Open Access

Identification of a PLCE1‑regulated competing endogenous RNA regulatory network for esophageal squamous cell carcinoma

  • Authors:
    • Zhihao Yang
    • Yi Hui
    • Hao Peng
    • Hongpan Zhang
    • Menglu Li
    • Lingxie Song
    • Feng Li
    • Xiaobin Cui
  • View Affiliations

  • Published online on: January 4, 2021     https://doi.org/10.3892/or.2021.7921
  • Pages: 857-868
  • Copyright: © Yang 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

Phospholipase C epsilon 1 (PLCE1) and the competing endogenous RNA (ceRNA) network are crucial for tumorigenesis and the progression of esophageal squamous cell carcinoma (ESCC). However, whether PLCE1 can regulate the ceRNA network in ESCC has not been clarified. In the present study, we aimed to identify the PLCE1‑regulated ceRNA network and further elucidate the regulatory mechanisms by which ESCC is promoted. Microarray analysis was used to identify differentially expressed lncRNAs (DELs) and differentially expressed genes (DEGs) from three pairs of samples of PLCE‑silenced Eca109 and control Eca109 cells. Next, the ceRNA regulatory network was established and visualized in Cytoscape, and functional enrichment analysis was performed to analyze DEGs from ceRNAs. Protein‑protein interaction (PPI) networks among the DEGs were established by the STRING database to screen hub genes. Kaplan‑Meier survival analysis was used to validate hub genes. Finally, PLCE1‑related hub gene/lncRNA/miRNA axes were also constructed based on the ceRNA network. A total of 105 DELs and 346 DEGs were found to be dysregulated in the microarray data (|log2FC| >1.5, adjusted P<0.05). We constructed a PLCE1‑regulated ceRNA network that incorporated 12 lncRNAs, 43 miRNAs, and 169 mRNAs. Functional enrichment analysis indicated that the DEGs might be associated with ESCC onset and development. A PPI network was established, and 9 hub genes [WD and tetratricopeptide repeats 1 (WDTC1), heat shock protein family A (Hsp70) member 5 (HSPA5), N‑ethylmaleimide sensitive factor, vesicle fusing ATPase (NSF), fibroblast growth factor 2 (FGF2), cyclin dependent kinase inhibitor 1A (CDKN1A or P21), bone morphogenetic protein 2 (BMP2), complement C3 (C3), GM2 ganglioside activator (GM2A) and discs large MAGUK scaffold protein 4 (DLG4)] were determined from the network. Kaplan‑Meier survival analysis validated four hub genes (BMP2, CDKN1A, GM2A, and DLG4) that were treated as prognostic factors. Ultimately, hub gene/lncRNA/miRNA subnetworks were obtained based on the 4 hub genes, 13 DEmiRNAs, and 10 DELs. In conclusion, the PLCE1‑regulated ceRNA contributes to the onset and progression of ESCC and the underlying molecular mechanisms may provide insights into personalized prognosis and new therapies for ESCC patients.

Introduction

Esophageal squamous cell carcinoma (ESCC), the most common subtype of esophageal cancer (ESCA), is the sixth most common cause of cancer-related death in the world (1). The treatment of ESCC has improved over the past few decades, with techniques that primarily include surgical resection, chemotherapy, and radiotherapy (2,3). Despite advances in treatment, the 5-year overall survival (OS) rate for ESCC patients remains unsatisfactory (1). Recent investigations have found that several prognostic factors, such as tumor stage, smoking, and alcohol, can be used to predict the OS of ESCC patients, yet the prognosis is still poor (47). Since we still do not fully understand how ESCC occurs and progresses, it is urgent to explore the underlying molecular mechanisms and identify novel prognostic biomarkers and potential therapeutic targets for ESCC.

Phospholipase C epsilon 1 (PLCE1), a new ESCC susceptibility locus at chromosome 10q236, has been identified by genome-wide association studies (GWAS) in Chinese Han populations (8). It has been reported that PLCE1 mediates different external signals that are relevant to tumor stage and survival in hepatocellular carcinoma, gastric carcinoma, bladder cancer and colorectal cancer (912). Our previous investigations also indicated that PLCE1 at the level of mRNA and protein is highly increased in ESCC from Han and Kazakh populations and that the overexpression of PLCE1 is correlated with metastasis and tumor aggressiveness (1316). More importantly, PLCE1 is also associated with poor prognosis of patients with ESCC (13). Zhai and coworkers also confirmed that when PLCE1 is knocked out, the transcriptional activity of SNAIL is particularly repressed (17). Finally, the invasion and migration of ESCC are inhibited when PLCE1 is knocked out, suggesting that PLCE1 could be regarded as a marker for the molecular typing of ESCC (16), and we have also confirmed that PLCE1 can function as an oncogene by enhancing ESCC cell proliferation and inhibiting apoptosis. Nevertheless, while PLCE1 is overexpressed in ESCC, the specific mechanism by which it promotes tumorigenesis has not been clarified.

Competitive endogenous RNA (ceRNA) is a newly proposed regulatory mechanism by which lncRNAs can adsorb microRNAs (miRNAs), to participate in the regulation of target genes (18), thus playing an essential role in many cancers, such as ESCC (19), lung cancer (20) and gallbladder cancer (21). Therefore, it is clear that lncRNA-miRNA-mRNA networks participate in the initiation and development of not only ESCC but also other types of cancers. Nevertheless, no report has indicated that PLCE1-regulated ceRNA is involved in ESCC progression according to microarray or sequencing data.

