Open Access

Potential five‑mRNA signature model for the prediction of prognosis in patients with papillary thyroid carcinoma

  • Authors:
    • Lin‑Kun Zhong
    • Xiao‑Xiong Gan
    • Xing‑Yan Deng
    • Fei Shen
    • Jian‑Hua Feng
    • Wen‑Song Cai
    • Qiong‑Yao Liu
    • Jian‑Hang Miao
    • Bing‑Xing Zheng
    • Bo Xu
  • View Affiliations

  • Published online on: June 26, 2020     https://doi.org/10.3892/ol.2020.11781
  • Pages: 2302-2310
  • 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

Although the mortality rate of papillary thyroid carcinoma (PTC) is relatively low, the recurrence rates of PTC remain high. The high recurrence rates are related to the difficulties in treatment. Gene expression profiles has provided novel insights into potential therapeutic targets and molecular biomarkers of PTC. The aim of the present study was to identify mRNA signatures which may categorize PTCs into high‑and low‑risk subgroups and aid with the predictions for prognoses. The mRNA expression profiles of PTC and normal thyroid tissue samples were obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed mRNAs were identified using the ‘EdgeR’ software package. Gene signatures associated with the overall survival of PTC were selected, and enrichment analysis was performed to explore the biological pathways and functions of the prognostic mRNAs using the Database for Visualization, Annotation and Integration Discovery. A signature model was established to investigate a specific and robust risk stratification for PTC. A total of 1,085 differentially expressed mRNAs were identified between the PTC and normal thyroid tissue samples. Among them, 361 mRNAs were associated with overall survival (P<0.05). A 5‑mRNA prognostic signature for PTC (ADRA1B, RIPPLY3, PCOLCE, TEKT1 and SALL3) was identified to classify the patients into high‑and low‑risk subgroups. These prognostic mRNAs were enriched in Gene Ontology terms such as ‘calcium ion binding’, ‘enzyme inhibitor activity’, ‘carbohydrate binding’, ‘transcriptional activator activity’, ‘RNA polymerase II core promoter proximal region sequence‑specific binding’ and ‘glutathione transferase activity’, and Kyoto Encyclopedia of Genes and Genomes signaling pathways such as ‘pertussis’, ‘ascorbate and aldarate metabolism’, ‘systemic lupus erythematosus’, ‘drug metabolism‑cytochrome P450 and ‘complement and coagulation cascades’. The 5‑mRNA signature model may be useful during consultations with patients with PTC to improve the prediction of their prognosis. In addition, the prognostic signature identified in the present study may reveal novel therapeutic targets for patients with PTC.

Introduction

Thyroid cancer (TC) is the most common endocrine malignancy worldwide; the incidence of TC has increased rapidly over the past decade, with an estimated 45,000 new cases diagnosed per year in the United States (1,2). Papillary thyroid carcinoma (PTC) is the most frequent form of TC, accounting for 80–90% of all thyroid malignancies (3). PTC variants include conventional, follicular, oncocytic, solid, tall cell, columnar cell, diffuse sclerosing and cribriform forms (4,5). Among these variants, conventional PTC is the main histological variant (6). PTC is a well-differentiated papillary carcinoma with a relatively low mortality rate compared with other types of TC, and the 5-year overall survival rate is >95% (7). However, the recurrence or persistence rate of PTC is relatively high (8,9). Potential tumor biomarkers for TC diagnosis and prognosis may serve a significant role in the development of new therapies for TC. Identification of low-risk subsets of patients with PTC may lead to the development of treatments to reduce morbidity. Additionally, identification of patients with PTC that are at a high risk of a poor prognosis may allow for more accurate pre/postoperative assessments and improve the surgical planning based on to the results of the prognostic biomarker tests. Therefore, novel biomarkers or prognostic models are urgently needed to devise new means for the prediction of survival of patients with PTC.

With the advances of genome sequencing technologies in recent years, accumulating evidence have demonstrated that molecular biomarkers, such as protein-coding genes and non-coding RNAs, are informative for cancer detection and classification. mRNAs have exhibited a great potential in both participating in the physiological and pathological processes as well as predicting prognosis of patients with various types of tumor, such as renal cell carcinoma and hepatic carcinoma (10,11). Thus, the dysregulated expression or mutation of mRNAs may be a promising predictor of poor prognosis in PTC. Previous studies have documented that numerous key mRNAs, such as checkpoint kinase 2 (CHEK2) and distal-less homeobox 6 (DLX6), are closely associated with the aggressive pathogenesis of PTC (1214). It is likely that multiple mRNAs may allow for the development of a signature model and provide more statistically predictive results of PTCs patients. Thus, a prospective clinical trial using a large cohort is required to investigate specific prognostic classifiers in patients with PTC.

The Cancer Genome Atlas (TCGA) database has publicized various mRNA sequence data, which may provide novel information to improve the understanding of the molecular mechanisms of action in the tumorigenesis and progression of PTC. The current study aimed to apply advanced RNA sequencing analysis to identify differentially expressed mRNAs (DEMs) between PTC samples and normal samples. In additional, the association between prognostic information and expression of these DEMs were evaluated. Functional enrichment analyses were also performed to investigate the mechanisms of action. Furthermore, the application value and reliability of the 5-mRNA signature model to predict PTC prognosis were investigated.

