Open Access

A putative competing endogenous RNA network in cisplatin‑resistant lung adenocarcinoma cells identifying potentially rewarding research targets

  • Authors:
    • Yepeng Li
    • Shiqing Huang
    • Zhongheng Wei
    • Bo Yang
  • View Affiliations

  • Published online on: March 27, 2020     https://doi.org/10.3892/ol.2020.11483
  • Pages: 4040-4052
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung adenocarcinoma (LUAD) is the most common type of non-small cell lung cancer and has a poor 5 year survival rate (<10%). Cisplatin is one of the most effective chemotherapeutic treatments for LUAD, even though it is of limited overall utility due to acquired drug resistance. To identify possible genetic targets for the mitigation of cisplatin resistance, gene expression data from cisplatin‑resistant cell lines were integrated with patient information. Expression data for cisplatin‑resistant and cisplatin‑sensitive A549 cell lines were obtained from the Gene Expression Omnibus database, while LUAD patient data was obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed mRNAs (DEmRNAs), microRNAs (DEmiRNAs) and long non‑coding RNAs (DElncRNAs) were identified between the cisplatin‑sensitive and cisplatin‑resistant cells. Using the TCGA patient data, 33 DEmRNAs associated with survival were identified. A total of 74 DElncRNAs co‑expressed with the survival‑associated DEmRNAs, and 11 DEmiRNAs that regulated the survival‑associated DEmRNAs, were also identified. A competing endogenous RNA (ceRNA) network was constructed based on the aforementioned results, which included 17 survival‑associated DEmRNAs, 9 DEmiRNAs and 16 DElncRNAs. This network revealed 8 ceRNA pathway axes possibly associated with cisplatin resistance in A549 cells. Specifically, the network suggested that the lncRNAs HOXD‑AS2, LINC01123 and FIRRE may act as ceRNAs to increase cisplatin resistance in human LUAD cells. Therefore, it was speculated that these lncRNAs represent potentially rewarding research targets.

Introduction

Lung cancer is a common malignancy and is the leading cause of cancer-associated mortality worldwide (1,2). Non-small cell lung cancer (NSCLC) accounts for ~85% of all lung cancers (3). Lung adenocarcinoma (LUAD) is the most common pathologic subtype of NSCLC in non-smoking males, and in all females (both smokers and non-smokers) (4,5). Although numerous resources have been directed towards the development of novel LUAD treatments, the prognosis of patients with advanced LUAD remains unsatisfactory, with a 5 year survival rate <10% in 2018 (6). LUAD is relatively sensitive to primary chemotherapy, but tumors rapidly acquire chemoresistance, leading to death for most patients (7,8).

Cisplatin is one of the most effective chemotherapeutic drugs and is used to treat various tumors, including testicular cancer, ovarian cancer, cervix carcinoma, breast cancer, prostate carcinoma, bladder cancer, lung cancer, melanoma and head-and-neck cancer (9,10). Cisplatin has a broad-spectrum anticancer activity, but its use is limited due to it causing severe side effects and due to a number of tumors acquiring cisplatin resistance (9). Although the side effects caused by cisplatin have been mildly alleviated by newly-developed antagonists (11), cisplatin resistance, which commonly originates from multiple cellular self-defense adaptations, often results in disease recurrence (12). Thus, the development of cisplatin resistance remains a substantial challenge for chemotherapeutics.

A major impediment to a comprehensive understanding of the molecular mechanisms underlying cisplatin-induced drug resistance is that most currently available results were generated using isolated cell lines. These studies can be misleading when extended to in vivo experiments and clinical trials (12,13). However, the integration of cell line data with clinical information, especially overall survival (OS) time, may improve this issue. For example, Zhao et al (14) used The Cancer Genome Atlas (TCGA) database to demonstrate that patients expressing high levels of the long non-coding RNA (lncRNA) HOMEOBOX A11 antisense RNA (HOXA11-AS) have shorter survival rates compared to the low expression level group; mechanistic experiments subsequently showed that the microRNA (miRNA/miR) targeted by HOXA11-AS affects cisplatin resistance in LUAD cells. The aforementioned study thus provides a framework for the identification of additional miRNAs associated with cisplatin resistance in LUAD cells.

In the present study, the framework of Zhao et al (14) was used to identify miRNA targets that may be useful for the mitigation of cisplatin resistance. The present study aimed to: i) Identify differentially expressed (DE) mRNAs (DEmRNAs), DEmiRNAs and DElncRNAs between two LUAD cell lines, namely A549 (cisplatin-sensitive) and A549-DDP (cisplatin-resistant), using data from the Gene Expression Omnibus (GEO) database (15); ii) quantify the expression levels of these DEmRNAs in samples of patients with LUAD using data downloaded from the TCGA database; iii) construct a competing endogenous RNA (ceRNA) network based on the aforementioned data; and iv) assess the associations between the elements of the ceRNA network and patient OS time to identify potential research targets.

Materials and methods

A549/A549-DDP data retrieval

Two miRNA and mRNA expression datasets were downloaded from the GEO database (16): GSE43249 (17), which was derived from the GPL14613 (miRNA-2) Affymetrix Multispecies miRNA-2 Array, and GSE43493 (18), which was derived from the GPL15314 Arraystar Human LncRNA microarray V2.0 (Agilent_033010 Probe Name version). Each dataset contained six samples, three that were cisplatin-sensitive and three that were cisplatin-resistant.

