Open Access

Five extracellular matrix‑associated genes upregulated in oral tongue squamous cell carcinoma: An integrated bioinformatics analysis

  • Authors:
    • Pingping Zhong
    • Liya Liu
    • Aling Shen
    • Zhongxin Chen
    • Xiaoyan Hu
    • Yichao Cai
    • Jie Lin
    • Bangyan Wang
    • Jiesen Li
    • Youqin Chen
    • Jun Peng
  • View Affiliations

  • Published online on: October 11, 2019     https://doi.org/10.3892/ol.2019.10982
  • Pages: 5959-5967
  • Copyright: © Zhong 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

Despite advancements in treatment regimens, the mortality rate of patients with oral tongue squamous cell carcinoma (OTSCC) is high. In addition, the signaling pathways and oncoproteins involved in OTSCC progression remain largely unknown. Therefore, the aim of the present study was to identify specific prognostic marker for patients at a high risk of developing OTSCC. The present study used four original microarray datasets to identify the key candidate genes involved in OTSCC pathogenesis. Expression profiles of 93 OTSCC tissues and 76 normal tissues from GSE9844, GSE13601, GSE31056 and GSE75538 datasets were investigated. Differentially expressed genes (DEGs) were determined, and gene ontology enrichment and gene interactions were analyzed. The four GSE datasets reported five upregulated and six downregulated DEGs. Five upregulated genes (matrix metalloproteinase 1, 3, 10 and 12 and laminin subunit gamma 2) were localized in the extracellular region of cells and were associated with extracellular matrix disassembly. Furthermore, analysis for The Cancer Genome Atlas database revealed that the aforementioned five upregulated genes were also highly expressed in OTSCC and head and neck squamous cell carcinoma tissues. These results demonstrated that the five upregulated genes may be considered as potential prognostic biomarkers of OTSCC and may serve at understanding OTSCC progression. Upregulated DEGs may therefore represent valuable therapeutic targets to prevent or control OTSCC pathogenesis.

Introduction

Head and neck squamous cell carcinoma (HNSC) is a common cancer worldwide and accounts for >600,000 new cases annually (1). Despite significant advancements in treatments, including reconstructive microvascular free tissue transfer, hyperfractionated radiotherapy and concurrent chemoradiation, the survival rate of patients with HNSC has not sufficiently improved over the last 50 years, with overall survival of ~50%, resulting in increased mortality rates worldwide every year (2). HNSC is a highly complex disease emerging from the oral cavity, tongue, pharynx or larynx (3). Each tumor harbors unique mutations, presents variable clinical outcomes and is associated with specific risk factors (4,5). For example, TP53 inactivation, either through somatic mutation or HPV infection, appears nearly universal in this malignancy (6). The present study particularly focused on one type of HNSC, the oral tongue squamous cell carcinoma (OTSCC), because of its poor diagnosis, high incidence rate, aggressive clinical behavior and poor outcome (710). A recent study reported a five-year survival rate of 63% for patients with OTSCC in The Netherlands (11). In 2017, nearly 16,400 new cases of tongue cancer were diagnosed and 2,400 tongue cancer-associated mortality cases were recorded in the United States (12). Several prognostic factors for OTSCC exist, including occult node positivity, tumor depth, lymphovascular invasion and perineural invasion (13). Nevertheless, robust and reliable molecular prognostic biomarkers need to be determined in order to identify patients with advanced stages of OTSCC.

Several thousands of tumor biomarkers have been discovered and are associated with the prognosis of various types of cancer (14). In particular, markers for OTSCC, including microtubule associated scaffold protein 1 and beta-parvin, have attracted much attention due to their crucial role in OTSCC pathogenesis (15). Furthermore, it was demonstrated that downregulation of these markers markedly decreases cancer cell survival (1519).

