Open Access

Cancer‑related long noncoding RNAs show aberrant expression profiles and competing endogenous RNA potential in esophageal adenocarcinoma

  • Authors:
    • Yang Yu
    • Xingxing Chen
    • Shundong Cang
  • View Affiliations

  • Published online on: September 5, 2019     https://doi.org/10.3892/ol.2019.10808
  • Pages: 4798-4808
  • Copyright: © Yu 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

Long non‑coding RNAs (lncRNAs) govern gene expression by competitively binding to microRNA response elements (MREs). Although they were initially considered as transcriptional noise, lncRNAs have attracted increased attention in oncology. Dysregulation of lncRNAs occurs in various types of human tumor, including esophageal adenocarcinoma (EAC). However, the functions of these cancer‑associated lncRNAs and of their related competitive endogenous RNA (ceRNA) network in EAC remains unknown. To determine the relevant potential mechanisms, the present study analyzed the transcriptome sequencing data and clinical information of 79 patients with EAC, including 79 tumor samples and 11 normal samples, which were obtained from The Cancer Genome Atlas esophageal cancer project. The edgeR v3.25.0 software was used for differential gene expression analysis. The results exhibited 561 cancer‑associated lncRNAs with a >2.0‑fold change and a false discovery rate‑adjusted P<0.01. Among these lncRNAs, 26 were significantly associated with patient overall survival. According to data from bioinformatics databases and differentially expressed RNAs, an lncRNA‑regulated ceRNA network for EAC was constructed. The results demonstrated that the aberrantly expressed lncRNA‑associated ceRNA network included 37 EAC cancer‑associated lncRNAs, five miRNAs and 13 mRNAs. In conclusion, the present study identified novel lncRNAs as candidate prognostic biomarkers and revealed a potential regulatory network of gene expression in EAC.

Introduction

Esophageal adenocarcinoma (EAC) is a highly lethal malignancy that occurs mainly in the distal esophagus and gastroesophageal junction (1). EAC is rare in China; however, it represents the predominant type of esophageal cancer in North America and Europe. In these continents, the overall incidence of EAC has rapidly increased over the past three decades at a rate (5–10%) greater than that of any other major cancer and the incidence rate is higher in white males compared with that in white females (24). The reason for this increase is not entirely understood. Previous studies have reported that EAC differs from esophageal squamous cell carcinoma (ESCC) in terms of genetic and environmental risk factors such as tobacco use, alcohol, obesity and germline mutations (5,6). Systematic therapy for EAC typically includes endoscopic mucosal resection, surgical resection, chemoradiotherapy and neoadjuvant chemotherapy; however, the mortality rate remains high and the overall 5-year survival rate is 17% in the United States (7). Although endoscopy can accurately diagnose early-stage EAC, most patients are diagnosed with regional metastasis or distant metastasis, which are positively correlated with a considerable decline in the 5-year survival rate (8). There is therefore an urgency to identify novel potential diagnostic and prognostic biomarkers for EAC.

Long non-coding RNAs (lncRNAs) represent a new class of non-coding RNAs (ncRNAs) and are defined as transcripts >200 nucleotides in length (9). Unlike their shorter counterparts, including microRNAs (miRNAs), the roles and underlying mechanisms of lncRNAs in human disease remain largely unknown. Due to improvements in DNA sequencing techniques, numerous lncRNAs have been discovered. In addition, an increasing number of lncRNAs have been identified in human cancer, such as HAGLR opposite strand lncRNA overexpression in gastric cancer (10). Previous studies have focused on the biological function and underlying molecular mechanism of lncRNAs in various types of cancer, including colorectal cancer, gastric cancer, hepatocellular carcinoma, renal cell carcinoma, prostate carcinoma and EAC (1117). Although studies reported that lncRNAs can be involved in the development and progression of ESCC (6,14), only a few studies have determined the function of lncRNAs in EAC (5,18).

