Differential expression profiles of long non-coding RNAs reveal potential biomarkers for identification of human gastric cancer

  • Authors:
    • Chengyun Li
    • Geyu Liang
    • Wenzhuo Yao
    • Jing Sui
    • Xian Shen
    • Yanqiu Zhang
    • Shumei Ma
    • Yancheng Ye
    • Zhiyi Zhang
    • Wenhua Zhang
    • Lihong Yin
    • Yuepu Pu
  • View Affiliations

  • Published online on: December 29, 2015     https://doi.org/10.3892/or.2015.4531
  • Pages: 1529-1540
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) is one of the most lethal malignancies worldwide. To reduce its high mortality, sensitive and specific biomarkers for early detection are urgently needed. Recent studies have reported that tumor-specific long non-coding RNAs (lncRNAs) seem to be potential biomarkers for the early diagnosis and treatment of cancer. In the present study, lncRNA and mRNA expression profiling of GC specimens and their paired adjacent non-cancerous tissues was performed. Differentially expressed lncRNAs and mRNAs were identified through microarray analysis. The function of differential mRNA was determined by gene ontology and pathway analysis and the functions of lncRNAs were studied by constructing a co-expression network to find the relationships with corresponding mRNAs. We connected the co-expression network, mRNA functions, and the results of the microarray profile differential expression and selected 14 significantly differentially expressed key lncRNAs and 21 key mRNAs. Quantitative RT-PCR (qRT-PCR) was conducted to verify these key RNAs in 50 newly diagnosed GC patients. The data showed that RP5-919F19, CTD-2541M15 and UCA1 was significantly higher expressed. AP000459, LOC101928316, RP11-167N4 and LINC01071 expression was significantly lower in 30 advanced GC tumor tissues than adjacent non-tumor tissues P<0.05. Then, we further validated the above significant differential expression candidate lncRNAs in 20 early stage GC patients. Results showed that CTD-2541M15 and UCA1 were significantly higher expressed, AP000459, LINC01071 and MEG3 expression was significantly lower in 20 early stage GC patient tumor tissues than adjacent non-tumor tissues (P<0.05). In addition, expression of these lncRNAs shows gradual upward trend from early stage GC to advanced GC. Furthermore, conditional logistic regression analysis revealed the aberrant expression of CTD-2541M15, UCA1 and MEG3 closely linked with GC. There is a set of differentially expressed lncRNAs in GC which may be associated with the progression and development of GC. The differential expression profiles of lncRNAs in GC may be promising biomarkers for the early detection and early screening of high‑risk populations.

Introduction

Each year ~990,000 people are diagnosed with gastric cancer (GC) and ~738,000 die from this disease worldwide, making GC the fourth most common cancer and the second most common cause of cancer death (1). The highest incidence rates are observed in South America, East Europe, and East Asia (particularly in Korea, Mongolia, Japan and China) (2). Detection of GC in the early stage is important to reduce mortality. Many Japanese series have consistently reported early diagnosis and treatment of GC, with a 5-year survival rate >90%. The key factor for preventing GC is early diagnosis, as confirmed by the good results obtained in surgical series in which early cases are frequent (3). Recent developments in the field of gastrointestinal (GI) endoscopy have been remarkable (4). However, endoscopic biopsy and pathological morphological observation cannot find all precancerous lesions and early GC. To improve this situation, there is an urgent need for novel and reliable biomarkers for early diagnosis of GC.

In recent years, several studies have confirmed that long non-coding RNAs (lncRNAs) have emerged as essential regulators in almost all aspects of biology (5). They are a group of RNA transcripts that are >200 nucleotides in length and lack significant open reading frames (6). Accumulating evidence suggests that lncRNAs play an important role in tumorigenesis (7). lncRNAs may function as oncogenes or tumor suppressors by altering the chromatin structure or by regulating the transcription of protein-coding genes (8). lncRNA is the functional end-product, and the level of lncRNA expression correlates directly with the level of the active molecule. Thus, the use of lncRNAs in diagnostics has intrinsic advantages over the use of protein-coding RNAs. In addition, lncRNAs show greater tissue specificity compared to protein-coding mRNAs and miRNAs, making them attractive in the search for novel diagnostic and prognostic cancer biomarkers (9). It is known that gastric adenocarcinoma accounts for 95% of gastric malignancies. However, there is little information to clarify the relationship between lncRNAs, mRNAs and validation in advanced and early stage GC (10). Therefore, further study of endoscopic biopsy of GC tissues may help to establish inner relationships between lncRNA, mRNA and GC.

In the present study, differential expression profiles of lncRNAs and mRNAs were detected from advanced GC tissues and adjacent non-tumor tissues by microarray analysis. We investigated the targeted and regulated genes of lncRNAs through construction of a co-expression network and Gene Ontology (GO) analysis. Then, we used real-time PCR validation of microarray differential expression lncRNAs and mRNAs in tumor tissues and adjacent non-tumor tissues from 30 newly diagnosed advanced GC patients and 20 newly diagnosed early stage GC patients.

Materials and methods

Specimen collection

The procedures used in the present study were approved by the Wuwei Tumor Hospital of Gansu (Wuwei, China) and conformed to the Helsinki Declaration, and current legislation. A total of 10 human advanced GC specimens and their paired adjacent non-cancerous tissue specimens were obtained for lncRNA and mRNA microarray analysis from the Wuwei Tumor Hospital of Gansu (Table I). In addition, 30 advanced GC patients and 20 newly diagnosed early stage GC patients were recruited from the Wuwei Tumor Hospital of Gansu, aged 45–70 years, for quantitative RT-PCR analysis between 2014–2015. All of these patients were assigned a diagnosis of GC based on histopathology and clinical history. Clinical information that was recorded for each specimen included age, tumor grade, cancer stage, tissue dimensions and date of resection. The pathologist assessed the tumor content by microscopic examination of cases where the percentage of tumor tissue was estimated to be ≥80%. None of the patients had received preoperative chemoradiation. Adjacent non-cancerous tissues were located ≥5 cm from the tumor edge. Tissue samples were immersed in RNAlater (Ambion, Inc., Austin, TX, USA) and stored at −80°C until use.

