Open Access

Circular RNA hsa_circ_0005114‑miR‑142‑3p/miR‑590‑5p-adenomatous polyposis coli protein axis as a potential target for treatment of glioma

  • Authors:
    • Bo Wei
    • Le Wang
    • Jingwei Zhao
  • View Affiliations

  • Published online on: November 19, 2020     https://doi.org/10.3892/ol.2020.12320
  • Article Number: 58
  • 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

Glioma is the most common type of brain tumor and is associated with a high mortality rate. Despite recent advances in treatment options, the overall prognosis in patients with glioma remains poor. Studies have suggested that circular (circ)RNAs serve important roles in the development and progression of glioma and may have potential as therapeutic targets. However, the expression profiles of circRNAs and their functions in glioma have rarely been studied. The present study aimed to screen differentially expressed circRNAs (DECs) between glioma and normal brain tissues using sequencing data collected from the Gene Expression Omnibus database (GSE86202 and GSE92322 datasets) and explain their mechanisms based on the competing endogenous (ce)RNA regulatory hypothesis. In total, 424 commonly downregulated DECs (with the Gene_symbol annotated in the circBase database) in these two datasets were identified. Using the CircInteractome and Starbase databases, 18 micro (mi)RNAs (miRs) were predicted to interact with DECs, while 22 glioma‑related genes obtained from the Comparative Toxicogenomics Database were predicted to be regulated by 15 miRNAs via the miRwalk 2.0 database. A ceRNA network was established based on 115 DECs, 15 miRNAs and 22 mRNAs. LinkedOmics online analysis using The Cancer Genome Atlas (TCGA) data showed that hsa-miR-142-3p/hsa‑miR‑590‑5p and their target gene adenomatous polyposis coli protein (APC) were all significantly associated with overall survival rate and their prognosis trend was opposite, revealing that high expression levels of hsa‑miR‑142‑3p/hsa‑miR‑590‑5 were associated with a poor overall survival rate, while high APC expression with a good overall survival rate. UALCAN analysis using TCGA data of glioblastoma multiforme and the GSE25632 and GSE103229 microarray datasets showed that hsa‑miR‑142‑3p/hsa‑miR‑590‑5p was upregulated and APC was downregulated. Thus, hsa-miR-142-3p/hsa-miR-590-5p-APC-related circ/ceRNA axes may be important in glioma, and hsa_circ_0005114 interacted with both of these miRNAs. Functional analysis showed that hsa_circ_0005114 was involved in insulin secretion, while APC was associated with the Wnt signaling pathway. In conclusion, hsa_circ_0005114-miR-142-3p/miR-590-5p‑APC ceRNA axes may be potential targets for the treatment of glioma.

Introduction

Glioma is the most common malignant primary brain tumor, with an incidence of 4.7–7.3/100,000 individuals (14) and a mortality rate of 1.2–4.0/100,000 individuals (5). Despite advances in treatment options (such as radical surgery and chemo-radiotherapy), the prognosis of most patients remains poor due to high recurrence rates and invasiveness, with the 5-year survival rate of only 3–6% for grade IV (1,4). Therefore, it is important to further resolve the mechanisms underlying glioma progression to identify novel targets and develop new therapeutic approaches.

Circular (circ)RNAs are a newly identified class of non-coding (nc)RNAs with a covalent loop structure lacking the 5′-end cap and 3′-end ploy A tail, which prevents degradation by RNA exonucleases and confers strong stability in specific cells, such as tumor cells (68). Therefore, abnormal expression of circRNAs may be a potential mechanism for the development and progression of cancer, including glioma. This hypothesis has been demonstrated in several studies. For example, Zhou et al (9) demonstrated that hsa_circ_0008344 was significantly upregulated in glioblastoma tissues compared with the adjacent normal brain tissue. Zhou et al also reported that knockdown of hsa_circ_0008344 suppressed glioblastoma cell proliferation, colony formation, migration and invasion, but facilitated apoptosis. Wang et al (10) found that hsa_circ_0001649 expression was decreased in glioma specimens and cell lines. In addition, downregulated hsa_circ_0001649 is significantly associated with advanced grade and poor prognosis for patients with glioma. The functions and mechanism of circRNAs remain unclear; however, accumulating evidence indicates circRNAs have micro (mi)RNA (miR) response elements (MREs) (68), while MREs are widely known to be located in the 3′-untranslated region (3′-UTR) of the target mRNAs and miRNAs inhibit mRNA translation by binding to MREs. Thus, circRNAs may directly bind to miRNAs, impacting the interaction between miRNAs and mRNAs and contributing to the progression of glioma. This is known as the competitive endogenous RNA (ceRNA) hypothesis (68). This mechanism of circRNA function has been reported in several studies. For example, circ-pituitary homeobox (PITX)1 is upregulated in cancerous tissues and four cell lines of glioma. Dual-luciferase reporter and rescue assays indicated that circ-PITX1 may exert its oncogenic functions by sponging miR-518a-5p, leading to the release of the miR-518a-5p-mediated repression of interleukin 17 receptor D (IL17RD) (11). Similarly, hsa_circ_0034642 levels are also increased in glioma tissues and cells. Mechanistically, hsa_circ_0034642 sponges miR-1205 to regulate basic leucine zipper ATF-like transcription factor (BATF)3 levels to facilitate cell proliferation, migration and invasion (12). Circ-epididymis-specific α-mannosidase (MAN2B2) regulates S100 calcium-binding protein A8 (S100A8) expression by inhibiting miR-1205 and increasing S100A8 expression rescues the tumor suppressor effects of knockout of circ-MAN2B2 (13). Upregulated hsa_circ_0074362 plays a role in glioma progression by regulating the miR-1236-3p/homeobox B7 pathway (14), and downregulated hsa_circ_0001946 may act as a ceRNA, inhibiting glioblastoma progression by modulating miR-671-5p and cerebellar degeneration-related protein 1 (15). Overall, these findings imply that circRNA/ceRNA axes may be potential targets for the treatment of glioma; however, few studies have investigated this.

The present study aimed to further screen crucial circRNA/ceRNA axes for glioma by using the circRNA sequencing data of Yuan et al (16) and Zhu et al (17). Only the common differentially expressed circRNAs (DECs) in these two datasets were used to construct the ceRNA network based on the interactions between circRNAs-miRNAs and miRNAs-mRNAs. The expression and prognostic ability of these hub miRNAs and mRNAs were also validated using The Cancer Genome Atlas (TCGA) database and microarray data, which indirectly illustrated the possible proto- or antioncogenic roles of circRNAs. The current study may provide some novel therapeutic targets for glioma.

Materials and methods

Data collection

In total, two circRNA datasets of glioblastoma with the accession numbers GSE86202 (16) and GSE92322 (17) were downloaded from the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/). Three glioma and paired normal brain tissues were used for RNA sequencing on Illumina HiSeq 2500 (Homo sapiens, GPL16791) in GSE86202 dataset (16), while 10 samples (five glioma and paired normal brain tissues) were included for circRNA sequencing on Illumina HiSeq 2000 (Homo sapiens, GPL11154) in GSE92322 dataset (17).