A549/A549-DDP data pre-processing

The raw microarray data were read using the package affy v1.52.0 (19) in R v3.4.3 (http://www.bioconductor.org/packages/release/bioc/html/affy.html), and was standardized using the robust multi-array average (20,21) method, with background adjustment, quantile normalization and summarization on a log2 scale. Using the platform annotation file, the probe was annotated and the unmatched probe was removed. To map different probes to the same mRNA or miRNA data, the mean value of each different probe was used as the final expression, and the genes were divided into mRNAs and lncRNAs following the guidelines of the HUGO Gene Nomenclature Committee (22).

Identification of DEmRNAs, DEmiRNAs and DElncRNAs

The DEmRNAs, DElncRNAs and DEmiRNAs were identified in the GEO datasets using the R package limma v3.34.9 (23). The classical Bayesian test was used to calculate P-values. mRNAs, lncRNAs and miRNAs were considered significantly differentially expressed if |log2 (fold change)|≥1 and P<0.05. To visualize the DEmRNAs, DElncRNAs and DEmiRNAs, heat maps and volcano maps were generated using the R packages ggplot2 (24) and heatmap2 (25), respectively.

TCGA patient data retrieval

RNA sequence data and clinical information (specifically, cisplatin treatment status and OS time) for 576 patients with LUAD were retrieved from the TCGA database (https://www.cancer.gov/tcga; accessed on August 29, 2017). The use of TCGA data in the present study is in accordance with TCGA publication guidelines (https://cancergenome.nih.gov/publications/publicationguidelines). Since the patient data used originated from the TCGA database, no further ethical approval was required.

Identification of DEmRNAs associated with patient survival

The expression levels of each of the identified DEmRNAs were quantified in each patient with LUAD. For each DEmRNA, patients were divided into a low- and a high-expression group based on mean gene expression. Kaplan-Meier survival curves were generated, and the DEmRNAs that were significantly associated with OS were identified using a log-rank test. P<0.05 was considered to indicate a statistically significant difference.

Functions and interactions of the survival-associated DEmRNAs

The Database for Annotation, Visualization and Integrated Discovery v.6.8 (26) was used to identify the Gene Ontology (GO) (27) terms and the Kyoto Encyclopedia of Genes and Genomes (KEGG) (28) pathways significantly enriched in the survival-associated DEmRNAs (i.e. those with P<0.05). The STRING database v10.5 (29) was used to predict protein-protein interactions (PPIs) of the survival-associated DEmRNAs. PPI scores ≤0.15 were considered of low confidence. Cytoscape v3.6.1 (30) was used to visualize the PPI network and to calculate node degrees.

Co-expression of DElncRNAs and survival-associated DEmRNAs

The Pearson correlation coefficient between each DElncRNA and each survival-associated DEmRNA was calculated. P-values were adjusted using the false discovery rate (FDR) to control for the effects of multiple comparisons. DElncRNAs and survival-associated DEmRNAs were considered to be co-expressed when |r|>0.95 and P<0.05 (FDR-adjusted). The functions of the co-expressed DElncRNAs were predicted based on the lncRNA-mRNA regulatory network; the R package clusterProfiler (31) was used to identify the pathways significantly enriched in the target genes of the co-expressed DElncRNAs. Pathways with Benjamini-Hochberg-adjusted P-values <0.05 were considered significantly enriched.

DEmiRNA regulatory networks and KEGG pathway enrichment

The target gene prediction module of miRWalk v2.0 (http://zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk2/miRretsys-self.html) (32) was used to identify possible target genes of the DEmiRNAs in eight databases miRWalk (http://mirwalk.umm.uni-heidelberg.de), Microt4 (http://mirtarbase.mbc.nctu.edu.tw/php/index.php), MiRanda (http://www.microrna.org/microrna/home.do), miRDB (http://www.mirdb.org/miRDB/policy.html), miRMap (https://mirmap.ezlab.org), PITA (http://genie.weizmann.ac.il/pubs/mir07/mir07_dyn_data.html), RNA22 (https://cm.jefferson.edu/rna22) and Targetscan (http://www.targetscan.org/vert_71/). To increase the reliability of the search results, only genes identified in ≥5 databases were used to construct the miRNA control network, which was visualized with Cytoscape v3.6.1 (30). The KEGG pathway enrichment of the predicted DEmiRNA target genes was investigated using clusterProfiler (31).

Construction of a ceRNA regulatory network

lncRNAs associated with the DEmiRNAs were identified using the prediction module of DIANA-LncBase v2 (33); only lncRNAs with scores >0.75 were included. Subsequently, a ceRNA network based on several data sources was constructed: The lncRNA-miRNA regulatory network; the miRNA-target mRNA regulatory network; and the DElncRNAs that were positively co-expressed with survival-associated DEmRNAs.

Association between OS time and the expression levels of selected lncRNA targets

Preliminary results demonstrated that the lncRNAs HOXD-AS2, LNC01123 and FIRRE appeared in one or more ceRNA axes. Therefore, the expression levels of these lncRNAs were quantified, as well as those of the co-expressed lncRNAs and survival-associated DEmRNAs, in non-LUAD tumors using the Gene Expression Profiling Interactive Analysis (GEPIA) server (34); GEPIA analyses RNA expression in 9,736 tumors and 8,587 normal samples from the TCGA and the Genotype-Tissue Expression projects.

Statistical analysis

The classical Bayesian test was used to test differentially expressed mRNAs, lncRNAs and miRNAs. DEmRNAs that were significantly associated with OS time were identified using the log-rank test. Fisher's exact test was applied for the GO enrichment of DEmRNAs associated with OS time. All comparisons were between cisplatin-resistant A549-DDP cells and cisplatin-sensitive A549 cells. P<0.05 was considered to indicate a statistically significant difference, unless otherwise specified. The statistical analysis was performed with R v.3.4.3 (35).

