Open Access

Integrated bioinformatics analysis revealing independent prognostic long non‑coding RNAs DNAH17‑AS1 and RP11‑400N13.2 and their potential oncogenic roles in colorectal cancer

  • Authors:
    • Wen Zhou
    • Boyu Pan
    • Liren Liu
  • View Affiliations

  • Published online on: August 7, 2019     https://doi.org/10.3892/ol.2019.10730
  • Pages: 3705-3715
  • Copyright: © Zhou 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

The aberrant expression of long non‑coding RNAs (lncRNAs) has been associated with a variety of malignancies, including colorectal cancer (CRC); however, the key lncRNAs associated with patient prognosis and their biological roles in CRC are yet to be determined. The aim of the present study was to determine the key lncRNAs associated with patient prognosis as well as their biological roles in CRC. Therefore, a dataset from The Cancer Genome Atlas containing the lncRNA expression data of 521 CRC and normal colorectal mucosal tissues, as well as the corresponding clinical data, were screened. A total of 1,180 significantly differentially expressed lncRNAs were associated with CRC as determined by t‑tests in edgeR. Kaplan‑Meier analysis revealed that 56 of the 1,180 lncRNAs were associated with overall survival (OS); 7 of the 56 lncRNAs were identified as key lncRNAs associated with the Tumor‑Node‑Metastasis stage of CRC by Kruskal‑Wallis test. Subsequent univariate and multivariate Cox regression analyses of the 7 lncRNAs revealed 2 lncRNAs, DNAH17‑AS1 and RP11‑400N13.2, as potential independent prognostic factors for the OS of patients with CRC. Furthermore, the expression levelsof these 2 lncRNAs were significantly upregulated in CRC compared with those in normal tissues, which suggested that they may serve an oncogenic role in CRC. In addition, networks comprising the 2 lncRNAs and their respective co‑expressed protein‑coding genes (PCGs) were constructed using cor.test in R. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses of these PCGs were conducted; DNAH17‑AS1‑ and RP11‑400N13.2‑associated PCGs were reported to be involved in G‑protein coupling‑related functions. Thus, these independent prognostic lncRNAs and their associated functions identified in the present study may provide novel insight into potential prognostic biomarkers and therapeutic targets for the treatment of CRC.

Introduction

Colon cancer is the most common type of tumor of the gastrointestinal tract, and ranks as the third highest cause of cancer-associated mortality worldwide (1). The etiology and pathogenesis of colon cancer are complex and are associated with various factors, such as diet- and lifestyle-associated genetic and epigenetic changes (2). Recent advances in the treatment of colon cancer have been reported, including surgery combined with chemotherapy, radiofrequency ablation or targeted therapy; however, the rate of postoperative recurrence remain at ~50%, leading to a poor overall survival (OS) for the patients with colon cancer (3). Therefore, there is an urgent need to identify novel biomarkers and potential therapeutic targets for this deadly disease (4).

Long non-coding RNAs (lncRNAs), which are >200 nucleotides in length, have been reported to act as key regulators of various biological processes; the aberrant expression of lncRNAs are associated with several diseases, including cancer (59). Accumulating evidence has suggested that lncRNAs could serve as potential biomarkers for the early diagnosis, prognosis and prediction of metastasis for various types of malignancy (1015).

In recent years, with advances in bioinformatics and interdisciplinary studies involving the development of a series of computational methods and software tools for the analysis of extensive biological data, numerous lncRNAs have been identified to be dysregulated in colon cancer. For instance, by a bioinformatic approach, a recent study classified Linc00659 as a novel oncogenic lncRNA involved in the tumorigenesis of colon cancer by modulating the progression of the cell cycle; downregulation of Linc00659 expression resulted in severe cell cycle arrest and enhanced the apoptosis of colon cancer cells (16). Similarly, based on bioinformatics analysis of The Cancer Genome Atlas (TCGA) and/or the Gene Expression Omnibus datasets, as well as subsequent experimental validation, metastasis-associated lung adenocarcinoma transcript 1 and small nuclear host gene 1 have been recently identified to be oncogenic lncRNAs, which may serve as potential diagnostic and therapeutic targets in colorectal cancer (CRC) (1720). These results suggest the potential clinical value of lncRNAs in CRC; however, the lncRNAs associated with the prognosis and survival of patients, as well as their biological roles, require further investigation.

Therefore, the present study aimed to identify the key lncRNAs associated with their prognostic and biological roles using a comprehensive bioinformatics process. The gene expression datasets downloaded from The Cancer Genome Atlas (TCGA) database, which includes the corresponding survival and Tumor-Node-Metastasis (TNM) stage (21) status of patients with CRC, were utilized to construct a prognostic prediction system.

