Identification of prognostic microRNA candidates for head and neck squamous cell carcinoma

  • Authors:
    • Lian Hui
    • Hua Wu
    • Ning Yang
    • Xing Guo
    • Xuejun Jang
  • View Affiliations

  • Published online on: March 23, 2016     https://doi.org/10.3892/or.2016.4698
  • Pages: 3321-3330
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to uncover potential prognostic microRNA (miRNA) markers in head and neck squamous cell carcinoma (HNSCC). miRNA expression profile and clinical data were obtained from The Cancer Genome Atlas (TCGA) database. Survival analysis was conducted by Kaplan-Meier method. Then, candidate prognostic miRNAs were screened via Cox proportional hazards regression analysis. Furthermore, target genes of miRNAs were predicted, and their interactions and functions were then analyzed. A total of 15 miRNAs were discovered to be significantly related to overall survival. Among them, miR-1251, miR-618 and miR-328 with p<0.05 were potentially significant prognostic factors of HNSCC. In the protein-protein interaction (PPI) network, the target gene of miR-328, MAX, interacted with multiple genes. The target gene of miR-618, E2F1, interacted with genes such as MAX, SMAD3 and IGF1. Furthermore, the target genes of miR-618 (e.g. E2F1 and SMAD3), and the target genes of miR-328 (e.g. MAX) were significantly enriched in the functions of transcriptional regulation. The target gene of miR-1251, IGF1, was associated with functions such as signal transduction. The above miRNAs (miR-1251, miR-618 and miR-328) may be potential prognostic markers in HNSCC.

Introduction

Head and neck squamous cell carcinoma (HNSCC) accounts for the major proportion of head and neck cancers and is the sixth most common non-skin cancer worldwide (1). The high mortality rate of HNSCC results from poor prognosis and lack of reliable prognostic signatures. Thus, it is urgent to reveal more reliable prognostic markers to achieve better outcomes for HNSCC patients.

MicroRNAs (miRNAs) are small non-protein-coding RNAs of ~22 nucleotides, and they play roles in various biological functions, such as cell growth, differentiation and the development of disease (2). Dysregulation of miRNAs is also closely related to the development, diagnosis and prognosis of many types of cancers including HNSCC (3). For instance, miR-218 inhibits cell migration and invasion by suppressing the expression of laminin-332 in HNSCC (4). By suppressing invadopodia activity, miR-375 impairs the tumor invasion of HNSCC and is implicated in the poor outcome of HNSCC (5,6). Moreover, poorer overall survival of HNSCC has been found to be linked with the promoter methylation of miR-137 (7). In addition, overexpression of miR-205 is able to mediate HNSCC growth and E2F1 signaling (8). Although more and more studies have focused on uncovering miRNAs that are involved in HNSCC, few miRNAs have been identified as potential prognostic markers for HNSCC.

In the present study, to identify potential prognostic miRNA markers for HNSCC, miRNA expression profile and clinical data of 397 HNSCC cases were obtained from The Cancer Genome Atlas (TCGA) database. Survival and Cox regression analyses were performed to identify the miRNAs that were potentially associated with the survival of HNSCC patients. Furthermore, target genes of those miRNAs were predicted and the functions of the target genes were analyzed. The results may provide new information for the further study of HNSCC, and the identified miRNAs may be used as prognostic markers for HNSCC, which may be useful to improve future clinical outcome of HNSCC.

Materials and methods

Data acquisition

The miRNA expression profiling and clinical data of HNSCC were obtained from the TCGA database (http://cancergenome.nih.gov/, the deadline of data downloading: July 20, 2014). The miRNA expression profiling data included a total of 513 cases, and they were produced by two platforms: BCGSC_IlluminaGA_miRNASeq (37 cases) and BCGSC_IlluminaHiSeq_miRNASeq (476 cases). The clinical data included 418 HNSCC cases. After data filtering, only cases with both miRNA expression profile and clinical survival data (397 cases) were retained for further analysis.

Survival analysis

The average expression value of each miRNA in the 397 cases was calculated, and the mean value was set as the critical value. Then, the cases were divided into two groups according to the miRNA expression values higher or lower than the critical value. Furthermore, survival estimates were calculated by the Kaplan-Meier (KM) method and the log-rank test based on a survival package in R (9). Only the miRNAs with p<0.05 were considered significant. The miRNAs that had a significant association with survival time were selected for subsequent analysis.

Cox regression analysis

The miRNAs having a significant impact on survival time and clinical survival data received Cox proportional hazards regression analysis (10) based on KMsurv package in R (11). Patients with a high risk score were considered to have poor survival. The miRNAs with p<0.05 were chosen for further analysis.