Results

DEmRNAs, DEmiRNAs and DEIncRNAs in the A549 and A549-DDP cell lines

A total of 842 mRNAs were identified to be differentially expressed between the A549 and A549-DDP cell lines. Among these DEmRNA, 245 (29.10%) were upregulated in the A549-DDP cell line compared with the A549 cell line, while 597 (70.90%) were downregulated (Fig. 1). In addition, 90 DElncRNAs and 18 DEmiRNAs were identified. Among these DEmiRNAs and DElncRNAs, 37 DElncRNAs (41.11%) and 8 DEmiRNAs (44.44%) were upregulated in the A549-DDP cell line compared with the A549 cell line, while 53 lncRNAs (58.89%) and 10 miRNAs (55.56%) were downregulated (Fig. 1; Table SI).

Survival-associated DEmRNAs

In the TCGA patient dataset, 86 patients treated with cisplatin were identified. These patients expressed 786 of the identified DEmRNAs. Among these, 33 DEmRNAs were significantly associated with OS time (Table I). Five upregulated DEmRNAs were associated with low OS time: Rh family B glycoprotein (RHBG), phosphatase orphan 2 (PHOSPHO2), activity regulated cytoskeleton associated protein (ARC), thioredoxin (TXN) and kinesin family member 26A (KIF26A; Fig. 2A-E; Table SII). Four other upregulated DEmRNAs were associated with high OS time: Zinc finger protein 417, neural cell adhesion molecule 1 (NCAM1), mediator complex subunit 12 (MED12) and ADP ribosylation factor 4 (ARF4; Fig. 2F-G; Table SII). These nine DEmRNAs comparing with the other 24 DEmRNAs, were more related to the prognosis of patients.

Table I.

Survival-associated mRNAs differentially expressed between cisplatin-resistant and cisplatin-sensitive cell lines.

Table I.

Survival-associated mRNAs differentially expressed between cisplatin-resistant and cisplatin-sensitive cell lines.

SymbolLog2 FCLow medianHigh medianDescription
MT1A−2.63 2.55Metallothionein 1A
VGF−2.504.152.55VGF nerve growth factor inducible
SARM1−2.222.864.15Sterile alpha and TIR motif containing 1
DPP4−2.212.864.15 Dipeptidyl-peptidase 4
SIRT4−1.852.87 Sirtuin 4
SH2B2−1.65 2.86SH2B adaptor protein 2
PER1−1.65 2.86PER1
FKBP1B−1.604.152.60FK506 binding protein 1B
FAM117A−1.552.86 Family with sequence similarity 117 member A
DIRAS3−1.50 2.60DIRAS family GTPase 3
STAC3−1.47 2.60SH3 and cysteine rich domain 3
MAGEH1−1.422.60 MAGE family member H1
RAB9B−1.402.87 RAB9B, member RAS oncogene family
SLC17A9−1.282.86 Solute carrier family 17 member 9
ADRA1D−1.214.152.55Adrenoceptor alpha 1D
ELOVL2−1.19 2.87ELOVL fatty acid elongase 2
DBP−1.192.87 D-box binding PAR bZIP transcription factor
NR1D1−1.102.87 Nuclear receptor subfamily 1 group D member 1
HSPA2−1.074.152.55Heat shock protein family A (Hsp70) member 2
GJA1−1.044.152.21Gap junction protein alpha 1
CEACAM6−1.034.152.55Carcinoembryonic antigen related cell adhesion molecule 6
ID4−1.014.152.60Inhibitor of DNA binding 4, HLH protein
NDST31.012.532.53N-deacetylase and N-sulfotransferase 3
ZNF4171.062.60 Zinc finger protein 417
PHOSPHO21.09 2.86Phosphatase, orphan 2
ARC1.11 2.55Activity regulated cytoskeleton associated protein
TXN1.134.152.60Thioredoxin
ARF41.152.86 ADP ribosylation factor 4
RHBG1.274.152.86Rh family B glycoprotein (gene/pseudogene)
EDEM11.282.86 ER degradation enhancing alpha-mannosidase like protein 1
KIF26A1.48 2.55Kinesin family member 26A
NCAM12.202.86 Neural cell adhesion molecule 1
MED122.462.534.15Mediator complex subunit 12

[i] Genes highlighted in bold are the five highly-expressed DEmRNAs associated with low OS time. The column ‘Symbol’ contains the gene name/ID; the column ‘LOG FC’ contains the log2 FC of up/downregulated genes; the column ‘Low median’ contains the median OS time of the low expression group; the column ‘High median’ contains the median OS time of the high expression group; and the column ‘Description’ contains the full gene name. OS time, overall survival; FC, fold change. If genes are without low median the overall survival time of their low expression group is out of the end of follow-up time point (e.g. five years). If genes are without high median the overall survival time of their high expression group is out of the end of follow-up time point.

Functional enrichment and PPIs of the survival-associated DEmRNAs

The GO terms most over-represented in the DEmRNAs annotations were ‘extracellular region’, ‘blood microparticle’, ‘extracellular space’ and ‘linoleic acid metabolic process’ (Fig. 3A; Table SIII). No KEGG pathways enriched in the DEmRNAs were identified (data not shown). The PPI network of the survival-associated DEmRNAs (Fig. 3B) contained 19 nodes and 26 interaction pairs, including 6 upregulated and 13 downregulated DEmRNAs.

Co-expression of DElncRNAs and survival-associated DEmRNAs

A total of 168 positively co-expressed pairs of DElncRNAs and survival-associated DEmRNAs were identified (74 DElncRNAs and 32 DEmRNAs). According to the DElncRNA-DEmRNA network, the target genes of the co-expressed DElncRNAs were over-represented in three KEGG pathways: ‘Protein processing in endoplasmic reticulum’, ‘mineral absorption’ and ‘circadian rhythm’ (Fig. 4; Table SIV).