In this study, microarray analysis was used to identify differentially expressed lncRNAs (DELs) and differentially expressed genes (DEGs) from three pairs of samples of PLCE-silenced Eca109 and control Eca109 cells. The DELs and DEGs from microarray data were used to establish a ceRNA regulatory network. Subsequently, we predicted the cellular functions and pathways of DEGs involved in the ceRNA network by GO and KEGG Pathway analysis. Protein-protein interaction (PPI) analysis of DEGs was performed to screen for crucial hub genes. Finally, PLCE1-related hub gene/lncRNA/miRNA axes were obtained from the ceRNA, which provide insights into the regulatory mechanisms of ESCC.

Materials and methods

Cell culture and PLCE1-silenced microarray

The Eca109 cells purchased from Shanghai Institute of Biochemistry and Cell Biology of the Chinese Academy of Sciences were cultured at 37°C in DMEM or RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc.), containing 10% FBS (Sigma-Aldrich; Merck KGaA) in moist air containing 5% CO2. Stable PLCE1 siRNA (siRNA-PLCE1, 5′-GGGUCUUGCCAGUCGACUATT-3′) transfected cells or control cells were previously constructed (16) by the transfection of PLCE1 siRNA or si-RNA control, respectively. Briefly, Eca109 cells were grown in 6-well plates and transfected with si-PLCE1 and siRNA control using HiPerFect Transfection Reagent (Qiagen) according to the manufacturer's instructions. At 72 h after transfection, cells were harvested for western blot analyses. In addition, three pairs of PLCE-silenced Eca109 cells and controls were selected for a full transcription chip, Affymetrix Clariom™ D Array (Affymetrix, Thermo Fisher Scientific, Inc.) which was performed by Compass Biotechnology Co., Beijing, China. Array signal intensities were analyzed with Expression Console Software (version 1.4.1, Affymetrix; Thermo Fisher Scientific, Inc.). Furthermore, we used another si-PLCE1 chip in EC9706 cells, the Affymetrix GeneChip Human genome 3 (Affymetrix, Thermo Fisher Scientific, Inc.), to validate results of the si-PLCE1 chip in Eca109 cells.

Western blot analysis

Total protein was extracted using radio immunoprecipitation assay (RIPA) buffer. Protein (50 µg) was loaded onto SDS-PAGE gels (8–10%) and electrophoresed (200 V, 2 h), transferred onto PVDF membranes (BD Biosciences) and hybridized with primary antibodies, against β-actin (diluted 1:1,000, Zhongshan), PLCE1 (diluted 1:200, Santa Cruz Biotechnology, Inc.) followed by incubation with the appropriate secondary antibodies diluted to 1:10,000 (Zhongshan).

Cell proliferation and apoptosis assay

Ethynyl-2′-deoxyuridine (EdU) assays were conducted to examine cell proliferation using a Beyotime Assay Kit (C0071S, Shihezi). Cells (1×105/well) were cultured using si-PLCE1 for 72 h, fixed by 4% formaldehyde for 25 min at 4°C and 1X Click Reaction Buffer (Beyotime Institute of Biotechnology) was used to process them for approximately 30 min in the dark, followed by DAPI staining. After being rinsed with PBS, images were obtained under a fluorescence microscope (×200 magnification).

Mitochondrial membrane potential was measured using JC-1. JC-1 can potential-dependently accumulate in mitochondria as indicated by a color change from green to red. Therefore, the decline in the red/green fluorescence intensity ratio indicates mitochondrial depolarization. After PLCE1 silencing treatment, the medium was replaced with 500 µl fresh medium, including 5 µg/ml JC-1 (C2005, Beyotime Institute of Biotechnology). One hour after labeling in the dark, the images were obtained under a fluorescence microscope (×200 magnification).

TUNEL assays applying the Beyotime Apoptosis Kit (C1086, Beyotime Institute of Biotechnology) were carried out on PLCE1-silenced cells. Cells (1×105/well) were cultured with si-PLCE1 for 72 h, fixed by 4% formaldehyde for 25 min at room temperature, and permeabilized with 0.3% Triton-X-100 for 5 min. Before being stained with DAPI for 5 min at room temperature and washed with PBS, cells were mixed with TUNEL reagent for 60 min at 37°C. Images were obtained under a fluorescence microscope (×200 magnification).

Identification of DELs and DEGs

We used Perl and R scripts (https://www.r-project.org/; http://www.perl.org/) for all data processing and normalized analysis and usage. Furthermore, the lncRNA and mRNA expression profiles were analyzed, respectively, to screen DELs and DEGs in the microarray. Dysregulated genes were identified by using the edgeR package of the R language (22). A |log2foldchange (log2FC)|>1.5 and adjusted P<0.05 were set as the thresholds to define differences.

ceRNA network

To better assess the relationships between PLCE1 and ceRNAs in ESCC, we constructed a ceRNA network based on the theory that miRNAs can lead to gene silencing by interacting with mRNAs, whereas lncRNAs can function as miRNA sponges to regulate gene expression by competitively binding to miRNAs. We used miRcode tools (23) (http://www.mircode.org/) to predict which DELs and DEmiRNAs could interact with each other. DEGs targeted by DEmiRNAs were retrieved based on several databases, including Targetscan (24), miRTarBase (25), and miRDB (26). Furthermore, we built the ceRNA network relationship through the above DELs, DEmiRNAs, and DEGs, and the established network was visualized by Cytoscape software (27).

