Open Access

A risk score system based on DNA methylation levels and a nomogram survival model for lung squamous cell carcinoma

  • Authors:
    • Ming Zhang
    • Libing Sun
    • Yi Ru
    • Shasha Zhang
    • Junjun Miao
    • Pengda Guo
    • Jinghuan Lv
    • Feng Guo
    • Biao Liu
  • View Affiliations

  • Published online on: April 27, 2020     https://doi.org/10.3892/ijmm.2020.4590
  • Pages: 252-264
  • Copyright: © Zhang 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

Lung squamous cell carcinoma (LSCC) is one of the primary types of non‑small cell lung carcinoma, and patients with recurrent LSCC usually have a poor prognosis. The present study was conducted to build a risk score (RS) system for LSCC. Methylation data on LSCC (training set) and on head and neck squamous cell carcinoma (validation set 2) were obtained from The Cancer Genome Atlas database, and GSE39279 (validation set 1) was retrieved from the Gene Expression Omnibus database. Differentially methylated protein‑coding genes (DMGs)/long non‑coding RNAs (DM‑lncRNAs) between recurrence‑associated samples and nonrecurrence samples were screened out using the limma package, and their correlation analysis was conducted using the cor.test() function. Following identification of the optimal combinations of DMGs or DM‑lncRNAs using the penalized package in R, RS systems were built, and the system with optimal performance was selected. Using the rms package, a nomogram survival model was then constructed. For the differentially expressed genes (DEGs) between the high‑ and low‑risk groups, pathway enrichment analysis was performed by Gene Set Enrichment Analysis. There were 335 DMGs and DM‑lncRNAs in total. Following screening out of the top 10 genes (aldehyde dehydrogenase 7 family member A1, chromosome 8 open reading frame 48, cytokine‑like 1, heat shock protein 90 alpha family class A member 1, isovaleryl‑CoA dehydrogenase, phosphodiesterase 3A, PNMA family member 2, SAM domain, SH3 domain and nuclear localization signals 1, thyroid hormone receptor interactor 13 and zinc finger protein 878) and 6 top lncRNAs, RS systems were constructed. According to Kaplan‑Meier analysis, the DNA methylation level‑based RS system exhibited the best performance. In combination with independent clinical prognostic factors, a nomogram survival model was built and successfully predicted patient survival. Furthermore, 820 DEGs between the high‑ and low‑risk groups were identified, and 3 pathways were identified to be enriched in this gene set. The 10‑DMG methylation level‑based RS system and the nomogram survival model may be applied for predicting the outcomes of patients with LSCC.

Introduction

Lung cancer is a common cancer worldwide, with the second highest mortality rate among females and the highest mortality rate among males (1,2). Non-small cell lung cancer (NSCLC) is the primary lung cancer type that accounts for ~85% of lung cancer cases and includes the following common pathological subtypes: Lung squamous cell carcinoma (LSCC), lung adenocarcinoma (LUAD), and lung large cell carcinoma (3,4). LSCC is more common among males, potentially due to smoking habits (5). LSCC has distinct clinical characteristics, and the prognosis of patients with recurrent LSCC is poor (6). Therefore, it is urgent to identify new molecular factors predictive of LSCC treatment outcomes.

Methylation changes in certain tumor-associated genes have been identified in previous studies examining tumorigenesis, suggesting that they are critical risk factors of tumorigenesis and molecular markers for early diagnosis (7). Promoter methylation of PR/SET domain 5 is significantly associated with lymph node metastasis and tumor differentiation status of LSCC; therefore, this gene is a candidate target for the diagnosis, prognosis and treatment of this cancer (8). The drought-repressed 4 gene has been proposed for methylation status analysis of LSCC cells, and its low expression correlates with a poor patient prognosis (9). Tripartite motif-containing 58/cg26157385 methylation is associated with expression of 8 prognosis-associated genes in LSCC, indicating its potential regulatory role in the progression of LSCC (10).

Long non-coding RNAs (lncRNAs) are a class of RNAs measuring >200 nucleotides and have pivotal functions in the progression of numerous cancer types (11). In addition, increasing numbers of lncRNAs with a prognostic value have been described, for example, urothelial cancer associated and long intergenic non-coding RNA-p21 (12-14). The lncRNAs cancer susceptibility 2 and surfactant associated 1, pseudogene participate in the regulation of tumor suppressor genes and oncogenes during the formation of LSCC and may be used for the diagnosis, prognosis and targeted treatment of this disease (15). Several lncRNA combination signatures have been identified to predict LSCC prognosis (16,17). In LUAD, DNA methylation has regulatory effects on the role of lncRNA (18); however, to the best of our knowledge, this type of regulation in LSCC has rarely been reported.

Despite these valuable data, the predictive performance of methylated DNA sequences or lncRNAs is rarely compared, and additional potential biomarkers should be investigated to improve cancer prognosis. In the present study, the methylation levels of DNA sequences and lncRNAs in patients with LSCC were compared, and the levels identified to be significantly correlated with LSCC prognosis were screened out. Furthermore, different risk score (RS) systems were built, and the one with the best predictive performance was selected.

Materials and methods

Data sources

Methylation data from patients with LSCC (downloaded on March 25, 2019; platform: Illumina Infinium Human Methylation 450 BeadChip) including 372 LSCC samples and 43 healthy tissue samples, were retrieved from The Cancer Genome Atlas (TCGA; https://cancergenome.nih. gov/) database. Following collation with clinical information, 293 LSCC samples with data recurrence were selected as the training set.