DEmiRNA target gene prediction and functional enrichment analysis

Using miRWalk v2.0 (32), 11 DEmiRNAs targets, 17 survival-associated DEmRNA targets and 52 DEmiRNA/survival-associated DEmRNA pairs were identified in the DEmiRNA regulatory network (Fig. 5; Table SV). Several KEGG pathways, including ‘cell adhesion molecules (CAMs)’ and ‘circadian rhythm’, were enriched in the target genes (Fig. 6; Table SVI).

ceRNA regulatory network

Using Cytoscape (30), the DElncRNA-DEmiRNA regulatory network was combined with the DEmiRNA-DEmRNA network to obtain a DElncRNA-DEmiRNA-DEmRNA ceRNA network (Fig. 7A; Table SVII). The ceRNA network included 9 DEmiRNAs, 16 DElncRNAs, 17 target DEmRNAs and 87 pairs with a regulatory association. The present putative ceRNA network (Fig. 7B) included eight axes: HOXD-AS2/hsa-miR-152-3p/MED12, HOXD-AS2/hsa-miR-152-3P/NCAM1, LINC01123/hsa-miR-152-3p/MED12, LINC01123/hsa-miR-152-3P/NCAM1, LINC01123/hsa-miR-152-3P/ARF4, LINC01123/hsa-miR-762/NCAM1, LINC01123/hsa-miR-762/RHBG and FIRRE/hsa-miR-1231/ARF4. HOXD-AS2/has-miR-152-3p/ARF4 was not included in the axes list as HOXD-AS2/hsa-miR-152-3p/ARF4 didn't form a triangle and there was no line to connect HOXD-AS2 and ARF4 (Fig. 7B). The putative ceRNA axes included three lncRNAs (HOXD-AS2, LINC01123 and FIRRE), three miRNAs (hsa-miR-152-3p, hsa-miR-762 and hsa-miR-1231) and four genes (MED12, RHBG, NCAM1 and ARF4).

Association between OS time and the expression of selected lncRNA targets

The GEPIA analysis identified RHBG and NCAM1 as co-expressed genes of HOXD-AS2 and LNC01123, respectively. LNC1123 was positively co-expressed with RHBG in LUAD (R=0.18; Fig. 8A), lung squamous cell carcinoma (LUSC; R=0.14; Fig. 8B) and testicular germ cell tumors (TGCT; R=0.63; Fig. 8C). LNC1123 was positively co-expressed with NCAM1 in mesothelioma (MESO; R=0.27; Fig. 8D), pheochromocytoma and paraganglioma (PCPG; R=0.34; Fig. 8E), and TGCT (R=0.41; Fig. 8F). In addition, LNC1123 upregulation was associated with shorter patient OS time in head and neck squamous cell carcinoma (HNSCC; Fig. 9A), and in cervical squamous cell carcinoma and endocervical adenocarcinoma (P=0.057; Fig. 9B).

HOXD-AS2 upregulation was associated with shorter patient OS time in colon adenocarcinoma (COAD; Fig. 9C), brain lower-grade glioma (LGG; Fig. 9D), LUSC (Fig. 9E) and uveal melanoma (Fig. 9F). HOXD-AS2 was positively co-expressed with NCAM1 in COAD (R=0.15; Fig. 10A), LUAD (R=0.17; Fig. 10B), LUSC (R=0.09; Fig. 10C), TGCT (R=0.6; Fig. 10D), uterine corpus endometrial carcinoma (R=0.25; Fig. 10E) and uterine carcinosarcoma (R=0.37; Fig. 10F). No positive associations were identified between HOXD-AS2 and RHBG (data not shown).

FIRRE was positively co-expressed with NCAM1 in glioblastoma multiforme (R=0.47; Fig. 11A), liver hepatocellular carcinoma (LIHC; R=0.12; Fig. 11B), PCPG (R=0.29; Fig. 11C) and TGCT (R=0.49; Fig. 11D). FIRRE was positively co-expressed with RHBG in prostate adenocarcinoma (R=0.24; Fig. 11E) and TGCT (R=0.44; Fig. 11F). In addition, FIRRE upregulation was associated with shorter patient OS time in kidney renal clear cell carcinoma (Fig. 12A), kidney renal papillary cell carcinoma (Fig. 12B), LGG (Fig. 12C), LIHC (Fig. 12D), MESO (Fig. 12E) and pancreatic adenocarcinoma (Fig. 12F).

Discussion

The present analysis of gene expression patterns (based on the GEO datasets) identified 33 genes differentially expressed in cisplatin-resistant A549-DDP cells compared with in cisplatin-sensitive A549 cells. Among these, nine were upregulated in the cisplatin-resistant cells and 24 were downregulated. By cross-referencing these results with patient data from the TCGA dataset, five of these upregulated genes (PHOSPHO2, ARC, TXN, RHBG and KIF26A) were identified to be associated with poor OS time outcomes. These five genes may be useful potential targets for the reversal of cisplatin resistance in LUAD.

RHBG was identified as being of particular interest, as this gene also appeared in one of the axes of the putative ceRNA network generated in the present study. RHBG is a non-erythroid membrane glycoprotein of the Rh antigen family, and the mechanisms regulating RHBG expression remain poorly studied. Consistent with the results of the present study, RHBG has been demonstrated to be expressed in LIHC and COAD cell lines (36). Additionally, RHBG has been implicated in the growth of brain tumors in mice (37). RHBG deserves further study both as a possible maker of poor LUAD outcomes and as a potential target for cisplatin-resistance reversal therapy.

