

Statistical analysis

SPSS software (version 16.0; SPSS, Inc.) was used for the statistical analysis. One-way ANOVA followed by Tukey's post hoc test was used to compare multiple groups. A difference of P<0.05 was considered to indicate a statistically significant difference. The receiver operating characteristic (ROC) curve was plotted to assess the prognostic value of hub genes and the area under the curve (AUC), specificity and sensitivity were calculated.

Results

Data preprocessing and DEG screening

The GSE85195 dataset contained 15 OL samples, 24 early OSCC samples and 10 late OSCC samples. Samples at the three stages were compared in pairs and 4,595, 6,042 and 2,738 DEGs were detected in early OSCC vs. OL, late OSCC vs. OL and late OSCC vs. early OSCC groups, respectively. Samples were clustered based on the expression levels of the identified DEGs. A total of 665 overlapping genes were identified after comparing the screened DEGs (Fig. 1A). Trend clustering was performed on the 665 overlapping DEGs using STEM software. These results demonstrated two significant clusters (cluster 1 and cluster 7; Fig. 1B). Cluster 1 included 336 DEGs with a decreasing expression trend, whereas cluster 7 included 165 DEGs with an increasing expression trend.

Functional annotation and KEGG pathway enrichment analysis

Genes interact with each other to serve numerous roles and participate in different signaling pathways and functions. Consequently, it is critical to investigate related functions and pathways. Using the DAVID tools, GO function annotation and KEGG pathway enrichment analyses of DEGs in clusters 1 and 7 were performed. DEGs in cluster 1 were mainly enriched in 30 GO functions and 5 pathways, including ‘epithelial cell differentiation’ (n=10; P=5.07×10−4), ‘fatty acid metabolic process’ (n=12; P=6.01×10−4), ‘epidermis development’ (n=11; P=0.001), ‘arachidonic acid metabolism’ (n=6; P=0.00017) and ‘linoleic acid metabolism’ (n=4; P=0.0093) (Fig. 2A and C). However, DEGs in cluster 7 were mainly enriched in 22 GO functions and 5 pathways, including ‘phosphorus metabolic process’ (n=16; P=0.036), ‘phosphate metabolic process’ (n=16; P=0.036), ‘cell adhesion’ (n=15; P=0.005), ‘pathways in cancer’ (n=12; P=0.0015) and ‘focal adhesion’ (n=11; P=1.15×10−4) (Fig. 2B and D).

PPI network construction and topological structural analysis

The roles of genes are intricate, with a number working together through interaction. The influence of different genes is different and the number of genes involved in a given process may also be different. Through the PPI network, genes with the highest degree of interaction were screened out. The interaction relationships between DEGs in cluster 1 and cluster 7 were searched against the STRING database, using a threshold of an interaction score >0.6, and a total of 440 interaction pairs were identified. All the relationships in the constructed interaction network (Fig. 3) are presented in Table SI. GO functional annotation and KEGG pathway enrichment analyses using the DAVID database were performed on the genes in the network (Fig. 4A and B). These genes were mainly enriched into 49 functions and 8 pathways, including ‘extracellular matrix organization’ (n=15; P=1.75×10−6), ‘collagen catabolic process’ (n=8; P=4.97×10−5), ‘peptide cross-linking’ (n=7; P=1.03×10−4), ‘ECM-receptor interaction’ (n=10; P=8.12×10−5) and ‘protein digestion and absorption’ (n=8; P=0.0087). The four critical topological structural parameters of the interaction network (degree, BC, CC and path length) were calculated to screen hub genes with higher degrees in network connection. The average shortest path length, BC, CC, clusters and degrees of DEGs in cluster 1 and cluster 7 are presented in Table SII. Hub genes with higher degrees were selected and the top 10 hub nodes were selected according to the degree (Table II). The top seven hub nodes included FN1 (degree, 20), amyloid β precursor protein (APP; degree, 20), STAT1 (degree, 13), syndecan 4 (SDC4; degree, 11), COL4A1 (degree, 11), COL2A1 (degree, 11) and COL6A5 (degree, 9). The FDRs of these genes among three groups were calculated (Table III).

Table II.

Topology parameter information for the top 10 nodes.

Table II.