With 'lung cancer' and 'Homo sapiens' as key words, a search for methylation datasets was conducted in the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih. gov/geo/) database (19) using the following criteria: i) Available histological data that could differentiate adenocarcinoma from squamous cell carcinoma; ii) total sample size was not <150; and iii) the clinical information associated with the LSCC samples contained data on actual future recurrence. Following this search, an eligible methylation dataset (accession no., GSE39279 (20); platform: Illumina HumanMethylation450 BeadChip; validation set 1) was selected and downloaded. There were 444 samples in the dataset, including 43 LSCC samples with data concerning future recurrence.

In addition, the methylation data on head and neck squamous cell carcinoma (HNSC), including 530 HNSC samples and 50 healthy tissue samples, were also retrieved from TCGA to investigate whether the predictive model for LSCC was applicable to other squamous carcinoma types. Following collation with the clinical information downloaded from TCGA simultaneously, 382 HNSC samples with information on future recurrence were selected as validation set 2.

Differential methylation analysis

According to the data corresponding to probe locations and IDs provided in the downloaded annotation files that contained information about protein-coding genes and non-coding RNAs, like gene and lncRNA function, DNA location site and pathway information, corresponding lncRNAs and genes in the methylation data were annotated using the HUGO Gene Nomenclature Committee (http://www.genenames.org/) database (21), which contains records on 4,112 lncRNAs and 19,201 genes.

On the basis of the recurrence data, the LSCC samples in the training set were classified into recurrence-associated (the tumor recurred) and nonrecurrence (the tumor did not recur) groups. Using the limma package (22) (v.3.34.7, https://bioconductor.org/packages/release/bioc/html/limma.html) in the R software (23), differentially methylated genes (DMGs) between the recurrence-associated and nonrecurrence samples were identified. The thresholds for significance were defined as |log2 fold change (FC)|>0.263 and false discovery rate (FDR) <0.05. Following the addition of data on methylation levels of the DMGs in the training set, bidirectional hierarchical clustering was performed on the DMGs via the pheatmap package (v.1.0.8, https://cran.r-project.org/web/packages/pheatmap/index.html) in R (24).

Analysis of correlation between the methylation levels and expression levels of the DMGs

Data on the methylation levels and the matched expression levels of the DMGs were extracted, and their Pearson correlation coefficients (PCCs) were calculated using the cor.test() function (https://stat.ethz.ch/R-manual/R-devel/library/stats/html/cor.test.html) in R (25). P<0.05 was set as the significance threshold. DMGs with negative PCCs were selected for further analyses.

Construction of RS systems

Using univariate and multivariate Cox regression analyses in R package survival (v3.1-8; https://www.rdocumentation.org/packages/survival/versions/3.1-8), differentially methylated lncRNAs (DM-lncRNAs) and DMGs significantly associated with overall survival and independent prognosis factors were next screened out. A log-rank P<0.05 was selected as the cutoff criterion.

Based on the DMGs correlating with independent prognosis, an optimal gene combination and optimal lncRNA combination were identified using the LASSO Cox regression model (26) in penalized package in R (27) (v.0.9.50; https://cran.r-project.org/web/packages/penalized/index.html). Optimal parameter 'lambda' in the model was calculated by the 1,000-fold cross-validation likelihood (cvl) method.

In combination with prognostic coefficients of the optimal DM-lncRNAs and DMGs, methylation status-based or methylation level-based RS systems were built.

For the optimal combinations, cutoff values of methylation levels of the optimal lncRNAs and genes were computed via the X-Tile Bio-Informatics Tool (https://medicine.yale.edu/lab/rimm/research/software.aspx) (28). The cutoff values were determined based on the following criterion: Monte Carlo P<0.05. The status of each sample at an lncRNA or DNA methylation level was defined in accordance with the cutoff value of each lncRNA or gene. When the lncRNA or DNA methylation level was increased compared with the cutoff value, its status was set to 1.0. Otherwise, the status was set to 0.

In combination with the regression coefficients of each optimal lncRNA or gene and their methylation status, a RS system was created, and the RS for each sample was calculated via the following formula:

StatusRS=∑βRNAn×StatusRNAn; where β and Status represent the regression coefficient of an lncRNA or gene and the methylation status variable, respectively. The status RS systems, based on the methylation status of the optimal genes and optimal lncRNAs, were constructed separately as two separate status RS systems.

RS systems based on the methylation levels of optimal lncRNAs or optimal genes were constructed. The formula for calculating the RS of each sample was as follows:

MethylationRS=∑βRNAn×MethylationRNAn; where β and MethylationRNAn represent the regression coefficient and methylation level of an lncRNA or gene, respectively. A methylation RS system based on the methylation levels of optimal genes and a methylation RS system based on the methylation levels of optimal lncRNAs were built separately.

Samples in the training set were subdivided into high- and low-risk groups based on the median of the RSs of the two status RS systems and the two methylation RS systems. Using the Kaplan-Meier (KM) survival curve constructed by means of R package survival (29), associations between the prognosis and RS systems were analyzed. Concomitantly, the two validation sets were used to validate the results obtained. Finally, from the four RS systems generated, the system with optimum performance was selected through comparison of the parameters in the training set and the two validation sets.

Construction of a nomogram survival model

In the training set, independent prognostic factors were selected by univariate and multivariate Cox regression analyses via the survival package (29). The cutoff criterion was log-rank P<0.05. According to the risk data determined by the RS system, the associations between the independent clinical prognostic factors and prognosis were then analyzed. Using the rms package (v.5.1-2, https://cran.r-project.org/web/packages/rms/index.html) (30) in R, a 5-year nomogram survival model involving the independent clinical prognostic factors was built.