Materials and methods

Data collection and pre-processing

mRNA expression information and the corresponding clinical data of 551 samples, including 58 normal and 493 PTC samples, were obtained from TCGA database using the keyword ‘thyroid cancer’ (https://tcga-data.nci.nih.gov) (15) prior to July 15th, 2019. As the clinical data and mRNA expression information were downloaded from TCGA database, the current study did not require ethical approval. The mRNAs that were differentially expressed between normal and PTC samples were assessed using R Studio software (RStudio version 1.1.463; http://www.r-project.org) (16) with the ‘limma’ package (http://www.bioconductor.org/packages). mRNAs that satisfied the criteria |log2 fold change (FC)|>2 and P<0.05 were considered for subsequent analysis (17).

Survival analysis

The Kaplan-Meier analysis and log-rank test were applied to identify the prognostic DEMs. The ‘survival’ package in the R software (http://www.bioconductor.org/packages) was used to construct the survival curves. The survival endpoint was defined as the overall survival time (OS). P<0.05 was considered to indicate a statistically significant difference. In addition, the median expression value was used as the classification cut-off value for high and low expression.

Risk stratification and receiver operating characteristic (ROC) curves

To further investigate the crucial DEMs closely associated to OS, univariate Cox models were performed. Subsequently, multivariate COX regression analysis of selected candidate DEMs was performed. DEMs with P<0.05 were defined as candidate genes for further multivariate Cox regression analysis. Owing to the data processing above, 5 key DEMs were eventually subsumed into the multivariate Cox regression model to calculate their respective coefficients. Then, the score model composed of DEMs' respective coefficients and expression value was constructed as follows:

Survival Risk Score=∑i=1k(C1×Vi)

Where k is the number of prognostic RNAs, Ci represents the coefficient of the RNA in the multivariate Cox regression analysis, and Vi is the expression value of the RNA. The RNAs with Ci >0 were defined as high-risk markers, whereas those with Ci <0 were defined as protective RNAs (18). The median risk score was used as the classification cut-off value between high- and low-risk PTC subgroups in 5-mRNA signature model as previously described (19). Between the high- and low-risk PTC groups, the difference in OS was compared and analyzed using Kaplan-Meier analysis with the log-rank test. The specificity and sensitivity of the 5-mRNA prognostic signature model in the present study were assessed using an ROC curve and area under the ROC curve (AUC). The ‘timeROC’ package (https://cran.r-project.org/web/packages/timeROC/) was used to conduct all the analyses.

Prediction of target genes and functional enrichment analysis

Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene ontology (GO) were used to perform functional enrichment analyses of DEMs and to elucidate the mechanisms of action in PTC tumorigenesis using The Database for Annotation, Visualization and Integrated Discovery (https://david.ncifcrf.gov/) (20).

Results

DEMs in PTCs

The mRNA expression profiles between PTC and normal samples from TCGA database were downloaded and analyzed. A total of 853 upregulated and 232 downregulated DEMs that met the criteria |log2FC|>2 and P<0.05 were identified in PTC compared with normal samples. The volcano plot and heatmap of the DEMs in PTC are presented in Fig. 1. The top 10 upregulated and downregulated DEMs were selected for further analysis based on the false discovery rate (Table I).

Table I.

Top 10 upregulated and downregulated differentially expressed mRNAs.

Table I.

Top 10 upregulated and downregulated differentially expressed mRNAs.

A, Upregulated mRNAs

Gene symbolLog2FCFDRP-value
ARHGAP368.670156159 1.72×10−34 5.05×10−36
SFTPA18.648973385 1.87×10−21 1.67×10−22
WIF17.858016530 8.73×10−13 1.70×10−13
SERPINB117.516327014 6.02×10−08 1.90×10−08
SLC18A37.315944162 8.45×10−19 9.47×10−20
SFTPA27.309149505 6.95×10−22 6.00×10−23
GABRB27.144823578 1.53×10−71 1.04×10−74
TMPRSS67.123312367 2.06×10−41 3.30×10−43
SLC22A317.123263970 9.85×10−57 3.97×10−59
DPRX7.030322665 2.35×10−07 7.92×10−08

B, Downregulated mRNAs

Gene symbolLog2FCFDR P-values

OR2V1−4.212774737 6.32×10−43 8.82×10−45
KCNA1−4.091346010 3.91×10−51 2.59×10−53
SLC6A15−3.985283867 1.61×10−48 1.33×10−50
GRIA1−3.470505834 9.76×10−57 3.87×10−59
VSTM2A−3.439852831 1.18×10−32 3.94×10−34
UGT2B11−3.421202686 1.48×10−78 6.70×10−82
RELN−3.414498465 2.42×10−64 5.35×10−67
MYOC−3.390338222 1.13×10−31 4.03×10−33
DPT−3.282655507 1.28×10−44 1.51×10−46
SEC14L3−3.276717193 1.48×10−34 4.33×10−36

[i] FC, fold-change; FDR, false discovery rate.