Table I

Clinical characteristics of the patients for lncRNA and mRNA microarray analysis.

Table I

Clinical characteristics of the patients for lncRNA and mRNA microarray analysis.

Histopathology of gastric cancers patientsAge (years)GenderTumor size (cm)TNM stages
Ulcer type ductal carcinoma57Male2.2×0.7T3N2M1
Ulcer type moderately differentiated adenocarcinoma60Female5×3.5T3N2M0
Ulcer type moderately differentiated adenocarcinoma49Male4×7T4aN3M1
Ulcer type moderately differentiated adenocarcinoma62Female12×8T4N3M0
Ulcer type moderately differentiated adenocarcinoma52Male8×4T3N2M0
Ulcer type ductal carcinoma58Male6×4.5T3N1M0
Infiltrating type moderately differentiated adenocarcinoma54Male7×9T4aN3aM1
Ulcer type moderately differentiated adenocarcinoma65Male2.5×1.6T4aN1M0
Mushroom umbrella poorly differentiated adenocarcinoma68Female6×3T3N2M0
Ulcer type low differentiated adenocarcinoma60Male10×8.5T4bN0M0
Isolation of RNA

Total RNA was isolated from 10 advanced gastric adenocarcinoma specimens and their paired adjacent non-cancerous tissues using TRIzol reagent (Invitrogen, Carlsbad, CA, USA). The quality of RNA was measured by Agilent Bioanalyzer, which produces an RNA integrity number (RIN) between 1 and 10, with 10 being the highest quality samples, showing the least degradation. The RIN estimates sample integrity using gel electrophoresis and analysis of the ratios of 28S to 18S ribosomal bands.

lncRNA and mRNA microarray analysis

Three pairs of RNA samples were obtained from 10 patients by pooling RNA from two, four and four patients within one group as 'one sample'. Thus, we generated three pairs of pooled RNA samples from GC specimens and their paired adjacent non-cancerous tissues from 10 patients. These three pairs of RNA samples were subjected to microarray analysis. RiboArray Custom Array 1*90K combined microarray was used, which could detect 32,987 lncRNAs from authoritative data sources, including NCBI RefSeq, H-invDB, UCSC, LncRNAdb, John Rinn and GENCODE LncRNA. In addition, the microarray also included Human Gene Expression, which could detect 65,535 mRNAs. Signals were normalized using the median center tool for genes. ANOVA was used to compare the differentially expressed lncRNAs and mRNAs.

Differential expression of mRNA function

Differentially expressed mRNAs were entered into the Database for Annotation, Visualization and Integrated Discovery (DAVID), which utilized GO to identify the molecular function represented in the gene profile and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to analyze the potential functions of these genes in the pathways (11).

Bioinformatics analysis of co-expression of lncRNAs and mRNAs

According to the relationship among miRNA, lncRNA and mRNA, the post-transcriptional regulation of mRNA transcripts bound by single-stranded miRNAs is basically established. In the present study we built lncRNA-mRNA co-expression network which is based on the theory that lncRNA can regulate miRNA abundance by sequestering and binding them, acting as so-called miRNA sponges. Firstly, the differentially expressed lncRNAs and mRNAs were selected from GC specimens and their paired adjacent non-cancerous tissues. Standard selection criteria to identify differentially expressed lncRNAs and mRNAs were established at P<0.05 and fold change >2. Then, lncRNA and mRNA co-expression network was constructed based on the correlation between the differentially expressed lncRNAs and mRNAs. The correlation of expression profiles between biological replicates and group conditions was demonstrated by unsupervised hierarchical cluster analysis (Figs. 1 and 2).

Quantitative real-time PCR validation

To confirm the microarray results, we performed qRT-PCR analysis on larger samples, including 30 newly diagnosed advanced GC and 20 early stage GC patients. Early stage GC patients were all diagnosed as high level intraepithelial neoplasia by pathological morphological observation. GC specimens and their paired adjacent non-cancerous tissues were collected and total RNA was isolated, then reverse-transcribed using AMV reverse transcriptase (Promega, Madison, WI, USA). Real-time PCR was done with GoTaq® qPCR Master Mix (Promega) on StepOnePlus™ PCR System (Applied Biosystems, Waltham, MA, USA). The relative fold change was calculated using the 2−ΔΔCt method (12), where ΔCt = (CtRNAs − CtGAPDH) and ΔΔCt = ΔCttumor tissues − ΔCtadjacent non-tumor tissues. GAPDH was chosen as the endogenous standard. The threshold cycle indicated the fractional cycle number at which the amount of amplified target reached a fixed threshold and the Ct value was negatively correlated with copy numbers. In addition, conditional logistic regression analysis was used to evaluate the association between differentially expressed RNAs and the risk of GC (13).

Statistical analysis

All the results were expressed as mean ± SD. Statistical analysis was carried out with the Student's t-test for comparison of two groups in microarray analysis and ANOVA for multiple comparisons. In both cases, differences with P<0.05 were considered to indicate a statistically significant result. The statistical significance of microarray analysis results was analyzed by fold change and Student's t-test. False discovery rate was calculated to correct the P-value. We used fold change to screen differentially expressed LncRNAs and mRNAs, the threshold values were (fold change ≥2.0 or ≤0.50) and (P<0.05).