Pathway enrichment analysis

RSs of the best RS system were used to classify the samples in the training set into high- and low-risk groups. Using the limma package (23), differential expression analysis of the high- and low-risk groups was performed to identify differentially expressed genes (DEGs) between the two risk groups. The DEGs were screened according to FDR <0.05 and |log2 FC|>0.263 cut-off values. Using Gene Set Enrichment Analysis (http://software.broadinstitute.org/gsea/index.jsp) (31), pathway enrichment analysis of the DEG set was conducted. The pathways with P<0.05 were identified as significant.

Results

Differential-methylation analysis

On the basis of the annotation files, 1,028 lncRNAs and 15,544 genes were selected from the methylation dataset. According to the recurrence data, samples in the training set were subdivided into the recurrence-associated (78 LSCC samples from patients who later experienced cancer recurrence) and nonrecurrence (215 LSCC samples) groups. There were 335 DMGs and DM-lncRNAs between the recurrence-associated and nonrecurrence groups, including 27 DM-lncRNAs, 4 hypo-methylated and 23 hypermethylated lncRNAs, and 308 DMGs, 139 hypomethylated and 169 hypermethylated genes (Fig. 1A). The bidirectional hierarchical clustering heatmap for the DMGs and GM-lncRNAs is presented in Fig. 1B.

Analysis of the correlation between the methylation levels and expression levels of the DMGs or DM-lncRNAs

As aforementioned, the matched methylation samples from the LSCC mRNA-seq samples were first selected, and the methylation and expression data on the DMGs and DM-lncRNAs were then extracted. The overall correlation between the methylation levels and expression levels of the DMGs/DM-lncRNAs were analyzed among the matched samples, and a significant negative correlation (PCC=-0.3857; P=0.574×10−13) was identified (Fig. 2). A total of 181 DMGs and DM-lncRNAs with negative PCCs, including 25 DM-lncRNAs and 156 DMGs, were identified for subsequent analyses.

Construction of the RS system

Among the 181 DMGs and DM-lncRNAs, 105 were identified to be associated with prognosis, including 91 DMGs and 14 DM-lncRNAs, by univariate Cox regression analysis. Then, 16 DMGs and 6 DM-lncRNAs independently associated with prognosis were identified from the 105 DMGs/DM-lncRNAs using multivariate Cox regression analysis. According to the LASSO Cox regression model, 10 optimal DMGs, including aldehyde dehydrogenase 7 family member A1 (ALDH7A1), chromosome 8 open reading frame 48 (C8orf48), cytokine-like 1 (CYTL1), heat shock protein 90 alpha family class A member 1 (HSP90AA1), isovaleryl-CoA dehydrogenase (IVD), phosphodiesterase 3A (PDE3A), PNMA family member 2 (PNMA2), SAM domain, SH3 domain and nuclear localization signals 1 (SAMSN1), thyroid hormone receptor interactor 13 (TRIP13) and zinc finger protein 878 (ZNF878), and 6 optimal DM-lncRNAs, including ATXN8 opposite-strand lncRNA (ATXN8OS), brain cytoplasmic RNA 1 (BCYRN1), family with sequence similarity 138 member D (FAM138D), HLA complex group 11 (HCG11), MIR155 host gene (MIR155HG) and testis-specific transcript, Y-linked 13 (TTTY13) were finally identified in the training dataset (Fig. 3; Table I).

Table I

Genes and lncRNAs involved in the optimal combinations.

Table I

Genes and lncRNAs involved in the optimal combinations.

TypeGeneLocusCoefficientHazard ratio95% CIP-valueCutoff
geneALDH7A1cg26327732−1.1070.4150.250-0.884 2.350×10−50.34
C8orf48cg24727311−0.5720.1910.180-0.457 4.610×10−40.34
CYTL1cg175630340.4961.5091.104-2.323 3.051×10−30.16
HSP90AA1cg239042471.0124.7692.041-6.111 1.080×10−40.53
IVDcg275299300.5701.9251.244-3.191 3.500×10−40.11
PDE3Acg265718140.8991.9771.745-3.979 9.310×10−50.34
PNMA2cg262682770.0111.1231.043-2.705 7.295×10−30.48
SAMSN1cg139516640.1801.2081.079-2.546 1.490×10−40.25
TRIP13cg175103850.1611.331.038-2.691 9.838×10−30.65
ZNF878cg26626525−0.1690.5970.336-0.906 1.630×10−30.06
lncRNAATXN8OScg25514273−1.2270.3120.088-0.704 7.080×10−30.24
BCYRN1ch.X.1084981F13.1985.3991.028-10.38 1.229×10−20.04
FAM138Dcg265231962.6252.8541.681-4.425 9.330×10−30.73
HCG11cg274903872.9533.1641.550-6.734 2.010×10−30.20
MIR155HGcg234338891.2581.4311.281-3.186 3.341×10−20.04
TTTY13cg259188491.8311.5951.492-3.413 6.200×10−30.24

[i] lncRNA, long non-coding RNA; CI, confidence interval.

The cutoff values of the methylation levels of the lncRNAs and genes in the aforementioned optimal combinations were calculated (Table I). In combination with the regression coefficients of the optimal genes or lncRNAs, RS systems based on the methylation status of optimal genes or lncRNAs were constructed. The formulas were as follows:

Gene status RS=−1.1070 × StatusALDH7A1-0.5721 × StatusC8orf48 + 0.4962 × StatusCYTL1 + 1.0118 × StatusHSP90AA1 + 0.5696 × StatusIVD + 0.8992 × StatusPDE3A + 0.0109 × StatusPNMA2 + 0.1803 × StatusSAMSN1 + 0.1612 × StatusTRIP13-0.1688 × StatusZNF878; and lncRNA status RS=−1.2274 × StatusATXN8OS + 13.1984 × StatusBCYRN1 + 2.6252 × StatusFAM138D + 2.9526 × StatusHCG11 + 1.2577 × StatusMIR155HG + 1.8308 × StatusTTTY13.