Topology parameter information for the top 10 nodes.

GeneAverage shortest path lengthBetweenness centralityCloseness centralityClusterDegree
FN13.748633880.230321730.26676385720
APP3.617486340.329707110.27643505720
STAT14.092896170.233431020.24432577713
SDC44.11475410.080227450.24302789711
COL4A14.491803280.015212870.22262774711
COL2A14.502732240.019561350.22208738711
COL6A54.961748630.002483190.2015418519
THBS14.016393440.028170620.2489795979
COL10A15.267759560.004451080.1898340279
COL4A64.961748630.002483190.2015418579

[i] FN1, fibronectin 1; STAT1, signal transducer and activator of transcription 1; COL2A1, collagen type II α1 chain; COL10A1, collagen type X α1 chain; COL4A6, collagen type IV α6 chain; SDC4, syndecan 4; APP, amyloid β precursor protein; COL4A1, collagen type IV α1 chain; COL6A5, collagen type VI α5 chain; THBS1, thrombospondin 1.

Table III.

FDRs of the top seven hub genes among the 3 groups.

Table III.

FDRs of the top seven hub genes among the 3 groups.

FDRs

GeneEarly OSCC vs. OLLate OSCC vs. OLLate OSCC vs. early OSCC
FN10.01120.0001160.017
APP0.0003860.0001550.0413
STAT10.000003480.000002340.0239
SDC40.000000002260.00002040.0111
COL2A10.00170.000005680.0104
COL10A10.003290.00001450.0329
COL4A60.002470.0002080.0481

[i] FDR, false discovery rate; OL, oral leukoplakia; OSCC, oral squamous cell carcinoma; FN1, fibronectin 1; STAT1, signal transducer and activator of transcription 1; COL2A1, collagen type II α1 chain; COL10A1, collagen type X α1 chain; COL4A6, collagen type IV α6 chain; SDC4, syndecan 4; APP, amyloid β precursor protein.

Assessment of the top seven hub genes in clinical samples

Bioinformatic analysis indicated the important genes through algorithms based on the GSE85195 dataset. The identified hub genes were assessed clinically. The mRNA expression levels, in the clinical samples, of the top seven hub genes were assessed using RT-qPCR. FN1, STAT1, COL2A1 and COL10A1 demonstrated a significant increase in their mRNA expression levels in the late OSCC group compared with the OL group (Fig. 5). However, COL4A6 mRNA expression levels exhibited a significant decrease in the late OSCC group compared with the OL group. There was no significant difference in the mRNA expression levels of APP or SDC4. Both APP and SDC4 belonged to cluster 7, which had demonstrated an increasing trend on bioinformatic analysis. There were still differences between the results obtained using bioinformatic analysis and the actual expression in patient tissues. Therefore, FN1, STAT1, COL2A1, COL10A1 and COL4A6 might serve as potential biomarkers for predicting the progression from OL to OSCC. ROC analysis was used to assess the prognostic value of these hub genes. FN1 (OL AUC=0.7604; OSCC AUC=0.8977), STAT1 (OL AUC=0.6950; OSCC AUC=0.7541), COL2A1 (OL AUC=0.6944; OSCC AUC=0.7803), COL10A1 (OL AUC=0.6338; OSCC AUC=0.7718) and COL4A6 (OL AUC=0.5916; OSCC AUC=0.6668) were the candidate independent prognostic factors for OL and OSCC (Fig. 6A and B). Based on the AUC, FN1 appeared to be the better prognostic factor.

Discussion

OL is a well-characterized oral mucosal precancerous lesion (27). The abnormal genetic changes in OL have been reported to lay the biological basis for the development of oral cancer (28). In the present study, DEGs in different developmental stages were screened to assess their ability to predict OSCC. These DEGs were enriched in numerous different signaling pathways and functions. Using the PPI network, genes with the highest degree of interaction were screened.