Results

Quality of microarray sample RNAs

Microarray sample RNAs were assessed by electrophoresis on a denaturing agarose gel. Total RNA run on a denaturing gel has sharp 28S and 18S rRNA bands, and the 28S rRNA band should be approximately twice as intense as the 18S rRNA band. This intensity ratio was observed for the RNA in the present study, indicating that the RNA was intact. The concentration of RNAs (OD260), protein contamination of RNAs (ratio OD260/OD280), and organic compound contamination of RNAs (ratio OD260/OD230) were measured with the NanoDrop ND-1000. All samples had OD260/OD280 ratios of total RNA higher than 1.8, indicating adequate RNA concentration.

Overview of lncRNA and mRNA expression profiles

The lncRNA expression levels of GC tissues and their paired adjacent non-cancerous tissues were compared. We found that 427 upregulated and 619 downregulated lncRNAs were significantly differentially expressed (fold change ≥2.0). Cluster analysis was used to arrange specimens into groups according to their expression levels (Fig. 2). The top 40 differentially expressed lncRNAs are listed (Fig. 3). RP11-706O15 (fold change, 68.16) was the most upregulated lncRNA, and MID1 (fold change, 140.85) was the most downregulated. There were more downregulated than upregulated lncRNAs according to the microarray data. In addition, in the GC tissues and their paired adjacent non-cancerous samples, mRNA expression profiling data showed that 647 upregulated and 2,349 downregulated mRNAs were significantly differentially expressed (fold change ≥2.0). Hierarchical cluster analysis was used to arrange specimens into groups according to their expression levels (Fig. 2). The top 40 differentially expressed mRNAs are listed (Fig. 4). ZNF737 (fold change, 81.73) was the most upregulated mRNA, and MGME1 (fold change, 227.27) was the most downregulated. We found that there were more downregulated than upregulated mRNAs in this microarray analysis.

Function analysis of differentially expressed genes

Differential expression of mRNAs in GC tissues and adjacent non-cancerous tissues was analyzed by related (GO) analysis performed by the DAVID functional annotation chart. We analyzed the enrichment of these differentially expressed mRNAs. Enrichment provides a measure of the significance of the function, and as the enrichment increases, the corresponding function is more specific, which helps us to identify GO with a more definitive functional description (14). The results showed that the highest enriched GOs targeted by upregulated transcripts were DNA-dependent transcription (GO:0006351), proteolysis (GO:0006508) and mitotic cell cycle (GO:0000278). The highest enriched GOs targeted by downregulated transcripts were DNA-dependent transcription (GO:0006351), transmembrane transport (GO:0055085) and regulation of DNA-dependent transcription (GO:0006355) (Fig. 5).

Pathway analysis indicated that 30 pathways corresponded to upregulated transcripts and that the most enriched network was small cell lung cancer, composed of 86 targeted genes. Moreover, pathway analysis also showed that 37 pathways corresponded to downregulated transcripts and that the most enriched network was metabolic pathways, composed of 1,193 targeted genes. Among these pathways, the gene category 'mTOR signaling pathway', is involved in the carcinogenesis of gastric cardia adenocarcinoma and gastric adenocarcinoma (15). The gene category 'gastric acid secretion' has been investigated as a cause of adenocarcinoma at the gastroesophageal junction and the distal esophagus (16). The gene category 'pathways in cancer' is involved in the development of gastric cancer (17) (Fig. 6).

Construction of the lncRNA-mRNA co-expression network

We used a coding and non-coding gene co-expression (CNC) network to evaluate the interactions among lncRNAs and identify the core regulatory lncRNAs in the network. The principle of co-expression network studies is that if the expression pattern of lncRNAs and mRNAs shows an identical or opposite curve to the other, there is likely to be some interaction between the two genes. By bioinformatics analysis, the co-expression network in GC and their paired adjacent non-tumor tissues was constructed, and by comparing the differences between them, potential core genes could be identified (18). Based on our previous results, we built CNC networks among the differentially expressed lncRNAs and mRNAs in GC specimens and their paired adjacent non-cancerous tissue specimens. The results showed network connections between 184 lncRNAs and 164 mRNAs. We combined the co-expression network, mRNAs function and microarray profile differential expression to select the key lncRNAs. We found that 14 lncRNAs and 21 mRNAs were strongly associated (Table II and Fig. 7). Most of the genes mentioned here are reported to be linked to GC.

Table II

Key lncRNAs and mRNAs screened by co-expression network and microarray profile comprehensive analysis.

Table II

Key lncRNAs and mRNAs screened by co-expression network and microarray profile comprehensive analysis.

Name (LncRNAs)Transcript-IDRegulationDegreeFold-changeRelated mRNAs
RP5-919F19URS0000515CACUpregulation736.924COL5A1, EIF4E, LAMA4, BCL2L1, RPS6KA5, PIAS2, NLGN1, CD99, SDC2
RP11-54O7URS00005B803EUpregulation732.520
RP11-20I23URS00002B7786Upregulation732.431
CTD-2541M15URS0000359EF8Upregulation328.039
AC010731 ENST00000543490Upregulation525.333
UCA1NR_015379Upregulation511.510
AP000459URS000048CBEDDownregulation8−23.256
LOC101928316XR_428890Downregulation6−22.727COL11A2, LAMC2, PLCB2, LRP2, PLCB2, MYLK2, ADH6, GOT1, TYR, CYP3A5, UGT2B10, SV2A
RP11-167N4 ENST00000537019Downregulation12−22.727
RP11-111K18URS00002FCA1ADownregulation10−18.519
LINC01071NR_104174Downregulation5−18.519
TTC28-AS1 ENST00000430525Downregulation7−17.857
MTOR-AS1NR_046600Downregulation8−16.667
MEG3NR_046473Downregulation9−2.190
Validation of key lncRNAs and mRNAs by quantitative RT-PCR in advanced GC