In addition, RS systems based on the methylation levels of optimal genes or lncRNAs were built. The formulas for calculating the RSs were:

Gene methylation (methy) RS = −1.10 70 × MethyALDH7A1-0.5721 × MethyC8orf48 + 0.4962 × MethyCYTL1 + 1.0118 × MethyHSP90AA1 + 0.5696 × MethyIVD + 0.8992 × MethyPDE3A + 0.0109 × MethyPNMA2 + 0.1803 × MethySAMSN1 + 0.1612 × MethyTRIP13-0.1688 × MethyZNF878; and lncRNA methy RS=−1.2274 × MethyATXN8OS + 13.1984 × MethyBCYRN1 + 2.6252 × MethyFAM138D + 2.9526 × MethyHCG11 + 1.2577 × MethyMIR155HG + 1.8308 × MethyTTTY13.

According to the median RS values, samples in the training set, validation set 1 and validation set 2 were separately subdivided into high- and low-risk groups. The associations between prognosis and the results of RS systems were analyzed. The KM curves for the methylation status-based RS systems and those for the methylation level-based RS systems are presented in Figs. 4 and 5, respectively. These RS systems demonstrated high levels of specificity, with the area under the receiver operating characteristic curve >0.6 (Figs. 4 and 5), indicating accurate levels of risk prediction. Of all the systems generated, the RS system based on the methylation level of the 10 optimal DEGs exhibited the best predictive performance (Table II).

Table II

Survival analysis of the methylation status-based RS system.

Table II

Survival analysis of the methylation status-based RS system.

A, KM survival analysis for the methylation status-based RS system
Gene product typeStatus model (log-rank P-value)
TrainingHR (95% CI)Validation 1 (GSE39279)HR (95% CI)Validation 2 (TCGA-HNSC)HR (95% CI)
IncRNA 2.628×10−42.370 (1.471-3.818) 1.446×10−11.734 (0.821-3.662) 1.385×10−11.397 (0.896-2.178)
DNA 2.004×10−126.112 (3.452-10.82) 3.217×10−22.213 (1.050-4.664) 2.742×10−21.638 (1.052-2.550)

B, KM survival analysis for the methylation level-based RS system
Methylation model (log-rank P-value)
Gene product typeTrainingHR (95% CI)Validation 1 (GSE39279)HR (95% CI)Validation 2 (TCGA-HNSC)HR (95% CI)

IncRNA 6.884×10−52.601 (1.596-4.241) 7.923×10−21.913 (0.916-3.995) 1.366×10−11.391 (0.899-2.150)
DNA 5.928×10−125.870 (3.325-10.37) 2.209×10−2 2.355(1.108-5.006) 4.833×10−21.551 (1.098-2.402)

C, AUC of the four RS systems
Status model (ROC)
Methylation model (ROC)
Gene product typeTrainingValidation 1 (GSE39279)Validation 2 (TCGA-HNSC)TrainingValidation 1 (GSE39279)Validation 2 (TCGA-HNSC)

IncRNA0.9630.8530.9080.9970.8240.967
DNA0.9950.7420.9610.9590.8460.912

[i] KM, Kaplan-Meier; RS, risk score; ROC, receiver operating characteristic; AUC, area under the ROC curve; IncRNA, long non-coding RNA; HR, Hazard Ratio; CI, confidence interval; TCGA, The Cancer Genome Atlas; HNSC, head and neck squamous cell carcinoma.

Construction of the nomogram survival model

Independent clinical factors associating with prognosis in the training set were identified by means of univariate and multivariate Cox regression analyses (Table III). For both the training and validation sets, pathological T stage, radiotherapy and DNA methylation level-based model RS status were selected as independent prognostic factors. The KM curves demonstrating the correlations between pathological T/radiotherapy and prognosis are presented in Fig. 6. The patients with low pathological T staging and those who underwent radiotherapy exhibited improved prognoses, which is consistent with the outcomes observed in clinical practice.

Table III

Screening of the independent prognostic factors using

Table III

Screening of the independent prognostic factors using

Clinical characteristicsTCGA (n=293)Univariate
Multivariate
HR (95% CI)P-valueHR (95% CI)P-value
Age, years'67.72±8.630.988 (0.962-1.014) 3.505×10−1--
Sex. male/female211/821.329 (0.783-2257) 2.903×10−1--
Pathological M stage. M0/M1/-227/2/64 0.303(0.151-1536) 6.074×10−1--
Pathological N stage, N0/N1/N2/-193/76/20/4 1.562(1.142-2.138) 4.719×l0−30.365 (0.803-2 582)2.210×10−1
Pathological T stage, T1/T2/T3/T479/159/48/71.639 (1209-2220) 1.426×10−3 1.560(1.096-2.444) 4.520×l0−2
Pathological stage. LTIHLIW-139/110/39/2/3 1.768(1.326-2.356) 7.566×10−51.191 (0.653-2.171) 5.681×10−1
Radiation therapy, Yes/No/-31/251/11 2.054(1.145-3.686) 1.366×10−2 2.244(1.191-4227) 1.230×10−2
Targeted molecular therapy. Yes/No/-94/188/11 1.238(0.774-1.981) 3.719×10−1--
mRNA methylation model RS status, High/Low146/147 5.870(3.325-10.37) 5.928×l0−128.348 (4.408-15.812) 7.420×10−11
Recurrence, Yes/No78/215----
Recurrence free survival time, months'28.28±27.45----

a Data are expressed as mean ± standard deviation. HR, Hazard Ratio; CI, confidence interval; TCGA, The Cancer Genome Atlas.