Functional enrichment analysis

To identify the function of DEGs, Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed using Metascape (28). P-value <0.05 was chosen as the cutoff criterion. Significant GO terms with similar functions were visualized as interaction networks using the Enrichment Map plugin in Cytoscape (27).

Establishment of the PPI network and hub gene subnetworks

The DEGs were extracted by the STRING database (29) (https://string-db.org/) to assess the interactive relationships of DEGs from ceRNA, the network was visualized by Cytoscape, and hub genes were obtained by screening the degree of connectivity of each node in the network. Lastly, we established hub gene/lncRNA/miRNA networks to explore the molecular mechanisms.

Statistics and analysis procedure

Statistical analyses were performed with R software (https://www.r-project.org/; version 3.6.1). Results are expressed as mean ± standard deviation. P<0.05 was regarded as a statistically significant difference. Differences between means were investigated by Student's t-test within two groups. The survival value of hub genes were analyzed using Kaplan-Meier (KM) survival analysis, followed by a log-rank test. Fig. 1 demonstrates the analysis procedure of the data mining processes.

Results

Validation of PLCE1 knockdown

To confirm the oncogene function of PLCE1, si-PLCE1 was made to interfere with its expression in Eca109 cells. The expression of PLCE1 was determined by western blot analysis after transfection. The results revealed that PLCE1 protein expression levels were highly reduced in the PLCE1-silenced (si-PLCE1) cells compared with the controls (si-con) (Fig. S1), suggesting that the expression of PLCE1 was successfully silenced after siRNA transfection of Eca109 cells.

Identification of DELs and DEGs

In this study, we used a microarray to study the expression of DELs and DEGs in three paired PLCE1-silenced ECa109 cells and controls (Fig. 2A-D). In total, 105 lncRNAs and 346 mRNAs were identified as dysregulated between two groups from the microarray (|log2FC|>1.5, adjusted P-value <0.05).

Functional enrichment analysis of PLCE1-related DEGs

Functional enrichment analysis was used to explore the role of DEGs (adjusted P-value <0.05). The GO analysis indicated that upregulated genes were related with negative regulation of cell growth (GO: 0030308), negative regulation of protein homodimerization activity (GO: 0090074), and cellular lipid catabolic processes (GO: 0044242) (Fig. 3A and B). In contrast, downregulated genes were related with positive regulation of protein serine/threonine kinase activity (GO: 0071902), positive regulation of neurogenesis (GO: 0050769) and I-kappaB kinase/NF-kappaB signaling (GO: 0007249) (Fig. 4A and B). Furthermore, the pathway analysis revealed that upregulated genes were involved in the metabolism of lipids, asparagine N-linked glycosylation, and constitutive signaling by AKT1 E17K in cancer (Fig. 3A and B); and downregulated genes were enriched in MAPK signaling pathways and the activation of anterior HOX genes in hindbrain development during early embryogenesis (Fig. 4A and B).

PLCE1 enhances proliferation and inhibits apoptosis in ESCC in vitro

To investigate the effect of PLCE1 on ESCC growth and apoptosis in vitro, several phenotype experiments were incorporated in PLCE1-silenced Eca109 cells. As shown in Fig. 5A-C, TUNEL and JC-1 staining experiments showed that the apoptosis rate of ESCC cells was markedly increased, whereas proliferation was significantly suppressed according to the EdU assays when PLCE1 was knocked down in the Eca109 ESCC cells. This indicated that PLCE1 in Eca109 cells can induce proliferation and attenuate apoptosis.

lncRNA/miRNA/mRNA ceRNA networks and functional assessment

To fully comprehend the role of DELs and DEGs by differentially expressed miRNAs (DEmiRNAs), a ceRNA network was constructed. A total of 566 interaction pairs were determined. After intersecting with the DEmiRNAs, 123 DEL/DEmiRNA pairs remained, containing 12 DELs and 43 DEmiRNAs. First, we searched for 105 DELs in the miRCode database and obtained 123 interacting pairs of lncRNAs and miRNAs using the Perl language, which contained 12 DELs and 43 DEmiRNAs. We further searched for mRNAs targeted by the 43 DEmiRNAs in TargetScan, miRDB, and miRanda databases, and selected those that overlapped with the identified DEGs. Finally, a total of 443 DEmiRNA/DEG interactions were added to the ceRNA network (Fig. 6). We also investigated the potential function of these DEGs in the ceRNA network using Metascape analysis (28) and found that they mainly participate in fatty acid metabolic processes, DNA damage response and negative regulation of signal transduction by p53 (Fig. S2; Table I). The pathway analysis indicated that DEGs were involved in metabolism of lipids and regulation of TP53 expression and degradation (Fig. S2; Table I).

Table I.

Functional enrichment analysis of DEGs involved in the ceRNA network.

Table I.

Functional enrichment analysis of DEGs involved in the ceRNA network.

