Open Access

Potential ceRNA networks involved in autophagy suppression of pancreatic cancer caused by chloroquine diphosphate: A study based on differentially‑expressed circRNAs, lncRNAs, miRNAs and mRNAs

  • Authors:
    • Dan‑Ming Wei
    • Meng‑Tong Jiang
    • Peng Lin
    • Hong Yang
    • Yi‑Wu Dang
    • Qiao Yu
    • Dan‑Yu Liao
    • Dian‑Zhong Luo
    • Gang Chen
  • View Affiliations

  • Published online on: December 10, 2018     https://doi.org/10.3892/ijo.2018.4660
  • Pages: 600-626
  • Copyright: © Wei 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

Autophagy has been reported to be involved in the occurrence and development of pancreatic cancer. However, the mechanism of autophagy‑associated non‑coding RNAs (ncRNAs) in pancreatic cancer remains largely unknown. In the present study, microarrays were used to detect differential expression of mRNAs, microRNAs (miRNAs), long ncRNAs (lncRNAs) and circular RNAs (circRNAs) post autophagy suppression by chloroquine diphosphate in PANC‑1 cells. Collectively, 3,966 mRNAs, 3,184 lncRNAs and 9,420 circRNAs were differentially expressed. Additionally, only two miRNAs (hsa‑miR‑663a‑5p and hsa‑miR‑154‑3p) were underexpressed in the PANC‑1 cells in the autophagy‑suppression group. Furthermore, miR‑663a‑5p with 9 circRNAs, 8 lncRNAs and 46 genes could form a prospective ceRNA network associated with autophagy in pancreatic cancer cells. In addition, another ceRNA network containing miR‑154‑3p, 5 circRNAs, 2 lncRNAs and 11 genes was also constructed. The potential multiple ceRNA, miRNA and mRNA associations may serve pivotal roles in the autophagy of pancreatic cancer cells, which lays the theoretical foundation for subsequent investigations on pancreatic cancer.

Introduction

Pancreatic cancer is a highly aggressive and fatal malignancy, with stably high incidence and mortality for the past 30 years globally (1-4). Patients with pancreatic adenocarcinoma account for ~85% of all patients with pancreatic cancer and have a poor prognosis, compared with patients with other common solid tumors (2,5-9). Pancreatic cancer is primarily treated through surgical resection, whereas postoperative patients and patients with advanced disease are frequently subjected to adjuvant therapies with radiotherapy, chemotherapy, and a combination of radiotherapy and chemotherapy that are associated with high recurrence rates and poor treatment effectiveness (10). The recent development of and research into molecular targeted drugs have great significance for the early diagnosis and prognosis of malignant tumors, and have ushered a new era of tumor therapy (11-15). However, the overall treatment effect of these drugs remains poor, and further investigation is required (16).

Autophagy is a process by which eukaryotic cells undergo self-degradation of intracellular damaged macromolecular proteins and organelles (17,18). Autophagy at the physiological level is necessary to maintain the stability of the internal environment, and the occurrence of a variety of diseases, including inflammation, tumors and degenerative diseases, is frequently accompanied by an abnormal autophagic level (19-25). Currently, well-characterized, autophagy-associated proteins include Beclin 1, microtubule-associated-protein-1-light-chain-3 (LC3) and P62, and well-studied pathways include mechanistic target of rapamycin (mTOR) and phosphoinositide 3-kinase (PI3K) pathways (26,27).

Recently, the important role of autophagy in tumors has been recognized (19,28). Studies demonstrated that pancreatic cancer cells have an increased autophagy level, compared with other tumor cells (29-34). The growth of PDAC had a distinct dependence on autophagy in vivo and in vitro (35). Inhibition of autophagy is effective for the treatment of pancreatic cancer through animal experiments (36). Another study revealed that autophagy can be activated by gemcitabine and ionizing radiation in the treatment of pancreatic cancer cells, and activated autophagy serves a role in cancer suppression (37). However, whether autophagy promotes the occurrence, development and prognosis of pancreatic cancer is controversial (38). Therefore, the role of autophagy in the occurrence, development, prognosis and treatment of pancreatic cancer and its mechanism requires further examination.

mRNA is a single-stranded RNA that is transcribed using one of the DNA chains as the template and carries the genetic information that guides protein synthesis. Thus, mRNA occupies an important position in the heredity dogma (39). mRNA serves a key role in various human diseases and is involved in autophagy pathways (40,41); for example, signal transducer and activator of transcription 3 could inhibit autophagy in pancreatic cancer cells (42) and ubiquitin specific peptidase 1 regulates autophagy by targeting Unc-51 like autophagy activating kinase 1 (ULK1) (43). Non-coding RNA (ncRNA) refers to RNA that does not encode a protein and includes ribosomal RNA (rRNA), transfer RNA (tRNA), small nuclear RNA (snRNA), long ncRNA (lncRNA), microRNA (miRNA) and circular RNA (circRNA) (44-50). The involvement of lncRNAs and miRNAs in cancer has been studied intensively, whereas the potential significance of circRNAs in tumors has attracted attention over the last two years (51). The post-transcriptional levels of circRNAs serve a notable regulatory role in gene expression (52). Furthermore, the sponge effect of circRNAs on miRNAs is an important mechanism (53). circRNAs can contain multiple miRNA binding sites, and attract miRNAs effectively to reduce miRNA-mediated mRNA inhibition (54-56). Based on the interaction between these three types of ncRNAs and the current research status in pancreatic cancer, it was considered that an investigation of the effect of lncRNAs and circRNAs as competitive endogenous RNAs (ceRNAs) on the targeting of miRNAs in the development of pancreatic cancer is beneficial.

Recently, the number of autophagy-associated lncRNAs has been determined to be limited (57-60), and investigations on circRNAs and their associations with autophagy have not been reported to date. However, due to autophagy being involved in the occurrence and development of a variety of diseases, including breast (61,62), gastric (63) and lung cancer (64), in the present study, the human pancreatic cancer cell line PANC-1 and gene chip technology was used to detect differential expression of mRNAs, miRNAs, lncRNAs and circRNAs under different autophagy levels, and to investigate the genes associated with autophagy in pancreatic cancer and their underlying molecular mechanisms.

Materials and methods

Cell treatment

The human pancreatic cancer cell line PANC-1 was obtained from the Cell Laboratory of Chinese Academy of Sciences (Shanghai, China) and cultured in complete Dulbecco’s modified Eagle’s medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C. Due to chloroquine diphosphate being frequently used as a classic autophagic inhibitor (65,66), the following concentration gradient of chloroquine diphosphate (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was designed as recommended by the manufacturer’s protocols: 0, 25, 50, 75 and 100 µM. When the number of cells in the culture bottle grew to ~5×106, the original medium was replaced by a complete DMEM containing chloroquine diphosphate. Following treating the cells for 12 h at 37°C with the drug, the cellular proteins were immediately extracted using Radioimmunoprecipitation Assay Lysis Buffer (Beyotime Institute of Biotechnology, Haimen, China).

Western blotting

The lysis solution (radioimmunoprecipitation assay buffer:phenylmethylsulfonyl fluoride; 100:1, Beyotime Institute of Biotechnology) was prepared. The protein concentration in the lysate was determined with a bicinchoninic acid kit (Beyotime Institute of Biotechnology), according to the manufacturer’s instructions. Western blotting was performed according to routine procedures, and conducted with a 12% separating gel and a 5% stacking gel. A total of 20 µg protein was added to the glue holes. The electrophoresis condition was set to 120 Ma for 1 h. Following the electrophoresis, the protein was transferred to the polyvinylidene (PVDF) membrane (Beyotime Institute of Biotechnology). Subsequently, the PVDF membrane was placed into the Bull Serum Albumin blocking buffer (Beyotime Institute of Biotechnology) for shaking at 37°C for 2 h. Primary antibodies were incubated at 4°C overnight and secondary antibodies were incubated at room temperature for 2 h. The primary antibodies were LC3 β-specific rabbit polyclonal (1:1,000; cat. no. 18725-1-AP) and P62 mouse monoclonal antibodies (1:2,000; cat. no. 66184-1-Ig), which represented the state of autophagy, and the GAPDH horseradish peroxidase-labeled internal reference antibody (1:10,000; cat. no. HRP-60004), andhe secondary antibodies used were goat anti-mouse IgG (H+L), allopregnanolone conjugate (1:1,000; cat. no. SA00002-1) (all from ProteinTech Group, Inc., Chicago, IL, USA). The gray scale values of the protein bands from the raw image were determined using the ImageJ 1.48 (National Institutes of Health, Bethesda, MD, USA) and the formula Final gray scale value (G) = [G × (target band) − G × (background)] / [G × (internal reference band of the group) − G × (background)]. The data were analyzed using SPSS software 22 (SPSS, Inc., Chicago, IL, USA), and the results were plotted using the GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA).

Sample grouping

Based on the results of the western blotting experiment, the human pancreatic cancer PANC-1 cells treated with 100 µM chloroquine diphosphate were selected as the autophagic inhibition group, and the cells cultured in normal DMEM medium were selected as the control group. Subsequent experiments were compared between the autophagic inhibition group and the control group. Following chloroquine diphosphate treatment, total RNA was prepared using the TRIzol® reagent (Beyotime Institute of Biotechnology) with three replicates per group.

ceRNA microarray and detection, and statistical analysis of the miRNA chip results

The quality control for the gene analyses in the present study was performed by Shanghai Biotechnology Corporation (Shanghai, China). The starting sample of the microarray test was total RNA, which was analyzed with the NanoDrop ND-2000 spectrophotometer and the Agilent Bioanalyzer 2100 (Agilent Technologies, Inc., Santa Clara, CA, USA) to conduct the quality control assay. Only RNA samples that passed the quality control proceeded to the subsequent microarray experiments. The qualifying sample standards were that the RNA integrity number of each sample was ≥7.0 and that the 28S/18S was ≥0.7.

The ceRNA microarray detection assay included the detection of three types of RNA (lncRNA, circRNA and mRNA). The cutoff values of the differentials were all set to a fold change (FC)>2 or FC<0.5 and P<0.05. The assays and data analyses are described below. The analyses primarily included the normalization of raw data, sample association analysis, screening of genes with differential expression, Gene Ontology (GO) enrichment analysis of the differentially-expressed mRNAs, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, PANTHER pathway analysis of the differentially-expressed mRNAs, prediction of lncRNA and circRNA target genes, and prediction of lncRNA and circRNA adsorption of miRNAs. GO and KEGG pathway were generated by DAVID (https://david.ncifcrf.gov/) while PANTHER was analyzed using the WebGestalt 2017 tool (http://www.webgestalt.org/option.php). The clinical roles of genes were analyzed through the online database of Gene Expression Profiling Interactive Analysis (GEPIA; http://gepia.cancer-pku.cn/) Which is based on the data obtained from The Cancer Genome Atlas and GTEx. The miRNA microarray analyses primarily included the normalization of raw data, sample association analysis, screening of differentially-expressed miRNAs and prediction of miRNA target genes, which was predicted by 12 online miRNA target prediction databases [DIANA-microTv4.0 (67), DIANA-microT-CDS (68), miRanda-rel2010 (69), mirbridge (70), miRDB4.0 (71), miRMap (72), miRNAMap (73), PicTar2 (74), PITA (75), RNA22v2 (76), RNAhybrid2.1 (77) and Targetscan6.2 (78)] (http://zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk2/). The crossed genes from target prediction and differentially-expressed mRNAs were depicted in Compendia expression profiles (Novartis; http://software.broadinstitute.org/gsea/msigdb/annotate.jsp). Network visualization was performed in Cytoscape 3.5.1 (The Cytoscape Consoritum, New York, NY, USA).

The raw data of ceRNA microarray has been deposited in the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/; accession number GSE115517).

Tissue samples

A total of 31 formalin-fixed paraffin-embedded (FFPE) tissues, including 18 cases of Pancreatic ductal adenocarcinoma and 13 paracancerous tissues, were collected from the Department of Pathology of the First Affiliated Hospital of Guangxi Medical University (Nanning, China) between June 2015 and June 2018. The present study was approved by the Ethics Committee of the First Affiliated Hospital of Guangxi Medical University. All cases were from patients who have not been treated with chemotherapy or radiation prior to resection and have signed informed consent. There were 11 male and 7 female aged 29-77 years (mean age, 56 years) in 18 patients with adenocarcinoma.

RNA extraction and Real-time fluorescent quantitative PCR (RT-qPCR)

According to the user guides provided by the manufacturer, the total RNA of tissues was extracted using an E.Z.N.A.® FFPE RNA kit (Omega Bio-Tek, Inc., Norcross, GA, USA). The complementary DNA (cDNA) was reverse transcribed through the kit of miRNA First Strand cDNA Synthesis [Tailing Reaction (79,80)] (Sangon Biotech Co., Ltd., Shanghai, China), and RT-qPCR was conducted by applying a MicroRNAs qPCR kit [SYBR® Green method (81,82)] (Sangon Biotech Co., Ltd.), according to the manufacturer’s protocols, on an ABI Prism 7500 (Applied Biosystems; Thermo Fisher Scientific, Inc.). The temperature of pre-denaturation and denaturation were both set at 95°C, and the temperature of annealing/extension was set at 60°C. Pre-denaturation was conducted for 10min, denaturation for 15 sec and annealing/extension for 60 sec. The number of cycles was set to 40. Subsequently, the expression of miRNA relative to U6 was calculated via the 2−ΔΔCq method on identical samples (83). The PCR primers included: i) miR-663a-5p for ward, 5′-ATAGGCGGGGCGCCGCGGGAC-3′; ii) miR-154-3p forward, 5′-CCGGGAATCATACACGGTTGA CCTATT-3′; and iii) U6 forward and universal PCR reverse primer were attached to the kits. Their primer sequences are 5′-CTCGCTTCGGCAGCACA-3′ and 5′-AACGCTTCACGA ATTTGCGT-3′, respectively.