The 5-year nomogram survival model was built to further analyze the correlation between the 3 independent prognostic factors and prognosis; this model integrated various clinical indicators by means of the 'points' axis in the first row to predict the survival of patients corresponding to the samples (Fig. 7A). In addition, the predicted 5-year survival probability was compared to the actual 5-year survival probability, and a high level of consistency was observed (Fig. 7B).

Pathway enrichment analysis

As aforementioned, following the addition of the RSs of the DNA methylation level-based RS system to the analysis, the samples in the training set were classified into either high- or low-risk groups. A total of 820 DEGs between the two groups were identified, including 412 upregulated genes and 408 downregulated genes. Following pathway enrichment analysis, 3 pathways were identified to be enriched in this set of DEGs: The calcium signaling pathway (P=0.040944); complement and coagulation cascades (P=0.0034123); and vascular smooth muscle contraction (P=0.0427049; Table IV).

Table IV

Pathways enriched for the differentially expressed genes.

Table IV

Pathways enriched for the differentially expressed genes.

TermSizeESNESP-valueGenes
hsa04020: Calcium signaling pathway10−0.396503−1.114227 4.094×10−2EDNRB, HRH1, CALML3, PLN, LTB4R2, RYR1, BDKRB2, CALML5, ITPR2, F2R
hsa04610: Complement and coagulation cascades100.58634982.0125692 3.412×10−3C3AR1, F12, FGG, C5, F8, SERPINA1, C4BPA, C2, BDKRB2, F2R
hsa04270: Vascular smooth muscle contraction7−0.537324−1.341176 4.270×10−2PTGIR, CALML3, CALML5, PRKG1, PLA2G3, PPP1R14A, ITPR2

[i] ES, enrichment score; NES, normalized enrichment score.

Discussion

In the present study, a total of 335 DMGs and DM-lncRNAs were identified between the recurrence-associated samples and nonrecurrence samples of tumor tissues from patients with LSCC. Among them, 16 DMGs and 6 DM-lncRNAs were significantly associated with independent prognosis, and 10 optimal DMGs (ALDH7A1, C8orf48, CYTL1, HSP90AA1, IVD, PDE3A, PNMA2, SAMSN1, TRIP13 and ZNF878) and 6 optimal DM-lncRNAs (ATXN8OS, BCYRN1, FAM138D, HCG11, MIR155HG and TTTY13) were used to construct the methylation status-based or methylation level-based RS systems. In the KM analysis, the 10-DMG methylation level-based RS system exhibited the best performance. The set of DEGs between high- and low-risk groups according to the RS system was identified to be enriched in 3 pathways: The calcium signaling pathway; complement and coagulation cascades; and vascular smooth muscle contraction. As the RS system based on the 10 DMGs exhibited an improved predictive performance compared with that based on the 6 lncRNAs, the present study focused on the functions of these 10 DMGs in LSCC.

Elevated expression of ALDH7A1 in prostate cancer has been previously described, and was suggested to predict disease progression and metastasis (32), suggesting that alteration of the expression of this gene may control tumor progression. However, to the best of our knowledge, studies investigating the involvement of this gene in lung cancer are scarce. Low expression of ALDH7A1 in tumors of surgically treated patients with NSCLC is associated with a decreased incidence of tumor recurrence, indicating that decreased expression of this gene may predict a good prognosis for these patients (33). Patients with LSCC with idiopathic pulmonary fibrosis indicate a significantly decreased methylation level of ALDH7A1 compared with those without fibrosis (34). This observation suggests that methylation status of ALDH7A1 may affect other clinical factors. In the present study, ALDH7A1 was a component of the 10-DMG methylation level-based RS system that exhibited optimal performance; therefore, the methylation level of this gene may predict LSCC recurrence. Nevertheless, as the data concerning this gene in lung cancer are limited, the specific mechanisms of action of the ALDH7A1 protein and ALDH7A1 DNA methylation in LSCC should be studied further.

CYTL1 has important roles in certain types of cancer and is regulated by DNA methylation in LSCC (35). The methylation patterns of CYTL1 are evidently different between early and late stages of LSCC, and hypermethylation is more common in the advanced stages (36). Although CYTL1 hypermethylation does not affect the repression activity mediated by histone deacetylases (36), this repression activity may be subject to the regulation between CYTL1 hypermethylation and histone deacetylases, as the DNA methylation level is considered to affect the binding of a histone deacetylase to a promoter region (36,37). CYTL1 is suggested to be a risk indicator of smoking-associated impairment of metabolic health, as it is hypomethylated and upregulated in non-lung tissues of smokers (38). According to the present study, the CYTL1 gene was often methylated in recurring LSCC tumors, suggesting that it may be a predictive factor of LSCC recurrence.

High expression levels of HSP90AA1, heat shock protein 90 alpha family class B member 1 and heat shock protein 90 β family member 1 are associated with adverse outcomes among patients with NSCLC, and therefore may serve as promising prognostic markers and therapeutic targets in NSCLC (39). HSP90AA1 is differentially expressed between LUAD and LSCC, and therefore its expression profile may be used to distinguish the two subtypes (40). Nevertheless, data concerning HSP90AA1 methylation or the effect of this gene on tumor recurrence are scarce. PDE3A expression is low in chemoresistant NSCLC cells due to DNA hypermethylation, and high PDE3A expression is associated with improved survival in patients with LUAD (41). According to the results of the present study, methylated PDE3A may be associated with the prognosis of LSCC.