Target gene prediction

The target genes of the miRNAs with p<0.05 in the Cox regression analysis were obtained based on the databases of miRecords (http://c1.accurascience.com/miRecords/) (12) and MiRWalk (http://www.umm.uni-heidelberg.de/apps/zmf/mirwalk/) (13). Only the regulatory relationships recorded at least in 3 of the databases including miRanda (14), MirTarget2 (15), PicTar (16), PITA (17) and TargetScan (18) were selected for further analysis.

Construction of protein-protein interaction (PPI) network for target genes

PPIs between target genes of the miRNAs were analyzed via the online database Search Tool for the Retrieval of Interacting Genes (STRING, http://string-db.org/) (19). Then, target gene pairs with a combined score >0.4 were chosen to construct PPI networks, which were visualized using the Cytoscape software (http://www.cytoscape.org/) (20). In the network, a node represents a protein (gene), and lines represent the interactions of the proteins. The 'degree' of each node is equal to the number of nodes that interact with this node. The larger the degree is, the closer the connections with other nodes are.

Functional analyses of target genes

The Database for Annotation, Visualization and Integrated Discovery (DAVID; https://david.ncifcrf.gov/) was applied to investigate the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of the target genes (21). p<0.05 was chosen as the cut-off criterion.

To further investigate the function of the target genes, the GenCLip tool (22) was utilized to perform functional clustering for genes in the PPI network based on literature mining, and the cut-off criteria were p<1E-04 and hit ≥9. Additionally, pathway enrichment analysis was performed for the genes in the PPI network, based on the Gene Set Enrichment Analysis (GSEA) database (http://software.broadinstitute.org/gsea/index.jsp). p<0.05 was set as the cut-off criterion.

Results

Survival analysis

In total, expression values of 1,046 miRNAs were obtained from the TCGA database. Among the miRNAs, a total of 15 miRNAs were discovered to be significantly related to survival time. Low expression of the 15 miRNAs was correlated with a higher survival rate based on the Kaplan-Meier survival analysis. Among the 15 miRNAs, miR-4292, miR-3685 and miR-1302.3 with no expression values in most of the samples were deleted and 12 miRNAs were finally screened.

Cox proportional hazards analyses

Cox proportional hazards analysis was performed with the clinical data [demographic characteristics, tumor-node-metastasis (TNM) classification, history of drinking and smoking] and the 12 miRNAs. Compared with the other clinical factors, miR-1251, miR-618 and miR-328 with p<0.05 were significantly associated with risk factors of HNSCC (Table I).

Table I

Results of the Cox proportional hazard regression analysis.

Table I

Results of the Cox proportional hazard regression analysis.

FactorβHRP-valueLower 95%Upper 95%
Gender0.0701.0720.660.78781.459
Smoke0.1941.2140.190.90941.620
Alcohol0.0891.0930.550.81711.462
Clinical_M0.5761.7780.140.82183.848
Clinical_N−0.0490.9520.320.86481.049
Clinical_T0.0231.0230.760.88291.186
Clinical_stage0.0641.0660.620.82691.374
hsa-mir-643−0.1230.8840.390.66761.171
hsa-mir-1260.00004181.0000.440.99991.000
hsa-mir-12510.2931.3410.011.08971.649
hsa-mir-521-2−0.0010.9991.000.29493.386
hsa-mir-6180.2041.2260.011.05581.423
hsa-mir-5020.01351.0140.430.97991.048
hsa-mir-3280.01471.0150.021.00241.027
hsa-mir-193a−0.0010.9990.050.99821.000
hsa-mir-1301−0.0160.9840.220.95951.010
hsa-mir-12310.7752.1700.090.89215.278
hsa-mir-548y0.2751.3170.060.99061.750
hsa-mir-519c0.0681.0700.600.82741.385

[i] β, regression coefficient; HR, hazard ratio. Lower 95% and upper 95% represent the lower limit and the upper limit of the confidence interval, respectively.

Analysis of PPI network of target genes

A total of 245 genes were discovered to be regulated by miR-1251, miR-618 and miR-328. miR-328 modulated most of the genes, such as MAX. miR-618 regulated genes such as E2F1 and SMAD3. miR-1251 targeted 6 genes such as IGF1 (Fig. 1).

To further analyze the interactions of the target genes, an interaction network of the target gene was further constructed based on the STRING database. The PPI network was composed of 97 nodes and 112 lines (Fig. 2). Several genes had strong interactions with the other genes. For example, MAX strongly interacted with 8 genes (e.g. E2F1); E2F1 interacted with MAX, SMAD3 and E2F1.