To confirm the reliability and validity of the microarray data, we selected 14 differentially expressed key lncRNAs (RP5-919F19, RP11-54O7, RP11-20I23, CTD-2541M15, AC010731, UCA1, AP000459, LOC101928316, RP11-167N4, LINC01071, RP11-111K18, TTC28-AS1, MTOR-AS1 and MEG3) and randomly selected four differentially expressed mRNAs (COL5A1, BCL2L1, COL11A2 and PLCB2), and analyzed their actual expression levels in 30 advanced GC samples and paired adjacent non-tumor tissue samples with qRT-PCR. The relative expression levels of lncRNAs and mRNAs were given as ratios of RNA to GAPDH transcript levels in the same RNA sample. We applied the paired t-test to evaluate the difference in gene expression between the tumor tissues and their adjacent non-tumor tissues. Results showed that, RP5-919F19, CTD-2541M15 and UCA1 expression was significantly higher in carcinoma than in adjacent non-tumor tissues (P<0.05) (Table III; Figs. 8 and 9). AP000459, LOC101928316, RP11-167N4 and LINC01071 expression was significantly lower in carcinoma than in adjacent non-tumor tissues (P<0.05) (Figs. 8 and 9). Expression levels of these 14 lncRNAs and mRNAs were approximately consistent with the microarray results (Table III; Figs. 10 and 11).

Table III

Relative expression of lncRNAs and mRNAs in 30 pairs of advanced GC tumor and non-tumor tissues.

Table III

Relative expression of lncRNAs and mRNAs in 30 pairs of advanced GC tumor and non-tumor tissues.

Gene symbolTypeGroupMean ± SD of ΔCtΔΔCta (mean ± SD) 2−ΔΔCtP-valuet-value
RP5-919F19LncRNATumor tissues
Adjacent non-tumor tissues
11.779±2.693
12.706±2.529
−0.927±2.2525.499 0.046b2.098
RP11-54O7LncRNATumor tissues
Adjacent non-tumor tissues
10.778±4.415
10.441±3.892
0.337±3.590−0.7070.611−0.514
RP11-20I23LncRNATumor tissues
Adjacent non-tumor tissues
9.671±4.178
9.674±4.287
−0.002±2.2580.5080.9950.006
CTD-2541M15LncRNATumor tissues
Adjacent non-tumor tissues
7.773±2.224
8.774±1.816
−1.000±1.9804.174 0.011b2.719
AC010731LncRNATumor tissues
Adjacent non-tumor tissues
12.468±3.943
12.947±3.598
−0.478±3.66119.7870.5030.687
UCA1LncRNATumor tissues
Adjacent non-tumor tissues
11.097±3.666
13.198±3.619
−2.100±2.77419.168 0.001b3.862
AP000459LncRNATumor tissues
Adjacent non-tumor tissues
16.231±4.497
14.331±4.256
1.900±3.335−15.215 0.014b2.672
LOC101928316LncRNATumor tissues
Adjacent non-tumor tissues
11.307±3.017
9.907±3.176
1.400±2.473−5.237 0.008b2.889
RP11-167N4LncRNATumor tissues
Adjacent non-tumor tissues
17.467±2.557
15.485±2.800
−0.184±3.284−8.343 0.027b0.978
LINC01071LncRNATumor tissues
Adjacent non-tumor tissues
15.928±3.744
15.038±5.068
0.747±3.638−17.388 0.029b1.085
RP11-111K18LncRNATumor tissues
Adjacent non-tumor tissues
9.045±2.270
8.491±2.653
0.554±1.839−1.5530.1371.536
TTC28-AS1LncRNATumor tissues
Adjacent non-tumor tissues
9.373±2.853
8.988±2.714
0.385±2.9303.3540.5090.671
MTOR-AS1LncRNATumor tissues
Adjacent non-tumor tissues
8.920±3.764
8.430±3.685
0.490±2.584−0.8380.3330.986
MEG3LncRNATumor tissues
Adjacent non-tumor tissues
9.099±5.087
8.101±4.767
0.997±2.801−4.6150.0761.849
COL5A1mRNATumor tissues
Adjacent non-tumor tissues
6.982±2.174
7.721±2.412
−0.429±2.0687.5540.1231.594
BCL2L1mRNATumor tissues
Adjacent non-tumor tissues
7.259±6.294
7.891±6.338
−0.607±2.1782.6350.1311.560
COL11A2mRNATumor tissues
Adjacent non-tumor tissues
12.879±4.909
13.307±5.514
−0.774±4.937−7.2490.6560.452
PLCB2mRNATumor tissues
Adjacent non-tumor tissues
9.717±2.441
9.226±2.096
0.495±2.273−3.0350.2911.081

a ΔCt = Cttarget gene−CtGAPDH; ΔΔCt = ΔCttumor tissues − ΔCtadjacent non-tumor tissues. bP<0.05.

Validation of candidate lncRNAs in early stage GC

Then, we chose significant differential expression candidate lncRNAs (RP5-919F19, CTD-2541M15, UCA1, AP000459, LOC101928316, RP11-167N4, LINC01071 and MEG3) to further validate 20 early stage GC patient samples by qRT-PCR. Results showed that, the expression levels of CTD-2541M15 and UCA1 were significantly higher in 20 early stage GC patient tumor tissues than in their adjacent non-tumor tissues (P<0.050). AP000459, LINC01071 and MEG3 expression was significantly lower in carcinoma than in adjacent non-tumor tissues P<0.05 (Table IV). In addition, we also analyzed the change regularity of the significant differential lncRNAs both on advanced GC and early stage GC. Based on our results, compared with adjacent non-tumor tissues, the expression of CTD-2541M15 and UCA1 showed a significant gradual upward trend from early stage GC to advanced GC, AP000459, LINC01071 and MEG3 showed a significant gradual downward trend from early stage GC to advanced GC (Fig. 12).