FN1 was identified as the hub node with the highest degree in the present study. FN1 is a high-molecular weight glycoprotein, which is widely distributed in blood, body fluids and numerous tissues (29). FN1 possesses a variety of biological activities and is extensively involved in numerous cell processes, such as cell migration, adhesion, proliferation, hemostasis and tissue repair (30). In a previous study, the inhibition of FN1 expression was suggested as a promising treatment strategy for OSCC (31). Furthermore, FN1 has been reported as a biomarker for OSCC, with sensitivity and specificity of 80 and 84%, respectively (32). Moreover, Suresh et al (33) performed high-throughput analysis of oral tongue cancer and reported FN1 as a molecular biomarker for drug resistance. Importantly, FN1 expression is markedly different between patients with early and late OSCC (34). In the present study, FN1 was indicated to have participated in extracellular matrix (ECM) organization and ECM-receptor interaction during tissue reconstruction. The ECM is key to triggering cell migration and fixation (35), and ECM components facilitate the attachment, proliferation and cytoskeletal organization of human oral epithelial cells (36). Therefore, it can be hypothesized that FN1 was involved in the carcinogenesis of OL by participating in the ECM function and pathways.

STAT1 belongs to the STAT protein family. In response to cytokines and growth factors, the STAT members are phosphorylated by receptor-related kinases to form homodimers or heterodimers (37). This protein can be activated by a variety of ligands, such as interferon-α, interferon-γ, epidermal growth factor, PDGF and IL6 (38). A previous study reported that STAT1 expression was positively associated with the occurrence of oral precancerous lesions (39). Furthermore, the activation of STAT1 has been reported to predict the risk of OSCC and is also associated with lymph node status (40). STAT1/ATF4/S1OOP is a critical signaling pathway associated with the development of oral cancer (41), and the results of the present study were consistent with this. In the present study, STAT1 expression increased from OL to OSCC status and it was also indicated to participate in drug response, the type I interferon signaling pathway. Previous studies reported that type I interferon induced the expression of MDA-5, which improved the immune response of oral disease (42). Furthermore, the type I interferon signaling pathway is an important process linked with the defense mechanism against oral viral infections (43). Based on the aforementioned evidence, it was hypothesized that STAT1 not only served a critical role in the carcinogenesis of OL, but also participated in drug response and immune defense.

COL2A1, COL10A1 and COL4A6 all belong to the collagen family. Collagen fibers are the most important ECM component in the oral mucosa and are an important component of the cancer interstitium (44). Any changes in their morphological distribution or quantity may be related to OSCC invasion (45). In 2016, Li et al (46) reported the use of microarray data analysis to screen oral tongue SCC (OTSCC)-related biomarkers and reported that numerous collagen proteins were differentially expressed in OTSCC samples. Notably, COL2A1, COL10A1 and COL4A6 were enriched in ECM organization, collagen catabolic process, proteinaceous ECM and the PI3K-Akt signaling pathway in the present study. The PI3K-Akt signaling pathway is widely involved in oral diseases such as periapical pulp disease, periodontal disease and oral tumors (47). This pathway has also been reported to demonstrate oncogenic properties and is related to the proliferation and migration of OSCC cells (48). Combing these previous studies and the results of the present study suggested that COL2A1, COL10A1 and COL4A6 may serve as prognostic biomarkers for OSCC.

Genes in the PPI network in the present study were enriched in 49 functions and 8 pathways, such as ‘extracellular matrix organization’, ‘collagen catabolic process’, ‘peptide cross-linking’, ‘ECM-receptor interaction’ and ‘protein digestion and absorption’. These functions and pathways are critical in the carcinogenesis of OL. Previous studies have reported certain theoretical foundations for these pathways. Sutinen et al (49) reported the expression of MMP-1 and MMP-2, and their inhibitors TIMP-1, TIMP-2 and TIMP-3, in dysplastic epithelium and squamous cell carcinoma, and demonstrated that the expression of MMP and TIMP increased significantly from leukoplakia to cancer. Katayama et al (50) applied immunohistochemistry to detect the expression of MMPs and TIMPs in 53 early oral cancer biopsy specimens and reported that the high expression of TIMP-2 was an independent factor for the poor prognosis of early OSCC. Furthermore, Arora et al (51) assessed the expression of type IV collagen in OL and reported that the inhibition of collagen decomposition delayed OL deterioration and tumor spread. The aforementioned evidence suggested that these functions and pathways have specific roles in the progression from OL to OSCC.