GODescriptionCountLog10(P)Log10(q)
GO:0045664Regulation of neuron differentiation15−4.65−0.84
GO:0010575Positive regulation of vascular endothelial growth factor production4−4.47−0.84
GO:0006631Fatty acid metabolic process11−4.39−0.84
GO:0019228Neuronal action potential4−4.19−0.84
R-HSA-6798695Neutrophil degranulation12−4.18−0.84
GO:0046466Membrane lipid catabolic process4−4.09−0.84
R-HSA-556833Metabolism of lipids15−4.07−0.84
GO:0021675Nerve development5−3.8−0.73
GO:0006978DNA damage response, signal transduction by p53 class mediator resulting in transcription of p213−3.78−0.73
GO:0051961Negative regulation of nervous system development9−3.68−0.73
GO:0007169Transmembrane receptor protein tyrosine kinase signaling pathway14−3.62−0.72
GO:0001763Morphogenesis of a branching structure7−3.55−0.7
GO:0098876Vesicle-mediated transport to the plasma membrane5−3.37−0.57
GO:0045807Positive regulation of endocytosis6−3.31−0.57
GO:1901797Negative regulation of signal transduction by p53 class mediator3−2.91−0.42
GO:0030308Negative regulation of cell growth6−2.89−0.42
GO:0051235Maintenance of location8−2.87−0.42
GO:0019216Regulation of lipid metabolic process9−2.86−0.42
GO:0001893Maternal placenta development3−2.84−0.41
R-HSA-6806003Regulation of TP53 expression and degradation3−2.77−0.37

[i] DEGs, differentially expressed genes; ceRNA, competing endogenous RNA.

Establishment of the PPI network and hub gene subnetwork

PPI network was constructed that contained 93 nodes and 107 edges. According to the network, there were 9 major hub genes (WD and tetratricopeptide repeats 1 (WDTC1), heat shock protein family A (Hsp70) member 5 (HSPA5), N-ethylmaleimide sensitive factor, vesicle fusing ATPase (NSF), fibroblast growth factor 2 (FGF2), cyclin dependent kinase inhibitor 1A (CDKN1A or P21), bone morphogenetic protein 2 (BMP2), complement C3 (C3), GM2 ganglioside activator (GM2A) and discs large MAGUK scaffold protein 4 (DLG4)], and the gene with the highest degree (degree=7), can be observed in Fig. 7A and B, indicating that these genes may be the key genes that can eventually elicit a significant regulatory role in these networks. In other words, they belong to the lncRNA-miRNA-hub gene network.

Kaplan-Meier survival analysis

There were 4 hub genes (BMP2, CDKN1A, GM2A, and DLG4) that were treated as prognostic factors by the Kaplan-Meier plotter analysis tool (P<0.05). However, the remaining genes were not related to prognosis. High expression levels of BMP2 (Fig. 8B) [hazard ratio (HR)=4.37, 95% CI: 1.43–7.95, P=0.0032], GM2A (Fig. 8D) (HR=4.39, 95% CI: 1.03–18.69, P=0.029), and CDKN1A (Fig. 8A) (HR=2.34, 95% CI: 1.03–5.34, P=0.038) were related to poor OS in ESCC patients, while low expression of DLG 4 (Fig. 8C) (HR=0.29, 95% CI: 0.09–0.98, P=0.035) was significantly associated with a favorable prognosis in ESCC patients.

Construction of a ceRNA network of 4 prognostic hub genes

To identify prognostic ceRNA networks, we constructed a prognostic hub gene/lncRNA/miRNA network (Fig. S3), including 4 hub genes, 13 miRNAs, and 10 lncRNAs.

Discussion

Esophageal squamous cell carcinoma (ESCC) is a malignant tumor that seriously threatens human health (30,31). To date, no biomarkers have been wholly proven to predict the survival of patients with ESCC. Therefore, to improve prognosis, it is imperative to look for reliable biomarkers that identify high risk patients with ESCC. Our previous research confirmed that phospholipase C epsilon 1 (PLCE1) is associated with poor prognosis in patients with ESCC (13) and can serve as an oncogene by improving cell proliferation and repressing apoptosis. Additionally, growing evidence has indicated that competitive endogenous RNA (ceRNA) mechanisms play a critical role in cancer initiation and progression. However, the underlying mechanisms by which PLCE1-regulated ceRNAs function in ESCC are still largely elusive.

Here, we constructed a PLCE1-related ceRNA network obtained from microarray. We also predicted the biological functions and pathways of DEGs involved in the ceRNA network by GO and KEGG pathway analysis. PPI networks among the DEGs from the ceRNA were constructed using the STRING database, and hub genes were screened from the PPI network. Finally, PLCE1-related lncRNA/miRNA/hub gene axes were obtained from the ceRNA network, which could potentially contribute to personalized prognosis and new therapies for ESCC patients.

