Open Access

Gene expression profiling via bioinformatics analysis reveals biomarkers in laryngeal squamous cell carcinoma

  • Authors:
    • Guo‑Fang Guan
    • Ying Zheng
    • Lian‑Ji Wen
    • De‑Jun Zhang
    • Duo‑Jiao Yu
    • Yan‑Qing Lu
    • Yan Zhao
    • Hui Zhang
  • View Affiliations

  • Published online on: April 29, 2015     https://doi.org/10.3892/mmr.2015.3701
  • Pages: 2457-2464
  • Copyright: © Guan et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

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


Abstract

The present study aimed to identify key genes and relevant microRNAs (miRNAs) involved in laryngeal squamous cell carcinoma (LSCC). The gene expression profiles of LSCC tissue samples were analyzed with various bioinformatics tools. A gene expression data set (GSE51985), including ten laryngeal squamous cell carcinoma (LSCC) tissue samples and ten adjacent non‑neoplastic tissue samples, was downloaded from the Gene Expression Omnibus. Differential analysis was performed using software package limma of R. Functional enrichment analysis was applied to the differentially expressed genes (DEGs) using the Database for Annotation, Visualization and Integrated Discovery. Protein‑protein interaction (PPI) networks were constructed for the protein products using information from the Search Tool for the Retrieval of Interacting Genes/Proteins. Module analysis was performed using ClusterONE (a software plugin from Cytoscape). MicroRNAs (miRNAs) regulating the DEGs were predicted using WebGestalt. A total of 461 DEGs were identified in LSCC, 297 of which were upregulated and 164 of which were downregulated. Cell cycle, proteasome and DNA replication were significantly over‑represented in the upregulated genes, while the ribosome was significantly over‑represented in the downregulated genes. Two PPI networks were constructed for the up‑ and downregulated genes. One module from the upregulated gene network was associated with protein kinase. Numerous miRNAs associated with LSCC were predicted, including miRNA (miR)‑25, miR‑32, miR‑92 and miR‑29. In conclusion, numerous key genes and pathways involved in LSCC were revealed, which may aid the advancement of current knowledge regarding the pathogenesis of LSCC. In addition, relevant miRNAs were also identified, which may represent potential biomarkers for use in the diagnosis or treatment of the disease.

Introduction

Laryngeal squamous cell carcinoma (LSCC) is the most common type of laryngeal cancer. It is able to spread to regional cervical lymph nodes, or to more distant tissues, for example the lung. Improvements in current therapies have resulted in an improved quality of life for patients with LSCC (1). However, survival rates have not been significantly improved, which identifies the need for a change in clinical approach, requiring novel biomarkers for diagnosis, prognostic assessment and drug design (2).

Certain achievements have been made in the identification of biomarkers of LSCC. Järvinen et al (3) performed high-resolution copy number and gene expression microarray analyses to identify 739 genes overexpressed in LSCC. Gajecka et al (4) reported that polymorphisms of CYP1A1, CYP2D6, CYP2E1, NAT2, GSTM1 and GSTT1 were associated with an increased risk of LSCC. HLA class I antigen downregulation was identified as a poor prognostic marker for LSCC, which may reflect the reduction in the extent of CD8(+) T cell infiltration in LSCC lesions (5). Overexpression of osteopontin enhances the proliferation and invasiveness of LSCC (6), suggesting that it may represent a potential therapeutic target.

MicroRNAs (miRNAs) are short regulatory RNAs that modulate gene expression at the post-transcriptional level, and are involved in the pathogenesis of numerous types of cancer (7). Multiple miRNAs have been associated with LSCC. Overexpression of miR-21 contributes to the malignant phenotype of LSCC via inhibition of BTG family member 2 (8). miR-203 inhibits the proliferation of laryngeal carcinoma cells by modulating their survival (9). In addition, let-7a (10), miR-16 (11) and hsa-miR-34c (12) also have roles in laryngeal carcinoma.