Table IV

Relative expression of candidate lncRNAs in 20 pairs of early stage GC tumor and non-tumor tissues.

Table IV

Relative expression of candidate lncRNAs in 20 pairs of early stage GC tumor and non-tumor tissues.

Gene symbolGroupMean ± SD of ΔCtΔΔCta (mean ± SD)2−ΔΔCtP-valuet-value
RP5-919F19Tumor tissues
Adjacent non-tumor tissues
12.958±3.244
13.205±2.870
−0.247±2.3901.5520.6580.451
CTD-2541M15Tumor tissues
Adjacent non-tumor tissues
8.714±2.510
9.670±2.020
−0.955±2.0003.5280.046b2.135
UCA1Tumor tissues
Adjacent non-tumor tissues
9.387±3.335
11.432±3.803
−2.045±2.31011.5020.000b4.618
AP000459Tumor tissues
Adjacent non-tumor tissues
14.781±4.333
13.046±4.708
1.783±2.551−12.9110.012b2.831
LOC101928316Tumor tissues
Adjacent non-tumor tissues
10.766±3.217
10.562±2.712
0.203±2.205−0.0360.7010.391
RP11-167N4Tumor tissues
Adjacent non-tumor tissues
15.681±5.507
15.227±5.919
0.454±2.9170.4500.6730.440
LINC01071Tumor tissues
Adjacent non-tumor tissues
13.886±2.595
12.795±3.415
1.092±2.919−9.3790.044b2.181
MEG3Tumor tissues
Adjacent non-tumor tissues
8.651±3.123
7.338±2.936
1.284±2.132−2.6800.026b2.449

a ΔCt = Cttarget gene − CtGAPDH; ΔΔCt = ΔCttumor tissues − ΔCtadjacent non-tumor tissues.

b P<0.05.

Association between verified lncRNAs and the GC

Conditional logistic regression analysis was used to evaluate the association between differentially expressed lncRNAs and the risk of GC. As shown in Table V, significantly increased risk for GC was associated with increased expression of CTD-2541M15 and UCA1 (OR=2.860 and 30193, respectively) and reduced expression of MEG3 (OR=0.132). This suggested that CTD-2541M15 and UCA1 may function as oncogenes, while MEG3 might act as tumor suppressors.

Table V

Aberrant expression of lncRNAs associated with GC by conditional logistic regression analysis.

Table V

Aberrant expression of lncRNAs associated with GC by conditional logistic regression analysis.

Gene symbolGroupβSEWaldP-valueOR95% CI
CTD-2541M15Tumor tissues
Adjacent non-tumor tissues
1.0510.4804.796 0.029a2.860
1.000
1.117–7.326
UCA1Tumor tissues
Adjacent non-tumor tissues
1.1610.5244.914 0.027a3.193
1.000
1.144–8.915
AP000459Tumor tissues
Adjacent non-tumor tissues
−0.0470.1840.0650.7981.048
1.000
0.731–1.502
LINC01071Tumor tissues
Adjacent non-tumor tissues
0.3720.2502.2090.1371.450
1.000
0.888–2.369

Discussion

Despite advances in therapeutic options, early diagnosis and treatment remains the most effective way to reduce GC mortality. Even if gastrectomy is performed well, most patients with GC experience recurrence or metastasis within 1–2 years of surgery (19). The aims of cancer studies are to characterize systematically the cellular and molecular mechanisms involved in disease progression, and consequently, to identify potential biomarkers for predicting high-risk populations (20). Therefore, finding novel molecular biomarkers of malignancy has always been important for cancer prevention and treatment. Like proteins, mRNAs and miRNAs, lncRNAs show potential as novel biomarkers and therapeutic targets in different cancer types. These RNAs are >200 nucleotides and do not encode any protein. Some of them are strongly correlated with poor prognosis, suggesting a potential role in cancer progression (21). lncRNAs in GC have predominantly been reported from western countries and Japan, with few studies from China (22). Although the molecular mechanism of GC has been extensively investigated, the exact pathogenesis of this disease is still unclear (23,24). Molecular pathology of GC may vary among populations, which is likely due to differential exposure to disease risk factors including customs and habits, Helicobacter pylori variants and medical conditions. In the present study, our aim was to establish a comprehensive lncRNA and mRNA expression profile for GC in the Chinese population, a known high-risk population in Wuwei, and to investigate the mechanisms underlying GC carcinogenesis.

The present study revealed differential lncRNA and mRNA expression profiles in GC tissues and adjacent non-tumor tissues samples using microarray analysis. Here, we generated three pairs of pooled RNA samples from 10 advanced GC patients to microarray analysis. In this way, we can obtain more information of microarray detection and avoid separate specimen unique genes. In addition, using tissue samples to microarray analysis, we could not guarantee that each piece of specimen contains the same cell types and quantity, especially in tumor tissues and their adjacent non-tumor tissues. Thus, pooled RNA samples will improve the proportion of the same cell type tissues in the total RNA and make the microarray results more meaningful to population based study. In the present study, microarray results showed that lncRNA and mRNA expression levels in GC samples differed from those in adjacent non-tumor tissues. The microarray expression profile showed significantly differential expression (fold change ≥2.0) of 427 upregulated and 619 downregulated lncRNAs in 10 GC samples. In addition, there was significantly differential expression (fold change ≥2.0) of 647 upregulated and 2,349 downregulated mRNAs. GO and pathway analysis, and a lncRNA and mRNA co-expression network were used to study the biological function and potential mechanism of these genes in GC. We selected key lncRNAs and mRNAs for validation by quantitative real-time PCR in 30 pairs advanced GC and 20 early stage GC tissues and their paired adjacent non-tumor tissues. Finally, we used logistic regression analysis to find the association between candidate RNAs and the GC.