Retrieval of data from GEO

Within GEO, ‘pancreas’ or ‘pancreatic’, and ‘adenocarcinoma’, ‘carcinoma’, ‘cancer’, ‘neoplasm’, ‘tumor’, ‘tumour’, ‘neoplas*’, ‘malignan*’, ‘PDAC’, ‘OR’, ‘PAAD’ or ‘PC’ were employed as a search strategy in GEO to determine the expression of miR-663a-5p and miR-154-3p. STATA 12.0 (StataCorp LLC, College Station, TX, USA) was applied to estimate the pooled effects, heterogeneity and publication bias.

Statistical analysis

The data of GEO and RT-qPCR were calculated using SPSS 22 software for mean and standard deviation (SD). Continuous data are presented as mean ± SD. The forest plots were produced by Stata 12.0. The heterogeneity test was used to analyze the existence of heterogeneity and the source of the occurrence of heterogeneity (I2>50% or P<0.05 is the existence of heterogeneity). Funnel plots were used for the analysis of publication bias. The comparison between the two groups was performed using two-sample Student’s t-test. The method for differential gene expression analysis in GEPIA was one-way analysis of variance, using pathological stage as variable for calculating differential expression. GEPIA performs overall survival analysis based on gene expression. GO analysis was performed in DAVID tool which used Fisher’s exact test and the techniques of Kappa statistics (84). PANTHER analysis was performed in WebGestalt, which used Hypergeometric and Fisher’s exact tests. P<0.05 was considered to indicate a statistically significant difference.

Results

Autophagy following chloroquine diphosphate treatment

The present study design of the whole investigation is depicted in Fig. 1. The LC3-II expression level was significantly reduced in the control group, compared with the group treated with 25 or 100 µM chloroquine diphosphate (P<0.05). However, the P62 expression level was significantly increased in the group treated with 100 µM chloroquine diphosphate, compared with the control group or the groups treated with 25, 50, 75 or 100 µM chloroquine diphosphate (P<0.05). Thus, the 100 µM concentration of chloroquine diphosphate had the greatest significant inhibitory effect on autophagy in human pancreatic cancer PANC-1 cells (Fig. 2).

Quality control of samples for the ceRNA microarray and miRNA microarray chips

Electrophoresis of the RNA samples indicated that the RNA integrity number of each sample was ≥7.0 and that the 28S/18S was ≥0.7, which reached the qualifying sample standards. Thus, the samples were qualified for use in the subsequent experiments.

Assays to identify differentially-expressed mRNAs, lncRNAs, circRNAs and miRNAs

The ceRNA microarray results determined 3,966 differentially-expressed mRNAs in total. The expression levels of 2,445 mRNAs in the PANC-1 cells from the chloroquine diphosphate treatment group (autophagy suppression group) were downregulated and 1,521 mRNAs were upregulated, compared with the control group (Fig. 3). Additionally, 3,184 differentially-expressed lncRNAs were observed, including 1,637 downregulated and 1,547 upregulated (Fig. 4), whereas 9,420 circRNAs were determined to be differentially expressed, including 4,223 downregulated and 5,197 upregulated (Fig. 5). The miRNA microarray results demonstrated that the expression levels of two miRNAs (hsa-miR-663a-5p and hsa-miR-154-3p) were underexpressed in the PANC-1 cells in the autophagy-suppression group, compared with the control group. No upregulated miRNA was determined. The cutoff values of the aforementioned differentials were all set to a fold change>2 or FC<0.5 and P<0.05.

Bioinformatics analysis of the functions of the differentially-expressed mRNAs

GO and KEGG pathway analyses were performed using the DAVID database. The 3,966 differentially-expressed mRNAs in the chloroquine diphosphate treatment group (autophagy suppression group), compared with the control group, were subjected to bioinformatics analysis of their functions. The results demonstrated that these genes were concentrated in the biological processes, molecular functions and cellular components categories in the GO analysis (Table I), and were involved in the regulation of multiple KEGG signaling pathways including pathways in cancer and the mitogen-activated protein kinase signaling pathway (Table II). Of these functions, the involvement of the autophagy-associated pathway (Autophagy) was notable. Additionally, the differentially-expressed genes in this pathway included a number of recognized autophagy-associated genes, including autophagy-related 12 (ATG12), GABA type A receptor associated protein like 1 (GABARAPL1) and ULK2, which were differentially expressed in the present ceRNA microarray results also (Fig. 6). Following analyzing the clinical roles of the three genes via the Gene Expression Profiling Interactive Analysis online database, which is based on the data obtained from The Cancer Genome Atlas and GTEx, it was determined that the expression of ATG12 was upregulated in pancreatic cancer tissue, compared with non-tumor tissue. Furthermore, high expression of ATG12 was associated with significantly reduced survival time (P<0.01). Notably, the expression of ATG12, GABARAPL1 and ULK2 were increased in pancreatic cancer, compared with the control group (Fig. 6). These results validated that these autophagy-associated genes were involved in the onset and progression of pancreatic cancer.

Figure 6

Expression level, stage and overall survival curves of ATG12, GABARAPL1 and ULK2 in patients with pancreatic cancer. (A) The fold change of ATG12 (fold change=2.34), GABARAPL1 (fold change=3.90) and ULK2 (fold change=2.29) in the chloroquine diphosphate treatment group, compared with the control group. (B) The expression of ATG12 was upregulated in pancreatic cancer tissues, compared with normal tissues [T, n=179; N, n=171]. (C) ATG12 expression in different stages of pancreatic cancer is not significantly different (P=0.6140). (D) Patients with pancreatic cancer and a low expression of ATG12 have an increased survival time, compared with those with a high expression of ATG12 (HR=1.8; P=0.0042). (E) GABARAPL1 has an increased expression in pancreatic cancer tissues, compared with normal tissues [T, n=179; N, n=171]. (F) The association between GABARAPL1 and stage of patients with pancreatic cancer (P=0.5940). (G) The effect of GABARAPL1 expression on the overall survival time of patients with pancreatic cancer is not significant (HR=0.72; P=0.1100). (H) ULK2 has an increased expression in pancreatic cancer tissues, compared with normal tissues [T, n=179; N, n=171]. (I) The association between ULK2 and stage of patients with pancreatic cancer (P=0.0248). (J) The effect of ULK2 expression on the overall survival time of patients with pancreatic cancer is not significant (HR=0.79; P=0.2500). ATG12, autophagy-related 12; GABARAPL1, GABA type A receptor associated protein like 1; ULK2, Unc-51 like autophagy activating kinase 2; HR, hazard ratio; TPM, transcript per million.

Table I

GO analysis of differentially-expressed mRNAs following autophagy inhibition.

Table I

GO analysis of differentially-expressed mRNAs following autophagy inhibition.

GO IDDescriptionTypeGenes
GO:2000969Positive regulation of α-amino-3-hydroxy-5-methyl-4-isoxazole propionate selective glutamate receptor activity Biological_processRELN, NLGN3, ARC and SHANK3
GO:0000727Double-strand break repair via break-induced replication Biological_processCDC45, GINS2, CDC7 and GINS4
GO:0038026Reelin-mediated signaling pathway Biological_processDAB2IP, RELN, LRP8 and VLDLR
GO:0003150Muscular septum morphogenesis Biological_processHEY2, FZD2, VANGL2 and FZD1
GO:0050915Sensory perception of sour taste Biological_processASIC3, PKD2L1, ASIC2, PKD1L3 and ASIC1
GO:0031052Chromosome breakage Biological_processHMGA2, BRCA2, BRCA1, CD19 and TP53
GO:0038171Cannabinoid signaling pathway Biological_processDAGLA, CNR2, MGLL, ABHD6 and CNR1
GO:0003149Membranous septum morphogenesis Biological_processID2, WHSC1, FZD2, VANGL2 and FZD1
GO:0061156Pulmonary artery morphogenesis Biological_processHEY2, STRA6, BMP4 and JAG1
GO:2001286Regulation of caveolin-mediated endocytosis Biological_processUNC119, NEDD4L, CAV3 and PROM2
GO:0000942Condensed nuclear chromosome outer kinetochore Cellular_componentPLK1, BUB1, CCNB1 and NDC80
GO:0031262Ndc80 complex Cellular_componentSPC24, NUF2, SPC25 and NDC80
GO:0031298Replication fork protection complex Cellular_componentCDC45, MCM10, GINS2, TOP1MT and GINS4
GO:0072557IPAF inflammasome complex Cellular_componentCASP1, CASP12, CASP5 and NLRC4
GO:0000778Condensed nuclear chromosome kinetochore Cellular_componentPLK1, CENPA, NDC80, BUB1B, BUB1, CCNB1 and MIS18BP1
GO:0042555MCM complex Cellular_componentMCM4, TONSL, MCM5, MMS22L, MCM8, MCM2, MCMBP and MCM3
GO:0000940Condensed chromosome outer kinetochore Cellular_componentPLK1, SKA3, CENPF, NDC80, BUB1, BUB1B, CENPE, CCNB1 and SPDL1
GO:0005663DNA replication factor c complex Cellular_componentRFC4, PCNA, RFC3 and RFC5
GO:0097169AIM2 inflammasome complex Cellular_componentCASP1, CASP12, CASP5 and PYCARD
GO:0043240Fanconi anemia nuclear complex Cellular_componentFANCB, FANCC, C19ORF40, STRA13, FANCG, FANCM, FANCA and FANCE
GO:0043142Single-stranded DNA-dependent ATPase activity Molecular_functionPIF1, RAD18, RAD51, DNA2 and POLQ
GO:0032405Mutlalpha complex binding Molecular_functionTREX1, PCNA, MUTYH, MSH6 and MSH2
GO:0086006Voltage-gated sodium channel activity involved in cardiac muscle cell action potential Molecular_functionSCN1B, SCN4B, SCN3B and SCN2B
GO:0008574Plus-end-directed microtubule motor activity Molecular_functionKIF3B, KIF18B, KIF5A, KIF14, KIF5C, KIF3A, KIF20B, KIF17, KIF11, KIF26A and KIF16B
GO:0004908Interleukin-1 receptor activity Molecular_functionIL1R1, IL1RL2, IL1RL1, IL18R1 and IL1R2
GO:0032027Myosin light chain binding Molecular_functionCXCR4, IQGAP3, MYOC and MYH8
GO:00353125′-3′ exodeoxyribonuclease activity Molecular_functionEXO1, DCLRE1B, EXO5 and DCLRE1A
GO:0048256Flap endonuclease activity Molecular_functionEXO1, MUS81, FEN1, DNA2 and FAN1
GO:0036310Annealing helicase activity Molecular_functionRECQL4, SMARCA1, SMARCAL1, RAD54L and BLM
GO:0097153Cysteine-type endopeptidase activity involved in apoptotic process Molecular_functionCASP7, CASP6, CASP12, PYCARD, CASP2, CASP8, CLC, CASP1 and CASP5

[i] Only the top 10 signaling pathways of each classification were listed as examples. GO, Gene Ontology.

Table II

Kyoto Encyclopedia of Genes and Genomes pathway analysis of differentially-expressed mRNAs when autophagy was inhibited.

Table II

Kyoto Encyclopedia of Genes and Genomes pathway analysis of differentially-expressed mRNAs when autophagy was inhibited.

Pathway IDDescriptionGenes
hsa05200Pathways in cancerBCR, PDGFB, WNT4, ETS1, CASP8, BRCA2, ARHGEF11, TRAF3, GNAI3 and BID
hsa04060Cytokine-cytokine receptor interactionCCL4, PDGFB, IL4R, CCL21, IL12A, CSF2, GH2, CCL27, ACVR1B and LEP
hsa04010MAPK signaling pathwayPDGFB, MAP2K6, CACNG8, RPS6KA2, PLA2G4A, MAP3K4, GADD45G, CACNB3, JUN and TNFRSF1A
hsa04144EndocytosisFOLR3, NEDD4L, PSD4, SH3KBP1, GRK5, ERBB4, EHD2, CAV2, ITCH and EPS15L1
hsa04080Neuroactive ligand- receptor interactionADRA1D, GH2, PTAFR, C5AR1, GRIN3B, HTR1B, LEP, S1PR5, LPAR2, CHRND, AGTR1, GH1, CHRM1, SSTR5, ADRA1B, GRIN3A, DRD5 and PTGER3
hsa04621NOD-like receptor signaling pathwayCASP8, ATG12, TRAF3, TRAF5, JUN, OAS3, OAS1, MAVS, RNASEL and NLRP7
hsa00230Purine metabolismPFAS, PDE6G, POLD3, NME7, PDE9A, POLR3H, POLR2L, ADCY7, NME6 and NPR2
hsa04110Cell cycleRAD21, CDC16, E2F2, SFN, GADD45G, CCNH, CDC20, CDC25A, CCNA1 and TP53
hsa04062Chemokine signaling pathwayCCL4, CCL21, GNAI3, PIK3R5, GRK5, CCL27, PIK3CB, SHC1, VAV1 and CCL4L1
hsa04210ApoptosisCASP8, BID, PIK3R5, CTSC, LMNB1, PIK3CB, EIF2AK3, GADD45G, CASP6 and TRADD

[i] Only 10 genes were listed as examples in each pathway. MAPK, mitogen-activated protein kinase.

Prediction of target genes of the differentially-expressed miRNAs and the bioinformatics analysis of their functions

These results were integrated with the differentially-expressed mRNAs that were actually measured. The co-expressed genes were subjected to GO and KEGG pathway analyses using the DAVID database.

Prediction of miR-663a-5p target genes and bioinformatics analysis of their functions

A total of 1,726 target genes of miR-663a-5p were detected by at least 6 platforms using the aforementioned 12 online miRNA target gene prediction databases. Following cross-checking with the differentially-expressed genes in the autophagy suppression group derived from chloroquine diphosphate treatment, 462 co-expressed genes were obtained (Fig. 7). These genes were subjected to GO and KEGG pathway analyses. The results demonstrated that these genes were concentrated in the biological processes, molecular functions and cellular components categories in the GO analysis (Fig. 8A, Table III) and were involved in the regulation of multiple KEGG signaling pathways (Table III), including the aldosterone-regulated sodium reabsorption and Wnt signaling pathways (Fig. 8B-D). However, there were no significant pathways determined by PANTHER analysis (P>0.05).