Microarray technology provides global patterns of gene expression and therefore facilitates biomarker discovery. Lian et al (13) investigated tumorigenesis and regional lymph node metastasis in LSCC, whereas the present study focused specifically on the discovery of biomarkers associated with tumorigenesis. In the present study, gene expression profiles of LSCC were analyzed with a variety of bioinformatics tools, including functional enrichment and network analyses, in order to identify novel potential biomarkers. Additionally, miRNAs targeting these genes were also predicted. The identification of such biomarkers may be useful in the diagnosis and/or treatment of LSCC.

Materials and methods

Gene expression data

A gene expression data set (accession number GSE51985), which included ten LSCC tissue samples and ten adjacent non-neoplastic tissue samples, was downloaded from the Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo) (13). Gene expression levels were measured using the Illumina HumanHT-12 V4.0 expression beadchip platform (Illumina, San Diego, CA, USA; http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GPL10558). Platform annotation files were also acquired.

Pretreatment and differential analysis

According to the annotation files, probes were initially mapped into the genes. If more than one probe was mapped into a single gene, levels of the probes were averaged as the final expression level for the specific gene. Following normalization, differential analysis was performed between the ten LSCC tissues and corresponding adjacent non-neoplastic tissues using the Linear Models for Microarray Analysis package (limma; http://www.bioconductor.org/packages/release/bioc/html/limma.html) (14) of R. P<0.05 and |log2 (fold change)|>1 were set as the threshold levels for the identification of differentially expressed genes (DEGs).

Functional enrichment analysis

Functional enrichment analysis facilitated the identification of altered biological functions. In the present study, functional enrichment analysis was performed on the DEGs using the Database for Annotation, Visualization and Integration Discovery, version 6.7 (DAVID; http://david.abcc.ncifcrf.gov/) (15), which is able to reveal enriched Gene Ontology (GO) terms; the Kyoto Encyclopedia of Genes and Genomes (KEGG) for pathways (16) and InterPro, version 34.0 for protein domains (17) based on the hypergeometric distribution. P<0.05 was set as the threshold.

Construction of protein-protein interaction (PPI) networks

Proteins ‘work together’ to exert certain biological functions, and the genome-wide identification of PPIs represents a significant step in the elucidation of the underlying molecular mechanisms. In present study, PPI networks were constructed for the protein products using information from the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING, version 9.1; http://string-db.org/) (18). Interactions with a score (i.e. required confidence) >0.4 were retained in the network.

The proteins in the network serve as the ‘nodes’, and each pairwise protein interaction is represented by an undirected link. The ‘degree’ of a node corresponds to the number of interactions of that particular protein. The most highly connected nodes (those of a high degree) were considered to be the network ‘hubs’.

Module analysis of the network

The PPI networks and regulatory associations between miRNAs and target genes were combined and subsequently visualized with Cytoscape, version 2.6.3 (http://cytoscape.org/) (19). Functional modules of the network were explored using ClusterONE, version 1.0 (http://www.paccanarolab.org/clusterone), a Cytoscape software plugin (20). P<0.01 was set as the cut-off value.

Prediction of miRNAs

The miRNAs regulating the identified DEGs were predicted using WEB-based Gene Set Analysis Toolkit, version 2.0 (WebGestalt; http://bioinfo.vanderbilt.edu/webgestalt/) (21). Count ≥2 and false discovery rate (false positive rate, multiple testing corrected P-value) <0.1 were set as the threshold values.

Results

DEGs in LSCC

According to the criteria outlined (|log2(fold change)|>1; P<0.05), a total of 461 DEGs were identified in LSCC, of which 297 were upregulated and 164 were downregulated.

Functional enrichment analysis

GO and KEGG pathway enrichment analyses were applied to the up- and downregulated genes using DAVID tools. The results are presented in Tables I and II.

Table I

GO enrichment analysis result for up- and downregulated genes in laryngeal squamous cell carcinoma.

Table I

GO enrichment analysis result for up- and downregulated genes in laryngeal squamous cell carcinoma.