Several association studies have identified a large number of lncRNAs that are differentially expressed in disease states, including oncogenesis (25). An increasing number of studies have documented a biological link between aberrant expression of lncRNAs and GC (26). Dysregulation of lncRNAs, such as CCAT1, CHET1, H19, HOTAIR, MEG3, PVTI and SPRY4-IT1, is regarded as an important feature of GC (2731). However, the expression and functional significance of lncRNAs in GC tumorigenesis have not been completely characterized. We also used different databases, including LncRNAdb, Gene Expression Atlas, Gene Expression Omnibus (GEO), and Array Express Databases, to analyze the expression level of the most recently cited lncRNAs in studies on GC (32,33). We found that differentially expressed lncRNAs and mRNAs, such as UCA1, MEG3, HOTAIR, LINC01071, COL5A1, BCL2L1, COL11A2 and PLCB2, were the same as in other studies of GC (34). In the present study, the GO results showed that the functions of differentially expressed mRNAs in GC and their paired adjacent non-tumor tissues were DNA-dependent transcription, proteolysis, mitotic cell cycle, transmembrane transport and regulation of transcription. Abnormal expression of these regulatory genes is closely related to tumor development (35,36). Furthermore, KEGG pathway analysis revealed 67 pathways that may play key roles in the different core epigenetic mechanisms of GC, including the mTOR signaling pathway, gastric acid secretion, pathways in cancer, apoptosis and transcriptional regulation in cancer. Li et al (17) and Fan et al (37) have characterized differentially expressed genes that are involved in pathways associated with GC. They found that BCL2A1, LAMA4, ICM1 and LAMC2 participated in cancer-related signaling pathways such as mTOR and gastric acid secretion.

To gain insight into the function of lncRNAs, we used a lncRNA-mRNA co-expression network, which may be used for predicting target genes of lncRNAs. The principle of co-expression networks is that, by comparing the similarity or difference in the expression pattern of lncRNAs and mRNAs, any correlations between them can be determined (38,39). With this method, we compiled a list of lncRNAs and mRNAs that contained some key transcripts closely related to the pathogenesis of GC. We combined the results of microarray analysis, gene function and the co-expression network. The differential expression of mRNAs that participated in cancer-related pathways and related lncRNAs were RP5-919F19, RP11-54O7, RP11-20I23, CTD-2541M15, AC010731, UCA1, AP000459, LOC101928316, RP11-167N4, LINC01071, RP11-111K18, TTC28-AS1, MTOR-AS1 and MEG33. The LncRNAdb and LncRNA Diseases database showed that UCA1 is associated with urothelial cancer (40) and MEG3 with GC (41). The function of other lncRNAs was not reported.

Then, we analyzed the expression of 14 lncRNAs and four mRNAs in 30 pairs of tumor tissues and adjacent non-tumor tissues by using qRT-PCR. After qRT-PCR verification, we demonstrated for the first time that RP5-919F19, CTD-2541M15, AC010731 and UCA1 were upregulated in tumor tissues compared with adjacent non-tumor tissues, whereas AP000459, LOC101928316, RP11-167N4 and LINC01071 were downregulated in tumor tissues. Moreover, expression of seven lncRNAs was significantly correlated with GC-related mRNAs, suggesting these lncRNAs may play important roles in GC by regulating their target genes. Although the other indicators of the lncRNA and mRNA expression results did not differ significantly, the increases and decreases were approximately consistent with the microarray results. In the next step, we chose significant differential expression candidate lncRNAs to further validate 20 early stage GC patients samples. Results suggest that CTD-2541M15 and UCA1 was significantly higher expressed, AP000459, LINC01071 and MEG3 expression was significantly lower in the 20 early stage GC patient tumor tissues than in their adjacent non-tumor tissues. In addition, these lncRNAs expression levels appear with gradual upward or downward trends, respectively. Other indicators did not differ significantly in qRT-PCR, the reason may be due to the tumor tissues in this study, which were analysed by matching with adjacent non-tumor tissues instead of normal tissues, some early lesions might have been confused with normal tissues, which led to some differences in lncRNA and mRNA expression levels between the tumor and non-tumor tissues. Furthermore, we found that expression of CTD-2541M15, UCA1, AP000459, LINC01071 and MEG3 were sensitive between tumor and adjacent non-tumor tissues. Zheng et al (42) also found that the levels of UCA1 in gastric juice from GC patients were significantly higher than in normal subjects. In addition, Sun et al (30) found that MEG3 levels were markedly decreased in GC tissues compared with adjacent normal tissues. MEG3 expression level was significantly correlated with TNM stage, depth of invasion and tumor size. However, functions of CTD-2541M15, AP000459 and LINC01071 have not been reported. The conditional logistic regression analysis results showed that CTD-2541M15 and UCA1 may function as carcinogenic genes, while MEG3 might act as tumor suppressor, but the mechanisms of gene regulation are unclear. In the next step we will launch cell validation and gene intervention experiments to further study related functions.

In conclusion, the present study provided preliminary data that have helped to understand the potential mechanisms of GC, via differentially expressed lncRNAs and mRNAs. There was significant differential expression of lncRNAs between the GC and paired non-cancerous tissues, suggesting that lncRNAs may play important roles in the tumorigenesis of GC. Further validating these significant differential expression candidate RNAs we found that CTD-2541M15, UCA1, AP000459, LINC01071 and MEG3 were closely related to the progression and development of GC. Our results suggest that these differentially expressed lncRNAs may be potential biomarkers for early diagnosis of GC, which is an interesting direction for further research.