Table III

GO and KEGG analysis of potential target genes of microRNA-663a-5p related to autophagy.

Table III

GO and KEGG analysis of potential target genes of microRNA-663a-5p related to autophagy.

TermDescriptionTypeCountP-valueGenes
GO:0048562Embryonic organ morphogenesisBiological process11 1.66×10−3DLX2, FOXL2, TBX15, MAFB, CHST11, NKX3-2, SOBP, TBX1, ZEB1 and PAX2
GO:0045449Regulation of transcriptionBiological process87 1.71×10−3ZNF451, NR6A1, ZNF250, CBX2, ZEB1, MED22, PAX2, CBFA2T3, ZNF345 and TGFB1
GO:0006350TranscriptionBiological process73 1.78×10−3NR6A1, ZNF451, ZNF250, CBX2, MED22, ZEB1, PAX2, CBFA2T3, ZNF345 and CRX
GO:0043583Ear developmentBiological process9 2.35×10−3KCNMA1, MAFB, NKX3-2, SOBP, TBX1, PAX2, SLC9A3R2, TGFB1 and CDH23
GO:0017015Regulation of transforming growth factor β receptor signaling pathwayBiological process6 2.54×10−3DAND5, CHST11, TGFB1I1, ZEB1, PRDM16 and TGFB1
GO:0048839Inner ear developmentBiological process8 3.27×10−3KCNMA1, MAFB, SOBP, TBX1, PAX2, SLC9A3R2, TGFB1 and CDH23
GO:0048568Embryonic organ developmentBiological process12 3.56×10−3DLX2, FOXL2, TBX15, MAFB, FOXF1, CHST11, NKX3-2, SOBP, TBX1 and ZEB1
GO:0006357Regulation of transcription from RNA polymerase II promoterBiological process31 3.82×10−3TADA3, CRTC1, NR6A1, CBX2, ZEB1, MED22, PAX2, PRDM16, ZNF345 and TGFB1
GO:0006355Regulation of transcription DNA-dependentBiological process62 3.88×10−3NR6A1, ZNF250, CBX2, MED22, ZEB1, CBFA2T3, PAX2, ZNF345, TGFB1 and CRX
GO:0048598Embryonic morphogenesisBiological process17 3.96×10−3FOXL2, TBX15, MAFB, ARFRP1, TP53, SOBP, TBX1, ZEB1, CELSR1 and PAX2
GO:0030054Cell junctionCellular component27 1.02×10−4ACHE, CLDN9, PANX2, CLDN6, GRIK5, ZNRF1, ITSN1, SYNGR1, CALB2 and RIMS3
GO:0045202SynapseCellular component21 1.45×10−4KCNMA1, DLGAP1, ACHE, CDK5R1, ARC, EFNA2, SYT11, GRIK5, BCAN and BSN
GO:0044459Plasma membrane partCellular component68 4.22×10−3KCNC1, PCDHA8, CLDN9, CD8A, CLDN6, GRIK5, ANPEP, SYNGR1, ITSN1 and ZNRF1
GO:0019717SynaptosomeCellular component7 1.19×10−2PVRL1, SNPH, DLG4, BSN, LGI3, ITSN1 and RNF40
GO:0030173Integral to golgi membraneCellular component5 1.53×10−2ST6GAL1, ST8SIA4, RER1, STEAP2 and ABO
GO:0031228Intrinsic to golgi membraneCellular component5 1.92×10−2ST6GAL1, ST8SIA4, RER1, STEAP2 and ABO
GO:0005794Golgi apparatusCellular component29 3.41×10−2SLC9A8, ACHE, APC2, C6ORF25, PPIL2, ARFRP1, RER1, VPS53, CBFA2T3 and NUFIP2
GO:0005886Plasma membraneCellular component99 4.57×10−2KCNC1, ALPPL2, PLXNA1, ATP1B2, C6ORF25, EFNA2, FGFRL1, GRIK5, ZNRF1, TLR6 and ITSN1
GO:0031012Extracellular matrixCellular component14 4.66×10−2ACHE, ADAMTS14, ADAMTSL2, ADAMTS15, OLFML2A, BCAN, COL5A1, MMP25, TGFB1 and WNT7B
GO:0005667Transcription factor complexCellular component10 4.72×10−2BRF1, TADA3, TBX2, MAFB, FOXF1, NR6A1, TP53, NPAS4, ZEB1 and CRX
GO:0046872Metal ion bindingMolecular function123 1.21×10−2KCNC1, SLC9A8, ALAD, FSTL4, ALPPL2, ATP1B2, ZNF451, ZNF250, RNF216 and ITSN1
GO:0008270Zinc ion bindingMolecular function74 1.44×10−2ALPPL2, ALAD, ZCCHC24, ZNF451, NR6A1, SOBP, ZNF250, ANPEP, RNF216 and ZEB1
GO:0043169Cation bindingMolecular function123 1.64×10−2KCNC1, SLC9A8, ALAD, ALPPL2, ATP1B2, ZNF451, FSTL4, ZNF250, RNF216 and ITSN1
GO:0019209Kinase activator activityMolecular function4 2.32×10−2CDK5R1, MADD, ITSN1 and TAB1
GO:0043167Ion bindingMolecular function123 2.58×10−2KCNC1, SLC9A8, ALAD, ALPPL2, ATP1B2, ZNF451, FSTL4, ZNF250, RNF216 and ITSN1
GO:0003677DNA bindingMolecular function72 3.34×10−2PRR12, NR6A1, ZNF451, ZNF250, CBX2, ZEB1, PAX2, CBFA2T3, ZNF345 and CRX
GO:0046914Transition metal ion bindingMolecular function84 3.45×10−2STEAP3, ALPPL2, ALAD, ZCCHC24, ZNF451, NR6A1, SOBP, ZNF250, ANPEP and RNF216
GO:0030955Potassium ion bindingMolecular function8 4.02×10−2KCNMA1, KCNC1, SLC12A7, PDXK, ATP1B2, KCNK5, HCN4 and KCNG1
GO:0030528Transcription regulator activityMolecular function49 4.27×10−2NR6A1, CBX2, ZEB1, MED22, CBFA2T3, ZNF345, TGFB1, CRX, FOXF1 and ZNF445
GO:0016419 S-malonyltransferase activityMolecular function2 4.90×10−2MCAT and FASN
hsa04960 Aldosterone-regulated sodium reabsorptionKEGG pathway5 9.51×10−3ATP1B2, PIK3R5, NEDD4L, SFN and SLC9A3R2
hsa04310Wnt signaling pathwayKEGG pathway8 2.92×10−2WNT7B, SOST, VANGL1, APC2, TP53, FZD1, FRAT1 and FZD4
hsa05217Basal cell carcinomaKEGG pathway5 3.03×10−2WNT7B, APC2, TP53, FZD1 and FZD4
hsa00061Fatty acid biosynthesisKEGG pathway3 3.08×10−2MCAT, FASN and ACSL6
hsa04151PI3K-Akt signaling pathwayKEGG pathway14 3.48×10−2FGF18, IL2RB, LAMB3, FLT1, IL4R, CSF1, EFNA2, TP53, TNN and EFNA5
hsa05205Proteoglycans in cancerKEGG pathway9 6.69E×10−2WNT7B, CTTN, EZR, ANK1, TP53, FZD1, PIK3R5, FZD4 and TGFB1
hsa04724Glutamatergic synapseKEGG pathway6 9.94×10−2DLGAP1, SLC1A7, DLG4, GRIK5, CACNA1D and SHANK3

[i] Only 10 genes were listed as examples in each pathway. GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; PI3K, phosphoinositide 3-kinase.

Prediction of miR-154-3p target genes and bioinformatics analysis of their functions

A total of 294 target genes of miR-154-3p were detected by at least 6 programs using the aforementioned 12 online miRNA target gene prediction databases. Following cross-checking with the differentially-expressed genes in the autophagy suppression group derived from chloroquine diphosphate treatment, 294 co-expressed genes were obtained (Fig. 9). These genes were subjected to GO and KEGG pathway analyses. The results demonstrated that these genes were concentrated in the biological processes, molecular functions and cellular components categories in the GO analysis (Fig. 10A, Table IV), and were involved in the insulin signaling, Forkhead Box O (FoxO) signaling and proteoglycans in cancer pathways (Table IV, Fig. 10B-D). Additionally, the results of PANTHER analysis revealed these genes were concentrated in a numbered of pathways, including the p53 pathway by glucose deprivation and the classical gastrin cholecystokinin receptor (CCKR) signaling map (Table V, Fig. 11).

Table IV

GO and KEGG analysis of potential target genes of microRNA-154-3p associated with autophagy.

Table IV

GO and KEGG analysis of potential target genes of microRNA-154-3p associated with autophagy.

TermDescriptionTypeCountP valueGenes
GO:0007605Sensory perception of soundBiological process11 1.67×10−3NAV2, ASIC2, DCDC2, SOBP, TBX1, TIMM13, CACNA1D, FZD4, SLC52A3 and DNM1
GO:0030512Negative regulation of transforming growth factor β receptor signaling pathwayBiological process7 4.81×10−3DAND5, ADAMTSL2, ZNF451, CHST11, TGFB1I1, PRDM16 and TGFB1
GO:0072207Metanephric epithelium developmentBiological process3 5.74×10−3WNT7B, OSR1 and PAX2
GO:0070507Regulation of microtubule cytoskeleton organizationBiological process4 6.52×10−3DIXDC1, CDK5R1, ATAT1 and EFNA5
GO:0045216Cell-cell junction organizationBiological process4 9.16×10−3CLDN9, CLDN6, MARVELD3 and TGFB1
GO:0008285Negative regulation of cell proliferationBiological process19 9.45×10−3TP53I11, NACC2, CLMN, KLF10, FGFRL1, TP53, GPER1, ZEB1, CBFA2T3 and TGFB1
GO:0031497Chromatin assemblyBiological process3 1.53×10−2CDAN1, TP53 and CHAF1A
GO:0006366Transcription from RNA polymerase II promoterBiological process22 1.62×10−2MAF, MAFF, FOXL2, POLR2L, MAFB, SOX12, TP53, ARID3B, FOSB and NPAS4
GO:0008104Protein localizationBiological process6 1.69×10−2TNS4, MALL, CDAN1, TP53, GRASP and AKAP3
GO:0000122Negative regulation of transcription from RNA polymerase II promoterBiological process28 1.98×10−2NR6A1, CBX2, ZEB1, PRDM16, ZNF345, TGFB1, KANK2, AHRR, EZR and OSR1
GO:0030054Cell junctionCellular component24 6.45×10−4SH3PXD2B, ARC, DLGAP1, ACHE, SH3PXD2A, CACNG8, SYT11, GRIK5, LRRC4B and BSN
GO:0014069Postsynaptic densityCellular component13 1.51×10−3DLGAP1, ARC, CDK5R1, CACNG8, SYT11, GRIK5, BSN, GPER1, SHANK3 and CNIH2
GO:0005737CytoplasmCellular component152 2.33×10−3RBPMS2, STIL, PLXNA1, ATP1B2, LRRC4B, MED22, CALB2, TGFB1, KANK2 and PACSIN1
GO:0043197Dendritic spineCellular component9 2.60×10−3CDK5R1, ARC, CTTN, CNIH2, SYT11, DLG4, ASIC2, SEZ6 and SHANK3
GO:0044297Cell bodyCellular component7 3.72×10−3GNAZ, EZR, SYT11, PACRG, FADD, DISC1 and EPO
GO:0005634NucleusCellular component151 1.30×10−2ALAD, PLXNA1, ZNF451, ZNF250, CBX2, RNF216, ITPKB, CALB2, TGFB1 and ANK1
GO:0030424AxonCellular component12 1.72×10−2CDK5R1, ATAT1, STMN3, SYT11, FKBP15, LMTK3, BSN, GPER1, LDLRAP1 and KIF21B
GO:0005884Actin filamentCellular component6 1.89×10−2CTTN, EZR, APC2, AIF1L, FKBP15 and MYO9B
GO:0005886Plasma membraneCellular component115 3.45×10−2KCNC1, SLC9A8, ALPPL2, PLXNA1, ATP1B2, EFNA2, C6ORF25, FGFRL1, GRIK5 and TLR6
GO:0048471Perinuclear region of cytoplasmCellular component23 4.02×10−2CDK5R1, ACHE, STC2, APC2, CSF1, BRSK2, VPS53, MYO9B, GPER1 and NDOR1
GO:0001228Transcriptional activator, activity RNA polymerase II transcription regulatory region sequence-specific bindingMolecular function8 8.50×10−3MAF, MAFF, DLX2, MAFB, FOXF1, TP53, NEUROD2 and ARID3B
GO:0046872Metal ion bindingMolecular function68 8.68×10−3GNAZ, STEAP3, SGSH, ALPPL2, ALAD, ZNF451, SOBP, ZNF250, RNF216 and ZEB1
GO:0000978RNA polymerase II core promoter proximal region sequence-specific DNA bindingMolecular function17 1.37×10−2FOXL2, TBX15, NACC2, TBX2, MAFB, NR6A1, ARID3B, FOSB, NPAS4 and DDN
GO:0048495Roundabout bindingMolecular function3 1.90×10−2MYO9B, TGFB1I1 and TPBGL
GO:0003779Actin bindingMolecular function14 1.90×10−2KCNMA1, DIXDC1, FMNL3, CLMN, MYO9B, SHANK3, TNS4, EPB41L1, EZR and PACRG
GO:0030295Protein kinase activator activityMolecular function4 2.46×10−2DUSP19, MADD, RPTOR and EPO
GO:0098641Cadherin binding involved in cell-cell adhesionMolecular function14 2.58×10−2EPS15L1, SFN, SLC9A3R2, TNKS1BP1, PAK6, CTTN, EPB41L1, EZR, FASN and SPTBN1
GO:0016874Ligase activityMolecular function13 3.31×10−2DTX3L, PPIL2, ZNF451, RNF216, ZNRF1, TTLL11, RNF165, NEURL1B, PELI2 and NEDD4L
GO:0030276Clathrin bindingMolecular function5 3.75×10−2TOM1L2, SYT11, RPH3AL, C2CD4C and LDLRAP1
GO:0004314 [acyl-carrier-protein] S-malonyltransferase activityMolecular function2 4.81×10−2MCAT and FASN
hsa04910Insulin signaling pathwayKEGG pathway8 2.79×10−3CBLB, SOS1, PRKAB2, MAPK8, RPS6KB1, SOCS4, PRKACB and PPARGC1A
hsa04068FoxO signaling pathwayKEGG pathway7 1.01×10−2NLK, SOS1, PRKAB2, MDM2, FBXO32, MAPK8 and FOXO3
hsa04931Insulin resistanceKEGG pathway6 1.62×10−2RPS6KA2, PRKAB2, MAPK8, RPS6KB1, OGT and PPARGC1A
hsa05205Proteoglycans in cancerKEGG pathway8 1.98×10−2CBLB, CTTN, ERBB4, SOS1, MDM2, RPS6KB1, PRKACB and FZD4
hsa04012ErbB signaling pathwayKEGG pathway5 3.14×10−2CBLB, ERBB4, SOS1, MAPK8 and RPS6KB1
hsa04010MAPK signaling pathwayKEGG pathway8 6.08×10−2RPS6KA2, ARRB1, NLK, SOS1, FGF13, MAPK8, PRKACB and MAP3K13
hsa00410β-Alanine metabolismKEGG pathway3 6.72×10−2ALDH6A1, ALDH1B1 and EHHADH
hsa00512Mucin type O-Glycan biosynthesisKEGG pathway3 6.72×10−2GALNT4, GALNT18 and B4GALT5
hsa04920Adipocytokine signaling pathwayKEGG pathway4 7.13×10−2PRKAB2, MAPK8, PPARGC1A and CAMKK2
hsa04917Prolactin signaling pathwayKEGG pathway4 7.13×10−2SOS1, MAPK8, SOCS4 and FOXO3