Survival analysis

Kaplan-Meier univariate survival analyses were performed in the present study to investigate gene expression profiles on OS. Among the 1,085 DEMs examined in the present study, 361 mRNAs, including adhesion G protein-coupled receptor D1(ADGRD1), ankyrin 2 (ANK2), anoctamin 5 (ANO5), activity regulated cytoskeleton associated protein (ARC), acid sensing ion channel subunit 1 (ASIC1), checkpoint kinase 2 (CHEK2), cytochrome c oxidase subunit 7A1 (COX7A1), casein beta (CSN2), DDB1 and CUL4 associated factor 8 like 1 (DCAF8L1), distal-less homeobox 6 (DLX6), dipeptidyl peptidase like 6 (DPP6), desmocollin 3 (DSC3), elastase, neutrophil expressed (ELANE), erythroferrone (ERFE), coagulation factor X (F10), ficolin 3 (FCN3), FEZ family zinc finger 1 (FEZF1), surfactant protein A1 (SFTPA1), surfactant protein A2 (SFTPA2) and solute carrier family 22 member 31 (SLC22A31), were associated OS in patients with PTC. The top 20 mRNAs based on false discovery rate (FDR) value are presented in Fig. S1.

Establishment of a five-mRNA signature model

Uni- and multivariate analyses were performed using Cox regression analysis to assess the association between OS and the expression levels of the identified DEMs in patients with PTC (Table SI; Table II). A predictive signature model was established based on five DEMs, namely ADRA1B, RIPPLY3, PCOLCE, TEKT1 and SALL3, that were selected from the multivariate Cox regression analysis. The signature model in the current study was defined as follows: Survival risk score=(−0.771870966 × VADRA1B) + (−0.782948964 × VRIPPLY3) + (0.708200312 × VPCOLCE2) + (0.241743456 × VTEKT1) + (0.378467099 × VSALL3), and the V was the expression value (Table II).

Table II.

Multivariate cox regression analysis between OS and the expression levels of DEMs.

Table II.

Multivariate cox regression analysis between OS and the expression levels of DEMs.

Gene symbolCoefP-value
ADRA1B−0.7718709662.18×10-3
RIPPLY3−0.7829489649.03×10-4
TEKT10.2417434569.43×10-2
PCOLCE20.7082003129.17×10-5
SALL30.3784670991.67×10-4

[i] Coef, regression coefficient; OS, overall survival; DEMs, differentially expressed mRNAs; ADRA1B, adrenoceptor α-1B; RIPPLY3, ripply transcriptional repressor 3; TEKT1, tektin 1; PCOLCE2, procollagen C-endopeptidase enhancer 2; SALL3, spalt like transcription factor 3.

Risk stratification and ROC curve analysis

The risk scores of 493 patients with PTC from TCGA database were calculated using the 5-mRNA prediction model. All patients with PTC were assigned to the low-risk (risk score >1; n=264) or the high-risk group (risk score ≤1; n=229; Fig. 2). The prognostic power of the 5-mRNA signature model was evaluated using the AUC value of the ROC curve (Fig. 3). The 3- and 5-year AUC values in the present study were 0.937 and 0.925, respectively, which demonstrated a high sensitivity and specificity of the 5-mRNA signature model for predicting the risk levels in patients with PTC. This signature model may be used to identify patients with high risk scores who may benefit from more radical, effective and individualized therapy.

Functional enrichment analysis

The GO analysis results demonstrated that prognostic mRNAs were mainly associated with ‘calcium ion binding’, ‘enzyme inhibitor activity’, ‘carbohydrate binding’, ‘transcriptional activator activity’, ‘RNA polymerase II core promoter proximal region sequence-specific binding’ and ‘glutathione transferase activity’ (Table III). Using KEGG analysis, five statistically significant pathways were identified, namely ‘pertussis’, ‘ascorbate and aldarate metabolism’, ‘systemic lupus erythematosus’, ‘drug metabolism-cytochrome P450’ and ‘complement and coagulation cascades’ (Table III). These results suggested that the prognostic mRNA are key players in in many cellular and biochemical processes.

Table III.

Top 10 enriched GO terms, KEGG pathways and target mRNAs.

Table III.

Top 10 enriched GO terms, KEGG pathways and target mRNAs.