Materials and methods

TCGA CRC data mining and screening

The level 3 normalized lncRNA expression data of CRC, CRC gene expression data and corresponding clinical data were obtained from the TCGA database (https://cancergenome.nih.gov). The expression profiling platform RNA-seqv2 was used. No further normalizations were applied to the level 3 lncRNA expression profile data. A total of 521 samples were obtained, of which 480 were CRC tissues and 41 were adjacent normal tissues. The lncRNA expression profile of tumor and normal tissues was determined to screen for differentially expressed lncRNAs using edgeR (http://www.bioconductor.org/packages/release/bioc/html/edgeR.html; R software; version 3.4.2; Bell Laboratories) with thresholds of |log2[fold-change (FC)]|>2.0 and adjusted P-value [false discovery rate (FDR)]<0.05. A volcano plot was generated using theplot function in R (https://www.rdocumentation.org/packages/graphics/versions/3.6.0/topics/pl; R software; version 3.4.2; Bell Laboratories).

Survival analysis

Kaplan-Meier analysis followed by a log-rank test was performed to assess the OS between low- and high-lncRNA expression groups using the R package ‘survival’ (https://cran.r-project.org/web/packages/survival/index.html; R software; version 3.4.2; Bell Laboratories). The Kruskal-Wallis test was used to evaluate the association between tumor stage. The staging system of colon cancer using UICC/AJCC (7th edition) (21), and the lncRNAs that were significantly associated with OS. Additionally, univariate and multivariate Cox regression analyses were used to evaluate the association between the expression levels of lncRNAs and the OS of patients with CRC, and to identify independent prognostic values of lncRNAs. P<0.05 was considered to indicate a statistically significant difference.

Analysis of co-expressed protein-coding genes (PCGs)

To determine the association between lncRNAs and co-expressed PCGs, the Pearson correlation coefficients (r) of the lncRNAs and PCGs were calculated using the cor.test function in R. The PCGs with |r|>0.4 and P<0.001 were considered as lncRNA-associated PCGs.

Functional and pathway enrichment analyses

The identified co-expressed PCGs were further investigated using clusterProfiler R package (http://www.bioconductor.org/packages/release/bioc/html/clusterProfiler.html; R software; version 3.4.2; Bell Laboratories), including functional Gene Ontology (GO) (22) and Kyoto Encyclopedia of Genes and Genomes (KEGG) (23) pathway enrichment analyses. P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of significantly differentially expressed lncRNAs in CRC

In a preliminary screening, the data of 480 CRC and 41 adjacent normal colorectal mucosal tissues were extracted from the symbol matrix. A total of 1,180 significant differentially expressed lncRNAs were identified with |logFC|>2 and FDR<0.05, of which 916 were upregulated and 264 were downregulated. A volcano plot of the identified lncRNAs was constructed (Fig. 1). The top 10 upregulated and downregulated lncRNAs are presented in Tables I and II.

Table I.

Top 10 upregulated lncRNAs with significantly different expression between tumor and normal tissues in The Cancer Genome Atlas colon cancer data.

Table I.

Top 10 upregulated lncRNAs with significantly different expression between tumor and normal tissues in The Cancer Genome Atlas colon cancer data.

lncRNAEnsembl_Gene_IDlogFCP-valueFDR
PVT1ENSG000002498592.58 1.70×10−67 4.23×10−65
CCAT1ENSG000002478444.47 1.68×10−59 2.51×10−57
BLACAT1 ENSG000002814065.12 3.88×10−59 5.59×10−57
LINC02163 ENSG000002510267.08 1.19×10−54 1.37×10−52
CRNDE ENSG000002456944.58 1.30×10−52 1.31×10−50
MAFG-AS1 ENSG000002656882.88 7.82×10−51 7.51×10−49
RP5-884M6.1 ENSG000002287426.31 1.04×10−50 9.69×10−49
CASC19 ENSG000002541664.51 3.30×10−50 3.02×10−48
RP5-1120P11.1 ENSG000002376864.41 7.64×10−50 6.65×10−48
AC007128.1 ENSG000002299704.48 2.64×10−48 2.17×10−46

[i] lncRNA, long non-coding RNA; FC, fold-change; FDR, false discovery rate.

Table II.

Top 10 downregulated lncRNAs with significantly different expression between tumor and normal tissues in The Cancer Genome Atlas colon cancer data.

Table II.

Top 10 downregulated lncRNAs with significantly different expression between tumor and normal tissues in The Cancer Genome Atlas colon cancer data.

lncRNA Ensembl_Gene_IDlogFCP-valueFDR
XXbac-B476C20.9 ENSG00000225335−2.72 2.14×10−173 1.60×10−169
CDKN2B-AS1 ENSG00000240498−5.21 9.05×10−140 3.39×10−136
LINC01645 ENSG00000224968−4.60 6.90×10−127 1.72×10−123
PP7080 ENSG00000188242−3.48 3.24×10−124 6.07×10−121
XXyac-YM21GA2.7 ENSG00000214888−5.69 7.06×10−120 1.06×10−116
RP11-1090M7.1 ENSG00000265489−4.48 1.49×10−116 1.85×10−113
RP11-396O20.2 ENSG00000254645−5.01 1.71×10−114 1.83×10−111
AC007182.6 ENSG00000224721−4.67 4.65×10−108 4.35×10−105
AC106869.2 ENSG00000226087−4.18 2.71×10−105 2.25×10−102
LINC00682 ENSG00000245870−4.55 1.18×10−96 8.82×10−94

[i] lncRNA, long non-coding RNA; FC, fold-change; FDR, false discovery rate.