[i] Only 10 genes were listed as examples in each pathway. GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; MAPK, mitogen-activated protein kinase; FoxO, Forkhead box O.

Table V

PANTHER pathway analysis of potential target genes of miR-663a-5p and miR-154-3p in pancreatic cancer.

Table V

PANTHER pathway analysis of potential target genes of miR-663a-5p and miR-154-3p in pancreatic cancer.

miRNAGenesetDescriptionCountP-valueEOverlapping genesURL
miR-663a-5pP00037Ionotropic glutamate receptor pathway30.08521.05GRIK5, SLC1A7 and SHANK3http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00037
P02743 Formyltetrahydroformate biosynthesis10.13190.14MTRhttp://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P02743
P04392P53 pathway feedback loops 110.13190.14TP53http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P04392
P00012Cadherin signaling pathway60.14123.56PCDHA8, CDH23, WNT7B, FZD1, FZD4 and CELSR1http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00012
P05726 2-arachidonoylglycerol biosynthesis10.15220.16DAGLAhttp://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P05726
P00020FAS signaling pathway20.16140.72DFFB and FADDhttp://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00020
P00029Huntington disease50.17022.96ARL4C, DLG4, GRIK5, PACSIN1 and TP53http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00029
P00057Wnt signaling pathway90.23806.84APC2, PCDHA8, CDH23, TP53, WNT7B, FZD1, FZD4, KREMEN1 and CELSR1http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00057
P00045Notch signaling pathway20.23830.93HEYL and HES2http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00045
P02746Heme biosynthesis10.24670.28ALADhttp://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P02746
miR-154-3pP00032Insulin/IGF pathway-MAPK kinase/MAPK cascade30.01160.48RPS6KA2, RPS6KB1 and SOS1http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00032
P06959CCKR signaling map70.02312.92FOXO3, CXCL2, PRKACB, MAPK8, RPS6KB1, SOS1 and TAC1http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P06959
P043725-Hydroxytryptamine degradation20.03510.30ALDH1L2 and ALDH1B1http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P04372
P00018EGF receptor signaling pathway50.03981.92ERBB4, MAPK8, SOS1, CBLB and PHLDB2http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00018
P00048PI3K pathway30.04200.78FOXO3, RPS6KB1 and SOS1http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00048
P04397p53 pathway by glucose deprivation20.04670.35PRKAB2 and RPS6KB1http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P04397
P00041Metabotropic glutamate receptor group I pathway20.05940.40GRIN2A and PRKACBhttp://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00041
P00001Adrenaline and noradrenaline biosynthesis20.07330.45SLC6A19 and SLC6A15http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00001
P00035Interferon-γ signaling pathway20.07810.47SOCS4 and MAPK8http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00035
P00058mRNA splicing10.08080.08SNRNP40http://www.pantherdb.org/pathway/pathwayDiagram.jsp?catAccession=P00058

[i] miR, microRNA; MAPK, mitogen-activated protein kinase; PI3K, phosphoinositide 3-kinase; IGF, interferon growth factor; CCKR, classical gastrin cholecystokinin receptor.

Prediction of ceRNAs of the differentially-expressed miRNAs

Different online platforms were used to predict the target genes of the differentially-expressed miRNAs. The target circRNAs and lncRNAs of the differentially-expressed miRNAs were searched based on the prediction of the target miRNAs of the differentially-expressed circRNAs and lncRNAs revealed by microarray detection, and the differentially-expressed miRNAs that were detected. The ceRNAs that were negatively associated with the expression of the miRNAs were selected.

Prediction of ceRNAs of miR-663a-5p

A total of 21 differentially-expressed circRNAs that targeted miR-663a-5p were determined, of which nine (hsa_circ_0003176, hsa_circ_0048579, hsa_circ_0063706, hsa_circ_0071922, hsa_circ_0078989, hsa_circ_0079319, hsa_circ_0083080, hsa_circ_0089643 and hsa_circ_0090372) indicated negative associations with miR-663a-5p expression. hsa_circ_0071922 had the highest number of binding sites for miR-663a-5p, with six. Simultaneously, 45 differentially-expressed lncRNAs that targeted miR-663a-5p were determined, of which 8 lncRNAs (RP11-59C5.3, RP13-516M14.8, RP11-196G18.24, AJ006995.3, AC024560.2, PPP1R1C, LINC00595 and HAGLROS) were negatively associated with miR-663a-5p expression. All of these lncRNAs had one miR-663a-5p binding site (Table VI). Subsequently, 144 common predicted targets from at least 8 among 12 programs were selected to overlap with the 3,966 differentially-expressed genes following autophagy inhibition. Collectively, 46 potential targets were determined. Thus, a ceRNA hypothesis figure with miR-663a-5p, 9 circRNAs, 8 lncRNAs and 46 genes was depicted in Fig. 12.

Table VI

ceRNAs and their sequences of miR-663a-5p and miR-154-3p.

Table VI

ceRNAs and their sequences of miR-663a-5p and miR-154-3p.

miRNAceRNASequences
miR-663a-5p hsa_circ_0003176 5′-ACATGGAGCTGCACAGAATGTCAAGAATGCAAAGAGCAGAACCTTGCGCGGGCACAGGGC-3′
hsa_circ_0048579 5′-TTCCTTAGGTTACAAAACAAAACAGGGAGAGAAAAAGCAAGTTCTCCGGGGCTTGGCGTA-3′
hsa_circ_0063706 5′-ACTGGTCAAGTGAGTGACCTGCAGGAAGAAGGCAAGAATGCCATCAACTCACCGATGTCC-3′
hsa_circ_0071922 5′-GACCTCTGTTGTACCTGTAATAAATATATAGAAAAAGCAAACACATACACCAATGCGGAT-3′
hsa_circ_0078989 5′-TGTGGATGCTGTCAAACTGGGTCTACCGATTCTTCGCTGCTGCTGCCTTACCGCCGAGAA-3′
hsa_circ_0079319 5′-TTGTCAAGTGTCAATAAAAGCATCATGTAATTTACCGAACGCCCGCAGCAGGGTCAGAAG-3′
hsa_circ_0083080 5′-TGCATTCATTTTTTACTTTTATTAAAGGTTCAAAACCAACACGCCCCTGGGCGGTGCGCG-3′
hsa_circ_0089643 5′-CGTCACGGCGCGCTACTACGGGAAGTCGCTGCCGTTCGGTGCGCAGTCCACGCAGCGCGG-3′
hsa_circ_0090372 5′-AGCACCTAATTAAAGGGGCTGAAAGTCTGAGCTCTGGCCGGCCCCGGCGATTGGTCACCG-3′
RP11-59C5.3 5′-CTTGTTTGTGCCAGGGAAAGATGGAAAATTTTAGTTTATATTAAACACGCCTTTGAAGAC-3′
RP13-516M14.8 5′-AAACACCTGTCAGATTTCTAAGACCTGAAGCCTTGAGAAAGCATCGAAAAGTATTCAAGT-3′
RP11-196G18.24 5′-CGCTACATAGTACGTATTATGACCAATGGATATCATTCAGAGTTCTAGTTTACATATGCT-3′
AJ006995.3 5′-GGGAACAATTTATCTATGTTAAAAAGATGTGTGTGTGTGAATGGATGTATATACGTGTGC-3′
AC024560.2 5′-TCACCGTGAAATATCTTCTGAGACACAACAATCTCCCTGAATTAAAACTACAGACTCTAA-3′
PPP1R1C 5′-ACTTTTCTGAGTATACCATGGAATTCCACTGCTTGACTTCCAGAAGCATCCTCCATCTCT-3′
LINC00595 5′-GCTGGCATCTAACGGCTTTTGTTAATTACTATATCCCAAAGAAAATGAAATAAAGGAGGG-3′
HAGLROS 5′-GAAGTAGGCTAAGACTGCTGTTGAACTTGAAGCTAAACTTGAATGCCATTTAAAATAACA-3′
miR-154-3p hsa_circ_0000156 5′-TGCTTTCAAGAATCATAGTGAAAGATTCGTTTTCATTGCAGAGTGGTATGATCCAAATGC-3′
hsa_circ_0004089 5′-CACCTGCAGTAGGAAAAAGAAGCAACTAACAAAACACTGTGATAATAAGGATTATTCAGT-3′
hsa_circ_0006461 5′-GTATGATGACATCATCCCCCCGAAGGAGTGGAAGCCGCGGCAGACGTATGATGACATCGA-3′
hsa_circ_0015157 5′-GGAAAGAAAAAATCATAGTGAAAGATTCGTTTTCATTGCAGAGTGGTATGATCCAAATGC-3′
hsa_circ_0038665 5′-TAAAAAATGTTTGGAGTGTAAATATACTGATGATAATGCTCTGATTCCTAAGAATTCTTC-3′
RP11-686O6.1 5′-CCCTGCTTATGATGTAAAAATGGAGGCAGAAGACCTAAATCTAAGGCAATATACAATTTA-3′
LINC01140 5′-CAGATGGGGTTGAAAACTCCCCTCAAAATGCAGTCTTGTTAAATTAAAAACTTGATCCAG-3′

[i] miR, microRNA; ceRNA, competitive endogenous RNA.

Prediction of ceRNAs of miR-154-3p

A total of 6 differentially-expressed circRNAs that targeted miR-154-3p were determined, of which five (hsa_circ_0000156, hsa_circ_0004089, hsa_circ_0006461, hsa_circ_0015157 and hsa_circ_0038665) indicated a negative association with miR-154-3p expression. All of these circRNAs had two miR-154-3p binding sites. Simultaneously, 16 differentially-expressed lncRNAs that targeted miR-154-3p were determined, of which two (RP11-686O6.1 and LINC01140) were negatively associated with miR-154-3p expression. These lncRNAs had one miR-154-3p binding site (Table VI). Similarly to miR-663a-5p, 67 common predicted targets of miR-154-3p from at least 8 among 12 platforms were collected, and then they were intersected into the 3,966 differentially-expressed genes following autophagy suppression. Eventually, 11 potential targets were collected. Hence, a ceRNA hypothesis network with miR-154-3p, 5 circRNAs, 2 lncRNAs and 11 genes was presented (Fig. 13).

Meta-analysis of miR-663a-5p and miR-154-3p expression based on RT-qPCR and GEO data

A comprehensive analysis for the expression of miR-663a-5p and miR-154-3p in pancreatic cancer tissues was performed based on RT-qPCR and GEO data. The combined standardized mean difference (SMD) values of miR-663a-5p was −0.203 [95% confidence interval (CI), −0.675-0.269; Fig. 14A)], which indicated that miR-663a had low expression in pancreatic cancer tissues; however, heterogeneity existed (I2=63.6%; P=0.011; Fig. 14B). Sensitivity analysis and publication bias of miR-663a-5p were depicted in Fig. 14C. The combined SMD values of miR-154-3p was −0.434 (95% CI, −1.079-0.212; Fig. 15A)], which indicated that miR-154-3p had low expression in pancreatic cancer tissues; however, heterogeneity existed (I2=76.0%; P=0.001; Fig. 15B). Sensitivity analysis and publication bias of miR-154-3p were depicted in Fig. 15C.