A, GO terms

TermIDFDRTarget mRNAs
Calcium ion binding00055094.705418 PCDHGA12/F10/ADGRE3/MYL3/ENPP2/REG4/CRTAC1/DMP1/MMP19/MMP27/CABP7/PCDH12/PCDH8/MMP28/PADI4/DNASE1L3/PCDH19/DSC3/RGN/CALML6/PCDHA12/SGCA/CSN2/AOC3
Enzyme inhibitor activity000485719.02155 SLN/SCG5/UGT1A1/CSN2
Carbohydrate binding003024626.01117 EMCN/CHIA/OLR1/FCN3/SFTPA2/CHODL/SFTPA1/CLEC4D/CLEC4M
Transcriptional activator activity, RNA polymerase II core promoter proximal region sequence-specific binding000107727.08023 TCF21/OTX1/DLX5/ONECUT3/SOX2/TP63/NOBOX/PITX3/GRHL1/FOXD1
Glutathione transferase activity000436427.42761 GSTA1/CLIC3/ALOX5AP/CLIC5
RNA polymerase II core promoter proximal region sequence-specific DNA binding000097848.17595 FEZF1/MUC1/FEZF2/OTX1/DLX5/NKX3-2/KLF17/NOBOX/PITX3/GRHL1/DDN/FOXD1
Interleukin-8 binding001995952.49444A2M/CXCR1
Heparin binding000820158.36638 CCL2/REG4/ELANE/APOH/SERPIND1/GREM2/SOD3
Structural molecule activity000519862.23154 KRT25/DES/KRT74/LAMC3/KRTAP3-2/SYNC/CLDN5/LCE2B/LELP1
Chloride channel activity000525462.51447 CLCA2/GABRR1/CLIC3/CLIC5

B, KEGG pathways

PathwayIDFDRTarget mRNAs

Ascorbate and aldarate metabolismhsa0005313.65207 ALDH2/RGN/UGT2B4/UGT1A1
Systemic lupus erythematosushsa0532232.92029 HIST1H2BO/ELANE/HIST1H2BH/C2/HIST1H2AJ/HIST1H3G/HIST1H4J
Drug metabolism-cytochrome P450hsa0098233.51738 GSTA1/MAOB/UGT2B4/ADH7/UGT1A1
Complement and coagulation cascadeshsa0461034.81476 A2M/F10/C2/SERPIND1/PLAU
Metabolism of xenobiotics by cytochrome P450hsa0098041.43999 GSTA1/SULT2A1/UGT2B4/ADH7/UGT1A1
Chemical carcinogenesishsa0520449.49964 GSTA1/SULT2A1/UGT2B4/ADH7/UGT1A1
Salmonella infectionhsa0513253.48304 PFN2/IL6/WASF1/CXCL2/NOS2
Morphine addictionhsa0503263.62918 ADCY4/GABRR1/PDE1B/PDE2A/GABRP
Alcoholismhsa0503470.92004 HIST1H2BO/HIST1H2BH/MAOB/CALML6/HIST1H2AJ/HIST1H3G/HIST1H4J

[i] GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; FDR, false discovery rate.

Discussion

PTC is a well-differentiated papillary carcinoma with a relatively low mortality rate among types of TC (14). However, the persistence and recurrence rates in patients with PTC are high, ≥30% in certain demographics (8,9). Investigation into prognostic biomarkers for predicting the risk for patients with PTC may help develop clear and effective treatment strategies. mRNAs have been considered as prognostic biomarkers for the diagnosis and prognosis for patients with PTC in recent years (21). For instance, zinc finger E-box binding homeobox 2 has been reported to be highly expressed in ovarian tumor tissues, which is significantly associated with a poor prognosis (22). The expression of Forkhead box Q1 is also negatively associated with patient survival (23). It has been reported that the mRNA PFKFB4 is associated with tumorigenesis in TC, with PFKFB4 overexpression promoting migration and invasion of TC cells (24). In addition, Huang et al (25) have demonstrated that the overexpression of JAZF zinc finger 1 mRNA significantly inhibited proliferation, caused cell cycle arrest at the G0/G1 phase and promoted apoptosis in PTC cells. A previous study has suggested that the upregulation of the cytokine receptor factor-like 1 gene is associated with aggressive clinicopathological features and poor disease-free survival rates in patients with PTC (26). Additionally, Vecchio et al (27) reported that the downregulation of α-L-fucosidase 1 is associated with the increased aggressiveness of TC. A previous study has also identified an association between high expression levels of endo-5′-nucleotidase and the development of metastatic lymph nodes as well as tumor microinvasion in PTC (28). However, various mRNA-chip platforms or small number of patients were the major limitations of the aforementioned studies. PTC is a multifactorial disease, in which numerous dysregulated mRNAs participate in the tumorigenesis and progression of the tumor (29). Therefore, a signature model including multiple mRNAs may provide a more accurate and detailed prediction system for the diagnosis and prognosis of PTC compared with the use of a single mRNA biomarker.

A total of 853 upregulated and 232 downregulated DEMs in PTC compared with normal thyroid tissues were identified in the present study. Among them, 361 DEMs were associated with OS in patients with PTC. A number of DEMs identified in the present study have been previously investigated in TC. For example, CHEK2 mutations predispose patients to TC, familial breast cancer and double primary cancers of the breast and thyroid (30). Variants in the ATM-CHEK2-BRCA1 axis determine genetic predispositions and clinical presentations of PTC (31). The ANK2 gene has been demonstrated to exhibit a decrease in expression levels in novel tumors that were identified in pediatric post-Chernobyl tumor cases (32). A previous study has demonstrated that ANO5 was downregulated in PTC and follicular thyroid cancer when compared to adjacent non-cancerous tissues (33). ANO5 knockdown was also demonstrated to promote thyroid cancer cell invasion and migration in vitro; overexpression of ANO5 inhibited these phenotypes (33). Tenbaum et al (34) detected two Alien-specific mRNAs and two Alien-specific proteins in vivo and in cell lines; one of the two forms represented the CSN2 subunit of the COP9 signalosome.