Analysis of significant differentially expressed lncRNAs in CRC samples associated with OS and pathological stages

To investigate the association between lncRNA expression and OS, the expression profile of the 1,180 lncRNAs in tumor samples were determined, of which 56 lncRNAs were associated with OS, as determined by Kaplan-Meier analysis (Table III). The top 10 lncRNAs significantly associated with OS (P<0.05) were RP11-108K3.2, RP11-815M8.1, LINC01836, AC079612.1, LINC01354, RBAKDN, RP11-400N13.2, RP1-142L7.9, AFAP1-AS1 and LINC01655 (Fig. 2).

Table III.

lncRNAs significantly associated with OS.

Table III.

lncRNAs significantly associated with OS.

lncRNA Ensembl_Gene_IDP-value
RP11-108K3.2 ENSG00000259306 1.98646×10−5
RP11-815M8.1 ENSG00000238042 2.78454×10−5
LINC01836 ENSG000002675300.000256940
AC079612.1 ENSG000001967580.000535749
LINC01354 ENSG000002317680.000985613
RBAKDN ENSG000002733130.001143877
RP11-400N13.2 ENSG000002284370.002268513
RP1-142L7.9 ENSG000002706610.003700521
AFAP1-AS1 ENSG000002726200.004000305
LINC01655 ENSG000002279250.004380952
RP11-10A14.5 ENSG000002485380.004693149
RP11-384P7.7 ENSG000002609470.004845978
RP11-434D9.2 ENSG000002498940.005561553
RP11-742B18.1 ENSG000002490010.007363036
CTC-327F10.4 ENSG000002513200.008786437
AC064834.1 ENSG000002240990.00890189
ARHGEF26-AS1 ENSG000002430690.008966797
RP3-380B8.4 ENSG000002330640.009485845
LINC01829 ENSG000002367800.009974597
GAS1RR ENSG000002262370.013908392
RP11-84A19.4 ENSG000002699670.014180465
LINC02043 ENSG000002322330.016339686
LINC00922 ENSG000002617420.016854765
RP11-278L15.2 ENSG000002438850.017325027
RP1-122P22.4 ENSG000002686280.018507105
RP11-366L20.2 ENSG000001973010.019632443
DUXAP8 ENSG000002061950.020361403
CTB-181H17.1 ENSG000002722190.020685373
MIR31HG ENSG000001718890.021365649
AC012531.25 ENSG000002605970.023942649
FOXD3-AS1 ENSG000002307980.024144909
AC007128.1 ENSG000002299700.027145232
DNAH17-AS1 ENSG000002674320.02735923
LINC01833 ENSG000002594390.027941465
RP11-429J17.5 ENSG000002545480.028089082
LL22NC03-N14H11.1 ENSG000002728720.031532618
RP1-79C4.4 ENSG000002718110.031571197
CTB-186G2.4 ENSG000002673750.031679272
RP11-114H23.2 ENSG000002580880.033221364
LINC00484 ENSG000002296940.035515205
RP1-29C18.10 ENSG000002129390.035961207
AC073326.3 ENSG000002285400.036298277
RP11-126H7.4 ENSG000002040490.036449014
CTD-2619J13.13 ENSG000002683070.037145687
LINC01748 ENSG000002264760.038383393
RP11-532F6.3 ENSG000002724630.042708274
RP11-728G15.1 ENSG000002560080.043149989
LINC01060 ENSG000002493780.045508319
KCNQ1OT1 ENSG000002698210.045529181
LINC01996 ENSG000002618630.046591159
ELFN1-AS1 ENSG000002360810.046625182
HOTAIR ENSG000002286300.047908786
LINC00461 ENSG000002455260.04796558
CTD-2600O9.1 ENSG000001871850.048650792
LEF1-AS1 ENSG000002320210.049375626
FLJ16779 ENSG000002756200.049920309

[i] lncRNA, long non-coding RNA.

The association between clinical stages (UICC/AJCC 7th Edition) (21) and the 56 lncRNAs associated with OS was determined via a Kruskal-Wallis test. The results demonstrated that 7 lncRNAs were identified as key lncRNAs associated with the Tumor-Node-Metastasis (TNM) stages of colon cancer, including DNAH17-AS1, RP11-429J17.5, RP11-742B18.1, RP11-400N13.2, LL22NC03-N14H11.1, LINC01836 and HOTAIR (Table IV; Fig. 3). The detailed information of the patients at each TNM stage is presented in Table SI. Notably, these 7 lncRNAs were upregulated in colon cancer tissues compared with adjacent normal tissues, suggesting that they may serve a tumorigenic role in the initiation and progression of CRC.