Emerging technologies have increased our ability to determine the functions of cancer-associated lncRNAs. Significant progress towards understanding the underlying molecular mechanism by which lncRNAs can regulate miRNA function has therefore been made. Salmena et al (19) proposed a competing endogenous RNA (ceRNA) language where protein coding genes, microRNAs and lncRNAs communicate with each other by competitively binding to shared miRNA response elements (MREs). Competing endogenous RNA networks comprise a new regulatory network of mRNAs and non-coding RNAs, which reveals a greatly expanded role for lncRNAs in human disease (20). This hypothesis has been experimentally validated. For example, Cesana et al (21) identified a muscle-specific lncRNA named linc-MD1, which regulates the expression of mastermind-like 1 and myocyte-specific enhancer factor 2C by serving as a ‘sponge’ for miR-133. Furthermore, Qu et al (22) demonstrated that lncARSR mediates sunitinib resistance in renal cell carcinoma by competitively binding to miR-34/miR-449 to promote AXL receptor tyrosine kinase and c-MET expression. Exploration of RNA cross-talk offers therefore insights into cancer diagnosis and therapy. An lncRNA-miRNA-mRNA ceRNA network has therefore been constructed for various types of human cancer, in particular for ESCC (2326); however, such a network has not yet been described for EAC.

In order to systematically describe EAC-associated pseudogenes and to construct a ceRNA network, the present study comprehensively analyzed RNA sequencing (RNA-Seq) transcript data that were obtained from The Cancer Genome Atlas (TCGA) esophageal cancer project (https://www.cancer.gov/types/esophageal). The database includes lncRNA, microRNA and mRNA data and clinical information from patients with EAC. The present study included 79 EAC tumor and 11 adjacent non-tumor esophagus tissue samples. By using publicly available RNA-Seq data from TCGA, some EAC-associated lncRNAs, mRNAs and miRNAs were identified based on the ceRNA hypothesis. Furthermore, 561 differentially expressed lncRNAs (DElncRNAs), 1,289 differentially-expressed mRNAs (DEmRNAs) and 44 differentially-expressed miRNAs (DEmiRNAs) were identified. Subsequently, five dysregulated lncRNAs, 13 mRNAs and 32 miRNAs were identified and included in a constructed ceRNA network based on lncRNA-miRNA interactions predicted by miRcode v11 (www.mircode.org/). Potential prognostic biomarkers were then identified by exploring the influence of dysregulated RNAs on overall survival using the univariate Cox proportional hazards regression model and Kaplan-Meier curve analysis. The results from this comprehensive analysis provided the foundation for deeper understanding of the cancer-associated lncRNA functions in EAC and revealed potential prognostic biomarkers.

Materials and methods

Patients and samples

Data for 187 patients with esophageal cancer were obtained from the TCGA data portal (https://portal.gdc.cancer.gov/). The exclusion criteria were as follows: i) Patients with ESCC or undetermined pathological classification; and ii) samples without corresponding RNA-Seq and miRNA-Seq data. Overall, data from 79 patients with EAC were enrolled in the present study. This study followed the publication guidelines provided by TCGA (http://cancergenome.nih.gov/publications/publicationguidelines).

RNA-Seq data

RNA-Seq and miRNA-Seq data (level 3) were downloaded from 90 tissue samples of the TCGA database, including 79 EAC samples and 11 adjacent normal samples. The gene expression profiles generated from Illumina Hiseq platforms (Illumina, Inc.) were all publicly available data.

Analysis of DEmRNAs, DElcnRNAs and DEmiRNAs

The raw count data were processed with edgeR v3.25.0 (Bioconductor), which is a package based on the R language (v3.5.0) (27) for differential gene expression analysis. For all P-values, a false discovery rate (FDR) was applied to correct the statistical significance of multiple testing. Genes with >2.0 fold change (FC) and FDR-adjusted P<0.01 were considered significant. The volcano plot and heat map were designed to visualize the results using ggplots v3.0, which is a package based on the R language.

Association between DElncRNAs and patient prognosis

All patients were classified into high or low lncRNA-expression groups according to the median. Kaplan-Meier and log-rank methods were used to test differences between the two groups. P<0.05 was considered to indicate a statistically significant difference.

Construction of the ceRNA network

Three calculations were performed to construct the ceRNA network as follows: i) Cancer-associated lncRNA filtration, where lncRNAs with FC>2.0 (either up- or downregulated) and FDR-adjusted P<0.01 were considered as cancer-associated lncRNAs [to improve data reliability, cancer-associated lncRNAs that were not annotated by GENCODE (http://www.gencodegenes.org/) were excluded]; ii) lncRNA-miRNA interactions were predicted by miRcode (http://www.mircode.org/) and starBase (http://starbase.sysu.edu.cn/); and iii) target mRNAs of DEmiRNAs were predicted using the three bioinformatics databases miRDB (http://mirdb.org/), miRTarBase (http://mirtarbase.mbc.nctu.edu.tw/php/index.php) and TargetScan (http://www.targetscan.org). Gene Oncology (GO) was analyzed using Database for Annotation, Visualization, and Integrated Discovery bioinformatics tools (DAVID; v6.8; http://david.ncifcrf.gov). In order to improve the consistency of the bioinformatics analysis, the target genes were retained. A network graph was constructed and visualized using Cytoscape v3.5.1 (https://cytoscape.org/).