Thanks to the rapid development and extensive application of microarrays for gene identification in various types of cancer (2022), a high number of differentially expressed genes (DEGs) in OTSCC have been identified (13,19). However, the results between studies are inconsistent, which might be due the variability of tissue samples used. In addition, no reliable biomarkers for OTSCC have been established. Subsequently, the combination of bioinformatics methods and expression profiling techniques may represent a novel approach to resolve these problems. The present study used four microarray datasets [GSE9844 (3), GSE13601 (23), GSE31056 (24), and GSE75538 (25)] from the National Center for Biotechnology Information (NCBI)-Gene Expression Omnibus (GEO) database and an mRNA sequencing (mRNA-seq) dataset from The Cancer Genome Atlas (TCGA). DEGs were filtered using the GEO2R tool according to conventional data processing standards, and gene ontology (GO) enrichment analysis was performed to screen for DEGs using The Database for Annotation, Visualization and Integrated Discovery (DAVID). Expression levels of DEGs in samples from TCGA were assessed using Gene Expression Profiling Interactive Analysis (GEPIA), and corrections were applied for DEGs is samples from TCGA-HNSC using cBioPortal to identify potential oncogenes in OTSCC. The identified DEGs and their associated pathways may be considered as robust and reliable tumor biomarkers for OTSCC and serve as precise therapeutic targets for the prevention of OTSCC progression at early stages.

Materials and methods

GEO datasets

GEO (https://www.ncbi.nlm.nih.gov/gds) is a public repository at NCBI for storing high-throughput gene expression data (26). The gene expression profiles of GSE9844, GSE13601, GSE31056, and GSE75538 were selected from the GEO database (http://www.ncbi.nlm.nih.gov/geo/). GSE9844 included 26 OTSCC and 12 normal tissues, GSE13601 included 31 OTSCC and 26 normal tissues, GSE31056 included 22 OTSCC and 24 normal tissues, and GSE75538 included 14 OTSCC and 14 normal tissues.

Identification of DEGs

GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r/) was used to detect DEGs between OTSCC and normal tissue samples (27). Adjusted P-values <0.01 and |log fold change (FC)|>2 were set as cutoffs. Co-expressed DEGs that were downregulated or upregulated in the two sets of gene expression profiles were identified using Venn diagrams (http://bioinformatics.psb.ugent.be/webtools/Venn/).

GO enrichment analysis of DEGs

Usually, genes and their products are annotated to identify characteristic biological function and processes of a high-throughput genome or transcriptome (28). DAVID (https://david.ncifcrf.gov/) is a web-based bioinformatics resource for gene annotation and visualization with an integrated discovery function. It is therefore useful for determining gene biological attributes (29). P<0.01 and the Benjamini corrected P<0.01 (30) were set as the cutoff. Sequential pathways, molecular and cellular components and biological functions of DEGs could be visualized by using DAVID (https://david.ncifcrf.gov/).

Comparison of gene expression in patients with OTSCC

The mRNA-seq data of the five OTSCC genes of interest from HNSC samples were obtained from the TCGA database (https://cancergenome.nih.gov/) (31). In addition, clinical data of OTSCC were downloaded using TCGA Assembler (Table I). mRNA-seq data of 147 OTSCC tissues and 15 adjacent normal tongue tissues were obtained on an Illumina HiSeq RNASeq platform (32). Since the TCGA dataset was developed as a community resource project, no additional approval was required for this study from Fujian Medical University. The present study complied with the TCGA publication guidelines and data access policies (33).

Table I.

Characteristics of 147 patients with tongue tumor from The Cancer Genome Atlas database.

Table I.

Characteristics of 147 patients with tongue tumor from The Cancer Genome Atlas database.

CharacteristicsPatient numberPercentage
Sex
  Female4631.29
  Male10168.71
Age at diagnosis, years
  Mean58.60±12.71
  <40106.80
  40–492013.61
  50–593825.85
  60–695336.05
  70–792114.29
  >8042.72
Tumor pathological stage
  T1-T27047.62
  T3-T47248.98
Nodal pathological stage
  N0-N17248.98
  N2-N47047.62
Smoking history
  Yes9866.67
  No/Never4731.97
  Unknown21.36
Alcohol history
  Yes9967.35
  No/Never4429.93
  Unknown42.72
Primary lymph node
  Yes12887.07
  No96.12
Analysis of gene expression patterns in all tumors from TCGA through GEPIA

The expression patterns of the five genes of interest in various types of cancers and normal tissues were determined using GEPIA (http://gepia.cancer-pku.cn) (34). The program was used to analyze RNA sequencing data of 33 types of cancer (including adrenocortical carcinoma, bladder urothelial carcinoma, breast invasive carcinoma (BRCA), cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), cholangiocarcinoma (CHOL), colon adenocarcinoma (COAD), lymphoid neoplasm diffuse large B-cell lymphoma, esophageal carcinoma (ESCA), glioblastoma multiforme, head and neck squamous cell carcinoma (HNSC), kidney chromophobe (KICH), kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, acute myeloid leukemia, brain lower grade glioma, liver hepatocellular carcinoma, lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), mesothelioma, ovarian serous cystadenocarcinoma, pancreatic adenocarcinoma, pheochromocytoma and paraganglioma, prostate adenocarcinoma, rectum adenocarcinoma (READ), sarcoma, skin cutaneous melanoma, stomach adenocarcinoma (STAD), testicular germ cell tumor, thyroid carcinoma, thymoma, uterine corpus endometrial carcinoma, uterine carcinosarcoma, uveal melanoma) and adjacent normal samples from TCGA following the standard processing pipeline. P-value <0.01 and |logFC|>2 were set as cutoffs. Box plots were generated to visualize the associations.