PNMA2 is aberrantly expressed in various types of tumor in patients with paraneoplastic syndromes; therefore, PNMA2 may be implicated in tumorigenesis (42). PNMA2 has been demonstrated to be a tissue marker of small intestine neuroendocrine tumors, and Ma2 autoantibodies in the blood are a valuable biomarker for the diagnosis and prediction of tumor recurrence (43). Conversely, to the best of our knowledge, no studies on the methylation of this gene have been published, and this parameter may be a new prognostic indicator in LSCC, based on the results of the present study.

Downregulation of SAMSN1 is detectable in lung cancer cell lines and may be involved in the development of this disease (44). Elevated TRIP13 expression contributes to the progression of LUAD and may be a candidate biomarker or therapeutic target in LUAD (45). In lung cancer in Xuanwei, TRIP13, cAMP-responsive element-binding protein 3-like 4 and cyclin E2 exhibit concordant upregulation and frequent copy number gains, and have been proposed as potential oncogenes in the pathogenesis of lung cancer in this region (46). In addition, silencing of TRIP13 can suppress cell growth and metastasis of hepatocellular carcinoma by activating TGF-β1/SMAD3 signaling (47). TRIP13 is 1 of the 7 hypomethylated genes in kidney renal cell carcinoma that have been suggested as a prognostic factor (48). Downregulation of ZNF878 has been suggested to be correlated with a poor prognosis of patients with LUAD (49). Although published data on SAMSN1, TRIP13, and ZNF878 methylation levels or their effects on LSCC are limited, when taken into consideration with the results of the present study, we hypothesize that these 3 genes may perform important functions in LSCC progression, and their methylation levels may help to determine an accurate prognosis of the disease.

Information on the participation of genes C8orf48 and IVD in LSCC is scare. According to the results of the present study, their methylation may be a prognostic indicator of LSCC recurrence.

Despite the identification of the 10-DMG signature that may predict the recurrence of LSCC, the present study contained certain limitations, such as the small sample size of the data retrieved from the database, and the lack of expression validation experiments. In addition, the clinical information of the LSCC samples in the training set was not compared to the validation sets, due to the difference in platforms. Nevertheless, this did preclude the predictive performance evaluation on risks of the identified clinical factors, as they could be distinguished from the AUC values. Further experiments are required to confirm the results.

In conclusion, 335 DMGs and DM-lncRNAs, including 27 DM-lncRNAs and 308 DMGs, were identified between recurrence-associated and nonrecurrence samples. The RS system based on the methylation levels of the 10 optimal DMGs (ALDH7A1, C8orf48, CYTL1, HSP90AA1, IVD, PDE3A, PNMA2, SAMSN1, TRIP13 and ZNF878) may help to predict the outcomes of patients with LSCC.

Funding

The present study was supported by Suzhou Science and Technology Development Project (grant no. SYS2018085) and the Construction Project of Suzhou Clinical Medical Center (grant no. Szzx201506).

Availability of data and materials

All the data used in the present study are available from the Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/; accession no., GSE39279) and The Cancer Genome Atlas public databases.

Authors' contributions

MZ, LS, YR, SZ, JM, PG and JL were responsible for data collection and statistical analysis. MZ, LS and YR participated in drafting and editing the manuscript. FG and BL were responsible for designing the work and reviewing the manuscript. BL obtained the funding. 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.

Acknowledgments

The results shown here are in whole or part based upon data generated by The Cancer Genome Atlas Research Network: https://www.cancer.gov/tcga.

Abbreviations:

AUC

area under the receiver operating characteristic curve

DEG

differentially expressed gene

DMG

differentially methylated gene

DM-lncRNA

differentially methylated lncRNA

FDR

false discovery rate

FC

fold change

HNSC

head and neck squamous cell carcinoma

KM

Kaplan-Meier analysis

lncRNA

long non-coding RNA

LSCC

lung squamous cell carcinoma

NSCLC

non-small cell lung cancer

LUAD

lung adenocarcinoma

PCC

Pearson correlation coefficient

RFS

recurrence-free survival

RS

risk score

ROC

receiver operating characteristic

TCGA

The Cancer Genome Atlas

References

1 

McGuire S: World cancer report 2014. Geneva, Switzerland: World health organization, international agency for research on cancer WHO press, 2015. Adv Nutr. 7:418–419. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Hirsch FR and Bunn PA Jr: Progressin research on screening and genetics in lung cancer. Lancet Respir Med. 2:19–21. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Travis WD: Update on small cell carcinoma and its differentiation from squamous cell carcinoma and other non-small cell carcinomas. Mod Pathol. 25(Suppl 1): S18–S30. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Yoon SM, Shaikh T and Hallman M: Therapeutic management options for stage III non-small cell lung cancer. World J Clin Oncol. 8:1–20. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Tian S: Classification and survival prediction for early-stage lung adenocarcinoma and squamous cell carcinoma patients. Oncol Lett. 14:5464–5470. 2017.PubMed/NCBI

6 

Toyokawa G, Kozuma Y, Matsubara T, Haratake N, Takamori S, Akamine T, Takada K, Katsura M, Shimokawa M, Shoji F, et al: Prognostic impact of controlling nutritional status score in resected lung squamous cell carcinoma. J Thorac Dis. 9:2942–2951. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Pfeifer G: Defining driver DNA methylation changes in human cancer. Int J Mol Sci. 19:pii: E1166. 2018. View Article : Google Scholar