CategoryTermCountP-value
Upregulated genes
Cluster 1Enrichment score: 14.33726443940616
 BPGO: 0007049~cell cycle63 3.98×10−27
 BPGO: 0000278~mitotic cell cycle46 4.67×10−27
 BPGO: 0022402~cell cycle process50 8.03×10−23
 BPGO: 0000279~M phase37 3.88×10−20
 BPGO: 0000280~nuclear division30 1.39×10−18
 BPGO: 0007067~mitosis30 1.39×10−18
 BPGO: 0000087~M phase of mitotic cell cycle30 2.30×10−18
 BPGO: 0048285~organelle fission30 4.23×10−18
 BPGO: 0022403~cell cycle phase38 1.02×10−17
 CCGO: 0043232~intracellular non-membrane-bounded organelle91 1.87×10−15
 CCGO: 0043228~non-membrane-bounded organelle91 1.87×10−15
 BPGO: 0051301~cell division29 3.35×10−14
 CCGO: 0005819~spindle21 1.24×10−13
 CCGO: 0015630~microtubule cytoskeleton35 4.61×10−12
 CCGO: 0044430~cytoskeletal part41 6.61×10−09
 BPGO: 0007017~microtubule-based process20 2.68×10−08
 CCGO: 0005856~cytoskeleton49 6.72×10−08
 CCGO: 0005815~microtubule organizing center17 2.28×10−06
 CCGO: 0005813~centrosome16 2.37×10−06
 CCGO: 0005874~microtubule14 3.79×10−04
Cluster 2Enrichment score: 10.562682054877438
 CCGO: 0031981~nuclear lumen64 11×10−14
 CCGO: 0070013~intracellular organelle lumen69 3.57×10−13
 CCGO: 0031974~membrane-enclosed lumen70 8.50×10−13
 CCGO: 0043233~organelle lumen69 1.07×10−12
 CCGO: 0005654~nucleoplasm38 2.84×10−08
 CCGO: 0005730~nucleolus29 4.12×10−06
Cluster 3Enrichment score: 9.848444253761741
 CCGO: 0005694~chromosome34 1.72×10−13
 CCGO: 0000775~chromosome, centromeric region19 7.04×10−13
 CCGO: 0044427~chromosomal part30 1.90×10−12
 CCGO: 0000793~condensed chromosome17 1.60×10−10
 CCGO: 0000779~condensed chromosome, centromeric region12 5.30×10−09
 CCGO: 0000777~condensed chromosome kinetochore11 1.91×10−08
 CCGO: 0000776~kinetochore11 3.10×10−07
Cluster 4Enrichment score: 7.286973695097799
 MFGO: 0005524~adenosine triphosphate binding52 9.82×10−09
 MFGO: 0032559~adenyl ribonucleotide binding52 1.54×10−08
 MFGO: 0030554~adenyl nucleotide binding53 3.18×10−08
 MFGO: 0001883~purine nucleoside binding53 5.25×10−08
 MFGO: 0001882~nucleoside binding53 6.58×10−08
 MFGO: 0017076~purine nucleotide binding59 8.65×10−08
 MFGO: 0032553~ribonucleotide binding57 1.19×10−07
 MFGO: 0032555~purine ribonucleotide binding57 1.19×10−07
 MFGO: 0000166~nucleotide binding65 1.29×10−07
Cluster 5Enrichment score: 5.683294675111861
 BPGO: 0007017~microtubule-based process20 2.68×10−08
 BPGO: 0000226~microtubule cytoskeleton organization15 1.06×10−07
 BPGO: 0007051~spindle organization7 7.88×10−05
 BPGO: 0007010~cytoskeleton organization20 8.29×10−05
Downregulated genes
Cluster 1Enrichment score: 2.991494823613603
 BPGO: 0006414~translational elongation9 3.72×10−07
 MFGO: 0003735~structural constituent of ribosome10 2.59×10−06
 CCGO: 0033279~ribosomal subunit9 4.39×10−06
 CCGO: 0022626~cytosolic ribosome7 2.80×10−05
 CCGO: 0005840~ribosome10 2.81×10−05
 BPGO: 0006412~translation11 7.45×10−05
 CCGO: 0015934~large ribosomal subunit6 1.30×10−04
 CCGO: 0044445~cytosolic part7 8.82×10−04
 CCGO: 0022625~cytosolic large ribosomal subunit4 2.70×10−03
 CCGO: 0030529~ribonucleoprotein complex11 4.49×10−03
 MFGO: 0005198~structural molecule activity12 4.68×10−03

[i] Cluster, functional cluster; enrichment score, score for a functional cluster reflecting clustering effect; BP, biological process; CC, cellular components; MF, molecular function; count, number differentially expressed in a specific pathway; GO, gene ontology.