Numerous studies have recently shown that the expression of lncRNAs is dysregulated in ESCC and represents opportunities for cancer prognosis, diagnosis, and therapeutics, suggesting that lncRNAs could act as tumor-associated biomarkers. For example, Liang et al showed that CASC9 is highly increased in ESCC tissues and cell lines and is associated with overall survival (OS) (32). More importantly, increasing evidence has uncovered that lncRNAs can serve as miRNA sponges during ESCC onset and development. For instance, Li et al (33) revealed that regulation of ZFAS1 by overexpression of miRNA-124 may mediate ESCC proliferation, invasion, migration, and apoptosis. In the present study, a total of 12 lncRNAs (SNORA9, SNORD3D, HCP5, LENG8-AS1, HCG15, CEACAM22P, SHC1P1, HOTAIRM1, TSPY25P, DNAJC3-AS1, ABCC6P2, and RAB43P1) were found to be involved in the ceRNA network. Among them, HCP5 serves as a ceRNA to negatively regulate miR-203 and enhance SNAI expression, thus it is a candidate therapeutic target for lung adenocarcinoma (34). Additionally, a previous report indicated that HOTAIRM1 functions as a ceRNA to regulate the expression of miR-129-5p and miR-495-3p in glioma progression and is positively correlated with OS in glioma patients (35). However, in our analysis, the PLCE1 expression level had a negative correlation with HOTAIRM1, but a positive relationship with HCG15 in Eca109 cells, which has been effectively validated by ESCA tissues based on the GEPIA (36) (Fig. S4A and B).

miRNAs, as a class of non-coding single-stranded RNA molecules encoded by endogenous genes with a length of about 22 nucleotides, are involved in regulating the degradation of targeted mRNAs and inhibiting mRNA translation (37), which allows them to further regulate cancer proliferation, differentiation, apoptosis, and migration (38). In our previous research, 14 DEmiRNAs in the ceRNA network were identified, among which hsa-miR-17-5p, hsa-miR-22-3p, and hsa-miR-1297 have been reported to be involved in tumorigenesis (3941); hsa-miR-301b-3p and hsa-miR-455-5p have also been verified to be highly expressed in ESCA from the TCGA database (23) (Fig. S5).

To further identify the role of the ceRNA network, we established a PPI network that identified nine hub genes [WDTC1, HSPA5, NSF, FGF2, CDKN1A (P21), BMP2, C3, GM2A, and DLG4], indicating that PLCE1 could regulate these genes by a ceRNA mechanism. In this study, the PLCE1 expression level had a strongly negative relationship with HSPA5, NSF, CDKN1A, BMP2, GM2A, C3 and DLG4, while it had a positive correlation with WDTC1 and FGF2 in Eca109 cells, Our previous si-PLCE1 chip results in EC9706 also indicated that the PLCE1 expression level had an inverse relationship with HSPA5, NSF, FGF2, CDKN1A, BMP2, C3, and DLG4, while it correlated positively with WDTC1 and GM2A (Table II) (42), suggesting that their results are highly consistent with each other. Numerous reports have also demonstrated that CDKN1A (P21) and BMP2 (43) in many cancers can inhibit the cell cycle at the G1 and G2 phases to elicit tumor suppression (4447). Intriguingly, our results also confirmed that PLCE1 inhibition may lead to an increase in CDKN1A (P21) and BMP2. And more importantly, CDKN1A-associated ceRNA network includes CDKN1A/hsa-miR-22-3p/HOTAIRMI and CDKN1A/hsa-miR-206-5p/HOTAIRMI; BMP2-associated ceRNA network harbors BMP2/hsa-miR-17-5p/HOTAIRMI, BMP2/hsa-miR-20b-5p /HOTAIRMI, BMP2/hsa-miR-17-5p/HCG15 and BMP2/hsa-miR-20b-5p/HCG15 in our research, revealing that PLCE1 triggers ESCC progression by decreasing CDKN1A (P21) and BMP2 expression via ceRNA. Functionally, DNA damage response, signal transduction by p53 class mediator resulting in transcription of p21 (GO: 0006978) and negative regulation of cortisol biosynthetic process (GO: 2000065) were enriched based on these two hub genes by means of Metascape (28). However, Kaplan-Meier plotter analysis showed that CDKN1A and BMP2 afforded poor OS in ESCC patients. One obvious reason for this is the difference between esophageal squamous cell carcinoma cells and tissue. In addition, too few samples were used for survival analysis. Collectively, these findings indicate that PLCE1 triggers ESCC progression by meditating these hub genes via ceRNA.

Table II.

Differentially expressed hub genes in PLCE1 siRNA-treated ESCC cells compared with control cells.

Table II.

Differentially expressed hub genes in PLCE1 siRNA-treated ESCC cells compared with control cells.

Gene symbol log2(Fold change)Attribute Gene symbolFold changeAttribute
Eca109-associatedWDTC1−2.64Down EC9706-associatedWDTC10.78Down
microarrayHSPA51.92UpmicroarrayHSPA52.31Up
NSF2.44Up NSF1.16Up
FGF2−2.29Down FGF22.22Up
CDKN1A3.86Up CDKN1A3.27Up
BMP21.85Up BMP21.87Up
GM2A2.25Up GM2A0.73Down
C32.24Up C31.45Up
DLG42.91Up DLG41.14Up

[i] ESCC, esophageal squamous cell carcinoma; PLCE1, phospholipase C epsilon 1; WDTC1, WD and tetratricopeptide repeats 1; HSPA5, heat shock protein family A (Hsp70) member 5; NSF, N-ethylmaleimide sensitive factor, vesicle fusing ATPase; FGF2, fibroblast growth factor 2; CDKN1A or P21, cyclin dependent kinase inhibitor 1A; BMP2, bone morphogenetic protein 2; C3, complement C3; GM2A, GM2 ganglioside activator; DLG4, discs large MAGUK scaffold protein 4.