Screening of DECs

The normalized data of each dataset was downloaded from the GEO database. The gene symbol and circID of circRNAs were annotated using the circBase database (http://www.circbase.org) (18) according to the chromosome location and information of the positive-negative chain. The DECs were identified using edgeR package (http://www.bioconductor.org/packages/release/bioc/html/edgeR.html) (19). |log2FC (fold-change) |>1 and P<0.05 were defined as the significance threshold values. The heatmap of DECs was created using pheatmap package (version 1.0.8; http://cran.r-project.org/web/packages/pheatmap) of R language (version 3.4.1; http://www.r-project.org/) (20).

Construction of circRNA-miRNA-mRNA ceRNA network

The miRNAs that interacted with common DECs in two datasets were predicted using the CircInteractome (https://circinteractome.nia.nih.gov) (21) and StarBase (http://starbase.sysu.edu.cn/starbase2/) (22) databases. Only the shared miRNAs predicted in these two databases were used for further analysis. The target genes of these miRNAs were subsequently predicted using the miRwalk 2.0 database (23), which included 12 prediction programs (miRWalk, MicroT4, miRanda, miRBridge, miRDB, miRMap, miRNAMap, PICTAR2, PITA, RNA22, RNAhybrid and Targetscan). Only the target genes predicted by at least six algorithms were retained, which were then compared with established glioma-related genes collected from the Comparative Toxicogenomics database (CTD) (24) to obtain glioma-associated miRNA-mRNA interaction pairs. The circRNA-miRNA-mRNA-ceRNA network was then established based on these interaction relationships between DECs-miRNAs and miRNAs-mRNAs using Cytoscape software (version 3.6.1; www.cytoscape.org/) (25).

Functional enrichment analysis for DECs and target genes in the ceRNA network

Enrichment analyses for Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) [including biological process (BP), cellular component (CC) and molecular function (MF)] terms were conducted using the ClusterProfiler tool (26) to predict the possible function of DECs and target genes in the ceRNA network. P<0.05 adjusted for multiple testing according to Benjamini and Hochberg (27) was regarded as the cut-off value.

Prognosis and expression validation for miRNAs and target genes in the ceRNA network

The LinkedOmics online database (http://www.linkedomics.org/) (28) was searched to explore the associations between miRNAs/mRNAs and overall survival (OS) rate for glioma based on a Cox regression, after which Kaplan-Meier survival curves were automatically provided. TCGA-glioblastoma multiforme-low grade glioma (GBMLGG) miRNASeq/RNA-seq and clinical data were selected for miRNAs/mRNAs analysis. The UALCAN web-portal (http://ualcan.path.uab.edu/index.html) (29) was used for validation of the expression of target genes of miRNAs between unmatched glioblastoma multiforme and normal control tissues using TCGA data, in which the statistical significance was estimated using unpaired Student's t-tests. GSE103229 (including five glioblastoma and five normal control tissues) and GSE25632 (including 82 glioblastoma and five normal controls) (30) microarray datasets in the GEO database were used to determine the expression of crucial miRNAs with GEO2R, an interactive web-based statistical tool (https://www.ncbi.nlm.nih.gov/geo/geo2r/). P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of DECs

In the GSE86202 dataset, 23,463/37,085 circRNAs were annotated to the circID in the circBase database and 6,131 had corresponding Gene_symbols. In the GSE92322 dataset, 12,863/59,386 circRNAs were annotated to the circID in the circBase database and 4,676 of them had corresponding Gene_symbol.

Using the edgeR package, a total of 22 upregulated (20 had circID and Gene_symbol) and 2,068 (1,874 had circID and 1,332 had Gene_symbol) DECs were identified between glioma and paired normal brain tissues in the GSE86202 dataset. Meanwhile, 29 upregulated (zero had circID and Gene_symbol) and 1,205 (1,196 had circID and 884 had Gene_symbol) DECs were identified in the GSE92322 dataset (Table I). The heat maps of these DECs are displayed in Fig. 1. These findings suggested that these DECs may be effective to classify the samples into two groups.

Table I.

circRNAs identified for glioma.

Table I.

circRNAs identified for glioma.

GSE92322GSE86202


circIDGene_symbollogFCP-valuelogFCP-value
hsa_circ_0001368KLHL24−7.03946 1.62×10−7−11.743 3.62×10−3
hsa_circ_0098551YAF2−6.11209 1.81×10−4−9.32971 1.72×10−2
hsa_circ_0100496DGKH−5.23572 6.47×10−3−8.82079 2.37×10−2
hsa_circ_0001367KLHL24−5.88609 5.50×10−4−10.9425 6.07×10−3
hsa_circ_0064615SLC4A7−5.87691 3.79×10−4−10.562 7.75×10−3
hsa_circ_0060425PTPRT−5.01633 1.54×10−2−7.7421 4.77×10−2
hsa_circ_0001936BRWD3−5.02396 1.54×10−2−8.73538 2.51×10−2
hsa_circ_0104727SH3GL3−5.67125 7.75×10−4−11.395 4.53×10−3
hsa_circ_0005114RIMS2−8.09913 3.48×10−11−6.44629 3.90×10−2
hsa_circ_0104726SH3GL3−4.80592 1.18×10−2−10.2547 9.46×10−3
hsa_circ_0000120MAN1A2−4.33712 3.20×10−2−11.2985 4.83×10−3
hsa_circ_0069718DCUN1D4−5.42038 2.74×10−3−10.1942 9.83×10−3
hsa_circ_0130887PEX3−5.26036 6.47×10−3−10.2911 9.24×10−3
hsa_circ_0004516C12orf24−4.78292 1.18×10−2−9.66325 1.38×10−2
hsa_circ_0117841SLC4A10−5.46976 2.74×10−3−10.8846 6.30×10−3
hsa_circ_0001369KLHL24−5.94584 2.61×10−4−10.6354 7.40×10−3

[i] Only the top 16 most significantly downregulated circRNAs that may act as a sponge of miR-142-3p/miR-590-5p are listed. FC, fold change; circ, circular.

Functional enrichment analysis for DECs

A comparison between the two aforementioned datasets reported that 424 downregulated DECs with Gene_symbol were shared. The functions of DECs were enriched according to their corresponding Gene_symbols. The results showed that 23 KEGG pathways were enriched (Table II and Fig. 2A), mainly including ‘retrograde endocannabinoid signaling’, ‘ErbB signaling pathway’ and ‘insulin secretion’ for hsa_circ_0005114. Meanwhile, KEGG pathways enriched for hsa_circ_0100496 were ‘calcium signaling pathway’, ‘cholinergic synapse’ and ‘choline metabolism in cancer’, and ‘N-Glycan biosynthesis’ was enriched for hsa_circ_0000120. Furthermore, 143 GO terms (including 87 BPs, 38 CCs and 18 MFs; Table III and Fig. 2B) were also enriched, including ‘regulation of cell morphogenesis’ (hsa_circ_0005114 and hsa_circ_0001936), ‘regulation of neuron projection development’ (hsa_circ_0005114), ‘central nervous system neuron differentiation’ (hsa_circ_0117841), ‘positive regulation of neuron differentiation’ (hsa_circ_0005114, hsa_circ_0104727) and ‘cell-cell junction’ (hsa_circ_0001368). These findings suggested that hsa_circ_0005114 may be crucial for the development of glioma since it was enriched in KEGG pathways and GO terms.