Table II

Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis result for up-regulated genes in laryngeal squamous cell carcinoma.

Table II

Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis result for up-regulated genes in laryngeal squamous cell carcinoma.

TermCountP-value
Upregulated genes
 Hsa04110: Cell cycle16 4.12×10−09
 Hsa03050: Proteasome11 6.84×10−09
 Hsa03030: DNA replication6 4.21×10−04
 Hsa04114: Oocyte meiosis8 3.54×10−03
 Hsa03040: Spliceosome8 7.43×10−03
Downregulated genes
 Hsa03010: Ribosome8 5.59×10−06
 Hsa00071: Fatty acid metabolism4 4.19×10−03

The top five gene clusters with enrichment scores >2 are listed in Table I. The following molecular functional terms were significantly over-represented amongst the upregulated genes: ATP binding (GO: 0005524), adenyl ribonucleotide binding (GO: 0032559), adenyl nucleotide binding (GO: 0030554), purine nucleoside binding (GO: 0001883), nucleoside binding (GO: 0001882), purine nucleotide binding (GO: 0017076), ribonucleotide binding (GO: 0032553), purine ribonucleotide binding (GO: 0032555) and nucleotide binding (GO: 0000166). Amongst the downregulated genes, structural constituent of ribosome (GO: 0003735) and structural molecule activity (GO: 0005198) were significantly enriched.

In addition, the cell cycle (hsa04110), proteasome (hsa03050) and DNA replication (hsa03030) pathways were significantly enriched in the upregulated genes, while ribosome (hsa03010) was significantly enriched in the downregulated genes.

PPI networks of the DEGs

A PPI network was constructed for the protein products of the DEGs using STRING. Interactions with a combined score of >0.4 were included in the network.

A network consisting of 213 proteins (nodes) and 2719 interactions (edges) was generated for the upregulated genes (Fig. 1). The top nodes, or hubs, characterized by a degree of >70, were aurora kinase B (AURKB), cyclin dependent kinase (CDK)1, cell division cycle (CDC) 20 homolog, AURKA, CDC45-like, budding uninhibited by benzimidazoles 1 homolog (BUB1), PDZ binding kinase (PBK), non-SMC condensin I complex subunit G (NCAPG), cell division cycle associated 8 (CDCA8), ubiquitin-conjugating enzyme E2T (UBE2T), minichromosome maintenance complex component 2 (MCM2), centromere protein F (CENPF), MCM4, NDC80 homolog kinetochore complex component (NDC80), CENPA, baculoviral IAP repeat-containing 5 (BIRC5), denticleless homolog (DTL), CHK1 checkpoint homolog (CHEK1), kinesin family member 2C (KIF2C), maternal embryonic leucine zipper kinase (MELK), topoisomerase (DNA) II α 170kDa (TOP2A) and cell division cycle associated 5 (CDCA5). Furthermore, a network comprised of 50 nodes and 80 edges was obtained for the downregulated genes. Few PPIs were observed amongst the downregulated genes, and therefore the subsequent analyses were focused on the upregulated genes.

Module analysis

Module analysis was performed using ClusterONE to predict protein complexes, and the results are presented in Fig. 2. Two modules were identified amongst the upregulated genes: Module 1 (P<0.000) and Module 2 (P=5.559×10−4). Protein domain enrichment analysis was applied to the genes in the two modules using InterPro and the results are displayed in Table III. Serine/threonine protein kinase, active site (IPR008271); protein kinase, ATP binding site (IPR017441) and protein kinase, core (IPR000719) were significantly enriched amongst the genes from Module 1. No significantly enriched term was identified in the genes from Module 2. One module was generated by the downregulated genes (Fig. 2), but no significantly enriched protein domain was identified.