Furthermore, NCAM1, MED12 and ARF4 appeared in one or more ceRNA network axes, but increased expression levels of these genes were associated with improved OS time. NCAM1 encodes a cellular adhesion protein and is a well-known potential target of antibody-based cancer immunotherapies (38). In addition, NCAM1 has been identified as an immunohistochemical marker for lung neuroendocrine tumors (39), and it was recently proposed that the NCAM1-180 splice variant might be a useful marker for NSCLC (40). Furthermore, NCAM1 may be a useful biomarker and therapeutic target for acute myeloid leukemia (41), the follicular variant of papillary thyroid carcinoma (42) and breast cancer (43). Although NCAM1 has been associated with cisplatin resistance in ovarian cancer (44,45), the in vitro expression of NCAM improved the response of multiple myeloma cells to Bortezomib (Btz) treatment (46). Consistent with this, the present study revealed that NCAM upregulation was associated with improved patient OS time outcomes.

MED12 is a component of the CDK8 subcomplex. MED12 mutations are associated with tumorigenesis (47). Indeed, somatic mutations in MED12 exon 2 have been observed in uterine leiomyosarcoma, colorectal cancer (CRC) (47), uterine leiomyoma, breast fibroadenoma, phyllodes tumors and prostate cancer (48). Additionally, inhibition of MED12 expression has been associated with resistance to cisplatin and other chemotherapy drugs (49,50). This is consistent with the results of the present study, in which patients with high levels of MED12 had improved OS time.

ARF4 is a small guanine-binding protein that serves a role in vesicular trafficking (51). Although the results of the present study suggested that ARF4 upregulation was associated with improved patient outcomes, it has been previously reported that high expression levels of ARF4 in patients with breast cancer are significantly associated with increased risk of distant metastasis and shorter OS time. Conversely, ARF4 silencing reduces the colonization of the lung by metastatic breast cancer cells in vivo (51). These contradictory results suggest that the role of ARF4 in LUAD deserves further investigation.

The present putative ceRNA network included three miRNAs (hsa-miR-152-3p, hsa-miR-762 and hsa-miR-1231) across the eight axes. In several types of cancer (including prostate, ovarian and breast), miR-152 expression has been shown to reduce tumor cell viability and proliferation (5254). In addition, the suppression of miR-152 biogenesis increases cisplatin resistance in epithelial ovarian cancer (55). However, the overexpression of miR-152 increases cisplatin resistance and proliferation of nasopharyngeal carcinoma cells (56), while the overexpression of miR-762 stimulates the development of various tumors, including ovarian (57) and breast cancer (58). Conversely, the expression of miR-762 (in combination with other miRNAs) leads to the apoptosis of breast cancer cells (59). In the present study, miR-152 and miR-762 were downregulated in the cisplatin-resistant LUAD cells. Overall, these results suggested that the behavior of these miRNAs may vary in different types of cancer.

By contrast, miR-1231 expression consistently negatively regulates the progression of various types of cancer, including glioma (60,61), pancreatic cancer (62) and papillary thyroid cancer (63). Additionally, miR-1231 has been identified as an independent prognostic factor; low expression of miR-1231 is associated with worse patient outcomes compared with high expression of miR-1231 in glioma and pancreatic cancer (6062). Consistent with these results, the present study revealed that miR-1231 upregulation was associated with improved patient OS time and cisplatin sensitivity.

The present putative ceRNA axes included three lncRNAs (HOXD-AS2, LINC01123 and FIRRE). Each of these three lncRNAs has been shown to be upregulated in one or more types of cancer, and each one is commonly associated with poor patient prognosis. For example, LINC01123 is upregulated in intrahepatic cholangiocarcinomas (64) and is associated with poor prognosis in prostate cancer (65). Similarly, HOXD-AS2 is upregulated in glioma cells and is associated with poor prognosis (66). Consistent with these previous studies, the present study revealed that LINC01123 and HOXD-AS2 were upregulated in numerous types of cancer and were associated with reduced patient OS time. Importantly, the HOXD-AS2/hsa-miR-152-3P/NCAM1 and LINC01123/hsa-miR-762/RHBG axes in the present putative ceRNA network were supported by the co-expression results, which showed that LINC01123 was co-expressed with RHBG and that HOXD-AS2 was co-expressed with NCAM1.

FIRRE upregulation is associated with poor OS time in diffuse large B-cell lymphoma, CRC and HNSCC (67). However, FIRRE upregulation is also associated with significantly improved OS time in CRC (68). The present study revealed that FIRRE was upregulated in numerous types of cancer, possibly indicating that this lncRNA behaves differently under different circumstances.

In combination with the aforementioned results, the analyses of the current study revealed that the mRNAs, miRNAs and lncRNAs that form the potential axes in the present putative ceRNA network serve various important roles in cancer pathogenesis and progression. Importantly, a number of these molecules may serve different roles in different types of cancer. Thus, the results of the present study suggest these molecules as important targets for future studies focused on cancer diagnosis, prognosis and therapy. NCAM1 and miR-152 are particularly intriguing targets with respect to cisplatin resistance, as NCAM1 increases Btz sensitivity and miR-152 reduces cisplatin-induced effects (46). However, further investigations are necessary to determine the ceRNA mechanisms underlying cisplatin resistance in LUAD.