Correlations among genes expression in HNSC samples from TCGA through cBioPortal

The correlations between expression patterns of the genes of interest in cancer tissues from TCGA-HNSC dataset were analyzed using Pearson and Spearman correlation coefficient via cBioPortal (http://www.cbioportal.org) (3536).

Statistical analysis

Statistical analyses were performed using Graphpad Prism 5.0 (GraphPad Software, Inc.). Mann-Whitney U test was performed to compare gene expression between tumor and adjacent normal tissues. A two-tailed P-value <0.01 was considered statistically significant.

Results

Some biomarkers distinguished OTSCC tissues from normal tissues

Four GEO datasets (GSE9844, GSE13601, GSE31056, and GSE75538) were imported into the GEO2R analysis tool. Based on DEGs between OTSCC and normal tissues, five upregulated genes [matrix metalloproteinases 1, 3, 10 and 12 (MMP1, MMP3, MMP10 and MMP12) and laminin subunit gamma 2 (LAMC2)] (Fig. 1A) and six downregulated genes (dermatopontin, cartilage intermediate layer protein, keratin 4, ATP binding cassette subfamily A member 8, alcohol dehydrogenase 1B and protein phosphatase 1 regulatory subunit 3C) (Fig. 1B) were highlighted in all four datasets. Whether the five upregulated genes could be considered as potential biomarkers for distinguishing OTSCC tissues from normal tissues was therefore assessed.

GO function enrichment analysis

GO enrichment analysis was performed using DAVID tool. The five specific upregulated genes MMP1, LAMC2, MMP3, MMP10 and MMP12 were uploaded into the DAVID software. GO results indicated that these genes were specifically involved in certain biological processes, including ‘extracellular matrix disassembly’, ‘collagen catabolism’ and ‘proteolysis’. Furthermore, with regards to the molecular function, the products of these five genes mainly comprised metalloendopeptidases, serine-type endopeptidases, calcium ion binders and zinc ion binders. Similarly, GO cell component analysis revealed that the proteins encoded by these genes were significantly enriched for functions associated with the proteinaceous extracellular matrix and extracellular region (P<0.01, Table II).

Table II.

Gene ontology analysis of upregulated genes associated with oral tongue squamous cell carcinoma.

Table II.

Gene ontology analysis of upregulated genes associated with oral tongue squamous cell carcinoma.

CategoryTermCountGenesP-valueBenjamini corrected P-value
GOTERM_BP_DIRECT GO:0022617~extracellular matrix disassembly5MMP10, LAMC2, MMP3, MMP12, MMP1 3.9×10−10 7.7×10−9
GOTERM_BP_DIRECTGO:0030574~collagen catabolic process4MMP10, MMP3, MMP12, MMP1 2.1×10−7 2.1×10−6
GOTERM_MF_DIRECTGO:0004222 ~metalloendopeptidase activity4MMP10, MMP3, MMP12, MMP1 1.2×10−6 9.3×10−6
GOTERM_CC_DIRECT GO:0005578~proteinaceous extracellular matrix4MMP10, MMP3, MMP12, MMP1 1.2×10−5 1.1×10−4
GOTERM_MF_DIRECT GO:0004252~serine-type endopeptidase activity4MMP10, MMP3, MMP12, MMP1 1.3×10−5 5.4×10−5
GOTERM_MF_DIRECT GO:0004175~endopeptidase activity3MMP3, MMP12, MMP1 6.0×10−5 1.6×10−4
GOTERM_CC_DIRECT GO:0005576~extracellular region5MMP10, LAMC2, MMP3, MMP12, MMP1 6.1×10−5 2.7×10−4
GOTERM_BP_DIRECT GO:0006508~proteolysis4MMP10, MMP3, MMP12, MMP1 1.0×10−4 6.8×10−4
GOTERM_MF_DIRECTGO:0005509~calcium ion binding4MMP10, MMP3, MMP12, MMP1 3.0×10−4 5.9×10−4
GOTERM_MF_DIRECTGO:0008270~zinc ion binding4MMP10, MMP3, MMP12, MMP1 1.3×10−3 2.0×10−3
Gene expression of MMP1, LAMC2, MMP3, MMP10 and MMP12 in OTSCC samples from TCGA