Functional analysis of target genes

To investigate the functions of the three miRNAs that were found to be potentially associated with risk factors of HNSCC, GO functional and KEGG pathway enrichment analyses of the target genes were performed. The target genes of hsa-mir-328 were mainly enriched in the GO terms of positive regulation of transcription (e.g. IL5 and FOXA2) and transcription activator activity (e.g. FOXA2 and MAX), as well as the pathways in cancer (e.g. MAX and SLC2A1) (Table II). Meanwhile, the target genes of hsa-mir-618 were mainly enriched in the GO terms of the negative regulation of transcription from the RNA polymerase II promoter (e.g. E2F1 and ID2) and transcription repressor activity (e.g. E2F1 and ID2), as well as the TGF-β signaling pathway (e.g. ID2 and SMAD3) (Table III). There were no significant GO and pathway terms enriched by the target genes of hsa-mir-1251.

Table II

The enriched GO and pathway terms for target genes of hsa-mir-328.

Table II

The enriched GO and pathway terms for target genes of hsa-mir-328.

CategoryTermCountP-valueTarget genes
GOTERM_BP_FATGO:0045941-positive regulation of transcription205.51E-06ESRRA, IL5, FOXA2, SOX11, ARID1B, NPAS4, SIX4, FOXO4, FOXP3, TCF7L2, NRIP1, CITED2, PRDM7, HAND2, EBF3, NEUROD1, HOXB9, PBX2, SERTAD2, ZNF423
GOTERM_BP_FATGO:0045935-positive regulation of nucleobase, nucleoside, nucleotide and nucleic acid metabolic process216.43E-06ESRRA, IL5, FOXA2, SOX11, ARID1B, NPAS4, SIX4, FOXO4, FOXP3, TCF7L2, NRIP1, CITED2, PRDM7, HAND2, EBF3, NEUROD1, HOXB9, H2AFX, PBX2, SERTAD2, ZNF423
GOTERM_BP_FATGO:0010628-positive regulation of gene expression208.43E-06ESRRA, IL5, FOXA2, SOX11, ARID1B, NPAS4, SIX4, FOXO4, FOXP3, TCF7L2, NRIP1, CITED2, PRDM7, HAND2, EBF3, NEUROD1, HOXB9, PBX2, SERTAD2, ZNF423
GOTERM_BP_FATGO:0051173-positive regulation of nitrogen compound metabolic process211.02E-05ESRRA, IL5, FOXA2, SOX11, ARID1B, NPAS4, SIX4, FOXO4, FOXP3, TCF7L2, NRIP1, CITED2, PRDM7, HAND2, EBF3, NEUROD1, HOXB9, H2AFX, PBX2, SERTAD2, ZNF423
GOTERM_BP_FATGO:0010557-positive regulation of macromolecule biosynthetic process211.28E-05ESRRA, IL5, FOXA2, SOX11, ARID1B, NPAS4, SIX4, FOXO4, FOXP3, TCF7L2, NRIP1, TGFB2, CITED2, PRDM7, HAND2, EBF3, NEUROD1, HOXB9, PBX2, SERTAD2, ZNF423
GOTERM_CC_FAT GO:0005829-cytosol220.018286095ARL6IP1, FGD1, ARHGEF3, WDTC1, DIAPH2, NR3C1, FOXO4, ACSS2, TCF7L2, CAPRIN1, PLCE1, TIAM2, SNRPB, SPTA1, UROC1, PLCB1, CRK, CLINT1, TRIM39, PSTPIP2, AGPAT1, EDARADD
GOTERM_CC_FATGO:0031981-nuclear lumen220.042046874DNMT3A, UBE2Z, SOX11, ADNP, TLE1, NR3C1, NPAS4, ARID1B, ACSS2, TCF7L2, NRIP1, HAND2, ANXA11, PHF15, DDB2, SNRPB, DHX33, H2AFX, DNAJB1, HIST1H4D, PBX2, TSEN15
GOTERM_CC_FAT GO:0000785-chromatin60.04651322DNMT3A, ARID4B, MECP2, H2AFX, HIST1H4D, CITED2
GOTERM_MF_FAT GO:0016563-transcription activator activity145.97E-04FOXA2, NPAS4, ARID1B, FOXP3, FOXO4, NRIP1, CITED2, MAX, HAND2, NKX3-1, NEUROD1, HOXB9, ZNF423, SERTAD2
GOTERM_MF_FAT GO:0030528-transcription regulator activity319.24E-04FOXA2, CNOT2, NR3C1, FOXO4, TCF7L2, CITED2, HIC1, MAX, HAND2, NKX3-1, SERTAD2, ZNF423, ESRRA, SOX11, ADNP, RUNX1T1, MECP2, TLE1, ARID1B, EN2, NPAS4, SIX4, FOXP3, NRIP1, DLX3, EBF3, ZRANB2, NEUROD1, NEUROD4, HOXB9, PBX2
GOTERM_MF_FAT GO:0008134-transcription factor binding150.001574619YWHAZ, MECP2, PIM1, TLE1, ARID1B, FOXP3, FOXO4, TCF7L2, NRIP1, CITED2, MAX, HAND2, NKX3-1, NEUROD1, SERTAD2
GOTERM_MF_FAT GO:0003700-transcription factor activity220.002276973ESRRA, FOXA2, SOX11, ADNP, RUNX1T1, NR3C1, EN2, SIX4, FOXO4, FOXP3, TCF7L2, HIC1, CITED2, DLX3, MAX, HAND2, ZRANB2, NKX3-1, NEUROD1, HOXB9, PBX2, ZNF423
GOTERM_MF_FATGO:0003677-DNA binding390.006490896FOXA2, ARID4B, LEMD3, RAG2, NR3C1, FOXO4, TCF7L2, HIC1, CITED2, MAX, HAND2, ZNF697, NKX3-1, H2AFX, HIST1H4D, ZNF423, DNMT3A, ESCO1, ESRRA, ZNF280B, CHTF8, SOX11, MECP2, RUNX1T1, ADNP, ARID1B, SIX4, EN2, NPAS4, FOXP3, DLX3, EBF3, ZRANB2, DDB2, NEUROD1, NEUROD4, HOXB9, PBX2, ZNF436
KEGG_PATHWAYhsa05200:pathways in cancer90.025852862MAX, RASSF1, SLC2A1, RUNX1T1, NKX3-1, RALA, CRK, TCF7L2, TGFB2