Results

Patient characteristics

The detailed clinical information and pathological characteristics of the patients included in the present study, including sex, age at diagnosis, metastasis status, lymph node status and tumor-node-metastasis stage, are presented in Table I. The median age for all patients was 69 years (range, 27–86 years). The median overall survival was 14.29 months (range 0.36–83.18 months).

Table I.

Clinical characteristics of patients with esophageal adenocarcinoma.

Table I.

Clinical characteristics of patients with esophageal adenocarcinoma.

CharacteristicsNo. cases%
Age at diagnosis, years
  <602632.9
  ≥605367.1
Sex
  Male1113.9
  Female6886.1
Metastasis
  M05772.2
  M11012.7
  MX1012.7
Lymph node status
  N02227.8
  N14455.7
  N256.3
  N356.3
  NX33.8
Stage
  I911.4
  II2227.8
  III2632.9
  IV56.3
T stage
  T011.3
  T11924.1
  T21012.7
  T34658.2
  T411.3
  TX22.6
Histological grade
  G111.3
  G22835.4
  G32430.4
  GX2632.9
Identification of DElncRNAs

lncRNAs with FDR-adjusted P<0.01 and FC >2.0 were considered to be differentially expressed. A total of 561 DElncRNAs were identified, of which 217 were upregulated and 344 were downregulated (Table SI). A volcano plot was therefore constructed (Fig. 1A) to visually describe the FDRs and FCs. In addition, a heat map was designed (Fig. 1B) to highlight the top 100 significant DElncRNAs according to the FDR-adjusted P-values.

To explore the potential lncRNAs that could possess prognostic abilities, the expression profiles of 561 lncRNAs and corresponding clinical data were analyzed using Kaplan-Meier Curve. The results demonstrated that 26 lncRNAs were positively correlated with overall survival (OS; P<0.05; Fig. 2). Among these 26, 12 lncRNAs, AC007128.1, AC079354.3, AC246680.1, AL009178.2, AL135924.2, AL138789.1, AP003356.1, AP0033469.2, GK-IT1, HOTAIR, LINC01114 and LINC01768, were negatively correlated with OS (Fig. 2A). Conversely, the remaining 14 lncRNAs, AC004585.1, AC016395.1, AC024337.2, AC087491.1, AC093583.1, AC104211.1, AL022316.1, AL031429.1, CYP1B1-AS1, LINC00163, LINC00906, LINC01695, SLCO4A1-AS1 and UG0898H09, were positively correlated with OS (Fig. 2B).

Identification of DEmRNAs

The RNA expression levels in 79 EAC tumor samples and 11 normal samples were analyzed. With a cut-off value of FDR-adjusted P<0.01 and FC >2.0, 367 upregulated and 922 downregulated mRNAs were identified (Table SII). A volcano plot was therefore constructed to visualize the results (Fig. 3A). The top 100 significant DEmRNAs were then highlighted by plotting FDR-adjusted P-values in a heat map (Fig. 3C).

To analyze the DEmRNA functions, enrichment analysis based on enriched functional GO modules was performed. The results demonstrated that the DEmRNAs were significantly enriched in the ‘chemokine-mediated signaling pathway’ (GO: 0070098), ‘plasma membrane’ (GO: 0005886) and ‘calcium ion binding’ (GO: 0005509) GO terms under ‘biological process’, ‘cellular component’ and ‘molecular function’, respectively (Fig. 3B).

Identification of DEmiRNAs

In order to design a ceRNA network for EAC, the miRNA expression profiles between tumor samples and normal samples were compared. Subsequently, 44 DEmiRNAs, including 28 upregulated and 16 downregulated were identified (Fig. 4A and C). The mRNAs that were targeted by the 44 DEmiRNAs from miRDB, miRTarBase and TargetScan were then screened. To improve data reliability, mRNAs that were not included in the set of 1,289 DEmRNAs were excluded. Eventually, 13 DEmiRNAs remained in the ceRNA network (Fig. 4B).