Table II.

Kyoto Encyclopedia of Genes and Genomes pathways enriched for differentially expressed circRNAs.

Table II.

Kyoto Encyclopedia of Genes and Genomes pathways enriched for differentially expressed circRNAs.

IDDescriptionAdjusted P-valueGene ID
hsa04723Retrograde endocannabinoid signaling 1.65×10−2 CACNA1A/CACNA1C/GABRB2/GABRG3/GNAQ/NAPEPLD/NDUFA10/NDUFS1/PRKACB/PRKCB/RIMS1
hsa04012ErbB signaling pathway 1.65×10−2 AKT2/AKT3/PAK1/PAK3/PIK3CA/PRKCB/PTK2/SOS2
hsa04911Insulin secretion 1.65×10−2 CACNA1C/GNAQ/KCNMA1/PCLO/PRKACB/PRKCB/RIMS2/RYR2
hsa04020Calcium signaling pathway 1.65×10−2 RYR2/CACNA1A/CACNA1C/CACNA1E/CAMK4/GNAQ/PDE1C/PHKB/PRKACB/PRKCB/RYR2/SLC8A1
hsa04725Cholinergic synapse 1.65×10−2 AKT2/AKT3/CACNA1A/CACNA1C/CAMK4/GNAQ/PIK3CA/PRKACB/PRKCB
hsa05211Renal cell carcinoma 1.65×10−2 AKT2/AKT3/CREBBP/PAK1/PAK3/PIK3CA/SOS2
hsa05205Proteoglycans in cancer 2.22×10−2 AKT2/AKT3/ANK2/ANK3/ARHGEF12/PAK1/PIK3CA/PRKACB/PRKCB/PTK2/SOS2/TIAM1
hsa05231Choline metabolism in cancer 2.27×10−2 AKT2/AKT3/DGKB/DGKH/DGKI/PIK3CA/PRKCB/SOS2
hsa04660T cell receptor signaling pathway 2.30×10−2 AKT2/AKT3/DGKI/4CACNA1C/PAK1/PAK3/PIK3CA/SOS2
hsa04810Regulation of actin cytoskeleton 2.58×10−2 APC/ARHGEF12/ARHGEF7/DOCK1/MYH10/PAK1/PAK3/PIK3CA/PIKFYVE/PTK2/SOS2/TIAM1
hsa04024cAMP signaling pathway 3.38×10−2 AKT2/AKT3/RYR2/CACNA1C/CAMK4/CREBBP/PAK1/PIK3CA/PRKACB/RYR2/TIAM1
hsa04720Long-term potentiation 3.38×10−2 CACNA1C/CAMK4/CREBBP/GNAQ/PRKACB/PRKCB
hsa04919Thyroid hormone signaling pathway 3.38×10−2 AKT2/AKT3/CREBBP/MED13L/PIK3CA/PRKACB/PRKCB/THRB
hsa05032Morphine addiction 3.38×10−2 CACNA1A/GABRB2/GABRG3/PDE1C/PDE8A/PRKACB/PRKCB
hsa04014Ras signaling pathway 3.38×10−2 AKT2/AKT3/NTRK2/PAK1/PAK3/PIK3CA/PRKACB/PRKCB/RAPGEF5/RASA2/SOS2/TIAM1
hsa04961Endocrine and other factor-regulated calcium reabsorption 3.38×10−2 RYR2/GNAQ/PRKACB/PRKCB/SLC8A1
hsa00280Valine, leucine and isoleucine degradation 3.48×10−2 HADHB/HMGCLL1/HMGCS1/MCCC1/PCCA
hsa00510N-Glycan biosynthesis 3.48×10−2 FUT8/MAN1A2/MAN2A1/ST6GAL2/TUSC3
hsa04662B cell receptor signaling pathway 3.48×10−2 AKT2/AKT3/4CACNA1C/PIK3CA/PRKCB/SOS2
hsa05170Human immunodeficiency virus 1 infection 3.94×10−2 AKT2/AKT3/CUL5/GNAQ/4CACNA1C/PAK1/PAK3/PDIA3/PIK3CA/PRKCB/PTK2
hsa04070 Phosphatidylinositol signaling system 3.94×10−2 DGKB/DGKH/DGKI/PIK3C3/PIK3CA/PIKFYVE/PRKCB
hsa05214Glioma 3.95×10−2 AKT2/AKT3/CAMK4/PIK3CA/PRKCB/SOS2
hsa04728Dopaminergic synapse 4.67×10−2 AKT2/AKT3/CACNA1A/CACNA1C/CLOCK/GNAQ/PRKACB/PRKCB

[i] APC, adenomatous polyposis coli protein.

Table III.

Significant GO terms enriched for differentially expressed circRNAs.

Table III.

Significant GO terms enriched for differentially expressed circRNAs.