Acknowledgments

The present study was financially supported by the National Natural Science Foundation of China (81472939 and 81182618), the Qing Lan Project (no. 2012), the 333 project of Jiangsu Province (no. 2012), the Liu Da Ren Cai Gao Feng Project of Jiangsu Province (no. 2013-WSW-053) and the Fundamental Research Funds for the Central Universities.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Tanaka N, Katai H, Taniguchi H, Saka M, Morita S, Fukagawa T and Gotoda T: Trends in characteristics of surgically treated early gastric cancer patients after the introduction of gastric cancer treatment guidelines in Japan. Gastric Cancer. 13:74–77. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Kurella RR, Ancha HR, Ancha HB, Lightfoot SA, Guild RT and Harty RF: Obscure GI bleeding due to gastrointestinal stromal tumor (GIST) diagnosed by capsule endoscopy. J Okla State Med Assoc. 101:35–37. 2008.PubMed/NCBI

5 

Cheetham SW, Gruhl F, Mattick JS and Dinger ME: Long noncoding RNAs and the genetics of cancer. Br J Cancer. 108:2419–2425. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Mercer TR, Dinger ME and Mattick JS: Long non-coding RNAs: Insights into functions. Nat Rev Genet. 10:155–159. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Tsai MC, Spitale RC and Chang HY: Long intergenic noncoding RNAs: New links in cancer progression. Cancer Res. 71:3–7. 2011. View Article : Google Scholar : PubMed/NCBI

8 

He Y, Meng XM, Huang C, Wu BM, Zhang L, Lv XW and Li J: Long noncoding RNAs: Novel insights into hepatocelluar carcinoma. Cancer Lett. 344:20–27. 2014. View Article : Google Scholar

9 

Hung T and Chang HY: Long noncoding RNA in genome regulation: Prospects and mechanisms. RNA Biol. 7:582–585. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Song W, Liu YY, Peng JJ, Liang HH, Chen HY, Chen JH, He WL, Xu JB, Cai SR and He YL: Identification of differentially expressed signatures of long non-coding RNAs associated with different metastatic potentials in gastric cancer. J Gastroenterol. Jun 5–2015.Epub ahead of print. PubMed/NCBI

11 

Zhang T, Jiang M, Chen L, Niu B and Cai Y: Prediction of gene phenotypes based on GO and KEGG pathway enrichment scores. Biomed Res Int. 2013:8707952013.PubMed/NCBI

12 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

13 

Vu HL, Troubetzkoy S, Nguyen HH, Russell MW and Mestecky J: A method for quantification of absolute amounts of nucleic acids by (RT)-PCR and a new mathematical model for data analysis. Nucleic Acids Res. 28:E182000. View Article : Google Scholar : PubMed/NCBI

14 

Li X, Chen H, Li J and Zhang Z: Gene function prediction with gene interaction networks: A context graph kernel approach. IEEE Trans Inf Technol Biomed. 14:119–128. 2010. View Article : Google Scholar

15 

Zhao Z, Han F, Yang S, Wu J and Zhan W: Oxamate-mediated inhibition of lactate dehydrogenase induces protective autophagy in gastric cancer cells: Involvement of the Akt-mTOR signaling pathway. Cancer Lett. 358:17–26. 2015. View Article : Google Scholar

16 

Vilkin A, Levi Z, Morgenstern S, Shmuely H, Gal E, Hadad B, Hardi B and Niv Y: Higher gastric mucin secretion and lower gastric acid output in first-degree relatives of gastric cancer patients. J Clin Gastroenterol. 42:36–41. 2008. View Article : Google Scholar

17 

Li H, Yu B, Li J, Su L, Yan M, Zhang J, Li C, Zhu Z and Liu B: Characterization of differentially expressed genes involved in pathways associated with gastric cancer. PLoS One. 10:e01250132015. View Article : Google Scholar : PubMed/NCBI

18 

Carlson MR, Zhang B, Fang Z, Mischel PS, Horvath S and Nelson SF: Gene connectivity, function, and sequence conservation: Predictions from modular yeast co-expression networks. BMC Genomics. 7:402006. View Article : Google Scholar : PubMed/NCBI

19 

Songun I, Putter H, Kranenbarg EM, Sasako M and van de Velde CJ: Surgical treatment of gastric cancer: 15-year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol. 11:439–449. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Cho JY: Molecular diagnosis for personalized target therapy in gastric cancer. J Gastric Cancer. 13:129–135. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Cogill SB and Wang L: Co-expression network analysis of human lncRNAs and cancer genes. Cancer Inform. 13(Suppl 5): 49–59. 2014.PubMed/NCBI

22 

Wang YY, Ye ZY, Zhao ZS, Tao HQ and Li SG: Systems biology approach to identification of biomarkers for metastatic progression in gastric cancer. J Cancer Res Clin Oncol. 136:135–141. 2010. View Article : Google Scholar

23 

Charvat H, Sasazuki S, Inoue M, Iwasaki M, Sawada N, Shimazu T and Yamaji T: Prediction of the 10-year probability of gastric cancer occurrence in the Japanese population: The JPHC study cohort II. Int J Cancer. Jul 28–2015. View Article : Google Scholar : Epub ahead of print. PubMed/NCBI

24 

Song H, Ekheden IG, Zheng Z, Ericsson J, Nyrén O and Ye W: Incidence of gastric cancer among patients with gastric precancerous lesions: Observational cohort study in a low risk Western population. BMJ. 351:h38672015. View Article : Google Scholar : PubMed/NCBI

25 

Li M, Qiu M, Xu Y, Mao Q, Wang J, Dong G, Xia W, Yin R and Xu L: Differentially expressed protein-coding genes and long noncoding RNA in early-stage lung cancer. Tumour Biol. Jul 16–2015.Epub ahead of print.