In order to identify the DEmRNAs that may have potential prognostic ability, the expression profiles of the 13 DEmRNAs included in the ceRNA network were analyzed using Kaplan-Meier curve. The results demonstrated that the expression profiles of three DEmRNAs were positively correlated with OS (P<0.05). Two of these DEmRNAs, angiopoietin 2 and interleukin 11 (IL11) were negatively correlated with OS, whereas neurotrophic receptor tyrosine kinase 2 (NTRK2) was positively correlated with OS (Fig. 4D).

To identify the lncRNA-miRNA interactions in EAC, the potential MREs in DElncRNAs were screened using miRcode. For miRNA-mRNA interactions, miRDB, miRTarBase, and TargetScan were used to identify the DEmRNAs targeted by DEmiRNAs. The results are listed in Tables II and III.

Table II.

Potential miRNAs that may target cancer-associated lncRNAs by miRNA response elements.

Table II.

Potential miRNAs that may target cancer-associated lncRNAs by miRNA response elements.

lncRNAsmiRNAs
LINC00525hsa-mir-301b, hsa-mir-383
PART1hsa-mir-301b, hsa-mir-145, hsa-mir-204
C2orf48hsa-mir-372, hsa-mir-372
LINC00483hsa-mir-372
C20orf166-AS1hsa-mir-301b, hsa-mir-372
C15orf54hsa-mir-301b, hsa-mir-372
C8orf31hsa-mir-372, hsa-mir-204
AP002478.1hsa-mir-372
LINC00314hsa-mir-372, hsa-mir-204
LINC00501hsa-mir-301b, hsa-mir-204
C1orf137hsa-mir-204, hsa-mir-383
MUC19hsa-mir-301b, hsa-mir-372, hsa-mir-145, hsa-mir-204
KRBOX1-AS1hsa-mir-301b, hsa-mir-372, hsa-mir-204
LINC00114hsa-mir-204
SNHG14hsa-mir-301b, hsa-mir-372, hsa-mir-145, hsa-mir-204
LINC00337hsa-mir-372, hsa-mir-145, hsa-mir-383
HOTAIRhsa-mir-301b, hsa-mir-204
LINC00332hsa-mir-204
CYP1B1-AS1hsa-mir-301b, hsa-mir-145, hsa-mir-204
AC110491.1hsa-mir-204
LINC00494hsa-mir-372, hsa-mir-145, hsa-mir-383
LINC00299hsa-mir-145, hsa-mir-204, hsa-mir-383
CADM2-AS1hsa-mir-301b, hsa-mir-145, hsa-mir-383
AC011374.1hsa-mir-372
ADAMTS9-AS1hsa-mir-301b, hsa-mir-145
AC002464.1hsa-mir-383
AC007834.1hsa-mir-301b
ADAMTS9-AS2hsa-mir-301b, hsa-mir-372, hsa-mir-145, hsa-mir-204
GK-AS1hsa-mir-145
LIFR-AS1hsa-mir-372, hsa-mir-204
ALDH1L1-AS2hsa-mir-301b, hsa-mir-372, hsa-mir-145
PVT1hsa-mir-372, hsa-mir-145, hsa-mir-383
LINC00536hsa-mir-204
AC006487.1hsa-mir-204
AC090206.1hsa-mir-204
FAM181A-AS1hsa-mir-204
LINC00524hsa-mir-204

[i] lncRNA, long non-coding RNA; miRNA, microRNA.

Table III.

Differentially expressed mRNAs targeted by differentially expressed miRNAs.

Table III.

Differentially expressed mRNAs targeted by differentially expressed miRNAs.

miRNAsmRNAs
hsa-mir-145ANGPT2, PDGFD, DDC, MEST
hsa-mir-148aHOXC8, NPTX1
hsa-mir-196bHOXB7, IGF2BP3, HOXC8
hsa-mir-204HOXC8, NTRK2, SLC22A6, CHRDL1, IL11, NPTX1, CDH2
hsa-mir-372TMEM100, CADM2

[i] miRNA, microRNA.

ceRNA network construction

In order to improve knowledge on DElncRNA function in EAC, a dysregulated lncRNA-miRNA-mRNA ceRNA network based on the aforementioned data (presented in Tables II and III) was constructed. The results demonstrated that in the ceRNA network, interaction of five DEmiRNAs with 37 DElncRNAs was predicted, according to the results retrieved from miRcode. The ceRNA network is presented in Fig 5.