[i] The GO terms listed in this Table are the top 5 ones with a lower p-value in BP and MF, as well as the significant ones in CC, respectively. GO, gene ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; BP, biological process; CC, cellular component; MF, molecular function.

Table III

The enriched GO and pathway terms for target genes of hsa-mir-618.

Table III

The enriched GO and pathway terms for target genes of hsa-mir-618.

CategoryTermCountP-valueTarget genes
GOTERM_BP_FATGO:0000122-negative regulation of transcription from RNA polymerase II promoter79.04E-06E2F1, ID2, KLF12, MECP2, SMAD3, POU4F2, NCOR1
GOTERM_BP_FATGO:0045892-negative regulation of transcription, DNA-dependent74.69E-05E2F1, ID2, KLF12, MECP2, SMAD3, POU4F2, NCOR1
GOTERM_BP_FATGO:0051253-negative regulation of RNA metabolic process75.15E-05E2F1, ID2, KLF12, MECP2, SMAD3, POU4F2, NCOR1
GOTERM_BP_FATGO:0016481-negative regulation of transcription71.91E-04E2F1, ID2, KLF12, MECP2, SMAD3, POU4F2, NCOR1
GOTERM_BP_FAT GO:0006355-regulation of transcription, DNA-dependent122.22E-04E2F1, ATRX, ID2, KLF12, KLF9, REL, MECP2, SMAD3, POU4F2, NFYB, NCOR1, RAB1A
GOTERM_CC_FAT GO:0044427-chromosomal part50.002702275ATRX, ID2, PPP2CB, MECP2, RCC1
GOTERM_CC_FAT GO:0000785-chromatin40.00353511ATRX, ID2, MECP2, RCC1
GOTERM_CC_FAT GO:0005694-chromosome50.00507017ATRX, ID2, PPP2CB, MECP2, RCC1
GOTERM_CC_FAT GO:0043228-non-membrane-bounded organelle100.01099599ATRX, ID2, LRP12, PPP2CB, MECP2, WIPF1, RCC1, NCOR1, NOVA1, MARK1
GOTERM_CC_FAT GO:0043232-intracellular non-membrane-bounded organelle100.01099599ATRX, ID2, LRP12, PPP2CB, MECP2, WIPF1, RCC1, NCOR1, NOVA1, MARK1
GOTERM_MF_FAT GO:0016564-transcription repressor activity66.08E-04E2F1, ID2, KLF12, MECP2, SMAD3, NCOR1
GOTERM_MF_FAT GO:0003700-transcription factor activity90.001015664E2F1, KLF12, KLF9, REL, SALL1, SMAD3, POU4F2, NFYB, NCOR1
GOTERM_MF_FAT GO:0030528-transcription regulator activity110.001104363E2F1, ID2, KLF12, KLF9, REL, SALL1, MECP2, SMAD3, POU4F2, NFYB, NCOR1
GOTERM_MF_FAT GO:0003714-transcription corepressor activity40.004030547E2F1, KLF12, MECP2, NCOR1
GOTERM_MF_FAT GO:0008134-transcription factor binding60.005109515E2F1, KLF12, MECP2, SMAD3, NCOR1, RAB1A
KEGG_PATHWAYhsa04350:TGF-beta signaling pathway30.005747228ID2, PPP2CB, SMAD3

[i] The GO terms listed in this Table are the top 5 ones with a lower p-value in BP, CC and MF, respectively. GO, gene ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; BP, biological process; CC, cellular component; MF, molecular function.