Certain limitations should be noted in the present study. Firstly, the present study conducted the preliminary verification of the identified DEGs, but the underlying mechanisms of these DEGs were not verified in vitro and the detailed molecular mechanisms were not evaluated. For an in vitro study, cell functional experiments, including analysis of the effect of FN1, STAT1, COL2A1, COL10A1 and COL4A6 on cell proliferation, apoptosis, migration and invasion, as well as elucidation of the underlying molecular mechanism, should be performed. For an in vivo study, animal models of oral leukoplakia and oral squamous cell carcinoma should be constructed to evaluate the effect of FN1, STAT1, COL2A1, COL10A1 and COL4A6 on tumor growth and regulation mechanisms. Secondly, due to the limitations of the online dataset, the expression levels were not compared with those in normal samples. Thirdly, due to the small sample size and short time period of clinical sample collection, clinical samples from patients who developed OSCC from OL were not involved in this study and the expression changes of DEGs during the progression from OL to OSCC were not assessed. Therefore, the DEGs among a greater number of healthy individuals and patients with OL and OSCC should be evaluated. Moreover, the protein levels of these mRNAs in the same samples should be assessed to evaluate the physiological relevance in further studies.

In conclusion, FN1, STAT1, COL2A1, COL10A1 and COL4A6 were identified as potential biomarkers for the carcinogenesis of OL, which may provide a theoretical basis for the clinical assessment of the prognosis of OSCC. More studies are required to evaluate the underlying molecular mechanism of these genes and related pathways.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the Youth Fund of The First Affiliated Hospital of Zhengzhou University (grant no. 2018YJ156).

Availability of data and materials

The GSE85195 dataset (https://ftp.ncbi.nlm.nih.gov/geo/series/GSE85nnn/GSE85195/matrix/) was downloaded from the NCBI Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/). The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LHY and BG were responsible for study design and data collection. BG, JNW and JLW contributed to data analysis. LHY and JLW were responsible for clinical data recording and data analysis. LHY, JNW and JLW wrote the manuscript and confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The project protocol was approved by the Ethics Committee of The First Affiliated Hospital of Zhengzhou University (approval number: 2018KJ06) and performed in accordance with the Guidelines of The First Affiliated Hospital of Zhengzhou University (Zhengzhou, China). Patients provided written, informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Madke B, Chougule BD, Kar S and Khopkar U: Appearances in clinical dermatology. Indian J Dermatol Venereol Leprol. 80:432–447. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Marija BB: The prevalence of precancerous oral lesions: Oral leukoplakia. Archive of Oncology. 8:107–109. 2000.

3 

Jha R and Parmar DV: A study of precancerous lesions for oral cancer in Jamnagar City. Journal of Indian Academy of Oral Medicine & Radiology. 23:S333–S335. 2011. View Article : Google Scholar

4 

Field JK: Oncogenes and tumor-suppressor genes in squamous cell carcinoma of the head and neck. Eur J Cancer B Oral Oncol. 28B:67–76. 1992. View Article : Google Scholar : PubMed/NCBI

5 

Kato K, Shimasaki M, Kato T, Segami N and Ueda Y: Expression of Sphingosine Kinase-1 is associated with invasiveness and poor prognosis of oral squamous cell carcinoma. Anticancer Res. 38:1361–1368. 2018.PubMed/NCBI

6 

Grimm M, Kraut W, Hoefert S, Krimmel M, Biegner T, Teriete P, Cetindis M, Polligkeit J, Kluba S, Munz A and Reinert S: Evaluation of a biomarker based blood test for monitoring surgical resection of oral squamous cell carcinomas. Clin Oral Investig. 20:329–338. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Xie L, Dang Y, Guo J, Sun X, Xie T, Zhang L, Yan Z, Amin H and Guo X: High KRT8 expression independently predicts poor prognosis for lung adenocarcinoma patients. Genes (Basel). 10:362019. View Article : Google Scholar : PubMed/NCBI