Discussion

lncRNAs represent a crucial category of non-coding genes in the transcriptome that act as pivotal regulators of cell physiology and pathology in human cancer by mediating gene expression through multiple mechanisms (28). The dysregulation of lncRNA expression is involved in the pathogenesis of various types of solid tumor (29,30). Numerous novel biological functions have been attributed to lncRNAs, which have become the focal point of many studies (10,17). The ceRNA hypothesis describes regulatory networks among protein-coding mRNAs and non-coding RNAs, including miRNAs and lncRNAs at the post-transcription level. According to this hypothesis, changes in the expression of one or multiple miRNA targets can alter the number of unbound miRNAs and lead to observable changes in miRNA activity. The various transcripts from the transcriptome communicate with one another by competitively binding to shared MREs (20). ceRNA networks in human cancer include cancer-associated lncRNAs, microRNAs and mRNAs. A previous study demonstrated a miRNA-lncRNA-mRNA interaction in ESCC (31). However, the ceRNA network in EAC remains poorly understood.

At present, since lncRNAs are able to regulate miRNA functions by competitively binding to shared MREs in mRNA, they are considered as diagnostic and prognostic biomarkers. Numerous well-studied lncRNAs have been identified as potential targets or powerful predictors in various types of cancer, including LINC00668, H19 and UCA1 (3234). However, studies on EAC remain rare. Based on the RNA-Seq data and clinical data from 79 patients with EAC, the present study demonstrated that 26 cancer-associated lncRNAs may affect the OS of patients with EAC. In particular, the results from this study reported that two DElncRNAs, CYP1B1-AS1 and HOTAIR, were not only identified as part of the ceRNA network, but were also positively and negatively correlated with OS, respectively, which suggested that these two lncRNAs may serve as essential oncogenes and as prognostic markers in EAC.

HOTAIR is a highly studied lncRNA. Previous studies demonstrated that it serves a role in the development and progression of various types of solid tumor, including renal cell carcinoma, colorectal cancer, breast cancer, gastric cancer, non-small cell lung cancer, cervical cancer and ovarian epithelial carcinoma (3545). In addition, Ren et al (46) demonstrated that HOTAIR can control the cell cycle by acting as a competing endogenous ‘sponge’ to downregulate miR-1 and upregulate cyclin D1 in ESCC. The present study reported that HOTAIR expression was upregulated in EAC tumor tissues. In addition, patients with highly expressed HOTAIR had worse survival outcomes. HOTAIR may therefore compete with miR-301b and miR-204 to regulate chordin like 1, NTRK2, IL11, neuronal pentraxin 1, homeobox C8 and solute carrier family 22 member 6 expression. Although these mRNAs have been identified as aberrantly expressed, their roles have not been fully investigated in EAC.

In addition to HOTAIR, LINC00163 and SLCO4A1-AS1 have also been reported to be associated with cancer prognosis. Guo et al (47) demonstrated that the LINC00163 level is significantly decreased in lung cancer tissues and cell lines following bioinformatics and reverse transcription-quantitative PCR analyses. LINC00163 expression was lower in metastatic tissues compared with non-metastatic tissues, and a higher LINC00163 expression in patients with lung cancer could predict a better prognosis. Yang et al (48) reported that SLCO4A1-AS1 expression was more upregulated in bladder cancer tissues compared with that in adjacent normal tissues, and that SLCO4A1-AS1 overexpression is associated with poor prognosis and tumor metastasis. Yu et al (49) demonstrated that a high SLCO4A1-AS1 expression level is associated with bladder cancer progression and that SLCO4A1-AS1 promotes malignant phenotypes of bladder cancer cells via the miRNA-335-5p/OCT4 axis.

To confirm the accuracy of the ceRNA network prediction presented in this study, interactions among lncRNAs, miRNAs and mRNAs in EAC were measured. Only cancer-associated lncRNAs and miRNAs with >2.0 FC and FDR <0.01 were selected. These non-coding genes were then annotated by GENCODE. Interactions among lncRNAs, miRNAs and mRNAs were predicted by experimentally conformed algorithms or by using miRDB, miRcode, miRTarBase and TargetScan databases. In the present study, cancer-associated lncRNAs in EAC were identified based on the RNA-Seq data of 79 EAC tissues and 11 normal tissues. Subsequently, cancer-associated miRNAs and mRNAs were identified. Eventually, interactions between lncRNAs, miRNAs and mRNAs was identified by constructing an lncRNA-miRNA-mRNA ceRNA network. A total of 37 DElncRNAs, five miRNAs and 13 mRNAs were selected to construct this newly-identified ceRNA-mediated gene regulatory network. This network included numerous active lncRNA-miRNA-mRNA interactions that may be used as prognostic biomarkers in EAC.