Table III

Significantly over-represented protein domains in the genes from the Module 1 network of upregulated genes.

Table III

Significantly over-represented protein domains in the genes from the Module 1 network of upregulated genes.

TermCountP-value
IPR008271: Serine/threonine protein kinase, active site11 1.61×10−06
IPR017441: Protein kinase, ATP binding site12 2.11×10−06
IPR000719: Protein kinase, core12 3.25×10−06
IPR002290: Serine/threonine protein kinase9 1.01×10−05
IPR017442: Serine/threonine protein kinase-related10 1.46×10−05
IPR018525: DNA-dependent ATPase MCM, conserved site3 4.52×10−04
IPR001208: DNA-dependent ATPase MCM3 5.80×10−04
IPR019821: Kinesin, motor region, conserved site4 6.66×10−04
IPR001752: Kinesin, motor region4 6.66×10−04
IPR007125: Histone core4 8.16×10−04
IPR009072: Histone-fold4 1.39×10−03
IPR002119: Histone H2A3 2.68×10ss03

[i] ATP, adenosine triphosphate; MCM, minichromosome maintenance complex component.

miRNA-target interactions

miRNAs regulating the DEGs were predicted by WebGestalt, and the results are exhibited in Table IV and Fig. 3. hsa_GTGCAAT, miR-25, miR-32, miR-92, miR-363, miR-367, hsa_TGCTGCT, miR-15A, miR-16, miR-15B, miR-195, miR-424, miR-497, hsa_TGGTGCT, miR-29A, miR-29B and miR-29C were included in the list of identified miRNAs.

Table IV

Predicted miRNAs regulating the DEGs.

Table IV

Predicted miRNAs regulating the DEGs.

miRNACountCOERrawPadjP
Hsa_GTGCAAT, miR-25, miR-32, miR-92, miR-363, miR-36712308123.423.510.00020.0123
Hsa_TGCTGCT, miR-15A, miR-16, miR-15B, miR-195, miR-424, miR-49718593186.592.730.00010.0123
Hsa_TGGTGCT, miR-29A, miR-29B, miR-29C15515155.722.620.00070.0287
Hsa_ACTACCT, miR-196A, miR-196B714371.594.410.00110.0338
Hsa_ATATGCA, miR-448820882.313.460.00240.0492
Hsa_TAATGTG, miR-323715871.753.990.00200.0492
Hsa_AACTGAC, miR-22359451.044.790.00400.0703
Hsa_TATCTGG, miR-48846240.695.810.00500.0724
Hsa_TGTTTAC, miR-30A-5P, miR-30C, miR-30D, miR-30B, miR-30E-5P14572146.352.200.00530.0724
Hsa_CAGCAGG, miR-370615361.703.530.00750.0922

[i] miRNA, micro RNA; DEGs, differentially expressed genes; miR, micro RNA; Count, number of DEGS regulated by a specific miRNA; C, number of reference genes in the category; O, number of genes in the gene set and also in the category; E, expected number in the category; R, ratio of enrichment; rawP, raw P-value calculated by WebGastalt; adjP, adjusted P-value.

Gene regulatory networks

The miRNA-target interactions were visualized by Cytoscape and thereby a gene regulatory network was established (Fig. 3).

Discussion

Through the comparative analysis of gene expression data of LSCC and adjacent non-neoplastic control tissues, a total of 461 DEGs were identified in LSCC, of which 297 were upregulated and 164 were downregulated. Functional enrichment analysis indicated that the cell cycle, proteasome and relevant biological pathways were over-represented amongst the upregulated genes. Cell cycle progression is closely associated with multiple types of cancer (2223), which indicated that the analysis results were of high confidence.