Table IV.

lncRNAs significantly associated with tumor clinical stage.

Table IV.

lncRNAs significantly associated with tumor clinical stage.

lncRNA Ensembl_Gene_IDP-value
FLJ16779 ENSG000002756200.049920309
DNAH17-AS1 ENSG00000267432 6.42×10−5
RP11-429J17.5 ENSG000002545480.000628536
RP11-742B18.1 ENSG000002490010.00141941
RP11-400N13.2 ENSG000002284370.003179863
LL22NC03-N14H11.1 ENSG000002728720.005376623
LINC01836 ENSG000002675300.011461646
HOTAIR ENSG000002286300.013000989

[i] lncRNA, long non-coding RNA.

Identification of independent prognostic lncRNAs in CRC

In order to detect potential independent prognostic lncRNAs in patients with CRC, univariate and multivariate Cox regression analyses of these 7 lncRNAs associated with TNM stage were performed. The association between the expression levels of lncRNAs and the OS of patients with colon cancer was explored using the R package ‘survival’; 2 lncRNAs, DNAH17-AS1 and RP11-400N13.2, were identified to be independent prognostic factors for OS in patients with CRC (P<0.05; Tables V and VI).

Table V.

Cox regression analyses of the association between DNAH17-AS1 and patient clinicopathological characteristics.

Table V.

Cox regression analyses of the association between DNAH17-AS1 and patient clinicopathological characteristics.

UnivariateMultivariate


CharacteristicP-valueHRHR (lower 0.95)HR (upper 0.95)P-valueHRHR (lower 0.95)HR (upper 0.95)
Age, years 1.54×10−21.0247051.004670061.045139527 8.62×10−51.0432931.0214527411.065600322
Sex 8.11×10−11.0559930.6753209041.651244946 5.15×10−10.8597530.5452339861.355701712
Stage 4.32×10−102.2605381.7498176462.92032165 3.09×10−11.4817530.6937529643.164805106
T 5.85×10−62.8421691.8090616844.465257511 4.32×10−21.7537151.0172446013.023378521
M 6.75×10−104.3482022.7264930716.934496948 3.83×10−11.5847560.5625330314.464537953
N 1.29×10−72.0140391.5530719572.611824311 2.84×10−11.2815190.8137880462.01808327
DNAH17-AS1 1.17×10−356.080874.924770723638.6214937 1.61×10−229.593421.872773849467.6327355

[i] HR, hazard ratio; HR (lower 0.95), hazard ratio (lower 95% CI); HR (upper 0.95), hazard ratio (upper 95%CI); T, tumor; N, node; M, metastasis.

Table VI.

Cox regression analyses of the association between RP11-400N13.2 and patient clinicopathological characteristics.

Table VI.

Cox regression analyses of the association between RP11-400N13.2 and patient clinicopathological characteristics.

UnivariateMultivariate


CharacteristicP-valueHRHR (lower 0.95)HR (upper 0.95)P-valueHRHR (lower 0.95)HR (upper 0.95)
Age, years 1.54×10−21.0247051.004670061.045139527 3.70×10−41.037411.0166471951.058596272
Sex 8.11×10−11.0559930.6753209041.651244946 7.37×10−10.9254320.5884642441.455355124
Stage 4.32×10−102.2605381.7498176462.92032165 2.61×10−11.546230.7225696963.308782317
T 5.85×10−62.8421691.8090616844.465257511 3.02×10−21.8423781.0599487913.202377507
M 6.75×10−104.3482022.7264930716.934496948 3.49×10−11.6287190.5867035774.521408077
N 1.29×10−72.0140391.5530719572.611824311 4.82×10−11.1815260.7416992341.882169148
RP11-400N13.2 2.49×10−41.146291.0655320181.233169628 6.46×10−31.1170751.0315232891.209722556

[i] HR, hazard ratio; HR (lower 0.95), hazard ratio (lower 95% CI); HR (upper 0.95), hazard ratio (upper 95% CI); T, tumor stage; N, node stage; M, metastasis stage.

Analyses of PCGs co-expressed with lncRNAs DNAH17-AS1 and RP11-400N13.2

Analysis of the PCGs co-expressed with DNAH17-AS1 and RP11-400N13.2 was conducted using the cor.test function with thresholds of |r|>0.4 and P<0.001. The results revealed that 1,048 PCGs were co-expressed with DNAH17-AS1 (Fig. 4A). Due to a large number of PCGs in DNAH17-AS1, only the top 100 genes with a lower P-value were presented in Fig. 4A. A total of 126 PCGs co-expressed with RP11-400N13.2 (Fig. 4B). The top 10 significant PCGs that were identified to be co-expressed with the 2 lncRNAs are listed in Table VII.

Table VII.

Co-expression analyses between DNAH17-AS1 and RP11-400N13.2 and the top 10 significant protein-coding genes.

Table VII.

Co-expression analyses between DNAH17-AS1 and RP11-400N13.2 and the top 10 significant protein-coding genes.