In conclusion, the present study identified some cancer-associated lncRNAs and revealed their potential use in prognosis prediction. In particular, some cancer-associated lncRNAs may serve as ceRNAs. The ceRNA network that was built in the present study may help understanding the mechanisms involved in the development and progression of EAC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

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

SC and YY conceived and designed the study. XC acquired the data. YY analyzed and interpreted the data. YY and XC wrote and revised the manuscript. SC supervised the study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Mannath J and Ragunath K: Role of endoscopy in early oesophageal cancer. Nat Rev Gastroenterol Hepatol. 13:720–730. 2016. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

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

4 

Pera M, Manterola C, Vidal O and Grande L: Epidemiology of esophageal adenocarcinoma. J Surg Oncol. 92:151–159. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Coleman HG, Xie SH and Lagergren J: The epidemiology of esophageal adenocarcinoma. Gastroenterology. 154:390–405. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Huang X, Zhou X, Hu Q, Sun B, Deng M, Qi X and Lü M: Advances in esophageal cancer: A new perspective on pathogenesis associated with long non-coding RNAs. Cancer Lett. 413:94–101. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Lagergren J and Lagergren P: Recent developments in esophageal adenocarcinoma. CA Cancer J Clin. 63:232–248. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Rustgi AK and El-Serag HB: Esophageal carcinoma. N Engl J Med. 371:2499–2509. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Chen JF, Wu P, Xia R, Yang J, Huo XY, Gu DY, Tang CJ, De W and Yang F: STAT3-induced lncRNA HAGLROS overexpression contributes to the malignant progression of gastric cancer cells via mTOR signal-mediated inhibition of autophagy. Mol Cancer. 17:62018. View Article : Google Scholar : PubMed/NCBI

11 

Zhou DD, Liu XF, Lu CW, Pant OP and Liu XD: Long non-coding RNA PVT1: Emerging biomarker in digestive system cancer. Cell Prolif. 50:2017. View Article : Google Scholar

12 

Song YX, Sun JX, Zhao JH, Yang YC, Shi JX, Wu ZH, Chen XW, Gao P, Miao ZF and Wang ZN: Non-coding RNAs participate in the regulatory network of CLDN4 via ceRNA mediated miRNA evasion. Nat Commun. 8:2892017. View Article : Google Scholar : PubMed/NCBI

13 

Parasramka MA, Maji S, Matsuda A, Yan IK and Patel T: Long non-coding RNAs as novel targets for therapy in hepatocellular carcinoma. Pharmacol Ther. 161:67–78. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Li W, Zheng J, Deng J, You Y, Wu H, Li N, Lu J and Zhou Y: Increased levels of the long intergenic non-protein coding RNA POU3F3 promote DNA methylation in esophageal squamous cell carcinoma cells. Gastroenterology. 146:1714–1726.e5. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Xiao ZD, Han L, Lee H, Zhuang L, Zhang Y, Baddour J, Nagrath D, Wood CG, Gu J, Wu X, et al: Energy stress-induced lncRNA FILNC1 represses c-Myc-mediated energy metabolism and inhibits renal tumor development. Nat Commun. 8:7832017. View Article : Google Scholar : PubMed/NCBI

16 

Martens-Uzunova ES, Böttcher R, Croce CM, Jenster G, Visakorpi T and Calin GA: Long noncoding RNA in prostate, bladder, and kidney cancer. Eur Urol. 65:1140–1151. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Wang P, Xu J, Wang Y and Cao X: An interferon-independent lncRNA promotes viral replication by modulating cellular metabolism. Science. 358:1051–1055. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Su W, Guo C, Wang L, Wang Z, Yang X, Niu F, Tzou D, Yang X, Huang X, Wu J, et al: LncRNA MIR22HG abrogation inhibits proliferation and induces apoptosis in esophageal adenocarcinoma cells via activation of the STAT3/c-Myc/FAK signaling. Aging (Albany NY). 11:4587–4596. 2019.PubMed/NCBI