Information regarding the regulatory role of ADGRD1, ARC, ASIC1, COX7A1, DCAF8L1, DLX6, DPP6, DSC3, ELANE, ERFE, F10, FCN3, FEZF1, SFTPA1, SFTPA2 and SLC22A31 in PTC is currently limited. However, these DEMs have been reported to be associated with the oncogenesis of other types of cancer in previous studies. For example, the ASIC1 gene is upregulated in pancreatic cancer tissues and expressed on the membrane of pancreatic cancer cells (35). Ceder et al (36) conducted a study that reported that high COX7A1 expression was associated with worse outcome in head and neck squamous cell carcinoma. The DLX6, HOXB7, ELF3, EN1, ETV1, IRX4, BARX1, FOXE1, IRX5 and SALL1 genes are potential oncogenic transcription factors in non-small cell lung cancer cells (37). Overexpression of DSC3 reduced cell colony formation and proliferative ability to promote apoptosis and induce G0/G1 cell cycle arrest in colorectal cancer cells (38). In addition, the ERFE gene was demonstrated to be associated with OS in colon cancer using univariate Cox regression analysis (39). Of note, the FEZF1 gene is an independent predictive factor for cervical cancer recurrence and promotes cervical cancer cell migration and proliferation (40). Of note, SLC22A31 mRNA expression levels are associated with OS in right-sided colon cancer, as determined using TCGA data (39).

GO and KEGG analyses of 361 prognostic DEMs was performed in the present study to identify the molecular mechanisms of action in PTC. GO and KEGG analyses results suggested that prognostic DEMs are key players in in numerous cellular and biochemical processes. Several signaling pathways and biological processes identified in the present study have been investigated in previous studies on PTC. The stress-activated MAPK signaling pathway is a common pro-oncogenic signaling pathway and tumor suppressor during the process of tumorigenesis in different malignancies including PTC, colorectal cancer, liver cancer and ovarian cancer (41,42). Of note, Li et al (43) reported that greater heparin binding was achieved by differentially expressed genes in PTC tissues compared with that in normal thyroid tissues. The results of the present study demonstrated that the signaling pathways associated with the complement and coagulation cascades as well as thyroid cancer may also serve important roles in the development of PTC. A previous study revealed that genes involved in a competing endogenous RNA network were significantly enriched during the metabolism of xenobiotics by cytochrome P450 in PTC (44). The aforementioned evidence was partially consistent with the results of the present GO and KEGG analyses.

Differential gene expression analysis results in PTC samples demonstrated that the intrathyroidal iodine metabolism machinery, methylation-induced gene silencing of tumor suppressor genes, upregulation of Glut-1 mRNA and pro-angiogenetic proteins such as VEGF were all associated with the impairment of the expression of BRAF genes (45). In the present study, it was demonstrated that the 5-mRNAs signature model constructed using TCGA data may be valuable prognostic tool to distinguish patients with PTC into low-risk and high-risk subgroups. Different therapeutic strategies may be selected according to the different risk levels based on the 5-mRNAs signature model, which may help improve the survival of patients with PTC. Patients with PTC in the high-risk group may demonstrate poor clinical outcomes, including aggressive tumor behavior, disease recurrence and disease-specific mortality; therefore, treatment strategies such as prophylactic central neck dissection, radioactive iodine therapy and close long-term follow-up may be applied. Wu et al (46) identified DEGs related to the progression-free interval (PFI) and applied lasso Cox regression to establish a prognostic gene signature which could predict the PFI of PTC. Similar multi-mRNA prognostic models have also been constructed in other tumor types. A previous study has demonstrated that a 3-mRNA signature (CLEC3B, C6 and CLCN1) model can successfully predict the survival of patients with oral squamous cell carcinoma (47). A 24-mRNA signature model was also constructed for early-relapse prediction of patients with hepatocellular carcinoma, which may help facilitate disease management for individual patients (48). A metastasis-related 4-mRNA gene signature model has been demonstrated to effectively classify patients with breast cancer into high- and low-risk groups (49). In addition, Cui et al (38) developed a 6-mRNA signature model that may potentially improve the risk stratification of patients with head and neck squamous cell carcinoma (50). Liang et al (39) reported an effective 6-gene prognostic signature model to act as a specific, robust and clinically practical risk stratification model for the survival outcome of patients with cytogenetically normal-acute myeloid leukemia (51). Additionally, a prognostic 9-mRNA gene signature model was constructed based on survival data from patients with glioma according to Chinese Glioma Genome Atlas database (52).

The present study had certain limitations. First, the mRNA expression data used for the signature model were downloaded from a single database (TCGA) rather than from a number of databases. Additional validation with independent external mRNA expression data is needed to investigate the performance of the 5-mRNAs signature model. Second, the 5-mRNA signature model was constructed using bioinformatics data. Further experimental and clinical studies are required to validate the functions and the predictive value of the 5-mRNA signature model.