A, Genes co-expressed with DNAH17-AS1

Co-expressed gene Ensembl_Gene_IDraP-value
DNAH17 ENSG000001877750.656 2.73×10−65
SSPO ENSG000001975580.564 4.89×10−45
TPTE ENSG000002743910.505 4.31×10−35
DPPA2 ENSG000001635300.473 1.96×10−30
GPR179 ENSG000002773990.473 2.22×10−30
RP11-505K9.4 ENSG000002603000.454 8.07×10−28
CFAP74 ENSG000001426090.453 1.15×10−27
POLG2 ENSG000002565250.451 1.99×10−27
KIF6 ENSG000001646270.449 3.41×10−27
STKLD1 ENSG000001988700.445 9.44×10−27

B, Genes co-expressed with RP11-400N13.2

Co-expressed gene Ensembl_Gene_IDraP-value

RP11-371E8.4 ENSG000002590660.439 5.20×10−26
RP11-507M3.1 ENSG000002760870.438 8.64×10−26
HIST3H3 ENSG000001681480.428 1.26×10−24
RP4-809F18.1 ENSG000002555950.428 1.34×10−24
OPRPN ENSG000001711990.426 2.51×10−24
RP11-385D13.1 ENSG000002515370.423 4.79×10−24
OR52B2 ENSG000002553070.422 5.87×10−24
OR52B6 ENSG000001877470.422 7.24×10−24
OR2AG2 ENSG000001881240.401 1.44×10−21
BDNF ENSG000001766970.397 3.93×10−21

a Pearson's correlation coefficient.

GO and KEGG enrichment analyses of the PCGs co-expressed with DNAH17-AS1 and RP11-400N13.2

To further investigate the potential roles of the two independent prognostic lncRNAs identified in the present study, functional enrichment analyses for their co-expressed PCGs were performed using the clusterProfiler R package. The results indicated that the PCGs co-expressed with DNAH17-AS1 were mainly enriched in ‘G-protein coupled receptor signaling pathway’, ‘detection of chemical stimulus involved in sensory perception of smell’, ‘integral component of membrane’, ‘integral component of plasma membrane’, ‘G-protein coupled receptor activity’ and ‘olfactory receptor activity’. Collectively, these PCGs were associated with G-protein coupling and cell membrane function (Fig. 5A). Additionally, the PCGs co-expressed with RP11-400N13.2 were mainly enriched in ‘G-protein coupled receptor signaling pathway’, ‘G-protein coupled receptor activity’, ‘spermatogenesis’, ‘negative regulation of endopeptidase activity’ and ‘endopeptidase inhibitor activity’, which are involved in G-protein coupling and endopeptidase function (Fig. 5B). Functional enrichment analysis revealed a high level of involvement of the associated PCGs inG-protein coupling, which suggested a crucial biological function of DNAH17-AS1 and RP11-400N13.2. The detailed information of the genes with G-protein-associated functions co-expressed with DNAH17-AS1 and RP11-400N13.2 is presented in Table SII. A total of 3 of these genes, 5′-hydroxytryptamine receptor 6, melanocortin 5 receptor and prokineticin receptor 2, were significantly associated with OS (P<0.05; Fig. S1).

KEGG pathway enrichment analysis of the PCGs co-expressed with the two independent prognostic lncRNAs was performed using clusterProfiler package in R with a threshold of P<0.05. The results revealed that the PCGs co-expressed with DNAH17-AS1 were involved in seven pathways, including ‘olfactory transduction’, ‘neuroactive ligand-receptor interaction’, ‘phototransduction’, ‘nicotine addiction’, ‘cocaine addiction’, ‘collecting duct acid secretion’ and ‘signaling pathways regulating pluripotency of stem cells’ (Fig. 6). This result provided novel insights into the potential associations between these pathways and CRC, which warrant further investigation. Of note, P>0.05 was reported for the enriched pathways of the PCGs co-expressed with RP11-400N13.2.

Discussion

In the present study, in silico analysis revealed 1,180 significantly differentially expressed lncRNAs that were associated with colorectal cancer, of which 56 and 7 genes were significantly associated with OS and TNM stage, respectively. Subsequent univariate and multivariate Cox regression analyses indicated that 2 of the 7 lncRNAs, DNAH17-AS1 and RP11-400N13.2, may be independent prognostic lncRNAs for the OS of patients with colorectal cancer.