Furthermore, to further reveal the functions of 97 genes in the PPI interaction network, functional clustering and pathway enrichment analysis were performed. According to the functional clustering, 19 clusters were enriched. A set of genes (e.g. E2F1, NR3C1, IGF1, SMAD3 and BDNF) were significantly enriched in multiple functional clusters, such as cell death and signal transduction (Fig. 3). Additionally, 26 genes in the network were markedly enriched in 9 pathway clusters, such as pathways in cancer (e.g. E2F1, MAX and SMAD3), cell cycle (e.g. E2F1 and SMAD3) and the Wnt signaling pathway (e.g. SMAD3) (Table IV).

Table IV

Enriched pathways of the target genes in the protein-protein interaction network.

Table IV

Enriched pathways of the target genes in the protein-protein interaction network.

ClusterPathwayCountP-valueGenes
1KEGG_Pathways in cancer113.74E-06CRK, E2F1, IGF1, MAX, RALA, RASSF1, RUNX1T1, SLC2A1, SMAD3, TCF7L2, TGFB2
KEGG_Chronic myeloid leukemia40.003836CRK, E2F1, SMAD3, TGFB2
KEGG_Renal cell carcinoma30.023754CRK, SLC2A1, TGFB2
2KEGG_Cell cycle50.000997E2F1, SMAD3, STAG1, TGFB2, YWHAZ
BIOCARTA_G1 pathway30.001847E2F1, SMAD3, TGFB2
KEGG_Pancreatic cancer40.003295E2F1, RALA, SMAD3, TGFB2
KEGG_Colorectal cancer30.017246SMAD3, TCF7L2, TGFB2
KEGG_TGFβ signaling pathway30.039070ID2, SMAD3, TGFB2
3KEGG_Acute myeloid leukemia30.013764PIM1, RUNX1T1, TCF7L2
4BIOCARTA_PPARA pathway30.014425CITED2, NCOR1, NRIP1
5KEGG_Wnt signaling pathway50.005182LRP6, PLCB1, SFRP1, SMAD3, TCF7L2
Wnt_Signaling30.043830LRP6, SFRP1, TLE1
6REACTOME_Clathrin derived vesicle budding30.016514CLINT1, CLTA, SNAP23
REACTOME_Membrane trafficking30.031420CLINT1, CLTA, SNAP23
7ST_Integrin signaling pathway30.031420CRK, ITGA5, RALA
8KEGG_Hypertrophic cardiomyopathy HCM30.036795IGF1, ITGA5, TGFB2
KEGG_Dilated cardiomyopathy30.045063IGF1, ITGA5, TGFB2
9KEGG_Prostate cancer30.043830E2F1, IGF1, TCF7L2

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes.

Discussion

HNSCC is a common type of non-skin cancer worldwide (1). In the present study, using the miRNA expression profile and clinical data of 397 cases in the TCGA database, miR-1251, miR-618 and miR-328 were screened out to be significantly associated with risk factors for HNSCC, based on the survival analysis and Cox regression analysis. A set of target genes of these three miRNAs were significantly enriched in a series of GO functions and pathways. For example, targets of miR-618, E2F1 and SMAD3, were significantly enriched in functions concerning regulation of transcription, and SMAD3 was also markedly enriched in the TGF-β signaling pathway. These two genes interacted with each other in the PPI network.

E2F1 encodes an E2F transcription factor, which mediates cell cycle and functions as a tumor-suppressor protein (23). Overexpression of E2F1 in vitro and in vivo suppresses HNSCC cell growth through induction of apoptosis (24). Additionally, single nucleotide polymorphisms (SNPs) of E2F1 are highly correlated with the risk of HNSCC (25). Moreover, as a transcriptional modulator and signal transducer, SMAD3 mediates multiple signaling pathways and plays a role in the regulation of carcinogenesis (26). In the present study, SMAD3 was predicted to interact with E2F1, which has been reported in a previous study (27). In HNSCC, SMAD3 is inactivated, which is associated with higher overall survival of patients (28). In the present study, SMAD3 was markedly enriched in the TGF-β signaling pathway. The association of SMAD3 and TGF-β signaling has been reported in numerous studies (2931). A previous study reported that overexpression of death-associated protein kinase-related apoptosis-inducing kinase 1 (DRAK1) binds to Smad3, and then inhibits TGF-β1 tumor suppressor activity in HNSCC (32). Additionally, SNPs in miR-618 are implicated in cancer risk (33). miR-618 has been discovered to be progressively expressed in esophageal adenocarcinoma (34). There is no evidence that miR-618 is associated with HNSCC risk or survival rate. Taken together, miR-618 may play a crucial role in the prognosis of HNSCC by regulating the genes E2F1 and SMAD3.