TCGA database was used to analyze the expression levels of the five genes of interest in OTSCC and adjacent normal tissues. The results demonstrated that the expression levels of MMP1, LAMC2, MMP3, MMP10 and MMP12 were significantly upregulated in OTSCC tissues compared with adjacent normal tissues (Fig. 2).

Gene expression of MMP1, LAMC2, MMP3, MMP10 and MMP12 in 23 cancer samples from TCGA

Consistent results were obtained for bladder urothelial carcinoma, BRCA, COAD, ESCA, HNSC, LUAD, LUSC, READ and STAD. MMP1 expression level was significantly increased in all cancer tissues compared with adjacent normal tissues (Fig. 3A). LAMC2 was upregulated in CESC, CHOL, ESCA, HNSC, STAD and uterine corpus endometrial carcinoma, whereas it was significantly downregulated in BRCA, kidney chromophobe and kidney renal clear cell carcinoma (Fig. 3B). Furthermore, MMP3 expression was upregulated in COAD, ESCA, HNSC, and READ (Fig. 3C). MMP10 expression was significantly upregulated in ESCA, HNSC and LUSC (Fig. 3D). In addition, MMP12 was upregulated in CESC, ESCA, HNSC, LUAD, LUSC and STAD, whereas it was downregulated in pancreatic adenocarcinoma (Fig. 3E). Overall, the expression level of MMP1, LAMC2, MMP3, MMP10 and MMP12 was significantly upregulated in HNSC tissues.

Correlations among genes expression in samples from the TCGA-HNSC database

To select the five genes of interest for further investigation, the correlations between gene expression patterns in samples from TCGA were analyzed using cBioPortal (3536). The results demonstrated that MMP1 expression level was positively correlated with LAMC2 (Fig. 4A), MMP3 (Fig. 4B) and MMP10 (Fig. 4C) expression levels. In addition, LAMC2 expression level was positively correlated with MMP10 expression level (Fig. 4D). Pearson correlation coefficients ranged from 0.33 to 0.69, and Spearman correlation coefficients ranged from 0.55 to 0.83. All together, these results suggested that MMP1, LAMC2, MMP3 and MMP10 expression levels were positively correlated in HNSC tissues.

Discussion

The American Joint Committee on Cancer has updated the Tumor-Node-Metastasis system for cancer staging system according to diagnostic class, treatment choices and prognosis for OTSCC (37). However, with the development of novel modern therapeutic approaches for cancer, genetic analysis is an outstanding tool for early diagnosis of cancer that can prolong patient survival (38). Previous clinical studies on OTSCC development and progression reported increased incidence and mortality rates due to the lack of knowledge on OTSCC (39,40). The present study identified 11 DEGs (five upregulated and six downregulated genes) from four mRNA expression profile datasets via GEO. These DEGs were significantly associated with OTSCC pathogenesis. The present study therefore focused on the five upregulated DEGs since they may be considered as potential reliable biomarkers. These five target genes were categorized into three classes according to their associated biological functions (molecular functions, biological processes and cellular components) thanks to the GO enrichment using multiple approaches. The results demonstrated that all genes were involved in extracellular matrix disassembly and were localized in the extracellular region during cancer development. Furthermore, expression levels of these genes were positively correlated with each other and upregulated in OTSCC and HNSC tissues compared with adjacent normal tissues, as confirmed using DAVID.