8 

Xie L, Li H, Zhang L, Ma X, Dang Y, Guo J, Liu J, Ge L, Nan F, Dong H, et al: Autophagy-related gene P4HB: A novel diagnosis and prognosis marker for kidney renal clear cell carcinoma. Aging (Albany NY). 12:1828–1842. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Rivera C, Oliveira AK, Costa RAP, De Rossi T and Paes Leme AF: Prognostic biomarkers in oral squamous cell carcinoma: A systematic review. Oral Oncol. 72:38–47. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Cssz V, Lábiscsák P, Kalló G, Márkus B, Emri M, Szabó A, Tar I, Tőzsér J, Kiss C and Márton I: Proteomics investigation of OSCC-specific salivary biomarkers in a Hungarian population highlights the importance of identification of population-tailored biomarkers. PLoS One. 12:e01772822017. View Article : Google Scholar : PubMed/NCBI

11 

Ali J, Sabiha B, Jan HU, Haider SA, Khan AA and Ali SS: Genetic etiology of oral cancer. Oral Oncol. 70:23–28. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Sathyan KM, Nalinakumari KR and Kannan S: H-Ras mutation modulates the expression of major cell cycle regulatory proteins and disease prognosis in oral carcinoma. Mod Pathol. 20:1141–1148. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Lord RV, Salonga D, Danenberg KD, Peters JH, DeMeester TR, Park JM, Johansson J, Skinner KA, Chandrasoma P, DeMeester SR, et al: Telomerase reverse transcriptase expression is increased early in the Barrett's metaplasia, dysplasia, adenocarcinoma sequence. J Gastrointest Surg. 4:135–142. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Zhou Q, Wang XH, Jiang YH, Li RW and Zhong M: Expression of telomerase genes hTRTmRNA in oral squamous cell carcinomas. Shanghai Kou Qiang Yi Xue. 15:259–262. 2006.(In Chinese). PubMed/NCBI

15 

Umbreit C, Tuch D, Hoffmann F, Gräfe C, Clement JH, Franz M, von Eggeling F, Berndt A and Guntinas-Lichius O: Characterization of phenotype changes after long-term inhibition of EGFR in OSCC and analysis by MALDI-MSI. Laryngorhinootologie. 97((S 02)): S137–S138. 2018.

16 

Matsuhira A, Noguchi S, Sato K, Tanaka Y, Yamamoto G, Mishima K and Katakura A: Cytokeratin 13, Cytokeratin 17, Ki-67 and p53 expression in upper layers of epithelial dysplasia surrounding tongue squamous cell carcinoma. Bull Tokyo Dent Coll. 56:223–231. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Bhosale PG, Cristea S, Ambatipudi S, Desai RS, Kumar R, Patil A, Kane S, Borges AM, Schäffer AA, Beerenwinkel N and Mahimkar MB: Chromosomal alterations and gene expression changes associated with the progression of leukoplakia to advanced gingivobuccal cancer. Transl Oncol. 10:396–409. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

19 

Wang L, Cao C, Ma Q, Zeng Q, Wang H, Cheng Z, Zhu G, Qi J, Ma H, Nian H and Wang Y: RNA-seq analyses of multiple meristems of soybean: Novel and alternative transcripts, evolutionary and functional implications. BMC Plant Biol. 14:1692014. View Article : Google Scholar : PubMed/NCBI

20 

Ghandhi SA, Sinha A, Markatou M and Amundson SA: Time-series clustering of gene expression in irradiated and bystander fibroblasts: An application of FBPA clustering. BMC Genomics. 12:22011. View Article : Google Scholar : PubMed/NCBI

21 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, et al: The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 45((D1)): D362–D368. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Bergholdt R, Brorsson C, Lage K, Nielsen JH, Brunak S and Pociot F: Expression profiling of human genetic and protein interaction networks in type 1 diabetes. PLoS One. 4:e62502009. View Article : Google Scholar : PubMed/NCBI

24 

Jeong H, Mason SP, Barabási AL and Oltvai ZN: Lethality and centrality in protein networks. Nature. 411:41–42. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Xu QS, Wang C, Li B, Li JZ, Mao MH, Qin LZ, Li H, Huang X, Han Z and Feng Z: Prognostic value of pathologic grade for patients with oral squamous cell carcinoma. Oral Dis. 24:335–346. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Nomura H, Sakamoto K, Sugihara T, Okamoto S, Aoki Y, Tanigawa T, Matoda M, Omatsu K, Kanao H, Kato K, et al: Oral leukoplakia, a precancerous lesion of squamous cell carcinoma, in patients with long-term pegylated liposomal doxorubicin treatment. Medicine (Baltimore). 97:e99322018. View Article : Google Scholar : PubMed/NCBI