According to the results of previous studies, specific DEGs including CDK4, CDK1, MCM2, MCM3 and MCM4, have been implicated in LSCC (13,24). CDK4 is required for cell cycle G1 phase progression (25). Dong et al (26) reported that CDK4 was overexpressed in LSCC and suggested that it may have a critical role in cell proliferation together with cyclin D1. MCM2 has been implicated in the initiation of eukaryotic genome replication, which has been proposed as a marker of dysplasia and malignancy (27). Chatrath et al (28) reported aberrant expression of MCM2 in LSCC, while Torres-Rendon et al (29) indicated that MCM2 may be an indicator of growth and may provide a useful prognostic tool for oral epithelial dysplasia. MCM3 and MCM4 were also identified as DEGs in the present study. These findings were consistent with the results presented in a study by Lian et al (13).

Additional potential biomarkers were discovered in the present study. There is emerging evidence that glycogen synthase kinase (GSK)3β may be a tumor suppressor in oral cancer (30). It was therefore hypothesized that GSK3β may function in a similar way in LSCC. Several subunits of the proteasome were also identified in LSCC, including proteasome subunit β type 4 (PSMB4), PSMB7, PSMB1 and PSMC3. The ubiquitin-proteasome system is a critical regulator of cell growth and apoptosis (31), and proteasome inhibitors have been developed for use in cancer therapy (3233). PSMB4 was identified as the first proteasomal subunit with oncogenic properties, promoting cancer cell survival and tumor growth in vivo (34). Elevated expression of PSMB4 is associated with poor prognosis in human cancer (34). PSMB7 was identified to be a prognostic biomarker in breast cancer (35). Therefore, further study of these subunits may reveal novel biomarkers for LSCC.

Network and module analyses were performed for the DEGs, and the identification of hub nodes and interactions may aid the elucidation of the underlying molecular mechanisms. AURKA and AURKB had a high degree in the PPI network for upregulated genes. Overexpression and hyperactivation of AURKA and AURKB have major roles in tumorigenesis, and therefore their inhibitors are already regarded as promising therapeutics for various types of cancer (36), including head and neck squamous-cell carcinoma (37). MCM2 and MCM4 also had a degree of >70, confirming their significant roles in the pathogenesis of LSCC.

Considering that miRNAs are closely involved in multiple types of cancer, a number of miRNAs targeting the DEGs were predicted by WebGestalt, including miR-15, miR-16, miR-25 and miR-195. Wu et al (11) reported that miR-16 was upregulated in LSCC and that it targets zyxin and promotes cell motility in human laryngeal carcinoma cell line HEp-2. Other miRNAs have been reported to have roles in various types of cancer. miR-15a forms a cluster with miR-16 at the chromosomal region 13q14, and functions as a putative tumor suppressor by targeting the oncogene BCL2 (38,39). miR-25 regulates apoptosis by targeting Bim in human ovarian cancer (40) and miR-195 is regarded as a predictor of poor prognosis in adrenocortical cancer (41). Future studies of these miRNAs may better describe the regulatory mechanisms underlying LSCC.

In conclusion, a number of key genes in LSCC were identified, which may represent novel biomarkers for diagnosis, prognosis and therapy. In addition, relevant miRNAs were also explored, and these may offer therapeutic targets for the modulation of abnormal gene expression in LSCC.

References

1 

Schorn VJ and Miles BA: Laryngeal squamous cell carcinoma. ENT Board Prep. Lin F and Patel Z: Springer; New York: pp. 227–233. 2014, View Article : Google Scholar

2 

Almadori G, Bussu F, Cadoni G, Galli J, Paludetti G and Maurizi M: Molecular markers in laryngeal squamous cell carcinoma: towards an integrated clinicobiological approach. Eur J Cancer. 41:683–693. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Järvinen AK, Autio R, Haapa-Paananen S, et al: Identification of target genes in laryngeal squamous cell carcinoma by high-resolution copy number and gene expression microarray analyses. Oncogene. 25:6997–7008. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Gajecka M, Rydzanicz M, Jaskula-Sztul R, Kujawski M, Szyfter W and Szyfter K: CYP1A1, CYP2D6, CYP2E1, NAT2, GSTM1 and GSTT1 polymorphisms or their combinations are associated with the increased risk of the laryngeal squamous cell carcinoma. Mutat Res. 574:112–123. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Ogino T, Shigyo H, Ishii H, et al: HLA class I antigen down-regulation in primary laryngeal squamous cell carcinoma lesions as a poor prognostic marker. Cancer Res. 66:9281–9289. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Celetti A, Testa D, Staibano S, et al: Overexpression of the cytokine osteopontin identifies aggressive laryngeal squamous cell carcinomas and enhances carcinoma cell proliferation and invasiveness. Clin Cancer Res. 11:8019–8027. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Zimmerman AL and Wu S: MicroRNAs, cancer and cancer stem cells. Cancer Lett. 300:10–19. 2011. View Article : Google Scholar