In addition, the present study presents some limitations, such as that the TCGA dataset included relatively few patients that met the set criteria and that the available clinical survival data was restricted to OS time. Future studies should recruit patients with lung cancer for cisplatin chemotherapy, collect lung lesion biopsy samples from patients with disease progression after three cycles of chemotherapy and then quantify the expression levels of the candidate lncRNAs (HOXD-AS2 and LINC01123), miRNAs (hsa-miR-152-3p and hsa-miR-762) and mRNAs (NCAM1, MED12 and ARF4) identified herein in the biopsy samples. Additionally, patients should be followed-up at 3 months, 6 months, 1 year and 3 years after chemotherapy to determine survival rates.

Despite these limitations, the results of the present study suggested that the integration of cell line experimental data with clinical information may be a valuable method to identify key cancer genes and potentially useful research targets.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present work was supported by grants from the Affiliated Hospital of Youjiang Medical University for Nationalities Outstanding Scholar Funding (Baise, Guangxi, China; grant no. R20196313) and Doctorate Awarding Unit Funding of the Affiliated Hospital of Youjiang Medical University for Nationalities [grant no. (2019)48]. The funders had no role in the design of the study, the collection, analyses or interpretation of data, the writing of the manuscript or the decision to publish the results.

Availability of data and materials

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

Authors' contributions

YL was involved in design of the study, analysis and interpretation of data and drafting the manuscript. BY gave final approval of the version of the manuscript to be published and was involved in data analysis. SH revised the manuscript critically for important intellectual content and was involved in the acquisition and analysis of data. ZW made substantial contributions to conception and design. All authors have 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 

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 

Molina JR, Yang P, Cassivi SD, Schild SE and Adjei AA: Non-Small cell lung cancer: Epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 83:584–594. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Liu L, Wu S, Yang Y, Cai J, Zhu X, Wu J, Wu J, Li M and Guan H: SOSTDC1 is down-regulated in non-small cell lung cancer and contributes to cancer cell proliferation. Cell Biosci. 6:242016. View Article : Google Scholar : PubMed/NCBI

5 

Li X, Shi Y, Yin Z, Xue X and Zhou B: An eight-miRNA signature as a potential biomarker for predicting survival in lung adenocarcinoma. J Transl Med. 12:1592014. View Article : Google Scholar : PubMed/NCBI

6 

Riaz SP, Lüchtenborg M, Coupland VH, Spicer J, Peake MD and Møller H: Trends in incidence of small cell lung cancer and all lung cancer. Lung Cancer. 75:280–284. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Murray N and Turrisi AT III: A review of first-line treatment for small-cell lung cancer. J Thorac Oncol. 1:270–278. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Sarvi S, Mackinnon AC, Avlonitis N, Bradley M, Rintoul RC, Rassl DM, Wang W, Forbes SJ, Gregory CD and Sethi T: CD133+ cancer stem-like cells in small cell lung cancer are highly tumorigenic and chemoresistant but sensitive to a novel neuropeptide antagonist. Cancer Res. 74:1554–1565. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Voigt W, Dietrich A and Schmoll HJ: Overview of development status and clinical action. Cisplatin and its analogues. Pharm Unserer Zeit. 35:134–143. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Silver DP, Richardson AL, Eklund AC, Wang ZC, Szallasi Z, Li Q, Juul N, Leong CO, Calogrias D, Buraimoh A, et al: Efficacy of neoadjuvant cisplatin in triple-negative breast cancer. J Clin Oncol. 28:1145–1153. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Santabarbara G, Maione P, Rossi A and Gridelli C: Pharmacotherapeutic options for treating adverse effects of cisplatin chemotherapy. Expert Opin Pharmacother. 17:561–570. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Yimit A, Adebali O, Sancar A and Jiang Y: Differential damage and repair of DNA-adducts induced by anti-cancer drug cisplatin across mouse organs. Nat Commun. 10:3092019. View Article : Google Scholar : PubMed/NCBI

13 

Eastman A: Improving anticancer drug development begins with cell culture: Misinformation perpetrated by the misuse of cytotoxicity assays. Oncotarget. 8:8854–8866. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Zhao X, Li X, Zhou L, Ni J, Yan W, Ma R, Wu J, Feng J and Chen P: LncRNA HOXA11-AS drives cisplatin resistance of human LUAD cells via modulating miR-454-3p/Stat3. Cancer Sci. 109:3068–3079. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Edgar R, Domrachev M and Lash AE: Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30:207–210. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Barrett T, Troup DB, Wilhite SE, Ledoux P, Rudnev D, Evangelista C, Kim IF, Soboleva A, Tomashevsky M and Edgar R: NCBI GEO: Mining tens of millions of expression profiles-database and tools update. Nucleic Acids Res. 35:D760–D765. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Yang Y, Li H, Liu J and Wang J: Noncoding RNA expression profiling of cisplatin-resistant cells derived from the A549 lung cell line (miRNA). NCBI. 2013, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE43249January 2–2013

18 

Yang Y, Li H, Liu J and Wang J: Noncoding RNA expression profiling of cisplatin-resistant cells derived from the A549 lung cell line (lncRNA and mRNA). NCBI. 2013, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE43493January 14–2013

19 

Gautier L, Cope L, Bolstad BM and Irizarry RA: Affy-Analysis of affymetrix genechip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Bolstad BM, Irizarry RA, Astrand M and Speed TP: A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatic. 19:185–193. 2003. View Article : Google Scholar

21 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Guidelines of HUGO Gene Nomenclature Committee (HGNC). European Bioinformatics Institute (EMBL-EBI). https://www.genenames.org/about/guidelines/

23 

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

24 

Wickham H: ggplot2: Elegant graphics for data analysis. New York. Springer–Verlag. 2016.