28 

Cohen HV and Quek SYP: The molecular biology of cancer-Part 1-cellular and genetic changes that lead to the development of cancer. J N J Dent Assoc. 82:22–25. 2011.PubMed/NCBI

29 

Ouaissi MA and Capron A: Fibronectins: Structure and functions. Ann Inst Pasteur Immunol. (1985). 136C:169–185. 1985.(In French). View Article : Google Scholar : PubMed/NCBI

30 

Nuttelman CR, Mortisen DJ, Henry SM and Anseth KS: Attachment of fibronectin to poly(vinyl alcohol) hydrogels promotes NIH3T3 cell adhesion, proliferation, and migration. J Biomed Mater Res. 57:217–223. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Chen Z, Tao Q, Qiao B and Zhang L: Silencing of LINC01116 suppresses the development of oral squamous cell carcinoma by up-regulating microRNA-136 to inhibit FN1. Cancer Manag Res. 11:6043–6059. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Yen CY, Huang CY, Hou MF, Yang YH, Chang CH, Huang HW, Chen CH and Chang HW: Evaluating the performance of fibronectin 1 (FN1), integrin α4β1 (ITGA4), syndecan-2 (SDC2), and glycoprotein CD44 as the potential biomarkers of oral squamous cell carcinoma (OSCC). Biomarkers. 18:63–72. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Suresh A, Vannan M, Kumaran D, Gümüs ZH, Sivadas P, Murugaian EE, Kekatpure V, Iyer S, Thangaraj K and Kuriakose MA: Resistance/response molecular signature for oral tongue squamous cell carcinoma. Dis Markers. 32:51–64. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Pavuluri S, Lefevre C, Sharp J, Vasireddi SP and Nicholas KR: Abstract C37: Gene profiling predicts biomarkers in oral squamous cell carcinoma with diagnostic and therapeutic importance. Mol Cancer Ther. 12 (11_Supplement):C372014. View Article : Google Scholar

35 

Stupack DG, Cho SY and Klemke RL: Molecular signaling mechanisms of cell migration and invasion. Immunol Res. 21:83–88. 2000. View Article : Google Scholar : PubMed/NCBI

36 

Park JC, Kim HM and Ko J: Effects of extracellular matrix constituents on the attachment of human oral epithelial cells at the titanium surface. Int J Oral Maxillofac Implants. 13:826–836. 1998.PubMed/NCBI

37 

Yang T, Lu P, Zhang Y, Zheng YB, Wang L, Zhou L and Liu J: Seeking colorectal carcinoma related genes based on regulation network. Afr J Pharm Pharmacol. 5:1467–1474. 2011. View Article : Google Scholar

38 

Bhat GJ and Baker KM: Cross-talk between angiotensin II and interleukin-6-induced signaling through Stat3 transcription factor. Basic Res Cardiol. 93 (Suppl 3):S26–S29. 1998. View Article : Google Scholar : PubMed/NCBI

39 

Mori K, Haraguchi S, Hiori M, Shimada J and Ohmori Y: Tumor-associated macrophages in oral premalignant lesions coexpress CD163 and STAT1 in a Th1-dominated microenvironment. BMC Cancer. 15:5732015. View Article : Google Scholar : PubMed/NCBI