19 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The rosetta stone of a hidden RNA language. Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Qi X, Zhang DH, Wu N, Xiao JH, Wang X and Ma W: ceRNA in cancer: Possible functions and clinical implications. J Med Genet. 52:710–718. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A and Bozzoni I: A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 147:358–369. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, Chen W, Liu F, Sun W, Li XF, et al: Exosome-transmitted lncARSR promotes Sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell. 29:653–668. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Cao C, Zhang T, Zhang D, Xie L, Zou X, Lei L, Wu D and Liu L: The long non-coding RNA, SNHG6-003, functions as a competing endogenous RNA to promote the progression of hepatocellular carcinoma. Oncogene. 36:1112–1122. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Conte F, Fiscon G, Chiara M, Colombo T, Farina L and Paci P: Role of the long non-coding RNA PVT1 in the dysregulation of the ceRNA-ceRNA network in human breast cancer. PLoS One. 12:e01716612017. View Article : Google Scholar : PubMed/NCBI

25 

Yang S, Ning Q, Zhang G, Sun H, Wang Z and Li Y: Construction of differential mRNA-lncRNA crosstalk networks based on ceRNA hypothesis uncover key roles of lncRNAs implicated in esophageal squamous cell carcinoma. Oncotarget. 7:85728–85740. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Li CY, Liang GY, Yao WZ, Sui J, Shen X, Zhang YQ, Peng H, Hong WW, Ye YC, Zhang ZY, et al: Integrated analysis of long non-coding RNA competing interactions reveals the potential role in progression of human gastric cancer. Int J Oncol. 48:1965–1976. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Team RC: R:, . A language and environment for statistical computingR Foundation for Statistical Computing; Vienna, Austria: 2013

28 

Marchese FP, Raimondi I and Huarte M: The multidimensional mechanisms of long noncoding RNA function. Genome Biol. 18:2062017. View Article : Google Scholar : PubMed/NCBI

29 

Alvarez-Dominguez JR and Lodish HF: Emerging mechanisms of long noncoding RNA function during normal and malignant hematopoiesis. Blood. 130:1965–1975. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Klingenberg M, Matsuda A, Diederichs S and Patel T: Non-coding RNA in hepatocellular carcinoma: Mechanisms, biomarkers and therapeutic targets. J Hepatol. 67:603–618. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Lin C, Zhang N, Wang Y, Wang Y, Nice E, Guo C, Zhang E, Yu L, Li M, Liu C, et al: Functional role of a novel long noncoding RNA TTN-AS1s in esophageal squamous cell carcinoma progression and metastasis. Clin Cancer Res. 24:486–498. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Zhang E, Yin D, Han L, He X, Si X, Chen W, Xia R, Xu T, Gu D, De W, et al: E2F1-induced upregulation of long noncoding RNA LINC00668 predicts a poor prognosis of gastric cancer and promotes cell proliferation through epigenetically silencing of CKIs. Oncotarget. 7:23212–23226. 2016.PubMed/NCBI

33 

Liu L, Liu L and Lu S: lncRNA H19 promotes viability and epithelial-mesenchymal transition of lung adenocarcinoma cells by targeting miR-29b-3p and modifying STAT3. Int J Oncol. 54:929–941. 2019.PubMed/NCBI

34 

Cui X, Zhao C, Yao X, Qian B, Su C, Ren Y, Yao Z, Gao X and Yang J: SND1 acts as an anti-apoptotic factor via regulating the expression of lncRNA UCA1 in hepatocellular carcinoma. RNA Biol. 15:1364–1375. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Katayama H, Tamai K, Shibuya R, Nakamura M, Mochizuki M, Yamaguchi K, Kawamura S, Tochigi T, Sato I, Okanishi T, et al: Long non-coding RNA HOTAIR promotes cell migration by upregulating insulin growth factor-binding protein 2 in renal cell carcinoma. Sci Rep. 7:120162017. View Article : Google Scholar : PubMed/NCBI