25 

Zhao S, Yin L, Guo Y, Sheng Q and Shyr Y: heatmap3: An Improved Heatmap Package. R package version 1.1.6. https://CRAN.R-project.org/package=heatmap3December 1–2018

26 

Huang DW, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS and Eppig JT: Gene ontology: Tool for the unification of biology. The gene ontology consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Kanehisa M: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

29 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43:D447–D452. 2014. View Article : Google Scholar : PubMed/NCBI

30 

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

31 

Yu G, Wang LG, Han Y and He QY: ClusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Dweep H and Gretz N: MiRWalk2.0: A comprehensive atlas of microRNA-target interactions. Nat Methods. 12:6972015. View Article : Google Scholar : PubMed/NCBI

33 

Paraskevopoulou MD, Vlachos IS, Karagkouni D, Georgakilas G, Kanellos I, Vergoulis T, Zagganas K, Tsanakas P, Floros E, Dalamagas T, et al: DIANA-LncBase v2: Indexing microRNA targets on non-coding transcripts. Nucleic Acids Res. 44:D231–D238. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Liao Y, Xie B, Zhang H, He Q, Guo L, Subramaniapillai M, Fan B, Lu C and Mclntyer RS: Efficacy of omega-3 PUFAs in depression: A meta-analysis. Transl Psychiatr. 9:1902019. View Article : Google Scholar

36 

Merhi A, De Mees C, Abdo R, Victoria Alberola J and Marini AM: Wnt/β-Catenin signaling regulates the expression of the ammonium permease gene RHBG in human cancer cells. PLoS One. 10:e01286832015. View Article : Google Scholar : PubMed/NCBI

37 

Johansson FK, Brodd J, Eklof C, Ferletta M, Hesselager G, Tiger CF, Uhrbom L and Westermark B: Identification of candidate cancer-causing genes in mouse brain tumors by retroviral tagging. Proc Natl Acad Sci USA. 101:11334–11337. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Jensen M and Berthold F: Targeting the neural cell adhesion molecule in cancer. Cancer Lett. 258:9–21. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Kashiwagi K, Ishii J, Sakaeda M, Arimasu Y, Shimoyamada H, Sato H, Sato H, Miyata C, Kamma H, Aoki I and Yazawa T: Differences of molecular expression mechanisms among neural cell adhesion molecule 1, synaptophysin, and chromogranin A in lung cancer cells. Pathol Int. 62:232–245. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Vander Borght A, Duysinx M, Broers JLV, Ummelen M, Falkenberg FW, Hahnel C and van der Zeijst BAM: The 180 splice variant of NCAM-containing exon 18-is specifically expressed in small cell lung cancer cells. Transl Lung Cancer Res. 7:376–388. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Sasca D, Szybinski J, Schüler A, Shah V, Heidelberger J, Haehnel PS, Dolnik A, Kriege O, Fehr EM, Gebhardt WH, et al: NCAM1 (CD56) promotes leukemogenesis and confers drug resistance in AML. Blood. 133:2305–2319. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Cho H, Kim JY and Oh YL: Diagnostic value of HBME-1, CK19, galectin 3, and CD56 in the subtypes of follicular variant of papillary thyroid carcinoma. Pathol Int. 68:605–613. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Ghaderi F, Ahmadvand S, Ramezan A, Montazer M and Ghaderi A: Production and characterization of monoclonal antibody against a triple negative breast cancer cell line. Biochem Biophys Res Commun. 505:181–186. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Zhao X, Tang DY, Zuo X, Zhang TD and Wang C: Identification of lncRNA-miRNA-mRNA regulatory network associated with epithelial ovarian cancer cisplatin-resistant. J Cell Physiol. 234:19886–19894. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Fang L, Wang H and Li P: Systematic analysis reveals a lncRNA-mRNA co-expression network associated with platinum resistance in high-grade serous ovarian cancer. Invest New Drugs. 36:187–194. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Yoshida T, Ri M, Kinoshita S, Narita T, Totani H, Ashour R, Ito A, Kusumoto S, Ishida T, Komatsu H and Iida S: Low expression of neural cell adhesion molecule, CD56, is associated with low efficacy of bortezomib plus dexamethasone therapy in multiple myeloma. PLoS One. 13:e01967802018. View Article : Google Scholar : PubMed/NCBI

47 

Kämpjärvi K, Mäkinen N, Kilpivaara O, Arola J, Heinonen HR, Böhm J, Abdel-Wahab O, Lehtonen HJ, Pelttari LM, Mehine M, et al: Somatic MED12 mutations in uterine leiomyosarcoma and colorectal cancer. Br J Cancer. 107:1761–1765. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Kämpjärvi K, Kim NH, Keskitalo S, Clark AD, von Nandelstadh P, Turunen M, Heikkinen T, Park MJ, Mäkinen N, Kivinummi K, et al: Somatic MED12 mutations in prostate cancer and uterine leiomyomas promote tumorigenesis through distinct mechanisms. Prostate. 76:22–31. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Huang S, Hölzel M, Knijnenburg T, Schlicker A, Roepman P, McDermott U, Garnett M, Grernrum W, Sun C, Prahallad A, et al: MED12 controls the response to multiple cancer drugs through regulation of TGF-β receptor signaling. Cell. 151:937–950. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Feliciano P: MED12 in cancer drug resistance. Nat Genet. 45:112012. View Article : Google Scholar

51 

Howley BV, Link LA, Grelet S, El-Sabban M and Howe PH: A CREB3-regulated ER-Golgi trafficking signature promotes metastatic progression in breast cancer. Oncogene. 37:1308–1325. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Ramalho-Carvalho J, Gonçalves CS, Graça I, Bidarra D, Pereira-Silva E, Salta S, Godinho MI, Gomez A, Esteller M, Costa BM, et al: A multiplatform approach identifies miR-152-3p as a common epigenetically regulated onco-suppressor in prostate cancer targeting TMEM97. Clin Epigenetics. 10:402018. View Article : Google Scholar : PubMed/NCBI