To the best of our knowledge, lncRNAs DNAH17-AS1 and RP11-400N13.2 have not been previously associated with colon cancer; however, a missense variant p.R3953Y of DNAH17, the related protein of DNAH17-AS1, was reported in undifferentiated embryonal sarcoma of the liver in a child (24). Additionally, p.R3953 of DNAH17 exhibited a high level of conservation among a variety of species, suggesting that this allele may be an important locus associated with protein function (24). A recent study revealed the mutational profile and a distinct mutation signature of T:A>A:T transversion in early-stage hepatocellular carcinoma (HCC) with hepatitis B virus (HBV) infection; thus, as a key gene of the mutational profile, DNAH17 was proposed to serve an important role in the HBV-mediated transformation of liver cells (25). Additionally, the hypomethylation status of DNAH17 has been reported in HCC, which is associated with several clinical characteristics and may serve as a potential biomarker of tumor thrombosis in patients with HCC (26). In the present study, the lncRNA expression level of DNAH17-AS1 in CRC samples was analyzed and compared with that in normal samples; however, the expression level and the methylation status of DNAH17 were not analyzed. Although the expression level of DNAH17, as well as its methylation status in CRC samples, may be informative to determine the role of DNAH17 in CRC, this was beyond the scope of the present study. Therefore, relevant studies will be performed in the future.

A limited number of studies have investigated RP11-400N13.2; however, other RP11 family members have been frequently reported to be dysregulated in CRC. RP11-708H21.4, an RP11 family lncRNA located in the 17q21 gene desert region, was proposed to serve a suppressive role in the tumorigenesis of colorectal cancer and act as a novel powerful diagnostic biomarker, as well as a therapeutic target for the treatment of CRC (27). The expression levels of RP11-462C24.1, another member of the RP11 family, were determined to be significantly correlated with distant metastasis in patients with CRC, and may serve as a potential prognostic marker for such patients (28). Additionally, the dysregulation of RP11 family members has been reported to be involved in other types of cancer. For instance, a recent study revealed that overexpression of lncRNA RP11-190D6.2 inhibited the proliferation, migration and invasion of epithelial ovarian cancer (EOC) cells and may be considered a novel biomarker and therapeutic target for EOC (29). Furthermore, lncRNA RP11-436H11.5 was identified to function as a competing endogenous RNA to promote the proliferation and invasion of renal cell carcinoma (RCC) cells, which suggests that RP11-436H11.5 may be a potential therapeutic target to suppress RCC tumorigenesis (30). Collectively, the RP11 family of lncRNAs serve important roles in carcinogenesis and may be used as potential diagnostic and prognostic biomarkers for various types of cancer.

Following the identification of two independent prognostic lncRNAs in colorectal cancer, the co-expressed PCGs were analyzed, and stepwise GO and KEGG enrichment analyses were conducted to determine the potential biological functions of these lncRNAs associated with CRC and the signaling pathways involved. The results of the functional enrichment analysis of DNAH17-AS1 and RP11-400N13.2 differed; however, these lncRNAs were determined to possess similar G-protein coupling-associated functions. G-protein coupled receptors have been previously reported to be associated with CRC tumorigenesis (3134). For example, the G-protein coupled receptor GPR55 may promote tumor progression by acting as an pro-oncogenic factor in CRC (31). In addition, GPR55 has been proposed to be involved in the migration of CRC cells and may serve as a potential target for the prevention of metastasis (32). On the contrary, orexin receptor type 1 and cholecystokinin A receptor, which belong to family A of the G-protein coupled receptors, serve opposing roles in the regulation of HT-29 CRC cell migration, but have also been reported to be involved in the pathogenesis of CRC metastasis (33). Furthermore, a recent study revealed that GPR109A, a G-protein coupled receptor for short-chain fatty acids, was silenced in CRC cells (34). In addition, the host immune system may employ interferon γ to counteract methylation-mediated silencing of GPR109A as a mechanism to suppress tumor development (34). Therefore, G-protein coupled receptors maybe associated with the carcinogenesis and metastasis of CRC; the roles of DNAH17-AS1 and RP11-400N13.2 in CRC, which may be mediated by these receptors, require further investigation.

In the present study, DNAH17-AS1 and RP11-400N13.2 were identified as potential independent prognostic lncRNAs for OS in patients with CRC. Further bioinformatics analyses revealed that these 2 lncRNAs may serve a pro-oncogenic role in CRC via G-protein coupling-related functions. Therefore, DNAH17-AS1 and RP11-400N13.2 may serve as prognostic biomarkers for CRC in the future. The detailed methodology of the present study is presented in Fig. S2.

Supplementary Material

Supporting Data
Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

This study was funded by the National Natural Science Foundation of China (grant no. 81572416), the National Key Technologies R&D Program of China (grant no. 2016YFC1303200), and the Tianjin Medical University Cancer Institute and Hospital Cancer Translational Medicine Seed Funds (grant no. 1701-1).

Availability of data and materials

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

Authors' contributions

LL designed and supervised the study and finalized the manuscript. WZ and BP made substantial contributions to the study design, performed the bioinformatics analysis and drafted the manuscript. 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.