To date, most research involved in ceRNAs in cancer development has comprised lncRNAs, miRNAs and coding genes. There are few reports investigating whether other genes regulate ceRNAs in cancer. However, Sun's group (48) found that TGF-β activates lncRNA-ATB and upregulates ZEB1 or ZEB2 by means of competitive binding of miR-200; thereby inducing epithelial-mesenchymal transition in vivo and in vitro in hepatocellular carcinoma. In our study, PLCE1 was also found to be involved in the regulation of ceRNAs to promote the development of ESCC. Despite revealing the relationship between PLCE1 and ceRNAs in ESCC, for the first time, there are some limitations to this investigation. The results were obtained from microarray data and have not yet been verified by experimental means. In addition, a limited number of samples were used for the bioinformatics analysis.

In conclusion, for the first time, we comprehensively revealed the relationship between PLCE1 and ceRNAs in ESCC and constructed a PLCE1-regulated ceRNA network, which highlights the role of PLCE1 in promoting ESCC. Consequently, PLCE1 is a promising biomarker and potential novel therapeutic target for ESCC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This study was supported by Grants from the National Natural Science Foundation of China (nos. 81460362, 81773116, 81760436, 81560399, 81860518, and 81360358), The Medical and Health Science and Technology Project of Suzhou High Tech Zone (no. 2017Z006), The International Science and Technology Cooperation Project of Shihezi University (GJHZ201702), the Applied Basic Research Projects of Xinjiang Production and Construction Corps (2016AG020), the Youth Science and Technology Innovation Leading Talents Project of Corps (2017CB004), The Major Science and Technology Projects of Shihezi University (gxjs2014-zdgg06), The High-Level Talent Project of Shihezi University (RCZX201533), The Foundation for Distinguished Young Scholars of Shihezi University (2015ZRKXJQ02) and the Scientific Research Project of Shihezi University (ZZZC201702A).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

The project was designed and conceived by HP, ZY and ML. HZ, LS and YH analyzed the data. ZY wrote the manuscript. FL and XC guided the execution of the study and revised the paper. All authors read and approved the final version of this 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:

ESCC

esophageal squamous cell carcinoma

PLCE1

phospholipase C epsilon 1

ceRNA

competing endogenous RNA

PPI

protein-protein interaction

lncRNAs

long non-coding RNAs

mRNAs

messenger RNAs

GWAS

genome-wide association studies

DEGs

differentially expressed genes

DELs

differentially expressed lncRNAs

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

KM

Kaplan-Meier

HR

hazard ratio

CI

confidence interval

References

1 

Stein HJ, Feith M, Bruecher BL, Naehrig J, Sarbia M and Siewert JR: Early esophageal cancer: Pattern of lymphatic spread and prognostic factors for long-term survival after surgical resection. Ann Surg. 242:566–575. 2005.PubMed/NCBI

2 

Bollschweiler E, Plum P, Mönig SP and Hölscher AH: Current and future treatment options for esophageal cancer in the elderly. Expert Opin Pharmacother. 18:1001–1010. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Suntharalingam M, Winter K, Ilson D, Dicker AP, Kachnic L, Konski A, Chakravarthy AB, Anker CJ, Thakrar H, Horiba N, et al: Effect of the addition of cetuximab to paclitaxel, cisplatin, and radiation therapy for patients with esophageal cancer: The NRG oncology RTOG 0436 Phase 3 randomized clinical trial. JAMA Oncol. 3:1520–1528. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Shao L, Zhang B, Wang L, Wu L, Kan Q and Fan K: MMP-9-cleaved osteopontin isoform mediates tumor immune escape by inducing expansion of myeloid-derived suppressor cells. Biochem Biophys Res Commun. 493:1478–1484. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Matsuda S, Tsubosa Y, Sato H, Takebayashi K, Kawamorita K, Mori K, Niihara M, Tsushima T, Yokota T, Onozawa Y, et al: Comparison of neoadjuvant chemotherapy versus upfront surgery with or without chemotherapy for patients with clinical stage III esophageal squamous cell carcinoma. Dis Esophagus. 30:1–8. 2017.

6 

Cheng YF, Chen HS, Wu SC, Chen HC, Hung WH, Lin CH and Wang BY: Esophageal squamous cell carcinoma and prognosis in Taiwan. Cancer Med. 7:4193–4201. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Sun Y, Zhang T, Wu W, Zhao D, Zhang N, Cui Y, Liu Y, Gu J, Lu P, Xue F, et al: Risk factors associated with precancerous lesions of esophageal squamous cell carcinoma: A screening study in a high risk chinese population. J Cancer. 10:3284–3290. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Wang LD, Zhou FY, Li XM, Sun LD, Song X, Jin Y, Li JM, Kong GQ, Qi H, Cui J, et al: Genome-wide association study of esophageal squamous cell carcinoma in Chinese subjects identifies susceptibility loci at PLCE1 and C20orf54. Nat Genet. 42:759–763. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Cheng Y, Xing SG, Jia WD, Huang M and Bian NN: Low PLCE1 levels are correlated with poor prognosis in hepatocellular carcinoma. Onco Targets Ther. 10:47–54. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Wang Q, Chen P, Chen D, Liu F and Pan W: Association between phospholipase C epsilon gene (PLCE1) polymorphism and colorectal cancer risk in a Chinese population. J Int Med Res. 42:270–281. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Liang P, Zhang W, Wang W, Dai P, Wang Q, Yan W, Wang W, Lei X, Cui D and Yan Z: PLCE1 polymorphisms and risk of esophageal and gastric cancer in a northwestern chinese population. Biomed Res Int. 2019:97651912019. View Article : Google Scholar : PubMed/NCBI