8 

Liu M, Wu H, Liu T, et al: Regulation of the cell cycle gene, BTG2, by miR-21 in human laryngeal carcinoma. Cell Res. 19:828–837. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Bian K, Fan J, Zhang X, et al: MicroRNA-203 leads to G1 phase cell cycle arrest in laryngeal carcinoma cells by directly targeting survivin. FEBS Lett. 586:804–809. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Long XB, Sun GB, Hu S, et al: Let-7a microRNA functions as a potential tumor suppressor in human laryngeal cancer. Oncol Rep. 22:1189–1195. 2009.PubMed/NCBI

11 

Wu H, Liu T, Wang R, et al: MicroRNA-16 targets zyxin and promotes cell motility in human laryngeal carcinoma cell line HEp-2. IUBMB Life. 63:101–108. 2011.PubMed/NCBI

12 

Cai KM, Bao XL, Kong XH, et al: Hsa-miR-34c suppresses growth and invasion of human laryngeal carcinoma cells via targeting c-Met. Int J Mol Med. 25:565–571. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Lian M, Fang J, Han D, et al: Microarray gene expression analysis of tumorigenesis and regional lymph node metastasis in laryngeal squamous cell carcinoma. PLoS One. 8:e848542013. View Article : Google Scholar

14 

Diboun I, Wernisch L, Orengo CA and Koltzenburg M: Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics. 7:2522006. View Article : Google Scholar : PubMed/NCBI

15 

Huang DW, Sherman BT, Tan Q, et al: The DAVID gene functional classification tool: a novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 8:R1832007. View Article : Google Scholar : PubMed/NCBI

16 

Aoki-Kinoshita KF and Kanehisa M: Gene annotation and pathway mapping in KEGG. Methods Mol Biol. 396:71–91. 2007.PubMed/NCBI

17 

Hunter S, Jones P, Mitchell A, et al: InterPro in 2011: new developments in the family and domain prediction database. Nucleic Acids Res. 40:D306–D312. 2012. View Article : Google Scholar :

18 

von Mering C, Huynen M, Jaeggi D, Schmidt S, Bork P and Snel B: STRING: a database of predicted functional associations between proteins. Nucleic Acids Res. 31:258–261. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Shannon P, Markiel A, Ozier O, et al: Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Nepusz T, Yu H and Paccanaro A: Detecting overlapping protein complexes in protein-protein interaction networks. Nat Methods. 9:471–472. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Zhang B, Kirov S and Snoddy J: WebGestalt: an integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res. 33:W741–W748. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Evan GI and Vousden KH: Proliferation, cell cycle and apoptosis in cancer. Nature. 411:342–348. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Kastan MB and Bartek J: Cell-cycle checkpoints and cancer. Nature. 432:316–323. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Cai K, Luo Y and Li L: Expression of MCM2 protein and its biological characteristic in human laryngeal squamous cell carcinoma. J Acta Academiae Medicinae Jiangxi. 3:58–60. 2008.