Glossary

Abbreviations

Abbreviations:

lncRNAs

long non-coding RNAs

CRC

colorectal cancer

OS

overall survival

PCGs

protein-coding genes

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Ghareeb AE, Moawed FSM, Ghareeb DA and Kandil EI: Potential prophylactic effect of berberine against rat colon carcinoma induce by 1,2-dimethyl hydrazine. Asian Pac J Cancer Prev. 19:1685–1690. 2018.PubMed/NCBI

3 

Sanoff HK, Sargent DJ, Campbell ME, Morton RF, Fuchs CS, Ramanathan RK, Williamson SK, Findlay BP, Pitot HC and Goldberg RM: Five-year data and prognostic factor analysis of oxaliplatin and irinotecan combinations for advanced colorectal cancer: N9741. J Clin Oncol. 26:5721–5727. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Dong Z, Zheng L, Liu W and Wang C: Association of mRNA expression of TP53 and the TP53 codon 72 Arg/Pro gene polymorphism with colorectal cancer risk in Asian population: A bioinformatics analysis and meta-analysis. Cancer Manag Res. 10:1341–1349. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Xue W, Li J, Wang F, Han P, Liu Y and Cui B: A long non-coding RNA expression signature to predict survival of patients with colon adenocarcinoma. Oncotarget. 8:101298–101308. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Dykes IM and Emanueli C: Transcriptional and posttranscriptional gene regulation by long non-coding RNA. Genomics Proteomics Bioinformatics. 15:177–186. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, et al: The landscape of long non-coding RNAs in the human transcriptome. Nat Genet. 47:199–208. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Prensner JR and Chinnaiyan AM: The emergence of lncRNAs in cancer biology. Cancer Discov. 1:391–407. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Zhou M, Wang X, Li J, Hao D, Wang Z, Shi H, Han L, Zhou H and Sun J: Prioritizing candidate disease-related long non-coding RNAs by walking on the heterogeneous lncRNA and disease network. Mol Biosyst. 11:760–769. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Lou Y, Jiang H, Cui Z, Wang X, Wang L and Han Y: Gene microarray analysis of lncRNA and mRNA expression profiles in patients with high-grade ovarian serous cancer. Int J Mol Med. 42:91–104. 2018.PubMed/NCBI

11 

Jin B, Jin H, Wu HB, Xu JJ and Li B: Long non-coding RNA SNHG15 promotes CDK14 expression via miR-486 to accelerate non-small cell lung cancer cells progression and metastasis. J Cell Physiol. 233:7164–7172. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Li S, Zhou J, Wang Z, Wang P, Gao X and Wang Y: Long noncoding RNA GAS5 suppresses triple negative breast cancer progression through inhibition of proliferation and invasion by competitively binding miR-196a-5p. Biomed Pharmacother. 104:451–457. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Liu Z, Yan Y, Cao S and Chen Y: Long non-coding RNA SNHG14 contributes to gastric cancer development through targeting miR-145/SOX9 axis. J Cell Biochem. 119:6905–6913. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Fan H, Lv P, Mu T, Zhao X, Liu Y, Feng Y, Lv J, Liu M and Tang H: lncRNA n335586/miR-924/CKMT1A axis contributes to cell migration and invasion in hepatocellular carcinoma cells. Cancer Lett. 429:89–99. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Ye XT, Huang H, Huang WP and Hu WL: lncRNA THOR promotes human renal cell carcinoma cell growth. Biochem Biophys Res Commun. 501:661–667. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Tsai KW, Lo YH, Liu H, Yeh CY, Chen YZ, Hsu CW, Chen WS and Wang JH: Linc00659, a long noncoding RNA, acts as novel oncogene in regulating cancer cell growth in colorectal cancer. Mol Cancer. 17:722018. View Article : Google Scholar : PubMed/NCBI

17 

Wu Q, Meng WY, Jie Y and Zhao H: lncRNA MALAT1 induces colon cancer development by regulating miR-129-5p/HMGB1 axis. J Cell Physiol. 233:6750–6757. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Yang H, Wang S, Kang YJ, Wang C, Xu Y, Zhang Y and Jiang Z: Long non-coding RNA SNHG1 predicts a poor prognosis and promotes colon cancer tumorigenesis. Oncol Rep. 40:261–271. 2018.PubMed/NCBI

19 

Xu Y, Zhang X, Hu X, Zhou W, Zhang P, Zhang J, Yang S and Liu Y: The effects of lncRNA MALAT1 on proliferation, invasion and migration in colorectal cancer through regulating SOX9. Mol Med. 24:522018. View Article : Google Scholar : PubMed/NCBI

20 

Xu M, Chen X, Lin K, Zeng K, Liu X, Pan B, Xu X, Xu T, Hu X, Sun L, et al: The long noncoding RNA SNHG1 regulates colorectal cancer cell growth through interactions with EZH2 and miR-154-5p. Mol Cancer. 17:1412018. View Article : Google Scholar : PubMed/NCBI

21 

Edge SB, Byrd DR, Compton CC, Fritz AG Greene, F and Trotti A: AJCC Cancer Staging Manual. 7th. Springer-Verlag; New York, NY: 2010

22 

Gene Ontology Consortium: The gene ontology (GO) project in 2006. Nucleic Acids Res. 34:D322–D326. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Altermann E and Klaenhammer TR: Pathwayvoyager: Pathway mapping using the Kyoto encyclopedia of genes and genomes (KEGG) database. BMC Genomics. 6:602005. View Article : Google Scholar : PubMed/NCBI

24 

Kim JH, Sio CA, Park H, Kim H, Shin HD and Jung K: Undifferentiated embryonal sarcoma of the liver in a child: A whole exome sequencing analysis. Dig Liver Dis. 49:944–946. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Zhan H, Jiang J, Sun Q, Ke A, Hu J, Hu Z, Zhu K, Luo C, Ren N, Fan J, et al: Whole-exome sequencing-based mutational profiling of hepatitis B virus-related early-stage hepatocellular carcinoma. Gastroenterol Res Pract 2017. 20293152017.