In conclusion, in the present study, a 5-mRNA model was constructed to predict the prognosis for patients with PTC. mRNAs with significantly altered expression patterns in PTC may act as prognostic biomarkers. The signature model developed in the present study may improve the prediction of PTC survival and progression.

Supplementary Material

Supporting Data

Acknowledgements

The authors would like to thank Professor TianXiong Shi (Department of General Surgery, Zhongshan City People's Hospital Affiliated to Sun Yat-sen University) for technical support.

Funding

This research was supported by Guangzhou Medicine and Healthcare Technology projects (grant nos. 20141A011011, 20151A011007 and 20161A011008).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

LKZ and BX conceived the study. LKZ, XXG and XYD designed the study, drafted and revised the manuscript. FS acquired the data. JHM, JHF, WSC, QYL and BXZ analyzed the data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, et al: SEER Cancer Statistics Review, 1975–2016. Bethesda, MD: National Cancer Institute; based on November 2018 SEER data submission, posted to the SEER web site. April. 2019

3 

Alzahrani AS and Xing M: Impact of Iymph node metastases identified on central neck dissection (CND) on the recurrence of papillary thyroid cancer: Potential role of BRAFV600E mutation in defining CND. Endocr Relat Cancer. 20:13–22. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Carcangiu ML, Zampi G, Pupi A, Castagnoli A and Rosai J: Papillary carcinoma of the thyroid. A clinicopathologic study of 241 cases treated at the University of Florence, Italy. Cancer. 55:805–828. 1985. View Article : Google Scholar : PubMed/NCBI

5 

Albores-Saavedra J and Wu J: The many faces and mimics of papillary thyroid carcinoma. Endocr Pathol. 17:1–18. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Shen X, Liu R and Xing M: A six-genotype genetic prognostic model for papillary thyroid cancer. Endocr Relat Cancer. 24:41–52. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 62:10–29. 2012. View Article : Google Scholar : View Article : Google Scholar : PubMed/NCBI

8 

Omry-Orbach G: Risk stratification in differentiated thyroid cancer: An ongoing process. Rambam Maimonides Med J. 7:e00032016. View Article : Google Scholar

9 

Luster M, Clarke SE, Dietlein M, Lassmann M, Lind P, Oyen WJ, Tennvall J and Bombardieri E; European Association of Nuclear Medicine (EANM), : Guidelines for radioiodine therapy of differentiated thyroid cancer. Eur J Nucl Med Mol Imaging. 35:1941–1959. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Feng G, Ma HM, Huang HB, Li YW, Zhang P, Huang JJ, Cheng L and Li GR: Overexpression of COL5A1 promotes tumor progression and metastasis and correlates with poor survival of patients with clear cell renal cell carcinoma. Cancer Manag Res. 11:1263–1274. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Tschirdewahn S, Panic A, Püllen L, Harke NN, Hadaschik B, Riesz P, Horváth A, Szalontai J, Nyirády P, Baba HA, et al: Circulating and tissue IMP3 levels are correlated with poor survival in renal cell carcinoma. Int J Cancer. 145:531–539. 2019.PubMed/NCBI

12 

Gąsior-Perczak D, Kowalik A, Walczyk A, Siołek M, Gruszczyński K, Pałyga I, Mikina E, Trybek T, Kopczyński J, Mężyk R, et al: Coexisting germline CHEK2 and somatic BRAFV600E mutations in papillary thyroid cancer and their association with clinicopathological features and disease course. Cancers (Basel). 11:17442019. View Article : Google Scholar

13 

Cancer Genome Atlas Research Network, . Integrated genomic characterization of papillary thyroid carcinoma. Cell. 159:676–690. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Zhao Y, Yu T, Chen L, Xie D, Wang F, Fu L, Cheng C, Li Y, Zhu X and Miao G: A germline CHEK2 mutation in a family with papillary thyroid cancer. Thyroid. 30:924–930. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Tomczak K, Czerwińska P and Wiznerowicz M: The Cancer Genome Atlas (TCGA): An immeasurable source of knowledge. Contemp Oncol (Pozn). 19:A68–A77. 2015.PubMed/NCBI

16 

R Core Team: R: A language and environment for statistical computing. Computing. 1:12–21. 2015.

17 

Gong Y, Zou B, Chen J, Ding L, Li P, Chen J, Chen J, Zhang B and Li J: Potential Five-MicroRNA signature model for the prediction of prognosis in patients with Wilms tumor. Med Sci Monit. 25:5435–5444. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Fan Q and Liu B: Identification of a RNA-Seq based 8-long non-coding RNA signature predicting survival in esophageal cancer. Med Sci Monit. 22:5163–5172. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Tian S, Meng G and Zhang W: A six-mRNA prognostic model to predict survival in head and neck squamous cell carcinoma. Cancer Manag Res. 11:131–142. 2018. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Tang J, Kong D, Cui Q, Wang K, Zhang D, Yuan Q, Liao X, Gong Y and Wu G: Bioinformatic analysis and identification of potential prognostic microRNAs and mRNAs in thyroid cancer. PeerJ. 6:e46742018. View Article : Google Scholar : PubMed/NCBI