Discussion

Pancreatic cancer is one of the common malignancy types of the digestive system. Due to the lack of an effective early diagnosis, numerous patients are already in the advanced stage of the cancer when diagnosed (85). Currently, pancreatic cancer treatment remains dominated by surgical resection, which has a low five-year survival rate (86-88). Therefore, there is an urgent requirement for a series of effective markers for pancreatic cancer to change the current status of the poor efficacy of individualized treatment. Recent studies demonstrated that although ncRNAs do not encode proteins, they serve a pivotal role in the regulation of a variety of malignant tumor types, including gastric (89), pancreatic (90), prostate (91) and breast cancer (92). Over the past two years, the extensively investigated ncRNAs, including lncRNAs and miRNAs, and circRNAs, which were newly revealed to have similar regulatory functions, particularly to miRNAs, were indicated to also serve crucial roles (44,93-97). Abnormal expression of a large number of miRNAs has been determined in malignant tumor tissues (98-100). For example, it has been reported that miR-221/222 and miR-15b serve a role in causing malignant tumors (101). Additionally, it was determined that miR-451 and miR-126 have an abnormal expression in lung cancer (102), and the targeted delivery of miRNA therapeutics was a promising strategy for cancer (103). This abnormal expression pattern affects the occurrence, development and prognosis of the tumor by directly regulating the biological functions of the targeted mRNAs (102,104-112). Studies have demonstrated that a variety of miRNAs exhibit abnormal expression patterns in pancreatic cancer cells that affect the occurrence, development and prognosis of pancreatic cancer; for example, the upregulated expression of miR-10b, miR-21, miR23a and miR-27a in pancreatic cancer affects cell growth, proliferation and apoptotic metastasis by targeting programmed cell death 4, BTG anti-proliferation factor 2, neural precursor cell expressed, developmentally downregulated 4-like, phosphatase and tensin homolog, HIV-1 Tat interactive protein 2 and p16 (113-117).

With the increased understanding of the regulatory mechanism of ncRNAs, ceRNAs have attracted increasing attention as important regulators of miRNA activity. ceRNA refers to lncRNAs and circRNAs that suppress miRNA expression through targeted binding to the miRNA, and thus regulate its activity, which affects the development, progression and prognosis of tumors (118-120). Studies have indicated that lncRNA-metastasis associated lung adenocarcinoma transcript 1 can target miR-124 to reduce its expression and activate cyclin-dependent kinase 4, thereby accelerating the progression of breast cancer (121-123). A recent study on pancreatic cancer demonstrated that lncRNA-urothelial cancer associated 1 was overexpressed in pancreatic cancer tissues and reduced miR-135a expression by adsorbing it, thereby inhibiting genes associated with tumor growth and metastasis (124). These examples highlight the significance of the interactions among lncRNAs, miRNAs and mRNAs for cancer diagnosis and treatment.

Therefore, our aim was to mine pancreatic cancer autophagy-associated ceRNAs and miRNAs through assays with ceRNA and miRNA microarrays, respectively, to reveal their targets using bioinformatics methods, construct ceRNA, miRNA and mRNA pathways, predict the potential molecular mechanisms of the ceRNA, miRNA and mRNA pathways in the autophagy of pancreatic cancer, and provide novel ideas and directions for further studies of pancreatic cancer.

Firstly, the differentially-expressed mRNAs were analyzed. The results demonstrated that PANC-1 cells treated with chloroquine diphosphate had multiple differentially-expressed mRNAs, compared with the control group, using the FC value FC>2 or FC<0.5 as the threshold. The bioinformatics analysis of these genes indicated that as the autophagic level changed, the differentially-expressed genes were primarily concentrated in tumor-associated and pancreatic cancer-associated pathways. Notably, the autophagy-associated pathway, in which three genes (ATG12, GABARAPL1 and ULK2) were abnormally expressed when the autophagic level, was inhibited. ATG12 is an autophagy-related protein that has been demonstrated to function as a key autophagy-associated target gene regulated by miR-23b, and suppression of ATG12 significantly increased the radiosensitivity of pancreatic cancer cells, whereas the miR-23b-induced radiosensitivity was eliminated by ATG12 overexpression (125). Studies indicated that ULK served an important role in the autophagy process (126-128). ULK2 and ULK1 are highly homologous, functionally complementary and indispensable for affecting autophagy (129-131). GABARAPL1 is considered as a target of miR-195 and regulates the proliferation, migration, angiogenesis and autophagy of endothelial progenitor cells (132). These three genes (ATG12, GABARAPL1 and ULK2) may serve pivotal roles in the autophagy suppressed by chloroquine diphosphate in pancreatic cancer cells.

To determine relevant miRNAs and elucidate their potential regulatory mechanisms in the autophagy of pancreatic cancer, differentially-expressed miRNAs were analyzed and their target genes were predicted. Subsequently, the results were compared with the target genes detected in the microarray analysis. The co-expressed genes were considered the putative target genes regulated by the abnormally-expressed miRNAs during the autophagy process in pancreatic cancer cells. Bioinformatics analyses were performed on these target genes to identify the signaling pathways and biological processes that were regulated. The results demonstrated that the expression of miR-663a-5p and miR-154-3p was downregulated in the PANC-1 cells treated with chloroquine diphosphate, compared with the control group. miR-663a-5p expression in pancreatic cancer tissues is significantly downregulated and negatively associated with eukaryotic translation elongation factor 1α 2 (eEF1A2) expression due to miR-663 reducing the proliferation and invasion of pancreatic cells by directly targeting eEF1A2 in vitro and in vivo (133). Additionally, miR-663a-5p has been reported to have a low expression in hepatocellular carcinoma (134), non-small cell lung cancer (135) and colorectal cancer (136). Similarly, the present comprehensive analysis indicated that the expression of miR-663a-5p was downregulated in pancreatic ductal adenocarcinoma (PDAC). In the present study, the bioinformatics analyses indicated that the putative autophagy pathways in the pancreatic cancer cells regulated by miR-663a-5p included the aldosterone-regulated sodium reabsorption, Wnt signaling, basal cell carcinoma, fatty acid biosynthesis and PI3K-Akt signaling pathways, of which the Wnt signaling and PI3K-Akt signaling pathways were considered to have a function in the regulation of autophagy (137-143). Currently, whether miR-154-3p is involved in the biological processes of pancreatic cancer cells, including occurrence, development, prognosis and autophagy, remains unknown; however, miR-154-3p was reported to be expressed at a low level in colorectal cancer and was associated with the degree of malignancy of colorectal cancer (144). Similarly, miR-154-3p exhibited low expression in breast cancer and affected the treatment outcome of breast cancer by regulating E2F transcription factor 5 (145). Additionally, the present comprehensive analysis indicated that the expression of miR-154-3p was downregulated in PDAC. Furthermore, the bioinformatics analyses indicated that the putative autophagy pathways in the pancreatic cancer cells regulated by miR-154-3p included the insulin signaling pathway, FoxO signaling, mitogen-activated protein kinase signaling, ErbB signaling, p53 pathway by glucose deprivation and CCKR signaling map pathways. Activation of insulin like growth factor 1 receptor (IGF-1R) signaling antagonizes the decrease in cell viability of human disc cells through the suppression of apoptosis and enhancement of autophagy (146). IGF-1R increases cell viability during hypoxia, which may be dependent on promoting autophagy by suppressing the PI3K/Akt/mTOR signaling pathway (147). The present study demonstrated that a number of genes, including suppressor of cytokine signaling 1 (SOCS1), plasma membrane intrinsic protein 3 (PIP3) and serine/threonine protein phosphatase type 1 α (PP1), which were involved in the insulin signaling pathway, were abnormally expressed when the autophagic level was decreased. Overexpression of SOCS1 suppressed the PI3K/PIP3/Akt signaling pathway with the subsequent PP1 activation, which has been demonstrated to induce autophagy (148,149). Autophagy is inhibited under the condition of oxygen-glucose deprivation (150). Therefore, we hypothesized that miR-154-3p influences the key genes in the insulin signaling or p53 pathways by glucose deprivation, thereby affecting autophagy. Additionally, due to the involvement of CCKR signaling in pancreatic enzyme secretion (151,152), miR-154-3p may serve a key role in the secretion process. Therefore, the data in the present study will provide novel directions for the investigation of pancreatic cancer and its autophagy.

To elucidate which genes inhibited the miRNAs regulating the autophagy of pancreatic cancer cells through competitive binding, potential target associations between circRNA and miRNA, and between lncRNA-miRNA were predicted based on the competitive expression of ceRNA and miRNA. When these results were combined with the differentially-expressed circRNAs and lncRNAs under different autophagic levels detected by the microarrays, numerous ceRNAs that exhibited target associations with miR-663a-5p and miR-154-3p, and negative associations with the expression of the targeted miRNAs under the same changes in the autophagic level were determined. AC024560.2 is an effective lncRNA residing on chromosome 3 that can be used to predict the metastasis of early cervical cancer to lymph nodes (153). In the present study, AC024560.2 was determined to competitively bind to miR-663a-5p and thus regulate the autophagic level of pancreatic cancer cells by inhibiting the expression of this miRNA. For the other ceRNAs, their associations with tumors have not been reported, and further investigations are required.

Since the molecular mechanisms of tumor occurrence and development are complex and the numerous different physiological processes, including differentiation (154) and aging (155), are frequently accompanied by an aberrant autophagic level (156,157), it is not surprising that the interference of autophagy is determined to be one of the therapeutic methods for tumors. The growth of PDAC could be repressed by hydroxychloroquine in vivo and in vitro (158). PDAC was dependent on autophagy and the use of an autophagy inhibitor may become the breakthrough point of treatment (35). However, in the treatment of PDAC with gemcitabine, a high concentration of gemcitabine is required following adding autophagy inhibitors (37). Therefore, autophagy may serve a dual role in the development of PDAC. Investigating the ceRNA network following inhibiting autophagy, of which these genes may serve role in inhibiting autophagy or promoting autophagy, will provide novel ideas for determining new ways to treat pancreatic cancer. On this basis, the biological function and action mechanism of these two miRNAs will be investigated in further studies.

In summary, the miRNAs that were significantly associated with the autophagy of pancreatic cancer included miR-663a-5p and miR-154-3p. These miRNAs were used as the intermediate regulatory sites, upstream to which multiple putative ceRNAs, including AC024560.2, were present and downstream to multiple targeted regulatory genes, including ATG12 and ULK1. The expression of these miRNAs was regulated through ceRNA, miRNA and mRNA interactions, which could serve important regulatory roles in pancreatic cancer autophagy.

In conclusion, autophagy serves an important role in the development and progression of various malignant tumor types, including lung (159), gastric (160) and renal cancer (161). The investigation of differentially-expressed genes in pancreatic cancer cells under different autophagic levels not only clarifies the regulatory mechanism of the autophagic process in pancreatic cancer cells, but also provides novel ideas for the effective diagnosis and treatment of pancreatic cancer. In the present study, microarray technology was employed to confirm that the change in the autophagic level of pancreatic cancer cells was accompanied by various differentially-expressed genes. Following establishing multiple ceRNA, miRNA and mRNA associations, there is more reason to consider that these genes will serve important roles in the diagnosis and treatment of pancreatic cancer, and thus lay the theoretical foundation for subsequent investigations on pancreatic cancer while indicating new research directions. Subsequently, the regulatory functions of these genes at the cellular and tissue levels will be investigated and their specificity and accuracy will be further validated through a series of in vivo and in vitro experiments.

Funding

The study was supported by the funds of the National Natural Science Foundation of China (grant no. NSFC81560448), the Natural Science Foundation of Guangxi, China (grant no. 2016GXNSFBA380039), Medical Excellence Award Funded by the Creative Research Development Grant from the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University Training Program for Distinguished Young Scholars, and the Promoting Project of Basic Capacity for University Young and Middle-aged Teachers in Guangxi (grant no. KY2016LX031). The funders had no role in the study design, the data collection and analysis, the decision to publish, or the preparation of the manuscript.

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

DMW and MTJ wrote the paper, performed the experiments and conducted bioinformatics analysis. PL, HY and YWD wrote the paper and conducted bioinformatics analysis. QY and DYL conducted bioinformatics analysis and statistical data analysis. DZL and GC designed the project, supervised the experiments and corrected the draft.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of the First Affiliated Hospital of Guangxi Medical University, and all patients provided signed informed consent.

Patient consent for publication

Consent for the publication of the pathological data was obtained from all patients who were involved in the present study.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank all publicly available data used in the present study.