25 

Sherr CJ: G1 phase progression: cycling on cue. J Cell. 79:551–555. 1994. View Article : Google Scholar

26 

Dong Y, Sui L, Sugimoto K, Tai Y and Tokuda M: Cyclin D1. CDK4 complex, a possible critical factor for cell proliferation and prognosis in laryngeal squamous cell carcinomas. Int J Cancer. 95:209–215. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Freeman A, Morris LS, Mills AD, et al: Minichromosome maintenance proteins as biological markers of dysplasia and malignancy. Clin Cancer Res. 5:2121–2132. 1999.PubMed/NCBI

28 

Chatrath P, Scott IS, Morris LS, et al: Aberrant expression of minichromosome maintenance protein-2 and Ki67 in laryngeal squamous epithelial lesions. Br J Cancer. 89:1048–1054. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Torres-Rendon A, Roy S, Craig G and Speight P: Expression of Mcm2, geminin and Ki67 in normal oral mucosa, oral epithelial dysplasias and their corresponding squamous-cell carcinomas. Br J Cancer. 100:1128–1134. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Mishra R: Glycogen synthase kinase 3 beta: can it be a target for oral cancer. Mol Cancer. 9:1442010. View Article : Google Scholar : PubMed/NCBI

31 

Almond JB and Cohen GM: The proteasome: a novel target for cancer chemotherapy. Leukemia. 16:433–443. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Orlowski RZ and Kuhn DJ: Proteasome inhibitors in cancer therapy: lessons from the first decade. Clin Cancer Res. 14:1649–1657. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Rajkumar SV, Richardson PG, Hideshima T and Anderson KC: Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol. 23:630–639. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Lee GY, Haverty PM, Li L, et al: Comparative oncogenomics identifies PSMB4 and SHMT2 as potential cancer driver genes. Cancer Res. 74:3114–3126. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Munkacsy G, Abdul-Ghani R, Mihaly Z, et al: PSMB7 is associated with anthracycline resistance and is a prognostic biomarker in breast cancer. Br J Cancer. 102:361–368. 2010. View Article : Google Scholar :

36 

Dar AA, Goff LW, Majid S, Berlin J and El-Rifai W: Aurora kinase inhibitors-rising stars in cancer therapeutics? Mol Cancer Ther. 9:268–278. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Mehra R, Serebriiskii IG, Burtness B, Astsaturov I and Golemis EA: Aurora kinases in head and neck cancer. Lancet Oncol. 14:e425–e435. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Bonci D, Coppola V, Musumeci M, et al: The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med. 14:1271–1277. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Cimmino A, Calin GA, Fabbri M, et al: miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA. 102:13944–13949. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Zhang H, Zuo Z, Lu X, Wang L, Wang H and Zhu Z: MiR-25 regulates apoptosis by targeting Bim in human ovarian cancer. Oncol Rep. 27:594–598. 2012.

41 

Soon PS, Tacon LJ, Gill AJ, et al: miR-195 and miR-483-5p identified as predictors of poor prognosis in adrenocortical cancer. Clin Cancer Res. 15:7684–7692. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2015
Volume 12 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guan GF, Zheng Y, Wen LJ, Zhang DJ, Yu DJ, Lu YQ, Zhao Y and Zhang H: Gene expression profiling via bioinformatics analysis reveals biomarkers in laryngeal squamous cell carcinoma. Mol Med Rep 12: 2457-2464, 2015
APA
Guan, G., Zheng, Y., Wen, L., Zhang, D., Yu, D., Lu, Y. ... Zhang, H. (2015). Gene expression profiling via bioinformatics analysis reveals biomarkers in laryngeal squamous cell carcinoma. Molecular Medicine Reports, 12, 2457-2464. https://doi.org/10.3892/mmr.2015.3701
MLA
Guan, G., Zheng, Y., Wen, L., Zhang, D., Yu, D., Lu, Y., Zhao, Y., Zhang, H."Gene expression profiling via bioinformatics analysis reveals biomarkers in laryngeal squamous cell carcinoma". Molecular Medicine Reports 12.2 (2015): 2457-2464.
Chicago
Guan, G., Zheng, Y., Wen, L., Zhang, D., Yu, D., Lu, Y., Zhao, Y., Zhang, H."Gene expression profiling via bioinformatics analysis reveals biomarkers in laryngeal squamous cell carcinoma". Molecular Medicine Reports 12, no. 2 (2015): 2457-2464. https://doi.org/10.3892/mmr.2015.3701