12 

Ou L, Guo Y, Luo C, Wu X, Zhao Y and Cai X: RNA interference suppressing PLCE1 gene expression decreases invasive power of human bladder cancer T24 cell line. Cancer Genet Cytogenet. 200:110–119. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Chen Y, Wang D, Peng H, Chen X, Han X, Yu J, Wang W, Liang L, Liu Z, Zheng Y, et al: Epigenetically upregulated oncoprotein PLCE1 drives esophageal carcinoma angiogenesis and proliferation via activating the PI-PLCepsilon-NF-κB signaling pathway and VEGF-C/ Bcl-2 expression. Mol Cancer. 18:12019. View Article : Google Scholar : PubMed/NCBI

14 

Cui XB, Li S, Li TT, Peng H, Jin TT, Zhang SM, Liu CX, Yang L, Shen YY, Li SG, et al: Targeting oncogenic PLCE1 by miR-145 impairs tumor proliferation and metastasis of esophageal squamous cell carcinoma. Oncotarget. 7:1777–1795. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Cui XB, Peng H, Li RR, Mu JQ, Yang L, Li N, Liu CX, Hu JM, Li SG, Wei Y, et al: MicroRNA-34a functions as a tumor suppressor by directly targeting oncogenic PLCE1 in Kazakh esophageal squamous cell carcinoma. Oncotarget. 8:92454–92469. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Chen Y, Xin H, Peng H, Shi Q, Li M, Yu J, Tian Y, Han X, Chen X, Zheng Y, et al: Hypomethylation-linked activation of PLCE1 impedes autophagy and promotes tumorigenesis through MDM2-mediated ubiquitination and destabilization of p53. Cancer Res. 80:2175–2189. 2020.PubMed/NCBI

17 

Zhai S, Liu C, Zhang L, Zhu J, Guo J, Zhang J, Chen Z, Zhou W, Chang T, Xu S, et al: PLCE1 promotes esophageal cancer cell progression by maintaining the transcriptional activity of snail. Neoplasia. 19:154–164. 2017. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Hu X, Li Y, Kong D, Hu L, Liu D and Wu J: Long noncoding RNA CASC9 promotes LIN7A expression via miR-758-3p to facilitate the malignancy of ovarian cancer. J Cell Physiol. 234:10800–10808. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Yang J, Qiu Q, Qian X, Yi J, Jiao Y, Yu M, Li X, Li J, Mi C, Zhang J, et al: Long noncoding RNA LCAT1 functions as a ceRNA to regulate RAC1 function by sponging miR-4715-5p in lung cancer. Mol Cancer. 18:1712019. View Article : Google Scholar : PubMed/NCBI

21 

Chen J, Yu Y, Li H, Hu Q, Chen X, He Y, Xue C, Ren F, Ren Z, Li J, et al: Long non-coding RNA PVT1 promotes tumor progression by regulating the miR-143/HK2 axis in gallbladder cancer. Mol Cancer. 18:332019. View Article : Google Scholar : PubMed/NCBI

22 

Robinson MD, McCarthy DJ and Smyth GK: edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Tomczak K, Czerwińska P and Wiznerowicz M: The cancer genome atlas (TCGA): An immeasurable source of knowledge. Contemp Oncol (Pozn). 19(1A):A68–A77. 2015.

24 

Arora A and Simpson DA: Simpson, Individual mRNA expression profiles reveal the effects of specific microRNAs. Genome Biol. 9:R822008. View Article : Google Scholar : PubMed/NCBI

25 

Huang HY, Lin YC, Li J, Huang KY, Shrestha S, Hong HC, Tang Y, Chen YG, Jin CN, Yu Y, et al: miRTarBase 2020: Updates to the experimentally validated microRNA-target interaction database. Nucleic Acids Res. 48:D148–D154. 2019.

26 

Chen Y and Wang X: miRDB: An online database for prediction of functional microRNA targets. Nucleic Acids Res. 48:D127–D131. 2019. View Article : Google Scholar

27 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C and Chanda SK: Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 10:15232019. View Article : Google Scholar : PubMed/NCBI

29 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47:D607–D613. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Zhang SW, Zheng RS, Zuo TT, Zeng HM, Chen WQ and He J: Mortality and survival analysis of esophageal cancer in China. Zhonghua Zhong Liu Za Zhi. 38:709–715. 2016.(In Chinese). PubMed/NCBI

31 

Domper Arnal MJ, Ferrández Arenas Á and Lanas Arbeloa Á: Esophageal cancer: Risk factors, screening and endoscopic treatment in Western and Eastern countries. World J Gastroenterol. 21:7933–7943. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Liang Y, Chen X, Wu Y, Li J, Zhang S, Wang K, Guan X, Yang K and Bai Y: LncRNA CASC9 promotes esophageal squamous cell carcinoma metastasis through upregulating LAMC2 expression by interacting with the CREB-binding protein. Cell Death Differ. 25:1980–1995. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Li Z, Qin X, Bian W, Li Y, Shan B, Yao Z and Li S: Exosomal lncRNA ZFAS1 regulates esophageal squamous cell carcinoma cell proliferation, invasion, migration and apoptosis via microRNA-124/STAT3 axis. J Exp Clin Cancer Res. 38:4772019. View Article : Google Scholar : PubMed/NCBI