Abbreviations:

rRNA

ribosomal RNA

tRNA

transfer RNA

snRNA

small nuclear RNA

lncRNA

long non-coding RNA

miRNA

microRNA

circRNA

circular RNA

ceRNAs

competitive endogenous RNAs

RIN

RNA integrity number

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

GEO

Gene Expression Omnibus

ATG12

autophagy-related 12

References

1 

Ryan DP, Hong TS and Bardeesy N: Pancreatic adenocarcinoma. N Engl J Med. 371:2140–2141. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Swords DS, Firpo MA, Scaife CL and Mulvihill SJ: Biomarkers in pancreatic adenocarcinoma: Current perspectives. OncoTargets Ther. 9:7459–7467. 2016. View Article : Google Scholar

3 

Siegel RL, Miller KD and Jemal A: Cancer Statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Song HY, Wang Y, Lan H and Zhang YX: Expression of Notch receptors and their ligands in pancreatic ductal adenocarcinoma. Exp Ther Med. 16:53–60. 2018.PubMed/NCBI

6 

Xu Q, Gao J and Li Z: Identification of a novel alternative splicing transcript variant of the suppressor of fused: Relationship with lymph node metastasis in pancreatic ductal adenocarcinoma. Int J Oncol. 49:2611–2619. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Li Q, Zhang L, Li X, Yan H, Yang L, Li Y, Li T, Wang J and Cao B: The prognostic significance of human epidermal growth factor receptor family protein expression in operable pancreatic cancer: HER14 protein expression and prognosis in pancreatic cancer. BMC Cancer. 16:9102016. View Article : Google Scholar

8 

Li H, Hao X, Wang H, Liu Z, He Y, Pu M, Zhang H, Yu H, Duan J and Qu S: Circular RNA Expression Profile of Pancreatic Ductal Adenocarcinoma Revealed by Microarray. Cell Physiol Biochem. 40:1334–1344. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Zhu L, Staley C, Kooby D, El-Rays B, Mao H and Yang L: Current status of biomarker and targeted nanoparticle development: The precision oncology approach for pancreatic cancer therapy. Cancer Lett. 388:139–148. 2017. View Article : Google Scholar :

10 

Kocaturk NM and Gozuacik D: Crosstalk Between Mammalian Autophagy and the Ubiquitin-Proteasome System. Front Cell Dev Biol. 6:1282018. View Article : Google Scholar : PubMed/NCBI

11 

Hong JC, Czito BG, Willett CG and Palta M: A current perspective on stereotactic body radiation therapy for pancreatic cancer. OncoTargets Ther. 9:6733–6739. 2016. View Article : Google Scholar

12 

Marsoner K, Haybaeck J, Csengeri D, Waha JE, Schagerl J, Langeder R, Mischinger HJ and Kornprat P: Pancreatic resection for intraductal papillary mucinous neoplasm - a thirteen-year single center experience. BMC Cancer. 16:8442016. View Article : Google Scholar

13 

Kang MJ, Jang JY and Kim SW: Surgical resection of pancreatic head cancer: What is the optimal extent of surgery. Cancer Lett. 382:259–265. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Ellerhoff TP, Berchtold S, Venturelli S, Burkard M, Smirnow I, Wulff T and Lauer UM: Novel epi-virotherapeutic treatment of pancreatic cancer combining the oral histone deacetylase inhibitor resminostat with oncolytic measles vaccine virus. Int J Oncol. 49:1931–1944. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Zuo C, Sheng X, Ma M, Xia M and Ouyang L: ISG15 in the tumorigenesis and treatment of cancer: An emerging role in malignancies of the digestive system. Oncotarget. 7:74393–74409. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Chang JH, Jiang Y and Pillarisetty VG: Role of immune cells in pancreatic cancer from bench to clinical application: An updated review. Medicine (Baltimore). 95:e55412016. View Article : Google Scholar

17 

Kim KH and Lee MS: Autophagy--a key player in cellular and body metabolism. Nat Rev Endocrinol. 10:322–337. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Tao Z, Li T, Ma H, Yang Y, Zhang C, Hai L, Liu P, Yuan F, Li J, Yi L, et al: Autophagy suppresses self-renewal ability and tumorigenicity of glioma-initiating cells and promotes Notch1 degradation. Cell Death Dis. 9:10632018. View Article : Google Scholar : PubMed/NCBI

19 

Mowers EE, Sharifi MN and Macleod KF: Autophagy in cancer metastasis. Oncogene. 36:1619–1630. 2017. View Article : Google Scholar :

20 

Tan YQ, Zhang J and Zhou G: Autophagy and its implication in human oral diseases. Autophagy. 13:225–236. 2017. View Article : Google Scholar :

21 

Green DR, Oguin TH and Martinez J: The clearance of dying cells: Table for two. Cell Death Differ. 23:915–926. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Sionov RV, Vlahopoulos SA and Granot Z: Regulation of Bim in Health and Disease. Oncotarget. 6:23058–23134. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Gafar AA, Draz HM, Goldberg AA, Bashandy MA, Bakry S, Khalifa MA, AbuShair W, Titorenko VI and Sanderson JT: Lithocholic acid induces endoplasmic reticulum stress, autophagy and mitochondrial dysfunction in human prostate cancer cells. PeerJ. 4:e24452016. View Article : Google Scholar : PubMed/NCBI

24 

Jawhari S, Ratinaud MH and Verdier M: Glioblastoma, hypoxia and autophagy: A survival-prone ‘ménage-à-trois’. Cell Death Dis. 7:e24342016. View Article : Google Scholar

25 

Mao S and Zhang J: Role of autophagy in chronic kidney diseases. Int J Clin Exp Med. 8:22022–22029. 2015.

26 

Mo He Y, Luo Q, Qiao B, Xu Y, Zuo R, Deng Z, Nong J, Peng X, He GW, et al: Induction of apoptosis and autophagy via mitochondria- and PI3K/Akt/mTOR-mediated pathways by E. adenophorum in hepatocytes of saanen goat. Oncotarget. 7:54537–54548. 2016.PubMed/NCBI

27 

Goulielmaki M, Koustas E, Moysidou E, Vlassi M, Sasazuki T, Shirasawa S, Zografos G, Oikonomou E and Pintzas A: BRAF associated autophagy exploitation: BRAF and autophagy inhibitors synergise to efficiently overcome resistance of BRAF mutant colorectal cancer cells. Oncotarget. 7:9188–9221. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Vera-Ramirez L, Vodnala SK, Nini R, Hunter KW and Green JE: Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence. Nat Commun. 9:19442018. View Article : Google Scholar : PubMed/NCBI

29 

Li SJ, Sun SJ, Gao J and Sun FB: Wogonin induces Beclin-1/PI3K and reactive oxygen species-mediated autophagy in human pancreatic cancer cells. Oncol Lett. 12:5059–5067. 2016. View Article : Google Scholar

30 

Klein K, Werner K, Teske C, Schenk M, Giese T, Weitz J and Welsch T: Role of TFEB-driven autophagy regulation in pancreatic cancer treatment. Int J Oncol. 49:164–172. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Kwon JJ, Willy JA, Quirin KA, Wek RC, Korc M, Yin XM and Kota J: Novel role of miR-29a in pancreatic cancer autophagy and its therapeutic potential. Oncotarget. 7:71635–71650. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Ranjan A and Srivastava SK: Penfluridol suppresses pancreatic tumor growth by autophagy-mediated apoptosis. Sci Rep. 6:261652016. View Article : Google Scholar : PubMed/NCBI

33 

Seo JW, Choi J, Lee SY, Sung S, Yoo HJ, Kang MJ, Cheong H and Son J: Autophagy is required for PDAC glutamine metabolism. Sci Rep. 6:375942016. View Article : Google Scholar : PubMed/NCBI

34 

Yan Y, Jiang K, Liu P, Zhang X, Dong X, Gao J, Liu Q, Barr MP, Zhang Q, Hou X, et al: Bafilomycin A1 induces caspase-independent cell death in hepatocellular carcinoma cells via targeting of autophagy and MAPK pathways. Sci Rep. 6:370522016. View Article : Google Scholar : PubMed/NCBI

35 

Yang S, Wang X, Contino G, Liesa M, Sahin E, Ying H, Bause A, Li Y, Stommel JM, Dell’antonio G, et al: Pancreatic cancers require autophagy for tumor growth. Genes Dev. 25:717–729. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Yang A, Herter-Sprie G, Zhang H, Lin EY, Biancur D, Wang X, Deng J, Hai J, Yang S, Wong KK, et al: Autophagy sustains pancreatic cancer growth through both cell autonomous and non-autonomous mechanisms. Cancer Discov. 8:276–287. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Mukubou H, Tsujimura T, Sasaki R and Ku Y: The role of autophagy in the treatment of pancreatic cancer with gemcitabine and ionizing radiation. Int J Oncol. 37:821–828. 2010.PubMed/NCBI

38 

Marinković M, Šprung M, Buljubašić M and Novak I: Autophagy Modulation in Cancer: Current Knowledge on Action and Therapy. Oxid Med Cell Longev. 2018.8023821:2018.

39 

Islam MA, Reesor EK, Xu Y, Zope HR, Zetter BR and Shi J: Biomaterials for mRNA delivery. Biomater Sci. 3:1519–1533. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Liu F, Gao S, Yang Y, Zhao X, Fan Y, Ma W, Yang D, Yang A and Yu Y: Antitumor activity of curcumin by modulation of apoptosis and autophagy in human lung cancer A549 cells through inhibiting PI3K/Akt/mTOR pathway. Oncol Rep. 39:1523–1531. 2018.PubMed/NCBI

41 

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

42 

Gong J, Muñoz AR, Chan D, Ghosh R and Kumar AP: STAT3 down regulates LC3 to inhibit autophagy and pancreatic cancer cell growth. Oncotarget. 5:2529–2541. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Raimondi M, Cesselli D, Di Loreto C, La Marra F, Schneider C and Demarchi F: USP1 (ubiquitin specific peptidase 1) targets ULK1 and regulates its cellular compartmentalization and autophagy. Autophagy. Oct 18–2018.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

44 

Pan Y, Li C, Chen J, Zhang K, Chu X, Wang R and Chen L: The Emerging Roles of Long Noncoding RNA ROR (lincRNA-ROR) and its Possible Mechanisms in Human Cancers. Cell Physiol Biochem. 40:219–229. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Bamodu OA, Huang WC, Lee WH, Wu A, Wang LS, Hsiao M, Yeh CT and Chao TY: Aberrant KDM5B expression promotes aggressive breast cancer through MALAT1 overexpression and downregulation of hsa-miR-448. BM. Cancer. 16:1602016.

46 

Zhang Y, He RQ, Dang YW, Zhang XL, Wang X, Huang SN, Huang WT, Jiang MT, Gan XN, Xie Y, et al: Comprehensive analysis of the long noncoding RNA HOXA11-AS gene interaction regulatory network in NSCLC cells. Cancer Cell Int. 16:892016. View Article : Google Scholar : PubMed/NCBI

47 

Peng L, Yuan XQ and Li GC: The emerging landscape of circular RNA ciRS-7 in cancer (Review). Oncol Rep. 33:2669–2674. 2015.Review. View Article : Google Scholar : PubMed/NCBI

48 

Deng T, Yuan Y, Zhang C, Zhang C, Yao W, Wang C, Liu R and Ba Y: Identification of Circulating MiR-25 as a Potential Biomarker for Pancreatic Cancer Diagnosis. Cell Physiol Biochem. 39:1716–1722. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Zhang YH, Fu J, Zhang ZJ, Ge CC and Yi Y: LncRNA-LINC00152 down-regulated by miR-376c-3p restricts viability and promotes apoptosis of colorectal cancer cells. Am J Transl Res. 8:5286–5297. 2016.

50 

Qu S, Yang X, Li X, Wang J, Gao Y, Shang R, Sun W, Dou K and Li H: Circular RNA: A new star of noncoding RNAs. Cancer Lett. 365:141–148. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Li X, Yang L and Chen LL: The Biogenesis, Functions, and Challenges of Circular RNAs. Mol Cell. 71:428–442. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Xuan L, Qu L, Zhou H, Wang P, Yu H, Wu T, Wang X, Li Q, Tian L, Liu M, et al: Circular RNA: A novel biomarker for progressive laryngeal cancer. Am J Transl Res. 8:932–939. 2016.PubMed/NCBI

53 

Jin X, Feng CY, Xiang Z, Chen YP and Li YM: CircRNA expression pattern and circRNA-miRNA-mRNA network in the pathogenesis of nonalcoholic steatohepatitis. Oncotarget. 7:66455–66467. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Liu Q, Zhang X, Hu X, Dai L, Fu X, Zhang J and Ao Y: Circular RNA Related to the Chondrocyte ECM Regulates MMP13 Expression by Functioning as a MiR-136 ‘Sponge’ in Human Cartilage Degradation. Sci Rep. 6:225722016. View Article : Google Scholar

55 

Xie H, Ren X, Xin S, Lan X, Lu G, Lin Y, Yang S, Zeng Z, Liao W, Ding YQ, et al: Emerging roles of circRNA_001569 targeting miR-145 in the proliferation and invasion of colorectal cancer. Oncotarget. 7:26680–26691. 2016.PubMed/NCBI

56 

Zhong Z, Lv M and Chen J: Screening differential circular RNA expression profiles reveals the regulatory role of circTCF25-miR-103a-3p/miR-107-CDK6 pathway in bladder carcinoma. Sci Rep. 6:309192016. View Article : Google Scholar : PubMed/NCBI

57 

Deng X, Feng N, Zheng M, Ye X, Lin H, Yu X, Gan Z, Fang Z, Zhang H, Gao M, et al: PM2.5 exposure-induced autophagy is mediated by lncRNA loc146880 which also promotes the migration and invasion of lung cancer cells. Biochim Biophys Acta, Gen Subj. 1861.112–125. 2017.

58 

Li C, Zhao Z, Zhou Z and Liu R: Linc-ROR confers gemcitabine resistance to pancreatic cancer cells via inducing autophagy and modulating the miR-124/PTBP1/PKM2 axis. Cancer Chemother Pharmacol. 78:1199–1207. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Chen ZH, Wang WT, Huang W, Fang K, Sun YM, Liu SR, Luo XQ and Chen YQ: The lncRNA HOTAIRM1 regulates the degradation of PML-RARA oncoprotein and myeloid cell differentiation by enhancing the autophagy pathway. Cell Death Differ. 24:212–224. 2017. View Article : Google Scholar :

60 

Pawar K, Hanisch C, Palma Vera SE, Einspanier R and Sharbati S: Down regulated lncRNA MEG3 eliminates mycobacteria in macrophages via autophagy. Sci Rep. 6:194162016. View Article : Google Scholar : PubMed/NCBI

61 

Vega-Rubín-de-Celis S, Zou Z, Fernández AF, Ci B, Kim M, Xiao G, Xie Y and Levine B: Increased autophagy blocks HER2-mediated breast tumorigenesis. Proc Natl Acad Sci USA. 115:4176–4181. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Zarzynska JM: The importance of autophagy regulation in breast cancer development and treatment. BioMed Res Int. 2014.710345:2014.