26 

Chen X, Sun J, Song Y, Gao P, Zhao J, Huang X, Liu B, Xu H and Wang Z: The novel long noncoding RNA AC138128.1 may be a predictive biomarker in gastric cancer. Med Oncol. 31:2622014. View Article : Google Scholar : PubMed/NCBI

27 

Mizrahi I, Mazeh H, Grinbaum R, Beglaibter N, Wilschanski M, Pavlov V, Adileh M, Stojadinovic A, Avital I, Gure AO, et al: Colon cancer associated transcript-1 (CCAT1) expression in adenocarcinoma of the stomach. J Cancer. 6:105–110. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Zhuang M, Gao W, Xu J, Wang P and Shu Y: The long non-coding RNA H19-derived miR-675 modulates human gastric cancer cell proliferation by targeting tumor suppressor RUNX1. Biochem Biophys Res Commun. 448:315–322. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Pan W, Liu L, Wei J, Ge Y, zhang J, Chen H, Zhou L, Yuan Q, Zhou C and Yang M: A functional lncRNA HOTAIR genetic variant contributes to gastric cancer susceptibility. Mol Carcinog. Jan 3–2015. View Article : Google Scholar : Epub ahead of print. PubMed/NCBI

30 

Sun M, Xia R, Jin F, Xu T, Liu Z, De W and Liu X: Downregulated long noncoding RNA MEG3 is associated with poor prognosis and promotes cell proliferation in gastric cancer. Tumour Biol. 35:1065–1073. 2014. View Article : Google Scholar

31 

Li PF, Chen SC, Xia T, Jiang XM, Shao YF, Xiao BX and Guo JM: Non-coding RNAs and gastric cancer. World J Gastroenterol. 20:5411–5419. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Niland CN, Merry CR and Khalil AM: Emerging roles for long Non-Coding RNAs in cancer and neurological disorders. Front Genet. 3:252012. View Article : Google Scholar : PubMed/NCBI

33 

Zhang H, Chen Z, Wang X, Huang Z, He Z and Chen Y: Long non-coding RNA: A new player in cancer. J Hematol Oncol. 6:372013. View Article : Google Scholar : PubMed/NCBI

34 

Wang J, Song YX and Wang ZN: Non-coding RNAs in gastric cancer. Gene. 560:1–8. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Wu XM, Shao XQ, Meng XX, Zhang XN, Zhu L, Liu SX, Lin J and Xiao HS: Genome-wide analysis of microRNA and mRNA expression signatures in hydroxycamptothecin-resistant gastric cancer cells. Acta Pharmacol Sin. 32:259–269. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Chen Z, Zhang L, Xia L, Jin Y, Wu Q, Guo H, Shang X, Dou J, Wu K, Nie Y, et al: Genomic analysis of drug resistant gastric cancer cell lines by combining mRNA and microRNA expression profiling. Cancer Lett. 350:43–51. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Fan H, Guo Z and Wang C: Combinations of gene ontology and pathway characterize and predict prognosis genes for recurrence of gastric cancer after surgery. DNA Cell Biol. 34:579–587. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Zhu YP, Bian XJ, Ye DW, Yao XD, Zhang SL, Dai B, Zhang HL and Shen YJ: Long noncoding RNA expression signatures of bladder cancer revealed by microarray. Oncol Lett. 7:1197–1202. 2014.PubMed/NCBI

39 

Pujana MA, Han JD, Starita LM, Stevens KN, Tewari M, Ahn JS, Rennert G, Moreno V, Kirchhoff T, Gold B, et al: Network modeling links breast cancer susceptibility and centrosome dysfunction. Nat Genet. 39:1338–1349. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Wang Y, Chen W, Yang C, Wu W, Wu S, Qin X and Li X: Long non-coding RNA UCA1a(CUDR) promotes proliferation and tumorigenesis of bladder cancer. Int J Oncol. 41:276–284. 2012.PubMed/NCBI

41 

Yan J, Guo X, Xia J, Shan T, Gu C, Liang Z, Zhao W and Jin S: MiR-148a regulates MEG3 in gastric cancer by targeting DNA methyltransferase 1. Med Oncol. 31:8792014. View Article : Google Scholar : PubMed/NCBI

42 

Zheng Q, Wu F, Dai WY, Zheng DC, Zheng C, Ye H, Zhou B, Chen JJ and Chen P: Aberrant expression of UCA1 in gastric cancer and its clinical significance. Clin Transl Oncol. 17:640–646. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 35 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li C, Liang G, Yao W, Sui J, Shen X, Zhang Y, Ma S, Ye Y, Zhang Z, Zhang W, Zhang W, et al: Differential expression profiles of long non-coding RNAs reveal potential biomarkers for identification of human gastric cancer. Oncol Rep 35: 1529-1540, 2016
APA
Li, C., Liang, G., Yao, W., Sui, J., Shen, X., Zhang, Y. ... Pu, Y. (2016). Differential expression profiles of long non-coding RNAs reveal potential biomarkers for identification of human gastric cancer. Oncology Reports, 35, 1529-1540. https://doi.org/10.3892/or.2015.4531
MLA
Li, C., Liang, G., Yao, W., Sui, J., Shen, X., Zhang, Y., Ma, S., Ye, Y., Zhang, Z., Zhang, W., Yin, L., Pu, Y."Differential expression profiles of long non-coding RNAs reveal potential biomarkers for identification of human gastric cancer". Oncology Reports 35.3 (2016): 1529-1540.
Chicago
Li, C., Liang, G., Yao, W., Sui, J., Shen, X., Zhang, Y., Ma, S., Ye, Y., Zhang, Z., Zhang, W., Yin, L., Pu, Y."Differential expression profiles of long non-coding RNAs reveal potential biomarkers for identification of human gastric cancer". Oncology Reports 35, no. 3 (2016): 1529-1540. https://doi.org/10.3892/or.2015.4531