Furthermore, in the PPI network, the target gene of miR-328, MAX, interacted with multiple genes, such as E2F1. MAX was significantly enriched in the function of transcription activator activity and the pathways in cancer. MYC associated factor X (MAX) encodes a transcription factor belonging to the helix-loop-helix leucine zipper (bHLHZ) family (35). It is able to form a homodimer and heterodimer with MYC, which is an oncoprotein correlated with cell proliferation, differentiation and apoptosis (35). There is no evidence to prove the association of MAX with HNSCC. However, overexpression of c-myc has been reported to be associated with the poor prognosis of HNSCC (36). Furthermore, c-myc promotes elevated levels of glutaminolysis and glutamic acid, which marker aggressive tumorigenesis in HNSCC (37). Additionally, miR-328 has been confirmed to be highly expressed in salinomycin-treated HNSCC cells (38). Additionally, the expression level of miR-328 was found to be significantly higher in early-stage non-small cell lung cancer (NSCLC), and shows good diagnostic accuracy in NSCLC patients (39). These results indicate an important role of miR-328 in cancer. Therefore, miR-328 may play a pivotal role in the progression of HNSCC via targeting MAX, and it may be associated with the survival time of HNSCC patients.

In addition, the target gene of miR-1251, IGF1, also interacted with E2F1 in the PPI network, and it was enriched in multiple function clusters, such as cell death and signal transduction. Insulin-like growth factor I (IGF1) is involved in regulating cell growth and development (40), and the activation of multiple signaling pathways in head and neck cancer (41). Previous studies have demonstrated that IGF1 is able to stimulate the expression of vascular endothelial growth factor, which is correlated with increased progression and poor prognosis of HNSCC (42,43). Moreover, miR-1251 has been discovered to be downregulated in nasopharyngeal carcinoma (44). There is no study to report the association of miR-1251 with HNSCC. Therefore, miR-1251 may play a crucial role in the progression of HNSCC via mediating the expression of IGF1, and it may be closely associated with the survival of HNSCC patients.

Despite the aforementioned results, there were several limitations in the present study. The predicted results should be confirmed by laboratory data. In our further studies, the expression levels of miR-1251, miR-618 and miR-328, as well as the regulatory relationships of them and their target genes, will be validated in a large scale of patients with HNSCC.

In conclusion, miR-1251, miR-618 and miR-328 were predicted to be associated with the survival time of HNSCC patients. They may play pivotal roles in the progression of HNSCC via a set of genes associated with transcriptional regulation or signal transduction, such as MAX, E2F1, SMAD3 and IGF1. The three miRNAs were considered as novel prognostic markers in HNSCC. These findings contribute to our further experimental study, and may provide new information for the estimation of the prognosis of HNSCC.

Acknowledgments

The present study was supported by the Natural Science Foundation of Liaoning Province (no. 201202287).

References

1 

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

2 

Krol J, Loedige I and Filipowicz W: The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet. 11:597–610. 2010.PubMed/NCBI

3 

Farazi TA, Hoell JI, Morozov P and Tuschl T: MicroRNAs in Human Cancer. MicroRNA Cancer Regulation, Advanced Concepts, Bioinformatics and Systems Biology Tools. Schmitz U, Wolkenhauer O and Vera J: Springer; pp. 1–20. 2013

4 

Kinoshita T, Hanazawa T, Nohata N, Kikkawa N, Enokida H, Yoshino H, Yamasaki T, Hidaka H, Nakagawa M, Okamoto Y, et al: Tumor suppressive microRNA-218 inhibits cancer cell migration and invasion through targeting laminin-332 in head and neck squamous cell carcinoma. Oncotarget. 3:1386–1400. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Jimenez L, Sharma VP, Lim J, Angeletti R, Condeelis J, Harris T, Prystowsky MB, Childs G and Segall JE: MicroRNA-375 impairs head and neck squamous cell carcinoma invasion by suppressing invadopodia activity. Cancer Res. 74(1452): 2014 View Article : Google Scholar

6 

Jimenez L, Harris T, Kawachi N, Belbin T, Schlecht N, Lim J, Angeletti R, Prystowsky MB, Childs G and Segall J: MicroRNA-375 regulates tumor invasion and metastasis phenotypes and is associated with poor outcome in head and neck squamous cell carcinoma. Cancer Res. 73(4939): 2013 View Article : Google Scholar

7 

Langevin SM, Stone RA, Bunker CH, Lyons-Weiler MA, LaFramboise WA, Kelly L, Seethala RR, Grandis JR, Sobol RW and Taioli E: MicroRNA-137 promoter methylation is associated with poorer overall survival in patients with squamous cell carcinoma of the head and neck. Cancer. 117:1454–1462. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Howard JD, Cheng H, Ratner E, Fertig EJ, Perez J, Quon H, Considine M, Ochs M, Weidhaas J and Chung CH: MicroRNA profiling reveals miR-205 upregulation is associated with head and neck squamous cell carcinoma and modulates E2F1 signaling. Cancer Res. 73(Suppl 8): 3100. 2013. View Article : Google Scholar