63 

Qin W, Li C, Zheng W, Guo Q, Zhang Y, Kang M, Zhang B, Yang B, Li B, Yang H, et al: Inhibition of autophagy promotes metastasis and glycolysis by inducing ROS in gastric cancer cells. Oncotarget. 6:39839–39854. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Cai J, Li R, Xu X, Zhang L, Lian R, Fang L, Huang Y, Feng X, Liu X, Li X, et al: CK1α suppresses lung tumour growth by stabilizing PTEN and inducing autophagy. Nat Cell Biol. 20:465–478. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Chen PM, Gombart ZJ and Chen JW: Chloroquine treatment of ARPE-19 cells leads to lysosome dilation and intracellular lipid accumulation: possible implications of lysosomal dysfunction in macular degeneration. Cell Biosci. 1:102011. View Article : Google Scholar : PubMed/NCBI

66 

Li ML, Xu YZ, Lu WJ, Li YH, Tan SS, Lin HJ, Wu TM, Li Y, Wang SY and Zhao YL: Chloroquine potentiates the anticancer effect of sunitinib on renal cell carcinoma by inhibiting autophagy and inducing apoptosis. Oncol Lett. 15:2839–2846. 2018.PubMed/NCBI

67 

Maragkakis M, Vergoulis T, Alexiou P, Reczko M, Plomaritou K, Gousis M, Kourtis K, Koziris N, Dalamagas T and Hatzigeorgiou AG: DIANA-microT Web server upgrade supports Fly and Worm miRNA target prediction and bibliographic miRNA to disease association. Nucleic Acids Res 39 (Web Server issue):. W145–W148. 2011. View Article : Google Scholar

68 

Paraskevopoulou MD, Georgakilas G, Kostoulas N, Vlachos IS, Vergoulis T, Reczko M, Filippidis C, Dalamagas T and Hatzigeorgiou AG: DIANA-microT web server v5.0: Service integration into miRNA functional analysis workflows. Nucleic Acids Res. 41(Web Server issue): W169–W173. 2013. View Article : Google Scholar : PubMed/NCBI

69 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2:e3632004. View Article : Google Scholar : PubMed/NCBI

70 

Tsang JS, Ebert MS and van Oudenaarden A: Genome-wide dissection of microRNA functions and cotargeting networks using gene set signatures. Mol Cell. 38:140–153. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Wang X: Improving microRNA target prediction by modeling with unambiguously identified microRNA-target pairs from CLIP-ligation studies. Bioinformatics. 32:1316–1322. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Vejnar CE, Blum M and Zdobnov EM: miRmap web: Comprehensive microRNA target prediction online. Nucleic Acids Res. 41(Web Server issue): W165–W168. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Hsu SD, Chu CH, Tsou AP, Chen SJ, Chen HC, Hsu PW, Wong YH, Chen YH, Chen GH and Huang HD: miRNAMap 2.0: Genomic maps of microRNAs in metazoan genomes. Nucleic Acids Res. 36(Database): D165–D169. 2008. View Article : Google Scholar :

74 

Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M, et al: Combinatorial microRNA target predictions. Nat Genet. 37:495–500. 2005. View Article : Google Scholar : PubMed/NCBI

75 

Kertesz M, Iovino N, Unnerstall U, Gaul U and Segal E: The role of site accessibility in microRNA target recognition. Nat Genet. 39:1278–1284. 2007. View Article : Google Scholar : PubMed/NCBI

76 

Miranda KC, Huynh T, Tay Y, Ang YS, Tam WL, Thomson AM, Lim B and Rigoutsos I: A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes. Cell. 126:1203–1217. 2006. View Article : Google Scholar : PubMed/NCBI

77 

Rehmsmeier M, Steffen P, Hochsmann M and Giegerich R: Fast and effective prediction of microRNA/target duplexes. RNA. 10:1507–1517. 2004. View Article : Google Scholar : PubMed/NCBI

78 

Lewis BP, Burge CB and Bartel DP: Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 120:15–20. 2005. View Article : Google Scholar : PubMed/NCBI

79 

Michelson AM and Orkin SH: Characterization of the homo-polymer tailing reaction catalyzed by terminal deoxynucleotidyl transferase. Implications for the cloning of cDNA. J Biol Chem. 257:14773–14782. 1982.PubMed/NCBI

80 

Hoshino T and Inagaki F: A comparative study of microbial diversity and community structure in marine sediments using poly(A) tailing and reverse transcription-PCR. Front Microbiol. 4:1602013. View Article : Google Scholar : PubMed/NCBI

81 

Tajadini M, Panjehpour M and Javanmard SH: Comparison of SYBR Green and TaqMan methods in quantitative real-time polymerase chain reaction analysis of four adenosine receptor subtypes. Adv Biomed Res. 3:852014. View Article : Google Scholar : PubMed/NCBI

82 

Mardis E and McCombie WR: Library Quantification Using SYBR Green-Quantitative Polymerase Chain Reaction (qPCR). Cold Spring Harb Protoc. 2017:pdb prot0947142017. View Article : Google Scholar

83 

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

84 

Huang W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar :

85 

Kang MJ, Jang JY, Chang YR, Kwon W, Jung W and Kim SW: Revisiting the concept of lymph node metastases of pancreatic head cancer: Number of metastatic lymph nodes and lymph node ratio according to N stage. Ann Surg Oncol. 21:1545–1551. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Georgiadou D, Sergenta nis T N, Sakellariou S, Vlachodimitropoulos D, Psaltopoulou T, Lazaris AC, Gounaris A and Zografos GC: Prognostic role of sex steroid receptors in pancreatic adenocarcinoma. Pathol Res Pract. 212:38–43. 2016. View Article : Google Scholar

87 

Lovecek M, Skalicky P, Klos D, Bebarova L, Neoral C, Ehrmann J, Zapletalova J, Svebisova H, Vrba R, Stasek M, et al: Long-term survival after resections for pancreatic ductal adeno-carcinoma. Single centre study. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 160:280–286. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Ramacciato G, Nigri G, Petrucciani N, Pinna AD, Ravaioli M, Jovine E, Minni F, Grazi GL, Chirletti P, Tisone G, et al: Pancreatectomy with Mesenteric and Portal Vein Resection for Borderline Resectable Pancreatic Cancer: Multicenter Study of 406 Patients. Ann Surg Oncol. 23:2028–2037. 2016. View Article : Google Scholar : PubMed/NCBI

89 

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

90 

Kishikawa T, Otsuka M, Ohno M, Yoshikawa T, Takata A and Koike K: Circulating RNAs as new biomarkers for detecting pancreatic cancer. World J Gastroenterol. 21:8527–8540. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Bolton EM, Tuzova AV, Walsh AL, Lynch T and Perry AS: Noncoding RNAs in prostate cancer: the long and the short of it. Clin Cancer Res. 20:35–43. 2014. View Article : Google Scholar

92 

Zhao Z, Li S, Song E and Liu S: The roles of ncRNAs and histone-modifiers in regulating breast cancer stem cells. Protein Cell. 7:89–99. 2016. View Article : Google Scholar :

93 

Lan PH, Liu ZH, Pei YJ, Wu ZG, Yu Y, Yang YF, Liu X, Che L, Ma CJ, Xie YK, et al: Landscape of RNAs in human lumbar disc degeneration. Oncotarget. 7:63166–63176. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Xu CZ, Jiang C, Wu Q, Liu L, Yan X and Shi R: A Feed-Forward Regulatory Loop between HuR and the Long Noncoding RNA HOTAIR Promotes Head and Neck Squamous Cell Carcinoma Progression and Metastasis. Cell Physiol Biochem. 40:1039–1051. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Dou C, Cao Z, Yang B, Ding N, Hou T, Luo F, Kang F, Li J, Yang X, Jiang H, et al: Changing expression profiles of lncRNAs, mRNAs, circRNAs and miRNAs during osteoclastogenesis. Sci Rep. 6:214992016. View Article : Google Scholar : PubMed/NCBI

96 

Huang M, Zhong Z, Lv M, Shu J, Tian Q and Chen J: Comprehensive analysis of differentially expressed profiles of lncRNAs and circRNAs with associated co-expression and ceRNA networks in bladder carcinoma. Oncotarget. 7:47186–47200. 2016.PubMed/NCBI

97 

Zhou X, Ye F, Yin C, Zhuang Y, Yue G and Zhang G: The Interaction Between MiR-141 and lncRNA-H19 in Regulating Cell Proliferation and Migration in Gastric Cancer. Cell Physiol Biochem. 36:1440–1452. 2015. View Article : Google Scholar : PubMed/NCBI

98 

Zeng JH, Xiong DD, Pang YY, Zhang Y, Tang RX, Luo DZ and Chen G: Identification of molecular targets for esophageal carcinoma diagnosis using miRNA-seq and RNA-seq data from The Cancer Genome Atlas: A study of 187 cases. Oncotarget. 8:35681–35699. 2017.PubMed/NCBI

99 

Massillo C, Dalton GN, Farré PL, De Luca P and De Siervi A: Implications of microRNA dysregulation in the development of prostate cancer. Reproduction. 154:R81–R97. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Murray MJ, Bell E, Raby KL, Rijlaarsdam MA, Gillis AJ, Looijenga LH, Brown H, Destenaves B, Nicholson JC and Coleman N: A pipeline to quantify serum and cerebrospinal fluid microRNAs for diagnosis and detection of relapse in paediatric malignant germ-cell tumours. Br J Cancer. 114:151–162. 2016. View Article : Google Scholar :

101 

Ouyang Q, Xu L, Cui H, Xu M and Yi L: MicroRNAs and cell cycle of malignant glioma. Int J Neurosci. 126:1–9. 2016. View Article : Google Scholar

102 

Del Vescovo V and Denti MA: microRNA and Lung Cancer. Adv Exp Med Biol. 889:153–177. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Rupaimoole R and Slack FJ: MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 16:203–222. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Zhang X, Tang W, Chen G, Ren F, Liang H, Dang Y and Rong M: An Encapsulation of Gene Signatures for Hepatocellular Carcinoma, MicroRNA-132 Predicted Target Genes and the Corresponding Overlaps. PLoS One. 11:e01594982016. View Article : Google Scholar : PubMed/NCBI

105 

Lan D, Zhang X, He R, Tang R, Li P, He Q and Chen G: MiR-133a is downregulated in non-small cell lung cancer: A study of clinical significance. Eur J Med Res. 20:502015. View Article : Google Scholar : PubMed/NCBI

106 

Gan TQ, Tang RX, He RQ, Dang YW, Xie Y and Chen G: Upregulated MiR-1269 in hepatocellular carcinoma and its clinical significance. Int J Clin Exp Med. 8:714–721. 2015.PubMed/NCBI

107 

Zhang X, Li P, Rong M, He R, Hou X, Xie Y and Chen G: MicroRNA-141 is a biomarker for progression of squamous cell carcinoma and adenocarcinoma of the lung: Clinical analysis of 125 patients. Tohoku J Exp Med. 235:161–169. 2015. View Article : Google Scholar : PubMed/NCBI

108 

Ren F, Ding H, Huang S, Wang H, Wu M, Luo D, Dang Y, Yang L and Chen G: Expression and clinicopathological significance of miR-193a-3p and its potential target astrocyte elevated gene-1 in non-small lung cancer tissues. Cancer Cell Int. 15:802015. View Article : Google Scholar : PubMed/NCBI

109 

Liu Y, Ren F, Luo Y, Rong M, Chen G and Dang Y: Down-Regulation of MiR-193a-3p Dictates Deterioration of HCC: A Clinical Real-Time qRT-PCR Study. Med Sci Monit. 21:2352–2360. 2015. View Article : Google Scholar : PubMed/NCBI

110 

Yang He R, Lin L, Chen X, Lin X, Wei X, Liang F, Luo X, Wu Y, Gan YT, et al: MiR-30a-5p suppresses cell growth and enhances apoptosis of hepatocellular carcinoma cells via targeting AEG-1. Int J Clin Exp Pathol. 8:15632–15641. 2015.

111 

Huang WT, Wang HL, Yang H, Ren FH, Luo YH, Huang CQ, Liang YY, Liang HW, Chen G and Dang YW: Lower expressed miR-198 and its potential targets in hepatocellular carcinoma: A clinicopathological and in silico study. OncoTargets Ther. 9:5163–5180. 2016. View Article : Google Scholar

112 

Zhang X, Tang W, Li R, He R, Gan T, Luo Y, Chen G and Rong M: Downregulation of microRNA-132 indicates progression in hepatocellular carcinoma. Exp Ther Med. 12:2095–2101. 2016. View Article : Google Scholar : PubMed/NCBI

113 

Frampton AE, Castellano L, Colombo T, Giovannetti E, Krell J, Jacob J, Pellegrino L, Roca-Alonso L, Funel N, Gall TM, et al: MicroRNAs cooperatively inhibit a network of tumor suppressor genes to promote pancreatic tumor growth and progression. Gastroenterology. 146:268-277e182014. View Article : Google Scholar

114 

Frampton AE, Krell J, Jamieson NB, Gall TM, Giovannetti E, Funel N, Mato Prado M, Krell D, Habib NA, Castellano L, et al: microRNAs with prognostic significance in pancreatic ductal adenocarcinoma: A meta-analysis. Eur J Cancer. 51:1389–1404. 2015. View Article : Google Scholar : PubMed/NCBI

115 

Giovannetti E, Funel N, Peters GJ, Del Chiaro M, Erozenci LA, Vasile E, Leon LG, Pollina LE, Groen A, Falcone A, et al: MicroRNA-21 in pancreatic cancer: Correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 70:4528–4538. 2010. View Article : Google Scholar : PubMed/NCBI

116 

Ouyang H, Gore J, Deitz S and Korc M: microRNA-10b enhances pancreatic cancer cell invasion by suppressing TIP30 expression and promoting EGF and TGF-β actions. Oncogene. 33:4664–4674. 2014. View Article : Google Scholar

117 

Nakata K, Ohuchida K, Mizumoto K, Kayashima T, Ikenaga N, Sakai H, Lin C, Fujita H, Otsuka T, Aishima S, et al: MicroRNA-10b is overexpressed in pancreatic cancer, promotes its invasiveness, and correlates with a poor prognosis. Surgery. 150:916–922. 2011. View Article : Google Scholar : PubMed/NCBI

118 

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

119 

Shao T, Wu A, Chen J, Chen H, Lu J, Bai J, Li Y, Xu J and Li X: Identification of module biomarkers from the dysregulated ceRNA-ceRNA interaction network in lung adenocarcinoma. Mol Biosyst. 11:3048–3058. 2015. View Article : Google Scholar : PubMed/NCBI