26 

Fan X, Guo H, Dai B, He L, Zhou D and Lin H: The association between methylation patterns of DNAH17 and clinicopathological factors in hepatocellular carcinoma. Cancer Med. 8:337–350. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Sun L, Jiang C, Xu C, Xue H, Zhou H, Gu L, Liu Y and Xu Q: Down-regulation of long non-coding RNA RP11-708H21.4 is associated with poor prognosis of colorectal cancer and promotes tumorigenesis through regulating AKT/mTOR pathway. Oncotarget. 8:27929–27942. 2017.PubMed/NCBI

28 

Shi D, Zheng H, Zhuo C, Peng J, Li D, Xu Y, Li X, Cai G and Cai S: Low expression of novel lncRNA RP11-462C24.1 suggests a biomarker of poor prognosis in colorectal cancer. Med Oncol. 31:312014. View Article : Google Scholar : PubMed/NCBI

29 

Tong W, Yang L, Yu Q, Yao J and He A: A new tumor suppressor lncRNA RP11-190D6.2 inhibits the proliferation, migration and invasion of epithelial ovarian cancer cells. Onco Targets Ther. 10:1227–1235. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Wang KF, Jin W, Song Y and Fei X: lncRNA RP11-436H11.5, functioning as a competitive endogenous RNA, upregulates BCL-W expression by sponging miR-335-5p and promotes proliferation and invasion in renal cell carcinoma. Mol Cancer. 16:1662017. View Article : Google Scholar : PubMed/NCBI

31 

Hasenoehrl C, Feuersinger D, Sturm EM, Bärnthaler T, Heitzer E, Graf R, Grill M, Pichler M, Beck S, Butcher L, et al: G protein-coupled receptor GPR55 promotes colorectal cancer and has opposing effects to cannabinoid receptor 1. Int J Cancer. 142:121–132. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Kargl J, Andersen L, Hasenöhrl C, Feuersinger D, Stančić A, Fauland A, Magnes C, El-Heliebi A, Lax S, Uranitsch S, et al: GPR55 promotes migration and adhesion of colon cancer cells indicating a role in metastasis. Br J Pharmacol. 173:142–154. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Bai B, Chen X, Zhang R, Wang X, Jiang Y, Li D, Wang Z and Chen J: Dual-agonist occupancy of orexin receptor 1 and cholecystokinin A receptor heterodimers decreases G-protein-dependent signaling and migration in the human colon cancer cell line HT-29. Biochim Biophys Acta Mol Cell Res 1864. 1153–1164. 2017. View Article : Google Scholar

34 

Bardhan K, Paschall AV, Yang D, Chen MR, Simon PS, Bhutia YD, Martin PM, Thangaraju M, Browning DD, Ganapathy V, et al: IFNγ Induces DNA methylation-silenced GPR109A expression via pSTAT1/p300 and H3K18 acetylation in colon cancer. Cancer Immunol Res. 3:795–805. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 18 Issue 4

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
Zhou W, Pan B and Liu L: Integrated bioinformatics analysis revealing independent prognostic long non‑coding RNAs DNAH17‑AS1 and RP11‑400N13.2 and their potential oncogenic roles in colorectal cancer. Oncol Lett 18: 3705-3715, 2019
APA
Zhou, W., Pan, B., & Liu, L. (2019). Integrated bioinformatics analysis revealing independent prognostic long non‑coding RNAs DNAH17‑AS1 and RP11‑400N13.2 and their potential oncogenic roles in colorectal cancer. Oncology Letters, 18, 3705-3715. https://doi.org/10.3892/ol.2019.10730
MLA
Zhou, W., Pan, B., Liu, L."Integrated bioinformatics analysis revealing independent prognostic long non‑coding RNAs DNAH17‑AS1 and RP11‑400N13.2 and their potential oncogenic roles in colorectal cancer". Oncology Letters 18.4 (2019): 3705-3715.
Chicago
Zhou, W., Pan, B., Liu, L."Integrated bioinformatics analysis revealing independent prognostic long non‑coding RNAs DNAH17‑AS1 and RP11‑400N13.2 and their potential oncogenic roles in colorectal cancer". Oncology Letters 18, no. 4 (2019): 3705-3715. https://doi.org/10.3892/ol.2019.10730