Expression levels of four MMP family members, including MMP1, MMP3, MMP10 and MMP12, were upregulated in HNSC tissues compared with adjacent normal tissues. These genes serve important functions in numerous physiological and pathological processes involved in tumor progression and can promote tumor-induced angiogenesis and extracellular matrix disassembly, enhancing therefore tumor invasion and metastasis (41,42). MMP1 is known to be significantly associated with poor clinical outcomes and is therefore a robust prognostic factor for various types of cancer, including colorectal cancer and breast cancer (4345). Furthermore, MMP3 has been proposed as a vital tumor oncogene in numerous cancers, including pancreatic, pulmonary, and mammary carcinoma (46). In addition, MMP10 has been demonstrated to be a potential clinical marker for cancer stem-like cells/cancer-initiating cells in epithelial ovarian cancer (EOC) and may serve as a therapeutic target in chemotherapy-resistant EOCs (47). MMP12 has been reported to be highly expressed in nasopharyngeal carcinoma both in vitro and in vivo, and is therefore a powerful tumor marker for cancer metastasis (48). Furthermore, LAMC2, which encodes the major component of laminin-5, was one of the upregulated genes in the present study. Highly expression levels of LAMC2 have been previously reported in numerous invasive tumors (49,50), including carcinomas of the lung (51), the colorectum (52,53) and the pancreas (54,55). In addition, it has been reported that LAMC2 overexpression in esophageal cancer is associated with poor survival (56,57). Subsequently, MMP1, MMP3, MMP10, MMP12 and LAMC2 may serve as novel promising prognostic factors in advanced metastasis of OTSCC. The protein expression of MMP1, MMP3, MMP10, MMP12 and LAMC2 should be further confirmed in OTSCC tissues. Furthermore, the correlation between MMP1, MMP3, MMP10, MMP12 and LAMC2 protein expression and the survival and clinic characteristics of patients with OTSCC should be further assessed. Tissue samples from patients with OSTCC are currently being collected for this purpose, and will be used for immunohistochemistry analysis to detect MMPs protein expression. Furthermore, the clinical significance, biological function and underlying mechanism of MMPs on the pathogenesis and progression of OTSCC will be further investigated.

In conclusion, the present study identified five upregulated genes that may be considered as robust and reliable biomarkers in OTSCC prognosis. The findings from this study may help discovering the role of oncogenes in cancer progression and determining the underlying pathways in order to develop novel therapeutic strategies for OTSCC.

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant no. 81673721) and the Natural Science Foundation of Fujian Province (grant no. 2017I0007).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

PZ, LL, YC and JP conceived and designed the experiments. PZ, AS, ZC and XH performed GEO and GEO2R analyses. LL, XH, YC and JL conducted gene ontology analyses using DAVID. BW, JL, YC and JP analyzed TCGA database using GEPIA. LL, AS, ZC and YC analyzed expressions of genes in OTSCC from the TCGA database. XH, JL, BW and JL assessed the correlations among gene expressions using cBioPortal. PZ, LL, YC and JP drafted the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interest

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

HNSC

head and neck squamous cell carcinoma

OTSCC

oral tongue squamous cell carcinoma

DEG

differentially expressed gene

TCGA

The Cancer Genome Atlas

GEO

Gene Expression Omnibus

GO

Gene Ontology

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Ledgerwood LG, Kumar D, Eterovic AK, Wick J, Chen K, Zhao H, Tazi L, Manna P, Kerley S, Joshi R, et al: The degree of intratumor mutational heterogeneity varies by primary tumor sub-site. Oncotarget. 7:27185–27198. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Ye H, Yu T, Temam S, Ziober BL, Wang J, Schwartz JL, Mao L, Wong DT and Zhou X: Transcriptomic dissection of tongue squamous cell carcinoma. BMC Genomics. 9:692008. View Article : Google Scholar : PubMed/NCBI

4 

Döbrossy L: Epidemiology of head and neck cancer: Magnitude of the problem. Cancer Metastasis Rev. 24:9–17. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Tímár J, Csuka O, Remenár E, Répássy G and Kásler M: Progression of head and neck squamous cell cancer. Cancer Metastasis Rev. 24:107–127. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Stransky N, Egloff AM, Tward AD, Kostic AD, Cibulskis K, Sivachenko A, Kryukov GV, Lawrence MS, Sougnez C, McKenna A, et al: The mutational landscape of head and neck squamous cell carcinoma. Science. 333:1157–1160. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Patel SC, Carpenter WR, Tyree S, Couch ME, Weissler M, Hackman T, Hayes DN, Shores C and Chera BS: Increasing incidence of oral tongue squamous cell carcinoma in young white women, age 18 to 44 years. J Clin Oncol. 29:1488–1494. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Ng JH, Iyer NG, Tan MH and Edgren G: Changing epidemiology of oral squamous cell carcinoma of the tongue: A global study. Head Neck. 39:297–304. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Bello IO, Soini Y and Salo T: Prognostic evaluation of oral tongue cancer: Means, markers and perspectives (I). Oral Oncol. 46:630–635. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Almangush A, Heikkinen I, Mäkitie AA, Coletta RD, Läärä E, Leivo I and Salo T: Prognostic biomarkers for oral tongue squamous cell carcinoma: A systematic review and meta-analysis. Br J Cancer. 117:856–866. 2017. View Article : Google Scholar : PubMed/NCBI