GO termIDDescriptionAdjusted P-valueGene ID
BPGO:0022604Regulation of cell morphogenesis 4.70×10−5 ARHGAP44/ARHGEF7/BRWD1/BRWD3/CAPRIN1/CORO1C/CUX1/DLG1/DOCK1/FMNL2/HECW1/HECW2/MKLN1/MYH10/NTRK2/PAK1/PAK3/PARP6/PHIP/PSEN1/PTK2/RHOBTB3/RIMS1/RIMS2/ROBO1/ROBO2/SYT1/TIAM1/TNIK/ZMYM4
BPGO:0031346Positive regulation of cell projection organization 4.70×10−5 APC/ARHGEF7/ATP8A2/AUTS2/CAPRIN1/CBFA2T2/CCDC88A/CORO1C/CPEB3/CUX1/HTT/KDM1A/LRRC7/MAGI2/NTRK2/PAK1/PAK3/PARP6/PSEN1/RIMS1/RIMS2/ROBO1/ROBO2/SYT1/TENM3/TIAM1
BPGO:0042391Regulation of membrane potential 1.09×10−4 AKAP6/AKT2/ANK2/ANK3/APP/CACNA1A/CACNA1C/CACNA1E/DGKI/DLG1/FGF14/GABRB2/GABRG3/GCLM/GNAQ/KCNH1/KCNK2/KCNMA1/NALCN/NDUFS1/NTRK2/PSEN1/RIMS1/RIMS2/RYR2/SLC8A1/SLMAP
BPGO:0010975Regulation of neuron projection development 2.93×10−4 ARHGAP44/ATP8A2/CAMSAP2/CAPRIN1/CBFA2T2/CCDC88A/CPEB3/CUX1/FBXO7/HECW1/HECW2/KDM1A/LRRC7/MAGI2/NTRK2/PAK1/PAK3/PARP6/PSEN1/PTK2/RIMS1/RIMS2/ROBO1/ROBO2/SYT1/TENM3/TIAM1/TNIK
BPGO:0034765Regulation of ion transmembrane transport BP 8.62×10−4 AKAP6/AKT2/ANK2/ANK3/APP/ATG5/CACNA1A/CACNA1C/CACNA1E/CRBN/DLG1/FGF14/HECW1/HECW2/HTT/KCNH1/KCNMA1/KLHL24/NALCN/PKD2/PSEN1/RYR2/SLC8A1/SLMAP/STK39/UBQLN1
BPGO:0021953Central nervous system neuron differentiation 9.64×10−4 ADARB1/AGTPBP1/DCLK1/DCLK2/GIGYF2/HERC1/LRP6/NFIB/NTRK2/PSEN1/PTK2/ROBO1/ROBO2/SATB2/SLC4A10
BPGO:0045666Positive regulation of neuron differentiation 1.17×10−4 ATP8A2/CAPRIN1/CBFA2T2/CPEB3/CUX1/KDM1A/LRRC7/MAGI2/NTRK2/PAK1/PAK3/PARP6/PSEN1/RIMS1/RIMS2/ROBO1/ROBO2/SH3GL3/SYT1/TCF4/TENM3/TIAM1
BPGO:0021954Central nervous system neuron development 4.84×10−3 ADARB1/DCLK1/DCLK2/NFIB/NTRK2/PTK2/ROBO1/ROBO2/SLC4A10
BPGO:0050769Positive regulation of neurogenesis 4.84×10−3 APP/ATP8A2/CAPRIN1/CBFA2T2/CPEB3/CUX1/KDM1A/LRRC7/MAGI2/MAN2A1/NTRK2/PAK1/PAK3/PARP6/PSEN1/RIMS1/RIMS2/ROBO1/ROBO2/SH3GL3/SYT1/TCF4/TENM3/TIAM1
BPGO:0097479Synaptic vesicle localization 5.75×10−3 AP3B2/CADPS2/DGKI/ERC2/MAGI2/PCLO/PRKCB/PSEN1/RIMS1/RIMS2/STXBP5L/SYN3/SYT1
CCGO:0097060Synaptic membrane 5.96×10−7 ANK2/ANK3/ANKS1B/ARHGAP32/ATP2B1/CACNA1C/CADPS2/CNKSR2/CPEB3/DENND1A/DGKI/DLG1/DLG2/ERC1/ERC2/GABRB2/GABRG3/KCNH1/KCNMA1/LRRC7/PICALM/PSD3/PSEN1/RIMS1/RIMS2/SLC8A1/STRN/SYNE1/SYT1/TIAM1
CCGO:0099572Postsynaptic specialization 5.96×10−7 ANKS1B/ARHGAP32/ARHGAP44/CACNA1C/CNKSR2/CPEB3/DCLK1/DGKI/DLG1/DLG2/GABRB2/KCNH1/LRRC7/MAGI2/MIB1/NTRK2/PAK3/PCLO/PSD3/SEPT11/SH3GL3/STRN/SYN3/TANC2/TIAM1/TNIK
CCGO:0098984Neuron to neuron synapse 5.96×10−7 ANKS1B/ARHGAP32/ARHGAP44/CACNA1C/CNKSR2/CPEB3/DCLK1/DGKI/DLG1/DLG2/KCNH1/LRRC7/MAGI2/MIB1/NTRK2/PAK3/PCLO/PSD3/SEPT11/SH3GL3/STRN/SYN3/SYT1/TANC2/TIAM1/TNIK
CCGO:0014069Postsynaptic density 1.84×10−6 ANKS1B/ARHGAP32/ARHGAP44/CACNA1C/CNKSR2/CPEB3/DCLK1/DGKI/DLG1/DLG2/KCNH1/LRRC7/MAGI2/MIB1/NTRK2/PAK3/PCLO/PSD3/SH3GL3/STRN/SYN3/TANC2/TIAM1/TNIK
CCGO:0098793Presynapse 5.86×10−5 APP/ARHGAP44/ATP2B1/CADPS2/DENND1A/DGKI/ERC1/ERC2/HTT/ICA1/KCNH1/KCNK2/MCTP1/NTRK2/PCLO/PICALM/PRKCB/PSEN1/RIMS1/RIMS2/RPH3A/SH3GL3/STX12/SYN3/SYT1/TNIK/WDR7
CCGO:0048786Presynaptic active zone 3.58×10−4 APP/ARHGAP44/ATP2B1/DGKI/ERC1/ERC2/PCLO/RIMS1/RIMS2
CCGO:0098978Glutamatergic synapse 2.05×10−3 ARHGAP44/ATP2B1/CADPS2/CAMK4/CNKSR2/DGKB/DGKI/DLG1/DLG2/ERC2/MYH10/PAK3/SEPT11/SH3GL3/SYN3/SYT1/TANC2/TIAM1/TNIK
CCGO:0005938Cell cortex 2.05×10−3 AKT2/ARHGAP32/ARHGEF7/ERC2/EXOC6B/FMN2/MKLN1/MYH10/PCLO/PKD2/PSEN1/PTK2/RHOBTB3/RIC8B/RIMS1/RIMS2/SEPT11
CCGO:0043025Neuronal cell body 2.28×10−3 AMFR/ARHGEF7/ATP2B1/CACNA1A/CACNA1C/CNKSR2/DENND1A/DGKI/GIGYF2/KCNH1/KCNK2/KLHL24/LMTK2/LRP6/MYH10/PDE1C/PICALM/PSEN1/SLC1A4/SLC4A10/STRN/TIAM1/TMEM50A
CCGO:0005911Cell-cell junction 8.24×10−3 AKAP6/ANK2/ANK3/APC/APP/ASH1L/DLG1/KLHL24/LRRC7/MAGI2/NFASC/PAK1/PIKFYVE/PKD2/SDCCAG8/SLC8A1/SPECC1L/STRN/TIAM1/TJP1
MFGO:0017137Rab GTPase binding 4.74×10−4 ACAP2/CLEC16A/DENND1A/DENND1B/DENND5B/ERC1/GAPVD1/PICALM/RHOBTB3/RIMS1/RIMS2/RPH3A/STXBP5L/TBC1D12/TBC1D1/TBCK
MFGO:0044325Ion channel binding 7.48×10−3 AKAP6/ANK2/ANK3/DLG1/HTT/KCNH1/PKD2/RIMS1/RIMS2/RYR2/SLC8A1
MFGO:0004143Diacylglycerol kinase activity 4.94×10−2DGKB/DGKH/DGKI

[i] Only terms enriched by significantly differentially expressed circRNAs that may act as a sponge of miR-142-3p/miR-590-5p are listed. The genes enriched in each term were corresponding to differentially expressed circRNAs. GO, Gene Ontology; BP, biological process; CC, cellular component; MF, molecular function; circ, circular; APC, adenomatous polyposis coli protein.

Construction of the ceRNA network and functional enrichment

After uploading the circID (197 common in the two datasets) to the CircInteractome database, 313 miRNAs were predicted to interact with DECs. Using Gene_symbol, 276 miRNAs were predicted to interact with DECs using Starbase. Among them, 18 miRNAs were shared, including hsa-miR-139-5p, −142-3p, −184, −217, −324-5p, −326, −330-5p, −338-3p, −346, −375, −384, −421, −496, −543, −590-5p, −599, −615-3p and −875-5p). Subsequently, the target genes were predicted for these 18 miRNAs using the miRwalk 2.0 database, which were then compared with 65 pre-established glioma-related genes collected from the CTD database. In total, 22 genes were shared, including BRD4, SUZ12, VEGFA, NOTCH3, NOTCH2, RUNX3, CTNND2, EGFR, CDK6, PML, BHLHE40, EGF, NOTCH1, TNFSF10, LZTR1, RUNX1, MDM4, MET, IL1B, JAG1, APC and RECK. Then, the ceRNA network was established based on 115 DECs, 15 miRNAs and the aforementioned 22 mRNAs (Fig. 3).