22 

Prislei S, Martinelli E, Zannoni GF, Petrillo M, Filippetti F, Mariani M, Mozzetti S, Raspaglio G, Scambia G and Ferlini C: Role and prognostic significance of the epithelial-mesenchymal transition factor ZEB2 in ovarian cancer. Oncotarget. 6:18966–18979. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Zhan HX, Xu JW, Wang L, Wu D, Zhang GY and Hu SY: FoxQ1 is a novel molecular target for pancreatic cancer and is associated with poor prognosis. Curr Mol Med. 15:469–477. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Lu H, Chen S, You Z, Xie C, Huang S and Hu X: PFKFB4 negatively regulated the expression of histone acetyltransferase GCN5 to mediate the tumorigenesis of thyroid cancer. Dev Growth Differ. 62:129–138. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Huang L, Cai Y, Luo Y, Xiong D, Hou Z, Lv J, Zeng F, Yang Y and Cheng X: JAZF1 suppresses papillary thyroid carcinoma cell proliferation and facilitates apoptosis via regulating TAK1/NF-κB pathways. Onco Targets Ther. 12:10501–10514. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Yu ST, Zhong Q, Chen RH, Han P, Li SB, Zhang H, Yuan L, Xia TL, Zeng MS and Huang XM: CRLF1 promotes malignant phenotypes of papillary thyroid carcinoma by activating the MAPK/ERK and PI3K/AKT pathways. Cell Death Dis. 9:3712018. View Article : Google Scholar : PubMed/NCBI

27 

Vecchio G, Parascandolo A, Allocca C, Ugolini C, Basolo F, Moracci M, Strazzulli A, Cobucci-Ponzano B, Laukkanen MO, Castellone MD and Tsuchida N: Human a-L-fucosidase-1 attenuates the invasive properties of thyroid cancer. Oncotarget. 8:27075–27092. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Bertoni APS, Bracco PA, de Campos RP, Lutz BS, Assis-Brasil BM, Meyer ELS, Saffi J, Braganhol E, Furlanetto TW and Wink MR: Activity of ecto-5′-nucleotidase (NT5E/CD73) is increased in papillary thyroid carcinoma and its expression is associated with metastatic lymph nodes. Mol Cell Endocrinol. 479:54–60. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Jendrzejewski JP, Liyanarachchi S, Nagy R, Senter L, Wakely PE, Thomas A, Nabhan F, He H, Li W, Sworczak K, et al: Papillary thyroid carcinoma: Association between germline DNA variant markers and clinical parameters. Thyroid. 26:1276–1284. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Siołek M, Cybulski C, Gąsior-Perczak D, Kowalik A, Kozak-Klonowska B, Kowalska A, Chłopek M, Kluźniak W, Wokołorczyk D, Pałyga I, et al: CHEK2 mutations and the risk of papillary thyroid cancer. Int J Cancer. 137:548–552. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Wójcicka A, Czetwertyńska M, Świerniak M, Długosińska J, Maciąg M, Czajka A, Dymecka K, Kubiak A, Kot A, Płoski R, et al: Variants in the ATM-CHEK2-BRCA1 axis determine genetic predisposition and clinical presentation of papillary thyroid carcinoma. Genes Chromosomes Cancer. 53:516–523. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Stein L, Rothschild J, Luce J, Cowell JK, Thomas G, Bogdanova TI, Tronko MD and Hawthorn L: Copy number and gene expression alterations in radiation-induced papillary thyroid carcinoma from chernobyl pediatric patients. Thyroid. 20:475–487. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Chang Z, Cai C, Han D, Gao Y, Li Q, Feng L, Zhang W, Zheng J, Jin J, Zhang H and Wei Q: Anoctamin5 regulates cell migration and invasion in thyroid cancer. Int J Oncol. 51:1311–1319. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Tenbaum SP, Juenemann S, Schlitt T, Bernal J, Renkawitz R, Muñoz A and Baniahmad A: Alien/CSN2 gene expression is regulated by thyroid hormone in rat brain. Dev Biol. 254:149–160. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Zhu S, Zhou HY, Deng SC, Deng SJ, He C, Li X, Chen JY, Jin Y, Hu ZL, Wang F, et al: ASIC1 and ASIC3 contribute to acidity-induced EMT of pancreatic cancer through activating Ca2+/RhoA pathway. Cell Death Dis. 8:e28062017. View Article : Google Scholar : PubMed/NCBI