8 

Tan SX, Hu RC, Tan YL, Liu JJ and Liu WE: Promoter methylation-mediated downregulation of PRDM5 contributes to the development of lung squamous cell carcinoma. Tumour Biol. 35:4509–4516. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Wang W, Qi X and Wu M: Effect of DR4 promoter methylation on the TRAIL-induced apoptosis in lung squamous carcinoma cell. Oncol Rep. 34:2115–2125. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Zhang W, Cui Q, Qu W, Ding X, Jiang D and Liu H: TRIM58/cg26157385 methylation is associated with eight prognostic genes in lung squamous cell carcinoma. Oncol Rep. 40:206–216. 2018.PubMed/NCBI

11 

Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and cancer: A new paradigm. Cancer Res. 77:3965–3981. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Tong J, Ma X, Yu H and Yang J: SNHG15: A promising cancer-related long noncoding RNA. Cancer Manag Res. 11:5961–5969. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Yao F, Wang Q and Wu Q: The prognostic value and mechanisms of lncRNA UCA1 in human cancer. Cancer Manag Res. 11:7685–7696. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Lu T, Wang Y, Chen D, Liu J and Jiao W: Potential clinical application of lncRNAs in non-small cell lung cancer. Onco Targets Ther. 11:8045–8052. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Huang GQ, Ke ZP, Hu HB and Gu B: Co-expression network analysis of long noncoding RNAs (IncRNAs) and cancer genes revealsSFTA1P and CASC2abnormalities in lung squamous cell carcinoma. Cancer Biol Ther. 18:115–122. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Li S, Teng Y, Yuan MJ, Ma TT, Ma J and Gao XJ: A seven long-noncoding RNA signature predicts prognosis of lung squamous cell carcinoma. Biomark Med. 14:53–63. 2020. View Article : Google Scholar

17 

Huang G, Huang Q, Xie Z, Zhou H, Cao J, Shi L and Yang M: A nine-long non-coding RNA signature for prognosis prediction of patients with lung squamous cell carcinoma. Cancer Biomark. 26:239–247. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Li R, Yang YE, Yin YH, Zhang MY, Li H and Qu YQ: Methylation and transcriptome analysis reveal lung adenocarcinoma-specific diagnostic biomarkers. J Transl Med. 17:3242019. View Article : Google Scholar : PubMed/NCBI

19 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets-update. Nucleic Acids Res. 41(Database Issue): D991–D995. 2013. View Article : Google Scholar

20 

Sandoval J, Mendez-Gonzalez J, Nadal E, Chen G, Carmona FJ, Sayols S, Moran S, Heyn H, Vizoso M, Gomez A, et al: A prognostic DNA methylation signature for stage I non-small-cell lung cancer. J Clin Oncol. 31:4140–4147. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Yates B, Braschi B, Gray KA, Seal RL, Tweedie S and Bruford EA: Genenames.orgurisimpleGenenames.org: The HGNC and VGNC resources in 2017. Nucleic Acids Res. 45(D1): D619–D625. 2017. View Article : Google Scholar

22 

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

23 

Law CW, Alhamdoosh M, Su S, Dong X, Tian L, Smyth GK and Ritchie ME: RNA-seq analysis is easy as 1-2-3 with limma, Glimma and edgeR. F1000Res. 5:14082016. View Article : Google Scholar

24 

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

25 

Kotthaus H, Korb I, Engel M and Marwedel P: Dynamic page sharing optimization for the R language. Acm Sigplan Notices. 50:79–90. 2014. View Article : Google Scholar

26 

Kong S and Nan B: Non-asymptotic oracle inequalities for the high-dimensional Cox regression via lasso. Stat Sin. 24:25–42. 2012.

27 

Goeman JJ: L1 penalized estimation in the Cox proportional hazards model. Biom J. 52:70–84. 2010.

28 

Camp RL, Marisa DF and Rimm DL: X-tile: A new bio-informatics tool for biomarker assessment and outcome-based cut-point optimization. Clin Cancer Res. 10:7252–7259. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Liu XR, Pawitan Y and Clements MS: Generalized survival models for correlated time-to-event data. Stat Med. 36:4743–4762. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Steyerberg EW and FRANK E HARRELL Jr: Regression modeling strategies: With applications, to linear models, logistic and ordinal regression, and survival analysis, 2nd ed. Heidelberg Springer. Biometrics. 72:1006–1007. 2016. View Article : Google Scholar

31 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI

32 

van den Hoogen C, van der Horst G, Cheung H, Buijs JT, Lippitt JM, Guzmán-Ramírez N, Hamdy FC, Eaton CL, Thalmann GN, Cecchini MG, et al: High aldehyde dehydrogenase activity identifies tumor-initiating and metastasis-initiating cells in human prostate cancer. Cancer Res. 70:5163–5173. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Giacalone NJ, Den RB, Eisenberg R, Chen H, Olson SJ, Massion PP, Carbone DP and Lu B: ALDH7A1 expression is associated with recurrence in patients with surgically resected non-small-cell lung carcinoma. Future Oncol. 9:737–745. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Hata A, Nakajima T, Matsusaka K, Fukuyo M, Morimoto J, Yamamoto T, Sakairi Y, Rahmutulla B, Ota S, Wada H, et al: A low DNA methylation epigenotype in lung squamous cell carcinoma and its association with idiopathic pulmonary fibrosis and poorer prognosis. Int J Cancer. 146:388–399. 2020. View Article : Google Scholar