Function analysis for the target genes of miRNAs in this ceRNA network showed that 44 KEGG pathways (Table IV and Fig. 4A) and 736 GO terms (including 713 BPs, three CCs and 20 MFs) (Table V and Fig. 4B) were enriched, such as ‘regulation of actin cytoskeleton’ (Table IV), various cancer pathways (including ‘miRNAs in cancer’, ‘breast cancer’, ‘endometrial cancer’, ‘gastric cancer’, ‘hepatocellular carcinoma’ and ‘colorectal cancer’; Fig. 4A), ‘Wnt signaling pathway’, ‘cell cycle arrest’, ‘cell fate commitment’ (Fig. 4B) and cell junction assembly (Table V), in all of which the APC gene was included. These findings suggested that the circRNAs-miRNAs that regulated APC may be crucial for the development of glioma.

Table IV.

Kyoto Encyclopedia of Genes and Genomes pathways enriched for genes in the competing endogenous RNA network.

Table IV.

Kyoto Encyclopedia of Genes and Genomes pathways enriched for genes in the competing endogenous RNA network.

IDDescriptionAdjusted P-valueGene ID
hsa05206microRNAs in cancer 3.02×10−8 APC/CDK6/EGFR/MDM4/MET/NOTCH1/NOTCH2/NOTCH3/RECK/VEGFA
hsa05224Breast cancer 3.50×10−8 APC/CDK6/EGF/EGFR/JAG1/NOTCH1/NOTCH2/NOTCH3
hsa05165Human papillomavirus infection 6.64×10−7 APC/CDK6/EGF/EGFR/JAG1/NOTCH1/NOTCH2/NOTCH3/VEGFA
hsa04658Th1 and Th2 cell differentiation 4.91×10−5 JAG1/NOTCH1/NOTCH2/NOTCH3/RUNX3
hsa01522Endocrine resistance 5.38×10−5 EGFR/JAG1/NOTCH1/NOTCH2/NOTCH3
hsa04330Notch signaling pathway 7.36×10−5 JAG1/NOTCH1/NOTCH2/NOTCH3
hsa05218Melanoma 3.22×10−4 CDK6/EGF/EGFR/MET
hsa05212Pancreatic cancer 3.31×10−4 CDK6/EGF/EGFR/VEGFA
hsa01521EGFR tyrosine kinase inhibitor resistance 3.62×10−4 EGF/EGFR/MET/VEGFA
hsa05219Bladder cancer 1.22×10−3EGF/EGFR/VEGFA
hsa05213Endometrial cancer 3.13×10−3APC/EGF/EGFR
hsa05226Gastric cancer 3.21×10−3 APC/EGF/EGFR/MET
hsa05223Non-small cell lung cancer 3.81×10−3CDK6/EGF/EGFR
hsa04010MAPK signaling pathway 3.81×10−3 EGF/EGFR/RUNX3/MET/VEGFA
hsa05225Hepatocellular carcinoma 4.05×10−3 APC/CDK6/EGFR/MET
hsa05214Glioma 4.59×10−3CDK6/EGF/EGFR
hsa05210Colorectal cancer 6.37×10−3APC/EGF/EGFR
hsa04510Focal adhesion 6.37×10−3 EGF/EGFR/MET/VEGFA
hsa04151PI3K-Akt signaling pathway 6.44×10−3 CDK6/EGF/EGFR/MET/VEGFA
hsa04015Rap1 signaling pathway 6.52×10−3 EGF/EGFR/MET/VEGFA
hsa04066HIF-1 signaling pathway 8.06×10−3EGF/EGFR/VEGFA
hsa05163Human cytomegalovirus infection 8.22×10−3 CDK6/EGFR/RUNX3/VEGFA
hsa04014Ras signaling pathway 8.80×10−3 EGF/EGFR/MET/VEGFA
hsa04919Thyroid hormone signaling pathway 1.08×10−2 NOTCH1/NOTCH2/NOTCH3
hsa05020Prion diseases 1.29×10−2RUNX3/NOTCH1
hsa04068FoxO signaling pathway 1.38×10−2 EGF/EGFR/TNFSF10
hsa05162Measles 1.38×10−2 CDK6/RUNX3/TNFSF10
hsa04934Cushing syndrome 2.02×10−2APC/CDK6/EGFR
hsa05160Hepatitis C 2.02×10−2CDK6/EGF/EGFR
hsa05144Malaria2.08E-02RUNX3/MET
hsa05164Influenza A 2.48×10−2 RUNX3/PML/TNFSF10
hsa05202Transcriptional misregulation in cancer 3.00×10−2MET/PML/RUNX1
hsa05230Central carbon metabolism in cancer 3.27×10−2EGFR/MET
hsa05221Acute myeloid leukemia 3.27×10−2PML/RUNX1
hsa05120Epithelial cell signaling in Helicobacter pylori infection 3.27×10−2EGFR/MET
hsa05205Proteoglycans in cancer 3.27×10−2EGFR/MET/VEGFA
hsa05211Renal cell carcinoma 3.27×10−2MET/VEGFA
hsa04115p53 signaling pathway 3.36×10−2CDK6/MDM4
hsa04520Adherens junction 3.36×10−2EGFR/MET
hsa04810Regulation of actin cytoskeleton 3.51×10−2APC/EGF/EGFR
hsa05220Chronic myeloid leukemia 3.55×10−2CDK6/RUNX1
hsa04012ErbB signaling pathway 4.28×10−2EGF/EGFR
hsa04540Gap junction 4.47×10−2EGF/EGFR
hsa05323Rheumatoid arthritis 4.55×10−2RUNX3/VEGFA

[i] APC, adenomatous polyposis coli protein.

Table V.

Significant GO terms enriched for genes in the competing endogenous RNA network.

Table V.

Significant GO terms enriched for genes in the competing endogenous RNA network.