120 

Xu J, Li Y, Lu J, Pan T, Ding N, Wang Z, Shao T, Zhang J, Wang L and Li X: The mRNA related ceRNA-ceRNA landscape and significance across 20 major cancer types. Nucleic Acids Res. 43:8169–8182. 2015. View Article : Google Scholar : PubMed/NCBI

121 

Feng T, Shao F, Wu Q, Zhang X, Xu D, Qian K, Xie Y, Wang S, Xu N, Wang Y, et al: miR-124 downregulation leads to breast cancer progression via LncRNA-MALAT1 regulation and CDK4/E2F1 signal activation. Oncotarget. 7:16205–16216. 2016.PubMed/NCBI

122 

Jin H, Li Q, Cao F, Wang SN, Wang RT, Wang Y, Tan QY, Li CR, Zou H, Wang D, et al: miR-124 Inhibits Lung Tumorigenesis Induced by K-ras Mutation and NNK. Mol Ther Nucleic Acids. 9:145–154. 2017. View Article : Google Scholar : PubMed/NCBI

123 

Wang M, Meng B and Liu Y, Yu J, Chen Q and Liu Y: MiR-124 Inhibits Growth and Enhances Radiation-Induced Apoptosis in Non-Small Cell Lung Cancer by Inhibiting STAT3. Cell Physiol Biochem. 44:2017–2028. 2017. View Article : Google Scholar : PubMed/NCBI

124 

Zhang X, Gao F, Zhou L, Wang H, Shi G and Tan X: UCA1 Regulates the Growth and Metastasis of Pancreatic Cancer by Sponging miR-135a. Oncol Res. 25:1529–1541. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Wang P, Zhang L, Chen Z and Meng Z: MicroRNA targets autophagy in pancreatic cancer cells during cancer therapy. Autophagy. 9:2171–2172. 2013. View Article : Google Scholar : PubMed/NCBI

126 

Ganley IG, Lam H, Wang J, Ding X, Chen S and Jiang X: ULK1.ATG13.FIP200 complex mediates mTOR signaling and is essential for autophagy. J Biol Chem. 284:12297–12305. 2009. View Article : Google Scholar : PubMed/NCBI

127 

Hosokawa N, Hara T, Kaizuka T, Kishi C, Takamura A, Miura Y, Iemura S, Natsume T, Takehana K, Yamada N, et al: Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 20:1981–1991. 2009. View Article : Google Scholar : PubMed/NCBI

128 

Jung CH, Jun CB, Ro SH, Kim YM, Otto NM, Cao J, Kundu M and Kim DH: ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol Biol Cell. 20:1992–2003. 2009. View Article : Google Scholar : PubMed/NCBI

129 

Yan J, Kuroyanagi H, Tomemori T, Okazaki N, Asato K, Matsuda Y, Suzuki Y, Ohshima Y, Mitani S, Masuho Y, et al: Mouse ULK2, a novel member of the UNC-51-like protein kinases: Unique features of functional domains. Oncogene. 18:5850–5859. 1999. View Article : Google Scholar : PubMed/NCBI

130 

Chan EY, Longatti A, McKnight NC and Tooze SA: Kinase-inactivated ULK proteins inhibit autophagy via their conserved C-terminal domains using an Atg13-independent mechanism. Mol Cell Biol. 29:157–171. 2009. View Article : Google Scholar :

131 

Kundu M, Lindsten T, Yang CY, Wu J, Zhao F, Zhang J, Selak MA, Ney PA and Thompson CB: Ulk1 plays a critical role in the autophagic clearance of mitochondria and ribosomes during reticulocyte maturation. Blood. 112:1493–1502. 2008. View Article : Google Scholar : PubMed/NCBI

132 

Mo J, Zhang D and Yang R: MicroRNA-195 regulates proliferation, migration, angiogenesis and autophagy of endothelial progenitor cells by targeting GABARAPL1. Biosci Rep. 36:362016. View Article : Google Scholar

133 

Zang W, Wang Y, Wang T, Du Y, Chen X, Li M and Zhao G: miR-663 attenuates tumor growth and invasiveness by targeting eEF1A2 in pancreatic cancer. Mol Cancer. 14:372015. View Article : Google Scholar : PubMed/NCBI

134 

Huang W, Li J, Guo X, Zhao Y and Yuan X: miR-663a inhibits hepatocellular carcinoma cell proliferation and invasion by targeting HMGA2. Biomed Pharmacother. 81:431–438. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Zhang Y, Xu X, Zhang M, Wang X, Bai X, Li H, Kan L, Zhou Y, Niu H and He P: MicroRNA-663a is downregulated in non-small cell lung cancer and inhibits proliferation and invasion by targeting JunD. BMC Cancer. 16:3152016. View Article : Google Scholar : PubMed/NCBI

136 

Kuroda K, Fukuda T, Krstic-Demonacos M, Demonacos C, Okumura K, Isogai H, Hayashi M, Saito K and Isogai E: miR-663a regulates growth of colon cancer cells, after administration of antimicrobial peptides, by targeting CXCR4-p21 pathway. BMC Cancer. 17:332017. View Article : Google Scholar : PubMed/NCBI

137 

Lin Y, Kuang W, Wu B, Xie C, Liu C and Tu Z: IL-12 induces autophagy in human breast cancer cells through AMPK and the PI3K/Akt pathway. Mol Med Rep. 16:4113–4118. 2017. View Article : Google Scholar : PubMed/NCBI

138 

Wu L, Zhang Q, Mo W, Feng J, Li S, Li J, Liu T, Xu S, Wang W, Lu X, et al: Quercetin prevents hepatic fibrosis by inhibiting hepatic stellate cell activation and reducing autophagy via the TGF-β1/Smads and PI3K/Akt pathways. Sci Rep. 7:92892017. View Article : Google Scholar

139 

Ren L, Han W, Yang H, Sun F, Xu S, Hu S, Zhang M, He X, Hua J and Peng S: Autophagy stimulated proliferation of porcine PSCs might be regulated by the canonical Wnt signaling pathway. Biochem Biophys Res Commun. 479:537–543. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Lin R, Feng J, Dong S, Pan R, Zhuang H and Ding Z: Regulation of autophagy of prostate cancer cells by β-catenin signaling. Cell Physiol Biochem. 35:926–932. 2015. View Article : Google Scholar

141 

Zhao S, Li L, Wang S, Yu C, Xiao B, Lin L, Cong W, Cheng J, Yang W, Sun W, et al: H2O2 treatment or serum deprivation induces autophagy and apoptosis in naked mole-rat skin fibroblasts by inhibiting the PI3K/Akt signaling pathway. Oncotarget. 7:84839–84850. 2016. View Article : Google Scholar : PubMed/NCBI

142 

Lu X, Lv S, Mi Y, Wang L and Wang G: Neuroprotective effect of miR-665 against sevoflurane anesthesia-induced cognitive dysfunction in rats through PI3K/Akt signaling pathway by targeting insulin-like growth factor 2. Am J Transl Res. 9:1344–1356. 2017.PubMed/NCBI

143 

Su N, Wang P and Li Y: Role of Wnt/β-catenin pathway in inducing autophagy and apoptosis in multiple myeloma cells. Oncol Lett. 12:4623–4629. 2016. View Article : Google Scholar

144 

Kai Y, Qiang C, Xinxin P, Miaomiao Z and Kuailu L: Decreased miR-154 expression and its clinical significance in human colorectal cancer. World J Surg Oncol. 13:1952015. View Article : Google Scholar : PubMed/NCBI

145 

Xu H, Fei D, Zong S and Fan Z: MicroRNA-154 inhibits growth and invasion of breast cancer cells through targeting E2F5. Am J Transl Res. 8:2620–2630. 2016.PubMed/NCBI

146 

Liu ZQ, Zhao S and Fu WQ: Insulin-like growth factor 1 antagonizes lumbar disc degeneration through enhanced autophagy. Am J Transl Res. 8:4346–4353. 2016.PubMed/NCBI

147 

Liu Q, Guan JZ, Sun Y, Le Z, Zhang P, Yu D and Liu Y: Insulin-like growth factor 1 receptor-mediated cell survival in hypoxia depends on the promotion of autophagy via suppression of the PI3K/Akt/mTOR signaling pathway. Mol Med Rep. 15:2136–2142. 2017. View Article : Google Scholar : PubMed/NCBI

148 

Ceperuelo-Mallafré V, Ejarque M, Serena C, Duran X, Montori-Grau M, Rodríguez MA, Yanes O, Núñez-Roa C, Roche K, Puthanveetil P, et al: Adipose tissue glycogen accumulation is associated with obesity-linked inflammation in humans. Mol Metab. 5:5–18. 2015. View Article : Google Scholar

149 

Ciarcia R, Damiano S, Montagnaro S, Pagnini U, Ruocco A, Caparrotti G, d’ Angelo D, Boffo S, Morales F, Rizzolio F, et al: Combined effects of PI3K and SRC kinase inhibitors with imatinib on intracellular calcium levels, autophagy, and apoptosis in CML-PBL cells. Cell Cycle. 12:2839–2848. 2013. View Article : Google Scholar : PubMed/NCBI

150 

Hua R, Han S, Zhang N, Dai Q, Liu T and Li J: cPKCγ-Modulated Sequential Reactivation of mTOR Inhibited Autophagic Flux in Neurons Exposed to Oxygen Glucose Deprivation/Reperfusion. Int J Mol Sci. 19:192018. View Article : Google Scholar

151 

Shivaram S, Crist KA, Chaudhuri B, Mucci SJ and Chaudhuri PK: Effect of CCK receptor antagonist on growth of pancreatic adenocarcinoma. J Surg Res. 53:234–237. 1992. View Article : Google Scholar : PubMed/NCBI

152 

Clerc P, Saillan-Barreau C, Desbois C, Pradayrol L, Fourmy D and Dufresne M: Transgenic mice expressing cholecystokinin 2 receptors in the pancreas. Pharmacol Toxicol. 91:321–326. 2002. View Article : Google Scholar

153 

Shang C, Zhu W, Liu T, Wang W, Huang G, Huang J, Zhao P, Zhao Y and Yao S: Characterization of long non-coding RNA expression profiles in lymph node metastasis of early-stage cervical cancer. Oncol Rep. 35:3185–3197. 2016. View Article : Google Scholar : PubMed/NCBI

154 

Bronietzki AW, Schuster M and Schmitz I: Autophagy in T-cell development, activation and differentiation. Immunol Cell Biol. 93:25–34. 2015. View Article : Google Scholar

155 

Martinez-Lopez N, Athonvarangkul D and Singh R: Autophagy and aging. Adv Exp Med Biol. 847:73–87. 2015. View Article : Google Scholar : PubMed/NCBI

156 

Tan Y, Gong Y, Dong M, Pei Z and Ren J: Role of autophagy in inherited metabolic and endocrine myopathies. Biochim Biophys Acta Mol Basis Dis. 1865.48–55. 2018.

157 

Chu CT: Autophagy in Neurological Diseases: An update. Neurobiol Dis. S0969–9961(18): 30728–9. 2018.

158 

Yang A, Rajeshkumar NV, Wang X, Yabuuchi S, Alexander BM, Chu GC, Von Hoff DD, Maitra A and Kimmelman AC: Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov. 4:905–913. 2014. View Article : Google Scholar : PubMed/NCBI

159 

Zhan Y, Wang K, Li Q, Zou Y, Chen B, Gong Q, Ho HI, Yin T, Zhang F, Lu Y, et al: The Novel Autophagy Inhibitor Alpha-Hederin Promoted Paclitaxel Cytotoxicity by Increasing Reactive Oxygen Species Accumulation in Non-Small Cell Lung Cancer Cells. Int J Mol Sci. 19:192018. View Article : Google Scholar

160 

Chen S, Wu J, Jiao K, Wu Q, Ma J, Chen D, Kang J, Zhao G, Shi Y, Fan D, et al: MicroRNA-495-3p inhibits multidrug resistance by modulating autophagy through GRP78/mTOR axis in gastric cancer. Cell Death Dis. 9:10702018. View Article : Google Scholar : PubMed/NCBI

161 

Verma SP and Das P: Monensin induces cell death by autophagy and inhibits matrix metalloproteinase 7 (MMP7) in UOK146 renal cell carcinoma cell line. In Vitro Cell Dev Biol Anim. 54:736–742. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 54 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wei DM, Jiang MT, Lin P, Yang H, Dang YW, Yu Q, Liao DY, Luo DZ and Chen G: Potential ceRNA networks involved in autophagy suppression of pancreatic cancer caused by chloroquine diphosphate: A study based on differentially‑expressed circRNAs, lncRNAs, miRNAs and mRNAs. Int J Oncol 54: 600-626, 2019
APA
Wei, D., Jiang, M., Lin, P., Yang, H., Dang, Y., Yu, Q. ... Chen, G. (2019). Potential ceRNA networks involved in autophagy suppression of pancreatic cancer caused by chloroquine diphosphate: A study based on differentially‑expressed circRNAs, lncRNAs, miRNAs and mRNAs. International Journal of Oncology, 54, 600-626. https://doi.org/10.3892/ijo.2018.4660
MLA
Wei, D., Jiang, M., Lin, P., Yang, H., Dang, Y., Yu, Q., Liao, D., Luo, D., Chen, G."Potential ceRNA networks involved in autophagy suppression of pancreatic cancer caused by chloroquine diphosphate: A study based on differentially‑expressed circRNAs, lncRNAs, miRNAs and mRNAs". International Journal of Oncology 54.2 (2019): 600-626.
Chicago
Wei, D., Jiang, M., Lin, P., Yang, H., Dang, Y., Yu, Q., Liao, D., Luo, D., Chen, G."Potential ceRNA networks involved in autophagy suppression of pancreatic cancer caused by chloroquine diphosphate: A study based on differentially‑expressed circRNAs, lncRNAs, miRNAs and mRNAs". International Journal of Oncology 54, no. 2 (2019): 600-626. https://doi.org/10.3892/ijo.2018.4660