53 

Ge S, Wang D, Kong Q, Gao W and Sun J: Function of miR-152 as a tumor suppressor in human breast cancer by targeting PIK3CA. Oncol Res. 25:1363–1371. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Li LW, Xiao HQ, Ma R, Yang M, Li W and Lou G: MiR-152 is involved in the proliferation and metastasis of ovarian cancer through repression of ERBB3. Int J Mol Med. 41:1529–1535. 2018.PubMed/NCBI

55 

Wang Y, Bao W, Liu Y, Wang S, Xu S, Li X, Li Y and Wu S: MiR-98-5p contributes to cisplatin resistance in epithelial ovarian cancer by suppressing miR-152 biogenesis via targeting dicer1. Cell Death Dis. 9:4472018. View Article : Google Scholar : PubMed/NCBI

56 

Huang S, Li X and Zhu H: MicroRNA-152 targets phosphatase and tensin homolog to inhibit apoptosis and promote cell migration of nasopharyngeal carcinoma cells. Med Sci Monit. 22:4330–4337. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Hou R, Yang Z, Wang S, Chu D, Liu Q, Liu J and Jiang L: MiR-762 can negatively regulate menin in ovarian cancer. Onco Targets Ther. 10:2127–2137. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Li Y, Huang R, Wang L, Hao J, Zhang Q, Ling R and Yun J: MicroRNA-762 promotes breast cancer cell proliferation and invasion by targeting IRF7 expression. Cell Prolif. 48:643–649. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Shi Y, Jia Y, Zhao W, Zhou L, Xie X and Tong Z: Histone deacetylase inhibitors alter the expression of molecular markers in breast cancer cells via microRNAs. Int J Mol Med. 42:435–442. 2018.PubMed/NCBI

60 

Wang H, Wu J, Luo WJ and Hu JL: Low expression of miR-1231 in patients with glioma and its prognostic significance. Eur Rev Med Pharmacol Sci. 22:8399–8405. 2018.PubMed/NCBI

61 

Zhang J, Zhang J, Qiu W, Zhang J, Li Y, Kong E, Lu A, Xu J and Lu X: MicroRNA-1231 exerts a tumor suppressor role through regulating the EGFR/PI3K/AKT axis in glioma. J Neurooncol. 139:547–562. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Chen SL, Ma M, Yan L, Xiong SH, Liu Z, Li S, Liu T, Shang S, Zhang YY, Zeng H, et al: Clinical significance of exosomal miR-1231 in pancreatic cancer. Zhonghua Zhong Liu Za Zhi. 41:46–49. 2019.(In Chinese; Abstract available in Chinese from the publisher). PubMed/NCBI

63 

Pan Y, Xu T, Liu Y, Li W and Zhang W: Upregulated circular RNA circ_0025033 promotes papillary thyroid cancer cell proliferation and invasion via sponging miR-1231 and miR-1304. Biochem Biophys Res Commun. 510:334–338. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Yang W, Li Y, Song X, Xu J and Xie J: Genome-Wide analysis of long noncoding RNA and mRNA co-expression profile in intrahepatic cholangiocarcinoma tissue by RNA sequencing. Oncotarget. 8:26591–26599. 2017.PubMed/NCBI

65 

Ma W, Chen X, Ding L, Ma J, Jing W, Lan T, Sattar H, Wei Y, Zhou F and Yuan Y: The prognostic value of long noncoding RNAs in prostate cancer: A systematic review and meta-analysis. Oncotarget. 8:57755–57765. 2017.PubMed/NCBI

66 

Qi Y, Wang Z, Wu F, Yin B, Jiang T, Qiang B, Yuan J, Han W and Peng X: Long noncoding RNA HOXD-AS2 regulates cell cycle to promote glioma progression. J Cell Biochem. 120:281172018.

67 

Shi X, Cui Z, Liu X, Wu S, Wu Y, Fang F and Zhao H: LncRNA FIRRE is activated by MYC and promotes the development of diffuse large B-cell lymphoma via Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun. 510:594–600. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Li M, Zhao LM, Li SL, Li J, Gao B, Wang FF, Wang SP, Hu XH, Cao J and Wang GY: Differentially expressed lncRNAs and mRNAs identified by NGS analysis in colorectal cancer patients. Cancer Med. 7:4650–4664. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2020
Volume 19 Issue 6

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
Li Y, Huang S, Wei Z and Yang B: A putative competing endogenous RNA network in cisplatin‑resistant lung adenocarcinoma cells identifying potentially rewarding research targets. Oncol Lett 19: 4040-4052, 2020
APA
Li, Y., Huang, S., Wei, Z., & Yang, B. (2020). A putative competing endogenous RNA network in cisplatin‑resistant lung adenocarcinoma cells identifying potentially rewarding research targets. Oncology Letters, 19, 4040-4052. https://doi.org/10.3892/ol.2020.11483
MLA
Li, Y., Huang, S., Wei, Z., Yang, B."A putative competing endogenous RNA network in cisplatin‑resistant lung adenocarcinoma cells identifying potentially rewarding research targets". Oncology Letters 19.6 (2020): 4040-4052.
Chicago
Li, Y., Huang, S., Wei, Z., Yang, B."A putative competing endogenous RNA network in cisplatin‑resistant lung adenocarcinoma cells identifying potentially rewarding research targets". Oncology Letters 19, no. 6 (2020): 4040-4052. https://doi.org/10.3892/ol.2020.11483