GO termIDDescriptionAdjusted P-valueGene ID
BPGO:0016055Wnt signaling pathway 2.09×10−5 APC/CTNND2/EGF/EGFR/MET/NOTCH1/RECK/RUNX1
BPGO:0198738Cell-cell signaling by wnt 2.09×10−5 APC/CTNND2/EGF/EGFR/MET/NOTCH1/RECK/RUNX1
BPGO:0030111Regulation of Wnt signaling pathway 2.17×10−5 APC/CTNND2/EGF/EGFR/NOTCH1/RECK/RUNX1
BPGO:0060828Regulation of canonical Wnt signaling pathway 7.32×10−5 APC/CTNND2/EGF/EGFR/NOTCH1/RECK
BPGO:0007050Cell cycle arrest 7.32×10−5 APC/CDK6/MDM4/NOTCH1/NOTCH2/PML
BPGO:0045165Cell fate commitment 9.34×10−5 APC/JAG1/NOTCH1/NOTCH2/NOTCH3/PML
BPGO:0060070Canonical Wnt signaling pathway 1.31×10−4 APC/CTNND2/EGF/EGFR/NOTCH1/RECK
BPGO:0042176Regulation of protein catabolic process 3.36×10−4 APC/EGF/EGFR/IL1B/MDM4/PML
BPGO:0048871Multicellular organismal homeostasis 7.99×10−4 APC/EGFR/IL1B/MET/NOTCH1/VEGFA
BPGO:0033044Regulation of chromosome organization 1.45×10−3 APC/BRD4/IL1B/PML/VEGFA
BPGO:0034329Cell junction assembly 3.62×10−3 APC/CTNND2/RUNX1/VEGFA
BPGO:1901652Response to peptide 3.98×10−3 APC/IL1B/JAG1/NOTCH1/TNFSF10
BPGO:0071900Regulation of protein serine/threonine kinase activity 4.29×10−3 APC/EGF/EGFR/IL1B/VEGFA
BPGO:0045930Negative regulation of mitotic cell cycle 1.45×10−3 APC/EGFR/MDM4/PML
BPGO:0034330Cell junction organization 4.78×10−3 APC/CTNND2/RUNX1/VEGFA
BPGO:0007043Cell-cell junction assembly 5.19×10−3 APC/CTNND2/RUNX1
BPGO:0010948Negative regulation of cell cycle process 5.30×10−3 APC/MDM4/PML/SUZ12
BPGO:0106106Cold-induced thermogenesis 6.58×10−3 APC/NOTCH1/VEGFA
BPGO:0045216Cell-cell junction organization 7.56×10−3 APC/CTNND2/RUNX1
MFGO:0008013Beta-catenin binding 3.47×10−2APC/CTNND2
MFGO:0031625Ubiquitin protein ligase binding 3.47×10−2APC/EGFR/PML
MFGO:0044389Ubiquitin-like protein ligase binding 3.78×10−2APC/EGFR/PML

[i] Only terms enriched by adenomatous polyposis coli protein are listed. GO, Gene Ontology; BP, biological process; MF, molecular function; APC, adenomatous polyposis coli protein.

Prognosis and expression analysis for miRNAs and target genes

LinkedOmics online analysis using TCGA data showed that 19 mRNAs and nine miRNAs were significantly associated with OS (Table VI and Fig. 5). These prognosis-related miRNAs and mRNAs constituted 20 interaction relationships in the ceRNA network. However, the prognosis trend between hsa-miR-217/hsa-miR-324-5p/hsa-miR-590-5p/hsa-miR-599/hsa-miR-875-5p and their target genes were consistent, with high expression levels of both miRNAs and their target genes indicating a poor prognosis, which is not in accordance with the negative regulatory mechanisms of miRNAs on mRNAs. Thus, only hsa-miR-139-5p-NOTCH1/RUNX1/CDK6/TNFSF10/VEGFA and hsa-miR-142-3p-APC-related ceRNA axes may be important. Furthermore, as all the circRNAs in the ceRNA network were downregulated, its interactive miRNAs should be upregulated according to the ceRNA theory (68); however, similarly to circRNAs, high expression levels of hsa-miR-139-5p were also associated with an excellent overall survival rate, indicating that hsa-miR-139-5p was also downregulated in glioma. Thus, hsa-miR-139-5p-related interaction relationships were excluded from further analyses and only hsa-miR-142-3p-APC-related ceRNA axes (in which high hsa-miR-142-3p expression was associated with poor OS, while high APC expression was associated with a good prognosis, as shown in Fig. 5) were crucial.

Table VI.

Overall survival-related mRNAs and miRNAs.

Table VI.

Overall survival-related mRNAs and miRNAs.

A, mRNA

RNACox regression testP-valueFDR (BH)
SUZ12 7.823×10−1 4.984×10−05 2.991×10−4
VEGFA 4.428×10−1 1.000×10−50 1.000×10−49
NOTCH3 5.14×10−1 2.21×10−11 6.64×10−11
RUNX3 5.539×10−1 1.000×10−38 1.000×10−37
CTNND2 −6.151×10−1 1.000×10−26 1.000×10−25
EGFR 1.815×10−1 2.405×10−6 1.443×10−5
CDK6 4.738×10−1 1.000×10−17 1.000×10−16
PML 8.536×10−1 3.663×10−12 2.198×10−11
BHLHE40 4.879×10−1 3.331×10−15 1.998×10−14
EGF 3.093×10−1 2.355×10−9 7.065×10−9
NOTCH1 5.332×10−1 1.000×10−52 1.000×10−52
TNFSF10 3.983×10−1 2.207×10−13 6.621×10−13
RUNX1 5.332×10−1 1.000×10−52 1.000×10−52
MDM4 3.306×10−1 1.131×10−7 6.784×10−7
MET 2.378×10−1 1.000×10−16 1.000×10−16
IL1B 1.159×10−1 1.164×10−4 2.329×10−4
JAG1 6.596×10−1 1.000×10−33 1.000×10−32
APC −7.618×10−1 1.000×10−36 1.000×10−35
RECK 4.158×10−1 6.612×10−3 1.984×10−2

B, miRNA
RNACox regression testP-valueFDR (BH)

hsa-miR-139-5p −1.909×10−1 3.418×10−3 1.025×10−2
hsa-miR-142-3p 4.080×10−1 6.370×10−9 3.822×10−8
hsa-miR-217 2.006×10−1 2.422×10−6 1.453×10−5
hsa-miR-324-5p 2.797×10−1 6.083×10−3 3.650×10−2
hsa-miR-346 −6.408×10−1 2.317×10−9 1.390×10−8
hsa-miR-590-5p 5.611×10−1 2.461×10−7 7.384×10−7
hsa-miR-599 4.453×10−1 9.471×10−7 2.841×10−6
hsa-miR-615-3p 9.771×10−1 1.000×10−8 1.000×10−7
hsa-miR-875-5p 1.369×10−1 5.275×10−4 1.361×10−3

[i] Only mRNAs and miRNAs significantly associated with overall survival P≤0.05 are shown. FDR, False Discovery Rate; miRNA, micro RNA; APC, adenomatous polyposis coli protein; hsa, Homo sapiens; BH, Benjamini and Hochberg.

Accordingly, hsa_circ_0000120/ 0001367/ 0001368/ 0001369/ 0001936/ 0004516/ 0005114/ 0060425/0064615/0130887-hsa-miR-142-3p-APC ceRNA axes may be potential targets for the development of glioma. Additionally, UALCAN analysis using TCGA data of glioblastoma multiforme demonstrated that APC was significantly downregulated in glioma compared with in normal tissues (P=0.0000822); while GSE25632 microarray analysis showed the expression of hsa-miR-142-3p was significantly higher in glioma compared with that in normal tissues (log2FC=1.21, P=0.00284), further confirming the creditability of these crucial ceRNA axes for glioma (data not shown).