9 

Therneau TM: Modeling survival data. Extending the Cox Model. Springer; 2000, View Article : Google Scholar

10 

Timsit JF, Alberti C and Chevret S: Cox proportional hazards regression analysis. Rev Mal Respir. 22:1058–1064. 2005.In French. View Article : Google Scholar : PubMed/NCBI

11 

Ferguson N, Datta S and Brock G: msSurv, an R package for nonparametric estimation of multistate models. J Stat Software. 50:1–24. 2012. View Article : Google Scholar

12 

Piriyapongsa J, Bootchai C, Ngamphiw C and Tongsima S: microPIR: An integrated database of microRNA target sites within human promoter sequences. PLoS One. 7:e338882012. View Article : Google Scholar : PubMed/NCBI

13 

Dweep H, Sticht C, Pandey P and Gretz N: miRWalk - database: Prediction of possible miRNA binding sites by 'walking' the genes of three genomes. J Biomed Inform. 44:839–847. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Enright AJ, John B, Gaul U, Tuschl T, Sander C and Marks DS: MicroRNA targets in Drosophila. Genome Biol. 5:R1. 2003. View Article : Google Scholar

15 

Wang X and El Naqa IM: Prediction of both conserved and nonconserved microRNA targets in animals. Bioinformatics. 24:325–332. 2008. View Article : Google Scholar

16 

Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M, et al: Combinatorial microRNA target predictions. Nat Genet. 37:495–500. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Thomas M, Lieberman J and Lal A: Desperately seeking microRNA targets. Nat Struct Mol Biol. 17:1169–1174. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP and Burge CB: Prediction of mammalian microRNA targets. Cell. 115:787–798. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, Lin J, Minguez P, Bork P, von Mering C, et al: STRING v9.1: Protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41:D808–D815. 2013. View Article : Google Scholar :

20 

Smoot ME, Ono K, Ruscheinski J, Wang PL and Ideker T: Cytoscape 2.8: New features for data integration and network visualization. Bioinformatics. 27:431–432. 2011. View Article : Google Scholar :

21 

Huang 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

22 

Huang ZX, Tian HY, Hu ZF, Zhou YB, Zhao J and Yao KT: GenCLiP: A software program for clustering gene lists by literature profiling and constructing gene co-occurrence networks related to custom keywords. BMC Bioinformatics. 9:3082008. View Article : Google Scholar : PubMed/NCBI

23 

Ting JH: Novel roles of cell cycle regulator E2F1 in the CNS: Implications for synaptic damage in HIV-associated neurocognitive disorders. University of Pennsylvania; Scholarly Commons: Publicly Accessible Penn Dissertations. Paper 1471. 2014, http://repository.upenn.edu/edissertations/1471.

24 

Liu TJ, Wang M, Breau RL, Henderson Y, El-Naggar AK, Steck KD, Sicard MW and Clayman GL: Apoptosis induction by E2F-1 via adenoviral-mediated gene transfer results in growth suppression of head and neck squamous cell carcinoma cell lines. Cancer Gene Ther. 6:163–171. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Lu M, Liu Z, Yu H, Wang LE, Li G, Sturgis EM, Johnson DG and Wei Q: Combined effects of E2F1 and E2F2 polymorphisms on risk and early onset of squamous cell carcinoma of the head and neck. Mol Carcinog. 51(Suppl 1): E132–E141. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Murata M, Yoshida K and Matsuzaki K: Early chronic inflammation and subsequent somatic mutations shift phospho-Smad3 signaling from tumor-suppression to fibro-carcinogenesis in human chronic liver diseases. Hepatocellular Carcinoma-Future Outlook. Kaseb A: InTech; 2013, View Article : Google Scholar : Available from: http://www.intechopen.com/books/hepatocellular-carcinoma-future-outlook/early-chronic-inflammation-and-subsequent-somatic-mutations-shift-phospho-smad3-signaling-from-tumor.

27 

Kowalik TF: Smad about E2F. TGFbeta repression of c-Myc via a Smad3/E2F/p107 complex. Mol Cell. 10:7–8. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Xie W, Aisner S, Baredes S, Sreepada G, Shah R and Reiss M: Alterations of Smad expression and activation in defining 2 subtypes of human head and neck squamous cell carcinoma. Head Neck. 35:76–85. 2013. View Article : Google Scholar

29 

Derynck R and Zhang YE: Smad-dependent and Smad-independent pathways in TGF-β family signalling. Nature. 425:577–584. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Massagué J and Wotton D: Transcriptional control by the TGF-β/Smad signaling system. EMBO J. 19:1745–1754. 2000. View Article : Google Scholar