34 

Jiang L, Wang R, Fang L, Ge X, Chen L, Zhou M, Zhou Y, Xiong W, Hu Y, Tang X, et al: HCP5 is a SMAD3-responsive long non-coding RNA that promotes lung adenocarcinoma metastasis via miR-203/SNAI axis. Theranostics. 9:2460–2474. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Liang Q, Li X, Guan G, Xu X, Chen C, Cheng P, Cheng W and Wu A: Long non-coding RNA, HOTAIRM1, promotes glioma malignancy by forming a ceRNA network. Aging (Albany NY). 11:6805–6838. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Tang Z, Kang B, Li C, Chen T and Zhang Z: GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 47:W556–W560. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Gebert LFR and MacRae IJ: Regulation of microRNA function in animals. Nat Rev Mol Cell Biol. 20:21–37. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Kong YW, Ferland-McCollough D, Jackson TJ and Bushell M: microRNAs in cancer management. Lancet Oncol. 13:e249–e258. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Li Z, Peng Z, Gu S, Zheng J, Feng D, Qin Q and He J: Global analysis of miRNA-mRNA interaction network in breast cancer with brain metastasis. Anticancer Res. 37:4455–4468. 2017.PubMed/NCBI

40 

Dong HX, Wang R, Jin XY, Zeng J and Pan J: LncRNA DGCR5 promotes lung adenocarcinoma (LUAD) progression via inhibiting hsa-mir-22-3p. J Cell Physiol. 233:4126–4136. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Chen Z, Zhang M, Qiao Y, Yang J and Yin Q: MicroRNA-1297 contributes to the progression of human cervical carcinoma through PTEN. Artif Cells Nanomed Biotechnol. 46 (Suppl 2):1120–1126. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Cui X, Xin H, Peng H and Chen Y: Comprehensive bioinformatics analysis of the mRNA profile of PLCE1 knockdown in esophageal squamous cell carcinoma. Mol Med Rep. 16:5871–5880. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Vishnubalaji R, Yue S, Alfayez M, Kassem M, Liu FF, Aldahmash A and Alajez NM: Bone morphogenetic protein 2 (BMP2) induces growth suppression and enhances chemosensitivity of human colon cancer cells. Cancer Cell Int. 16:772016. View Article : Google Scholar : PubMed/NCBI

44 

Zou J, Li S, Chen Z, Lu Z, Gao J, Zou J, Lin X, Li Y, Zhang C and Shen L: A novel oral camptothecin analog, gimatecan, exhibits superior antitumor efficacy than irinotecan toward esophageal squamous cell carcinoma in vitro and in vivo. Cell Death Dis. 9:6612018. View Article : Google Scholar : PubMed/NCBI

45 

Shimizu H, Shiozaki A, Ichikawa D, Fujiwara H, Konishi H, Ishii H, Komatsu S, Kubota T, Okamoto K, Kishimoto M and Otsuji E: The expression and role of Aquaporin 5 in esophageal squamous cell carcinoma. J Gastroenterol. 49:655–666. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Qin YR, Tang H, Xie F, Liu H, Zhu Y, Ai J, Chen L, Li Y, Kwong DL, Fu L and Guan XY: Characterization of tumor-suppressive function of SOX6 in human esophageal squamous cell carcinoma. Clin Cancer Res. 17:46–55. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Li L, Zhang C, Li X, Lu S and Zhou Y: The candidate tumor suppressor gene ECRG4 inhibits cancer cells migration and invasion in esophageal carcinoma. J Exp Clin Cancer Res. 29:1332010. View Article : Google Scholar : PubMed/NCBI

48 

Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC, et al: A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 25:666–681. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 45 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
Yang Z, Hui Y, Peng H, Zhang H, Li M, Song L, Li F and Cui X: Identification of a PLCE1‑regulated competing endogenous RNA regulatory network for esophageal squamous cell carcinoma. Oncol Rep 45: 857-868, 2021
APA
Yang, Z., Hui, Y., Peng, H., Zhang, H., Li, M., Song, L. ... Cui, X. (2021). Identification of a PLCE1‑regulated competing endogenous RNA regulatory network for esophageal squamous cell carcinoma. Oncology Reports, 45, 857-868. https://doi.org/10.3892/or.2021.7921
MLA
Yang, Z., Hui, Y., Peng, H., Zhang, H., Li, M., Song, L., Li, F., Cui, X."Identification of a PLCE1‑regulated competing endogenous RNA regulatory network for esophageal squamous cell carcinoma". Oncology Reports 45.3 (2021): 857-868.
Chicago
Yang, Z., Hui, Y., Peng, H., Zhang, H., Li, M., Song, L., Li, F., Cui, X."Identification of a PLCE1‑regulated competing endogenous RNA regulatory network for esophageal squamous cell carcinoma". Oncology Reports 45, no. 3 (2021): 857-868. https://doi.org/10.3892/or.2021.7921