In addition, both of GSE25632 (log2FC=5.75, P=0.0226) and GSE103229 (log2FC=1.65, P=0.0208) microarray analyses revealed that hsa-miR-590-5p was upregulated in glioma compared with that in normal tissues (data not shown). In fact, the interaction between hsa-miR-590-5p and APC was also predicted by RNA22 and RNAhybrid using miRwalk2.0 analysis (data not shown). Therefore, circRNAs interacting with hsa-miR-590-5p were also important, including hsa_circ_0005114, 0069718, 0098551, 0100496, 0104726, 0104727 and 0117841 (Fig. 3), among which hsa_circ_0005114 interacted with hsa-miR-142-3p. Therefore, the hsa_circ_0005114-miR-142-3p/miR-590-5p-APC ceRNA axis may represent a potential mechanism for the development of glioma.

Discussion

In the present study, 16 crucial downregulated circRNAs were identified that may sponge miR-142-3p/miR-590-5p to suppress glioma progression via regulating the APC gene. Most of them, to the best of our knowledge, were newly reported circRNAs associated with cancer, except for hsa_circ_0001368 that has a tumor-suppression role in gastric cancer by regulating the miR-6506-5p/forkhead box (FOX) protein O3 axis (31). However, hsa_circ_0005114 may be particularly important as it was a circRNA interacting with both miR-142-3p and miR-590-5p.

The present analysis did not identify the roles of hsa_circ_0005114 in cancer; however, hsa_circ_0005114 is derived from the regulating synaptic membrane exocytosis (RIMS)2 gene. Thus, the function of hsa_circ_0005114 may be similar to that of RIMS2. Notably, Mukasa et al (32) observed that RIMS2 expression is significantly higher in normal brain tissues compared with glioblastoma. In addition, it was reported that RIMS2 mediates the cAMP-guanidine nucleotide exchange factor II pathway to promote incretin-potentiated insulin secretion (33,34). The downregulation of RIMS2 may lead to the lower levels of insulin and the development of diabetes and obesity, as while 11.1% of patients with glioma (35) and 30% of those with brain tumor are diagnosed with obesity and insulin resistance/impaired glucose tolerance (36). Therefore, hsa_circ_0005114 may be involved in glioma by influencing the insulin secretion pathway, which was also predicted in the present function enrichment analysis. However, how hsa_circ_0005114 influences insulin secretion needs further investigation. A recent study proposed that high insulin index is positively associated with a high risk of glioma development (37). However, another study reported that decreased insulin receptor expression impairs cellular functions and represses orthotopic glioblastoma (38). These conflicting reports may be attributed to the dual roles of insulin (39).

In addition to insulin secretion mechanisms, the present study predicted that hsa_circ_0005114 may exert tumor suppressor functions by upregulating miR-142-3p and miR-590-5p, which subsequently inhibited the expression of APC. Studies have demonstrated that miR-590-5p is upregulated in liver (40), colorectal (41), gastric (42) and cervical cancer (43). miR-590-5p promotes cancer cell proliferation, invasion and therapy resistance by targeting FOXO1 (34), TGF-β R2 (44), matrix metalloproteinases (41), reversion-inducing cysteine-rich protein with Kazal motifs (36) and cell adhesion molecule L1-like (43). Furthermore, a previous study also showed that miR-590-3p is upregulated in human glioma tissues (especially high grade) and radioresistant human glioblastoma cells (45). The use of anti-miR-590-3p suppresses cell viability, decreases colony formation capacity, increases the apoptotic rate and enhances the radiosensitivity. A luciferase reporter assay demonstrated that leucine-rich repeats and immunoglobulin-like domains protein 1 is the direct target of miR-590-3p. Similarly, other studies using quantitative PCR also detected that miR-142-3p expression is significantly upregulated in renal cell carcinoma (46) and nasopharyngeal carcinoma (47) compared with adjacent tissues. Downregulation of miR-142-3p significantly suppresses cell proliferation, migration and cell cycle progression, promotes apoptosis in vitro (46) and blocks tumor growth in a mouse model via upregulating its targeted genes, including suppressor of cytokine signaling 6 (47) and Ras-related C3 botulinum toxin substrate 1 in colorectal cancer (48). High expression of miR-142-3p is correlated with histological differentiation and a poor prognosis for patients with esophageal squamous cell carcinoma (49). In line with the aforementioned findings, the present study reported that miR-590-5p and miR-142-3p were upregulated in glioblastoma tissues compared with normal controls and were associated with a worse prognosis. However, the targeted genes of these two miRNAs were not completely validated in cancer. It was predicted that APC, a regulator of the WNT signaling pathway (50), may be a direct target for both miR-590-5p and miR-142-3p. Their negative regulatory relationship was indirectly investigated by Naseri et al (51) who showed that delivery of anti-miR-142-3p by exosomes to breast cancer cells leads to a significant increase in APC mRNA levels. In addition, Wu et al (52) used dual-luciferase and western blot analysis in mesenchymal stem cells to demonstrate that miR-590-3p binds to the 3′UTR of APC mRNA. Notably, APC encodes a protein that acts as an antagonist of the Wnt signaling pathway (53), which is often activated in cancer (54). Thus, APC may be downregulated during carcinogenesis, which has been confirmed in various cancer types, including glioma (5557). For example, Cole et al (55) reported that APC expression is decreased in pancreatic ductal adenocarcinoma tissues, and that APC siRNA treatment promotes cell proliferation and migration. Wang et al (56) reported that the expression of APC mRNA is significantly decreased in ovarian tumor cells and tissues compared with in normal ovarian cells and tissues. In addition, overexpression of APC induces increased apoptosis of ovarian tumor cells by decreasing the ATP binding cassette subfamily B member 1 (also known as multidrug resistance gene 1)/chemokine (C-X-C motif) ligand 1 signaling pathway. Zhang et al (57) demonstrated that APC expression is downregulated in colorectal cancer tissues, which is associated with the expression of miR-494. Overexpression of miR-494 promotes colorectal cancer cell proliferation by inhibiting APC and consequently inducing Wnt/β-catenin signaling. Western blot assays of Li et al (58) also suggested that miR-106a-5p reduces APC protein levels and upregulates target proteins of the Wnt/β-catenin pathway, resulting in the invasion of glioblastoma cells (58). In accordance with these studies, the present study reported that APC expression was decreased in glioblastoma tissues compared with normal controls and high expression of APC was associated with a good prognosis. Functional enrichment analysis showed APC was involved in Wnt signaling pathway, cell fate commitment, and cell junction.

Overall, the present study suggested that hsa_circ_0005114-miR-142-3p/miR-590-5p-APC ceRNA axes may be mechanisms for the development and progression of glioma. These axes may also have potential as novel targets for the treatment of glioma. However, additional in vitro and in vivo experiments, such as gene interaction experiments involving knockout/overexpression of hsa_circ_0005114-miR-142-3p/miR-590-5p-APC and their influence on tumor cells (using cell proliferation, apoptosis, invasion and migration assays) and tumor growth, are required to validate these conclusions in the future, and the lack of these experiments is a limitation of the present study.

Acknowledgements

Not applicable.

Funding

The present study was funded by grants from The Natural Science Foundation of China (grant no. 81400404) and The Science and Technology of Jilin Province (grant no. 20200201470JC).