31 

Heldin CH, Miyazono K and ten Dijke P: TGF-β signalling from cell membrane to nucleus through SMAD proteins. Nature. 390:465–471. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Park Y, Kim W, Lee J, Park J, Cho JK, Pang K, Lee J, Kim D, Park SW, Yang KM, et al: Cytoplasmic DRAK1 overexpressed in head and neck cancers inhibits TGF-β1 tumor suppressor activity by binding to Smad3 to interrupt its complex formation with Smad4. Oncogene. 34:5037–5045. 2015. View Article : Google Scholar

33 

Pelletier C and Weidhaas JB: MicroRNA binding site polymorphisms as biomarkers of cancer risk. Expert Rev Mol Diagn. 10:817–829. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Wu X, Ajani JA, Gu J, Chang DW, Tan W, Hildebrandt MA, Huang M, Wang KK and Hawk E: MicroRNA expression signatures during malignant progression from Barrett's esophagus to esophageal adenocarcinoma. Cancer Prev Res. 6:196–205. 2013. View Article : Google Scholar

35 

Blackwood EM and Eisenman RN: Max: A helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc. Science. 251:1211–1217. 1991. View Article : Google Scholar : PubMed/NCBI

36 

Field JK, Spandidos DA, Stell PM, Vaughan ED, Evan GI and Moore JP: Elevated expression of the c-myc oncoprotein correlates with poor prognosis in head and neck squamous cell carcinoma. Oncogene. 4:1463–1468. 1989.PubMed/NCBI

37 

Banda AD, Cortes MB, Kamarajan P, Rajendiran T, Chinnaiyan AM and Kapila YL: Glutamic acid and glutaminolysis mark aggressive tumorigenesis in head and neck squamous cell carcinoma. Cancer Res. 75(Suppl 15): 11922015. View Article : Google Scholar

38 

Kuo SZ, Blair KJ, Rahimy E, Kiang A, Abhold E, Fan JB, Wang-Rodriguez J, Altuna X and Ongkeko WM: Salinomycin induces cell death and differentiation in head and neck squamous cell carcinoma stem cells despite activation of epithelial-mesenchymal transition and Akt. BMC Cancer. 12:5562012. View Article : Google Scholar : PubMed/NCBI

39 

Ulivi P, Foschi G, Mengozzi M, Scarpi E, Silvestrini R, Amadori D and Zoli W: Peripheral blood miR-328 expression as a potential biomarker for the early diagnosis of NSCLC. Int J Mol Sci. 14:10332–10342. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Woods KA, Camacho-Hübner C, Savage MO and Clark AJ: Intrauterine growth retardation and postnatal growth failure associated with deletion of the insulin-like growth factor I gene. N Engl J Med. 335:1363–1367. 1996. View Article : Google Scholar : PubMed/NCBI

41 

Rosenzweig SA and Holmes CO: Insulin-like growth factor-1 receptors in head and neck cancer. Molecular Determinants of Head and Neck Cancer. Springer. 113–130. 2014. View Article : Google Scholar

42 

Slomiany MG, Black LA, Kibbey MM, Day TA and Rosenzweig SA: IGF-1 induced vascular endothelial growth factor secretion in head and neck squamous cell carcinoma. Biochem Biophys Res Commun. 342:851–858. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Jameson MJ, Beckler AD, Taniguchi LE, Allak A, Vanwagner LB, Lee NG, Thomsen WC, Hubbard MA and Thomas CY: Activation of the insulin-like growth factor-1 receptor induces resistance to epidermal growth factor receptor antagonism in head and neck squamous carcinoma cells. Mol Cancer Ther. 10:2124–2134. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Plieskatt JL, Rinaldi G, Feng Y, Levine PH, Easley S, Martinez E, Hashmi S, Sadeghi N, Brindley PJ, Bethony JM, et al: Methods and matrices: Approaches to identifying miRNAs for nasopharyngeal carcinoma. J Transl Med. 12:32014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 35 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hui L, Wu H, Yang N, Guo X and Jang X: Identification of prognostic microRNA candidates for head and neck squamous cell carcinoma. Oncol Rep 35: 3321-3330, 2016
APA
Hui, L., Wu, H., Yang, N., Guo, X., & Jang, X. (2016). Identification of prognostic microRNA candidates for head and neck squamous cell carcinoma. Oncology Reports, 35, 3321-3330. https://doi.org/10.3892/or.2016.4698
MLA
Hui, L., Wu, H., Yang, N., Guo, X., Jang, X."Identification of prognostic microRNA candidates for head and neck squamous cell carcinoma". Oncology Reports 35.6 (2016): 3321-3330.
Chicago
Hui, L., Wu, H., Yang, N., Guo, X., Jang, X."Identification of prognostic microRNA candidates for head and neck squamous cell carcinoma". Oncology Reports 35, no. 6 (2016): 3321-3330. https://doi.org/10.3892/or.2016.4698