36 

Ceder R, Haig Y, Merne M, Hansson A, Zheng X, Roberg K, Nees M, Iljin K, Bloor BK, Morgan PR, et al: Differentiation-promoting culture of competent and noncompetent keratinocytes identifies biomarkers for head and neck cancer. Am J Pathol. 180:457–472. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Zhang DL, Qu LW, Ma L, Zhou YC, Wang GZ, Zhao XC, Zhang C, Zhang YF, Wang M, Zhang MY, et al: Genome-wide identification of transcription factors that are critical to non-small cell lung cancer. Cancer Lett. 434:132–143. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Cui T, Yang L, Ma Y, Petersen I and Chen Y: Desmocollin 3 has a tumor suppressive activity through inhibition of AKT pathway in colorectal cancer. Exp Cell Res. 378:124–130. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Liang L, Zeng JH, Qin XG, Chen JQ, Luo DZ and Chen G: Distinguishable prognostic signatures of left- and right-sided colon cancer: A study based on sequencing data. Cell Physiol Biochem. 48:475–490. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Lan Y, Xiao X, Luo Y, He Z and Song X: FEZF1 is an independent predictive factor for recurrence and promotes cell proliferation and migration in cervical cancer. J Cancer. 9:3929–3938. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Burotto M, Chiou VL, Lee JM and Kohn EC: The MAPK pathway across different malignancies: A new perspective. Cancer. 120:3446–3456. 2015. View Article : Google Scholar

42 

Nakagawa H, Hirata Y, Takeda K, Hayakawa Y, Sato T, Kinoshita H, Sakamoto K, Nakata W, Hikiba Y, Omata M, et al: Apoptosis signal-regulating kinase 1 inhibits hepatocarcinogenesis by controlling the tumor-suppressing function of stress-activated mitogen-activated protein kinase. Hepatology. 54:185–195. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Li S, Yin Y and Yu H: Genetic expression profile-based screening of genes and pathways associated with papillary thyroid carcinoma. Oncol Lett. 16:5723–5732. 2018.PubMed/NCBI

44 

Chen S, Fan X, Gu H, Zhang L and Zhao W: Competing endogenous RNA regulatory network in papillary thyroid carcinoma. Mol Med Rep. 18:695–704. 2018.PubMed/NCBI

45 

Puxeddu E and Moretti S: Clinical prognosis in BRAF-mutated PTC. Arq Bras Endocrinol Metabol. 51:736–747. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Wu M, Yuan H, Li X, Liao Q and Liu Z: Identification of a five-gene signature and establishment of a prognostic nomogram to predict progression-free interval of papillary thyroid carcinoma. Front Endocrinol (Lausanne). 10:7902019. View Article : Google Scholar : PubMed/NCBI

47 

Cao R, Wu Q, Li Q, Yao M and Zhou H: A 3-mRNA-based prognostic signature of survival in oral squamous cell carcinoma. PeerJ. 7:e73602019. View Article : Google Scholar : PubMed/NCBI

48 

Cai J, Tong Y, Huang L, Xia L, Guo H, Wu H, Kong X and Xia Q: Identification and validation of a potent multi-mRNA signature for the prediction of early relapse in hepatocellular carcinoma. Carcinogenesis. 40:840–852. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Xie X, Wang J, Shi D, Zou Y, Xiong Z, Li X, Zhou J, Tang H and Xie X: Identification of a 4-mRNA metastasis-related prognostic signature for patients with breast cancer. J Cell Mol Med. 23:1439–1447. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Guo W, Chen X, Zhu L and Wang Q: A six-mRNA signature model for the prognosis of head and neck squamous cell carcinoma. Oncotarget. 8:94528–94538. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Chuang MK, Chiu YC, Chou WC, Hou HA, Tseng MH, Kuo YY, Chen Y, Chuang EY and Tien HF: An mRNA expression signature for prognostication in de novo acute myeloid leukemia patients with normal karyotype. Oncotarget. 6:39098–39110. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Bao ZS, Li MY, Wang JY, Zhang CB, Wang HJ, Yan W, Liu YW, Zhang W, Chen L and Jiang T: Prognostic value of a nine-gene signature in glioma patients based on mRNA expression profiling. CNS Neurosci Ther. 20:112–118. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 20 Issue 3

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 LK, Gan XX, Deng XY, Shen F, Feng JH, Cai WS, Liu QY, Miao JH, Zheng BX, Xu B, Xu B, et al: Potential five‑mRNA signature model for the prediction of prognosis in patients with papillary thyroid carcinoma. Oncol Lett 20: 2302-2310, 2020
APA
Zhong, L., Gan, X., Deng, X., Shen, F., Feng, J., Cai, W. ... Xu, B. (2020). Potential five‑mRNA signature model for the prediction of prognosis in patients with papillary thyroid carcinoma. Oncology Letters, 20, 2302-2310. https://doi.org/10.3892/ol.2020.11781
MLA
Zhong, L., Gan, X., Deng, X., Shen, F., Feng, J., Cai, W., Liu, Q., Miao, J., Zheng, B., Xu, B."Potential five‑mRNA signature model for the prediction of prognosis in patients with papillary thyroid carcinoma". Oncology Letters 20.3 (2020): 2302-2310.
Chicago
Zhong, L., Gan, X., Deng, X., Shen, F., Feng, J., Cai, W., Liu, Q., Miao, J., Zheng, B., Xu, B."Potential five‑mRNA signature model for the prediction of prognosis in patients with papillary thyroid carcinoma". Oncology Letters 20, no. 3 (2020): 2302-2310. https://doi.org/10.3892/ol.2020.11781