11 

van Dijk BA, Brands MT, Geurts SM, Merkx MA and Roodenburg JL: Trends in oral cavity cancer incidence, mortality, survival and treatment in the Netherlands. Int J Cancer. 139:574–583. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Thangaraj SV, Shyamsundar V, Krishnamurthy A, Ramani P, Ganesan K, Muthuswami M and Ramshankar V: Molecular portrait of oral tongue squamous cell carcinoma shown by integrative meta-analysis of expression profiles with validations. PLoS One. 11:e01565822016. View Article : Google Scholar : PubMed/NCBI

14 

Ding X, Zhang N, Cai Y, Li S, Zheng C, Jin Y, Yu T, Wang A and Zhou X: Down-regulation of tumor suppressor MTUS1/ATIP is associated with enhanced proliferation, poor differentiation and poor prognosis in oral tongue squamous cell carcinoma. Mol Oncol. 6:73–80. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Eslami A, Miyaguchi K, Mogushi K, Watanabe H, Okada N, Shibuya H, Mizushima H, Miura M and Tanaka H: PARVB overexpression increases cell migration capability and defines high risk for endophytic growth and metastasis in tongue squamous cell carcinoma. Br J Cancer. 112:338–344. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Zhao T, Ding X, Chang B, Zhou X and Wang A: MTUS1/ATIP3a down- regulation is associated with enhanced migration, invasion and poor prognosis in salivary adenoid cystic carcinoma. BMC Cancer. 15:2032015. View Article : Google Scholar : PubMed/NCBI

17 

Rogler A, Hoja S, Giedl J, Ekici AB, Wach S, Taubert H, Goebell PJ, Wullich B, Stöckle M, Lehmann J, et al: Loss of MTUS1/ATIP expression is associated with adverse outcome in advanced bladder carcinomas: Data from a retrospective study. BMC Cancer. 14:2142014. View Article : Google Scholar : PubMed/NCBI

18 

Bravou V, Antonacopoulou A, Papanikolaou S, Nikou S, Lilis I, Giannopoulou E and Kalofonos HP: Focal adhesion proteins α- and β-parvin are overexpressed in human colorectal cancer and correlate with tumor progression. Cancer Invest. 33:387–397. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Qiu Z, Sun W, Gao S, Zhou H, Tan W, Cao M and Huang W: A 16-gene signature predicting prognosis of patients with oral tongue squamous cell carcinoma. PeerJ. 5:e40622017. View Article : Google Scholar : PubMed/NCBI

20 

Bullinger L, Döhner K, Bair E, Fröhling S, Schlenk RF, Tibshirani R, Döhner H and Pollack JR: Use of gene-expression profiling to identify prognostic subclasses in adult acute myeloid leukemia. N Engl J Med. 350:1605–1616. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Valk PJ, Verhaak RG, Beijen MA, Erpelinck CA, Barjesteh van Waalwijk van Doorn-Khosrovani S, Boer JM, Beverloo HB, Moorhouse MJ, van der Spek PJ, Löwenberg B and Delwel R: Prognostically useful gene-expression profiles in acute myeloid leukemia. N Engl J Med. 350:1617–1628. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr and Kinzler KW: Cancer genome landscapes. Science. 339:1546–1558. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Estilo CL, O-charoenrat P, Talbot S, Socci ND, Carlson DL, Ghossein R, Williams T, Yonekawa Y, Ramanathan Y, Boyle JO, et al: Oral tongue cancer gene expression profiling: Identification of novel potential prognosticators by oligonucleotide microarray analysis. BMC Cancer. 9:112009. View Article : Google Scholar : PubMed/NCBI

24 

Reis PP, Waldron L, Perez-Ordonez B, Pintilie M, Galloni NN, Xuan Y, Cervigne NK, Warner GC, Makitie AA, Simpson C, et al: A gene signature in histologically normal surgical margins is predictive of oral carcinoma recurrence. BMC Cancer. 11:4372011. View Article : Google Scholar : PubMed/NCBI