36 

Li M, Li X, Zhuang Y, Flemington EK, Lin Z and Shan B: Induction of a novel isoform of the lncRNA HOTAIR in Claudin-low breast cancer cells attached to extracellular matrix. Mol Oncol. 11:1698–1710. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Li P, Zhang X, Wang L, Du L, Yang Y, Liu T, Li C and Wang C: lncRNA HOTAIR contributes to 5FU resistance through suppressing miR-218 and activating NF-κB/TS signaling in colorectal cancer. Mol Ther Nucleic Acids. 8:356–369. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Ma J, Fan Y, Feng T, Chen F, Xu Z, Li S, Lin Q, He X, Shi W, Liu Y, et al: HOTAIR regulates HK2 expression by binding endogenous miR-125 and miR-143 in oesophageal squamous cell carcinoma progression. Oncotarget. 8:86410–86422. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Chi S, Shen L, Hua T, Liu S, Zhuang G, Wang X, Zhou X, Wang G and Wang H: Prognostic and diagnostic significance of lncRNAs expression in cervical cancer: A systematic review and meta-analysis. Oncotarget. 8:79061–79072. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Fan CN, Ma L and Liu N: Systematic analysis of lncRNA-miRNA-mRNA competing endogenous RNA network identifies four-lncRNA signature as a prognostic biomarker for breast cancer. J Transl Med. 16:2642018. View Article : Google Scholar : PubMed/NCBI

41 

Gao S, Zhao ZY, Wu R, Zhang Y and Zhang ZY: Prognostic value of long noncoding RNAs in gastric cancer: A meta-analysis. OncoTargets Ther. 11:4877–4891. 2018. View Article : Google Scholar

42 

Guo F, Cao Z, Guo H and Li S: The action mechanism of lncRNA-HOTAIR on the drug resistance of non-small cell lung cancer by regulating Wnt signaling pathway. Exp Ther Med. 15:4885–4889. 2018.PubMed/NCBI

43 

Qiu JJ, Lin YY, Ye LC, Ding JX, Feng WW, Jin HY, Zhang Y, Li Q and Hua KQ: Overexpression of long non-coding RNA HOTAIR predicts poor patient prognosis and promotes tumor metastasis in epithelial ovarian cancer. Gynecol Oncol. 134:121–128. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Wang WJ, Li HT, Yu JP, Han XP, Xu ZP, Li YM, Jiao ZY and Liu HB: A competing endogenous RNA network reveals novel potential lncRNA, miRNA, and mRNA biomarkers in the prognosis of human colon adenocarcinoma. J Surg Res. 235:22–33. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Zhang H, Wang Z, Wu J, Ma R and Feng J: Long noncoding RNAs predict the survival of patients with colorectal cancer as revealed by constructing an endogenous RNA network using bioinformation analysis. Cancer Med. 8:863–873. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Ren K, Li Y, Lu H, Li Z, Li Z, Wu K, Li Z and Han X: Long noncoding RNA HOTAIR controls cell cycle by functioning as a competing endogenous RNA in esophageal squamous cell carcinoma. Transl Oncol. 9:489–497. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Guo X, Wei Y, Wang Z, Liu W, Yang Y, Yu X and He J: LncRNA LINC00163 upregulation suppresses lung cancer development though transcriptionally increasing TCF21 expression. Am J Cancer Res. 8:2494–2506. 2018.PubMed/NCBI

48 

Yang Y, Wang F, Huang H, Zhang Y, Xie H and Men T: lncRNA SLCO4A1-AS1 promotes growth and invasion of bladder cancer through sponging miR-335-5p to upregulate OCT4. OncoTargets Ther. 12:1351–1358. 2019. View Article : Google Scholar

49 

Yu J, Han Z, Sun Z, Wang Y, Zheng M and Song C: LncRNA SLCO4A1-AS1 facilitates growth and metastasis of colorectal cancer through β-catenin-dependent Wnt pathway. J Exp Clin Cancer Res. 37:2222018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 18 Issue 5

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
Yu Y, Chen X and Cang S: Cancer‑related long noncoding RNAs show aberrant expression profiles and competing endogenous RNA potential in esophageal adenocarcinoma. Oncol Lett 18: 4798-4808, 2019
APA
Yu, Y., Chen, X., & Cang, S. (2019). Cancer‑related long noncoding RNAs show aberrant expression profiles and competing endogenous RNA potential in esophageal adenocarcinoma. Oncology Letters, 18, 4798-4808. https://doi.org/10.3892/ol.2019.10808
MLA
Yu, Y., Chen, X., Cang, S."Cancer‑related long noncoding RNAs show aberrant expression profiles and competing endogenous RNA potential in esophageal adenocarcinoma". Oncology Letters 18.5 (2019): 4798-4808.
Chicago
Yu, Y., Chen, X., Cang, S."Cancer‑related long noncoding RNAs show aberrant expression profiles and competing endogenous RNA potential in esophageal adenocarcinoma". Oncology Letters 18, no. 5 (2019): 4798-4808. https://doi.org/10.3892/ol.2019.10808