40 

Laimer K, Spizzo G, Obrist P, Gastl G, Brunhuber T, Schäfer G, Norer B, Rasse M, Haffner MC and Doppler W: STAT1 activation in squamous cell cancer of the oral cavity: A potential predictive marker of response to adjuvant chemotherapy. Cancer. 110:326–333. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Wu TS, Tan CT, Chang CC, Lin BR, Lai WT, Chen ST, Kuo MY, Rau CL, Jaw FS and Chang HH: B-cell lymphoma/leukemia 10 promotes oral cancer progression through STAT1/ATF4/S100P signaling pathway. Oncogene. 34:1207–1219. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Matsumiya T, Hayakari R, Narita N, Ito R, Kon T, Kubota K, Sakaki H, Yoshida H, Imaizumi T, Kobayashi W and Kimura H: Role of type I- and type II-interferon in expression of melanoma differentiation-associated gene-5 in HSC-3 oral squamous carcinoma cells. Biomed Res. 35:9–16. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Yokota S, Saito H, Kubota T, Yokosawa N, Amano K and Fujii N: Measles virus suppresses interferon-alpha signaling pathway: Suppression of Jak1 phosphorylation and association of viral accessory proteins, C and V, with interferon-alpha receptor complex. Virology. 306:135–146. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Carretero A, Soares da Costa D, Reis RL and Pashkuleva I: Extracellular matrix-inspired assembly of glycosaminoglycan-collagen fibers. J Mater Chem B. 5:3103–3106. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Benbow U, Schoenermark MP, Mitchell TI, Rutter JL, Shimokawa K, Nagase H and Brinckerhoff CE: A novel host/tumor cell interaction activates matrix metalloproteinase 1 and mediates invasion through type I collagen. J Biol Chem. 274:25371–25378. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Li Y, Chai Z, Wan L and Ding Y: Analysis of the gene expression profile for Oral tongue squamous cell carcinoma. Int J Clin Exp Med. 9:7471–7480. 2016.

47 

Wang H, Wu Q, Liu Z, Luo X, Fan Y, Liu Y, Zhang Y, Hua S, Fu Q, Zhao M, et al: Downregulation of FAP suppresses cell proliferation and metastasis through PTEN/PI3K/AKT and Ras-ERK signaling in oral squamous cell carcinoma. Cell Death Dis. 5:e11552014. View Article : Google Scholar : PubMed/NCBI

48 

Zheng Y, Wang Z, Xiong X, Zhong Y, Zhang W, Dong Y, Li J, Zhu Z, Zhang W, Wu H, et al: Membrane-tethered Notch1 exhibits oncogenic property via activation of EGFR-PI3K-AKT pathway in oral squamous cell carcinoma. J Cell Physiol. 234:5940–5952. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Sutinen M, Kainulainen T, Hurskainen T, Vesterlund E, Alexander JP, Overall CM, Sorsa T and Salo T: Expression of matrix metalloproteinases (MMP-1 and −2) and their inhibitors (TIMP-1, −2 and −3) in oral lichen planus, dysplasia, squamous cell carcinoma and lymph node metastasis. Br J Cancer. 77:2239–2245. 1998. View Article : Google Scholar : PubMed/NCBI

50 

Katayama A, Bandoh N, Kishibe K, Takahara M, Ogino T, Nonaka S and Harabuchi Y: Expressions of matrix metalloproteinases in early-stage oral squamous cell carcinoma as predictive indicators for tumor metastases and prognosis. Clin Cancer Res. 10:634–640. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Arora KS, Nayyar A, Kaur P, Arora KS, Goel A and Singh S: Evaluation of collagen in leukoplakia, oral submucous fibrosis and oral squamous cell carcinomas using polarizing microscopy and immunohistochemistry. Asian Pac J Cancer Prev. 19:1075–1080. 2018.PubMed/NCBI

Related Articles

Journal Cover

April-2023
Volume 25 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yao L, Guo B, Wang J and Wu J: Analysis of transcriptome expression profiling data in oral leukoplakia and early and late‑stage oral squamous cell carcinoma. Oncol Lett 25: 156, 2023
APA
Yao, L., Guo, B., Wang, J., & Wu, J. (2023). Analysis of transcriptome expression profiling data in oral leukoplakia and early and late‑stage oral squamous cell carcinoma. Oncology Letters, 25, 156. https://doi.org/10.3892/ol.2023.13742
MLA
Yao, L., Guo, B., Wang, J., Wu, J."Analysis of transcriptome expression profiling data in oral leukoplakia and early and late‑stage oral squamous cell carcinoma". Oncology Letters 25.4 (2023): 156.
Chicago
Yao, L., Guo, B., Wang, J., Wu, J."Analysis of transcriptome expression profiling data in oral leukoplakia and early and late‑stage oral squamous cell carcinoma". Oncology Letters 25, no. 4 (2023): 156. https://doi.org/10.3892/ol.2023.13742