25 

Krishnan NM, Dhas K, Nair J, Palve V, Bagwan J, Siddappa G, Suresh A, Kekatpure VD, Kuriakose MA and Panda B: A minimal DNA methylation signature in oral tongue squamous cell carcinoma links altered methylation with tumor attributes. Mol Cancer Res. 14:805–819. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Clough E and Barrett T: The gene expression omnibus database. Methods Mol Biol. 1418:93–110. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Davis S and Meltzer PS: GEOquery: A bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics. 23:1846–1847. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

29 

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

30 

Mandel-Brehm C, Retallack H, Knudsen GM, Yamana A, Hajj-Ali RA, Calabrese LH, Tihan T, Sample HA, Zorn KC, Gorman MP, et al: Exploratory proteomic analysis implicates the alternative complement cascade in primary CNS vasculitis. Neurology. 93:e433–e444. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Zhu Y, Qiu P and Ji Y: TCGA-assembler: Open-source software for retrieving and processing TCGA data. Nat Methods. 11:599–600. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Doecke JD, Chekouo TT, Stingo F and Do KA: miRNA target gene identification: Sourcing miRNA target gene relationships for the analyses of TCGA illumina MiSeq and RNA-Seq Hiseq platform data. Int J Hum Genet. 14:17–22. 2017. View Article : Google Scholar

33 

Chen WJ, Tang RX, He RQ, Li DY, Liang L, Zeng JH, Hu XH, Ma J, Li SK and Chen G: Clinical roles of the aberrantly expressed lncRNAs in lung squamous cell carcinoma: A studybased on RNA-sequencing and microarray data mining. Oncotarget. 8:61282–61304. 2017.PubMed/NCBI

34 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

37 

Amit M, Tam S, Takahashi H, Choi KY, Zafereo M, Bell D and Weber RS: Prognostic performance of the American Joint Committee on Cancer 8th edition of the TNM staging system in patients with early oral tongue cancer. Head Neck. 41:1270–1276. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Cancer Genome Atlas Network, . Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 517:576–582. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Ashley EA: The precision medicine initiative: A new national effort. JAMA. 313:2119–2120. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Pitiyage G, Tilakaratne WM, Tavassoli M and Warnakulasuriya S: Molecular markers in oral epithelial dysplasia: Review. J Oral Pathol Med. 38:737–752. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Duffy MJ: Use of biomarkers in screening for cancer. Adv Exp Med Biol. 867:27–39. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Kerkelä E and Saarialho-Kere U: Matrix metalloproteinases in tumor progression: Focus on basal and squamous cell skin cancer. Exp Dermatol. 12:109–125. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Stamenkovic I: Matrix metalloproteinases in tumor invasion and metastasis. Semin Cancer Biol. 10:415–433. 2000. View Article : Google Scholar : PubMed/NCBI

44 

Li H, Zhong A, Li S, Meng X, Wang X, Xu F and Lai M: The integrated pathway of TGFβ/Snail with TNFα/NFκB may facilitate the tumor-stroma interaction in the EMT process and colorectal cancer prognosis. Sci Rep. 7:49152017. View Article : Google Scholar : PubMed/NCBI

45 

Shen CJ, Kuo YL, Chen CC, Chen MJ and Cheng YM: MMP1 expression is activated by Slug and enhances multi-drug resistance (MDR) in breast cancer. PLoS One. 12:e01744872017. View Article : Google Scholar : PubMed/NCBI

46 

Lu X, Wang Q, Hu G, Van Poznak C, Fleisher M, Reiss M, Massagué J and Kang Y: ADAMTS1 and MMP1 proteolytically engage EGF-like ligands in an osteolytic signaling cascade for bone metastasis. Genes Dev. 23:1882–1894. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Mehner C, Miller E, Nassar A, Bamlet WR, Radisky ES and Radisky DC: Tumor cell expression of MMP3 as a prognostic factor for poor survival in pancreatic, pulmonary, and mammary carcinoma. Genes Cancer. 6:480–489. 2015.PubMed/NCBI

48 

Mariya T, Hirohashi Y, Torigoe T, Tabuchi Y, Asano T, Saijo H, Kuroda T, Yasuda K, Mizuuchi M, Saito T and Sato N: Matrix metalloproteinase-10 regulates stemness of ovarian cancer stem-like cells by activation of canonical Wnt signaling and can be a target of chemotherapy-resistant ovarian cancer. Oncotarget. 7:26806–26822. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Chung IC, Chen LC, Chung AK, Chao M, Huang HY, Hsueh C, Tsang NM, Chang KP, Liang Y, Li HP and Chang YS: Matrix metalloproteinase 12 is induced by heterogeneous nuclear ribonucleoprotein K and promotes migration and invasion in nasopharyngeal carcinoma. BMC Cancer. 14:3482014. View Article : Google Scholar : PubMed/NCBI