35 

Zhu S, Kuek V, Bennett S, Xu H, Rosen V and Xu J: Protein Cytl1: Its role in chondrogenesis, cartilage homeostasis, and disease. Cell Mol Life Sci. 76:3515–3523. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Kwon YJ, Lee SJ, Koh JS, Kim SH, Lee HW, Kang MC, Bae JB, Kim YJ and Park JH: Genome-wide analysis of DNA methylation and the gene expression change in lung cancer. J Thorac Oncol. 7:20–33. 2012. View Article : Google Scholar

37 

Kim SN, Kim NH, Lee W, Seo DW and Kim YK: Histone deacetylase inhibitor induction of P-glycoprotein transcription requires both histone deacetylase 1 dissociation and recruitment of CAAT/enhancer binding protein beta and pCAF to the promoter region. Mol Cancer Res. 7:735–744. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Tsai PC, Glastonbury CA, Eliot MN, Bollepalli S, Yet I, Castillo-Fernandez JE, Carnero-Montoro E, Hardiman T, Martin TC, Vickers A, et al: Smoking induces coordinated DNA methylation and gene expression changes in adipose tissue with consequences for metabolic health. Clin Epigenetics. 10:1262018. View Article : Google Scholar : PubMed/NCBI

39 

Liu K, Kang M, Li J, Qin W and Wang R: Prognostic value of the mRNA expression of members of the HSP90 family in non-small cell lung cancer. Exp Ther Med. 17:2657–2665. 2019.PubMed/NCBI

40 

Lu C, Chen H, Shan Z and Yang L: Identification of differentially expressed genes between lung adenocarcinoma and lung squamous cell carcinoma by gene expression profiling. Mol Med Rep. 14:1483–1490. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Tian FM, Zhong CY, Wang XN and Meng Y: PDE3A is hyper-methylated in cisplatin resistant non-small cell lung cancer cells and is a modulator of chemotherapy response. Eur Rev Med Pharmacol Sci. 21:2635–2641. 2017.PubMed/NCBI

42 

Lee YH, Pang SW and Tan KO: PNMA2 mediates heterodimeric interactions and antagonizes chemo-sensitizing activities mediated by members of PNMA family. Biochem Biophys Res Commun. 473:224–229. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Cui T, Hurtig M, Elgue G, Li SC, Veronesi G, Essaghir A, Demoulin JB, Pelosi G, Alimohammadi M, Öberg K and Giandomenico V: Paraneoplastic antigen Ma2 autoantibodies as specific blood biomarkers for detection of early recurrence of small intestine neuroendocrine tumors. PLoS One. 5:e160102010. View Article : Google Scholar

44 

Yamada H, Yanagisawa K, Tokumaru S, Taguchi A, Nimura Y, Osada H, Nagino M and Takahashi T: Detailed characterization of a homozygously deleted region corresponding to a candidate tumor suppressor locus at 21q11-21 in human lung cancer. Genes Chromosomes Cancer. 47:810–818. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Li W, Zhang G, Li X, Wang X, Li Q, Hong L, Shen Y, Zhao C, Gong X, Chen Y and Zhou J: Thyroid hormone receptor interactor 13 (TRIP13) overexpression associated with tumor progression and poor prognosis in lung adenocarcinoma. Biochem Biophys Res Commun. 499:416–424. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Zhang Y, Xue Q, Pan G, Meng QH, Tuo X, Cai X, Chen Z, Li Y, Huang T, Duan X and Duan Y: Integrated analysis of genome-wide copy number alterations and gene expression profiling of lung cancer in Xuanwei, China. PLoS One. 12:e01690982017. View Article : Google Scholar : PubMed/NCBI

47 

Yao J, Zhang X, Li J, Zhao D, Gao B, Zhou H, Gao S and Zhang L: Silencing TRIP13 inhibits cell growth and metastasis of hepatocellular carcinoma by activating of TGF-β1/smad3. Cancer Cell Int. 18:2082018. View Article : Google Scholar

48 

Hu F, Zeng W and Liu X: A gene signature of survival prediction for kidney renal cell carcinoma by multi-omic data analysis. Int J Mol Sci. 20:pii: E5720. 2019. View Article : Google Scholar

49 

Liu H and Zhao H: Prognosis related miRNAs, DNA methylation, and epigenetic interactions in lung adenocarcinoma. Neoplasma. 66:487–493. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 46 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang M, Sun L, Ru Y, Zhang S, Miao J, Guo P, Lv J, Guo F and Liu B: A risk score system based on DNA methylation levels and a nomogram survival model for lung squamous cell carcinoma. Int J Mol Med 46: 252-264, 2020
APA
Zhang, M., Sun, L., Ru, Y., Zhang, S., Miao, J., Guo, P. ... Liu, B. (2020). A risk score system based on DNA methylation levels and a nomogram survival model for lung squamous cell carcinoma. International Journal of Molecular Medicine, 46, 252-264. https://doi.org/10.3892/ijmm.2020.4590
MLA
Zhang, M., Sun, L., Ru, Y., Zhang, S., Miao, J., Guo, P., Lv, J., Guo, F., Liu, B."A risk score system based on DNA methylation levels and a nomogram survival model for lung squamous cell carcinoma". International Journal of Molecular Medicine 46.1 (2020): 252-264.
Chicago
Zhang, M., Sun, L., Ru, Y., Zhang, S., Miao, J., Guo, P., Lv, J., Guo, F., Liu, B."A risk score system based on DNA methylation levels and a nomogram survival model for lung squamous cell carcinoma". International Journal of Molecular Medicine 46, no. 1 (2020): 252-264. https://doi.org/10.3892/ijmm.2020.4590