50 

Koshikawa N, Moriyama K, Takamura H, Mizushima H, Nagashima Y, Yanoma S and Miyazaki K: Overexpression of laminin gamma 2 chain monomer in invading gastric carcinoma cells. Cancer Res. 59:5596–5601. 1999.PubMed/NCBI

51 

Pyke C, Salo S, Ralfkiaer E, Rømer J, Danø K and Tryggvason K: Laminin-5 is a marker of invading cancer cells in some human carcinomas and is coexpressed with the receptor for urokinase plasminogen activator in budding cancer cells in colon adenocarcinomas. Cancer Res. 55:4132–4139. 1995.PubMed/NCBI

52 

Kagesato Y, Mizushima H, Koshikawa N, Kitamura H, Hayashi H, Ogawa N, Tsukuda M and Miyazaki K: Sole expression of laminin gamma 2 chain in invading tumor cells and its association with stromal fibrosis in lung adenocarcinomas. Jpn J Cancer Res. 92:184–192. 2001. View Article : Google Scholar : PubMed/NCBI

53 

Hlubek F, Jung A, Kotzor N, Kirchner T and Brabletz T: Expression of the invasion factor laminin gamma2 in colorectal carcinomas is regulated by beta-catenin. Cancer Res. 61:8089–8093. 2001.PubMed/NCBI

54 

Sordat I, Bosman FT, Dorta G, Rousselle P, Aberdam D, Blum AL and Sordat B: Differential expression of laminin-5 subunits and integrin receptors in human colorectal neoplasia. J Pathol. 185:44–52. 1998. View Article : Google Scholar : PubMed/NCBI

55 

Soini Y, Määttä M, Salo S, Tryggvason K and Autio-Harmainen H: Expression of the laminin gamma 2 chain in pancreatic adenocarcinoma. J Pathol. 180:290–294. 1996. View Article : Google Scholar : PubMed/NCBI

56 

Takahashi S, Hasebe T, Oda T, Sasaki S, Kinoshita T, Konishi M, Ochiai T and Ochiai A: Cytoplasmic expression of laminin gamma2 chain correlates with postoperative hepatic metastasis and poor prognosis in patients with pancreatic ductal adenocarcinoma. Cancer. 94:1894–1901. 2002. View Article : Google Scholar : PubMed/NCBI

57 

Yamamoto H, Itoh F, Iku S, Hosokawa M and Imai K: Expression of the gamma(2) chain of laminin-5 at the invasive front is associated with recurrence and poor prognosis in human esophageal squamous cell carcinoma. Clin Cancer Res. 7:896–900. 2001.PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

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
Zhong P, Liu L, Shen A, Chen Z, Hu X, Cai Y, Lin J, Wang B, Li J, Chen Y, Chen Y, et al: Five extracellular matrix‑associated genes upregulated in oral tongue squamous cell carcinoma: An integrated bioinformatics analysis. Oncol Lett 18: 5959-5967, 2019
APA
Zhong, P., Liu, L., Shen, A., Chen, Z., Hu, X., Cai, Y. ... Peng, J. (2019). Five extracellular matrix‑associated genes upregulated in oral tongue squamous cell carcinoma: An integrated bioinformatics analysis. Oncology Letters, 18, 5959-5967. https://doi.org/10.3892/ol.2019.10982
MLA
Zhong, P., Liu, L., Shen, A., Chen, Z., Hu, X., Cai, Y., Lin, J., Wang, B., Li, J., Chen, Y., Peng, J."Five extracellular matrix‑associated genes upregulated in oral tongue squamous cell carcinoma: An integrated bioinformatics analysis". Oncology Letters 18.6 (2019): 5959-5967.
Chicago
Zhong, P., Liu, L., Shen, A., Chen, Z., Hu, X., Cai, Y., Lin, J., Wang, B., Li, J., Chen, Y., Peng, J."Five extracellular matrix‑associated genes upregulated in oral tongue squamous cell carcinoma: An integrated bioinformatics analysis". Oncology Letters 18, no. 6 (2019): 5959-5967. https://doi.org/10.3892/ol.2019.10982