Availability of data and materials

The GSE86202, GSE92322, GSE103229 and GSE25632 datasets were downloaded from the Gene Expression Omnibus repository (http://www.ncbi.nlm.nih.gov/geo/). LinkedOmics and UALCAN database used the miRNA and mRNA sequencing data of The Cancer Genome Atlas (https://gdc-portal.nci.nih.gov/) for analysis.

Authors' contributions

BW, LW and JWZ conceived and designed the study. BW collected and analyzed the data. LW and JWZ contributed to the interpretation of data. BW and LW drafted the manuscript. JWZ revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ryan CS, Juhn YJ, Kaur H, Wi CI, Ryu E, King KS and Lachance DH: Long-term incidence of glioma in Olmsted County, Minnesota, and disparities in postglioma survival rate: A population-based study. Neurooncol Pract. 7:288–298. 2020.PubMed/NCBI

2 

Sehmer EA, Hall GJ, Greenberg DC, O'Hara C, Wallingford SC, Wright KA and Green AC: Incidence of glioma in a northwestern region of England, 2006–2010. Neuro Oncol. 16:971–974. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Larjavaara S, Mäntylä R, Salminen T, Haapasalo H, Raitanen J, Jääskeläinen J and Auvinen A: Incidence of gliomas by anatomic location. Neuro Oncol. 9:319–325. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Rasmussen BK, Hansen S, Laursen RJ, Kosteljanetz M, Schultz H, Nørgård BM, Guldberg R and Gradel KO: Epidemiology of glioma: Clinical characteristics, symptoms, and predictors of glioma patients grade I–IV in the the Danish Neuro-Oncology Registry. J Neurooncol. 135:571–579. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Piñeros M, Sierra MS, Izarzugaza MI and Forman D: Descriptive epidemiology of brain and central nervous system cancers in Central and South America. Cancer Epidemiol. 44 (Suppl 1):S141–S149. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Ebbesen KK, Kjems J and Hansen TB: Circular RNAs: Identification, biogenesis and function. Biochim Biophys Acta. 1859:163–168. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Yu CY and Kuo HC: The emerging roles and functions of circular RNAs and their generation. J Biomed Sci. 26:292019. View Article : Google Scholar : PubMed/NCBI

8 

Belousova EA, Filipenko ML and Kushlinskii NE: Circular RNA: New regulatory molecules. Bull Exp Biol Med. 164:803–815. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Zhou J, Wang H, Chu J, Huang Q, Li G, Yan Y, Xu T, Chen J and Wang Y: Circular RNA hsa_circ_0008344 regulates glioblastoma cell proliferation, migration, invasion, and apoptosis. J Clin Lab Anal. 32:e224542018. View Article : Google Scholar : PubMed/NCBI

10 

Wang Y, Sui X, Zhao H, Cong L, Li Y, Xin T, Guo M and Hao W: Decreased circular RNA hsa_circ_0001649 predicts unfavorable prognosis in glioma and exerts oncogenic properties in vitro and in vivo. Gene. 676:117–122. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Zhan L, Mu Z, Yang M, Zhang T and Qian L: Elevation of circ-ITX1 upregulates interleukin 17 receptor D expression via sponging miR8a and facilitates cell progression in glioma. J Cell Biochem. 120:16495–16502. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Yi C, Li H, Li D, Qin X, Wang J, Liu Y, Liu Z and Zhang J: Upregulation of circular RNA circ_0034642 indicates unfavorable prognosis in glioma and facilitates cell proliferation and invasion via the miR-1205/BATF3 axis. J Cell Biochem. 120:13737–13744. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Xiong J, Wang T, Tang H, Lv Z and Liang P: Circular RNA circMAN2B2 facilitates glioma progression by regulating the miR-1205/S100A8 axis. J Cell Physiol. 234:22996–23004. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Duan X, Liu D, Wang Y and Chen Z: Circular RNA hsa_circ_0074362 promotes glioma cell proliferation, migration, and invasion by attenuating the inhibition of miR-1236-3p on HOXB7 expression. DNA Cell Biol. 37:917–924. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Li X and Diao H: Circular RNA circ_0001946 acts as a competing endogenous RNA to inhibit glioblastoma progression by modulating miR-671-5p and CDR1. J Cell Physiol. 234:13807–13819. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Yuan Y, Li J, Xiang W, Liu Y and Mao Q: Analyzing the interactions of mRNAs, miRNAs, lncRNAs and circRNAs to predict competing endogenous RNA networks in glioblastoma. J Neurooncol. 137:493–502. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Zhu J, Ye J, Zhang L, Xia L, Hu H, Jiang H, Wan Z, Sheng F, Ma Y, Li W, et al: Differential expression of circular RNAs in glioblastoma multiforme and its correlation with prognosis. Transl Oncol. 10:271–279. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Glažar P, Papavasileiou P and Rajewsky N: circBase: A database for circular RNAs. RNA. 20:1666–1670. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Nikolayeva O and Robinson MD: edgeR for Differential RNA-seq and ChIP-seq analysis: An application to stem cell biology. Methods Mol Biol. 1150:45–79. 2014. View Article : Google Scholar : PubMed/NCBI

20 

R Development Core Team, . R: A language and environment for statistical computing. 2015, simplehttp://www.r-project.org/February 10–2015

21 

Dudekula DB, Panda AC, Grammatikakis I, De S, Abdelmohsen K and Gorospe M: CircInteractome: A web tool for exploring circular RNAs and their interacting proteins and microRNAs. RNA Biol. 13:34–42. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Li JH, Liu S, Zhou H, Qu LH and Yang JH: Starbase v2.0: Decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale clip-seq data. Nucleic Acids Res. 42((Database issue)): D92–D97. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Dweep H and Gretz N: miRWalk2.0: A comprehensive atlas of microRNA-target interactions. Nat Methods. 12:6972015. View Article : Google Scholar : PubMed/NCBI

24 

Davis AP, Grondin CJ, Johnson RJ, Sciaky D, McMorran R, Wiegers J, Wiegers TC and Mattingly CJ: The Comparative Toxicogenomics Database: Update 2019. Nucleic Acids Res. 47:D948–D954. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networks. Methods Mol Biol. 696:291–303. 2011. View Article : Google Scholar : PubMed/NCBI

26 

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

27 

Ferreira JA: The Benjamini-Hochberg method in the case of discrete test statistics. Int J Biostat. 3:112007. View Article : Google Scholar

28 

Vasaikar SV, Straub P, Wang J and Zhang B: LinkedOmics: Analyzing multi-omics data within and across 32 cancer types. Nucleic Acids Res. 46((Database issue)): D956–D963. 2017.

29 

Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi BVSK and Varambally S: UALCAN: A portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 19:649–658. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Zhang W, Zhang J, Hoadley K, Kushwaha D, Ramakrishnan V, Li S, Kang C, You Y, Jiang C, Song SW, et al: miR-181d: A predictive glioblastoma biomarker that downregulates MGMT expression. Neuro Oncol. 14:712–719. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Lu J, Zhang PY, Li P, Xie JW, Wang JB, Lin JX, Chen QY, Cao LL, Huang CM and Zheng CH: Circular RNA hsa_circ_0001368 suppresses the progression of gastric cancer by regulating miR-6506-5p/FOXO3 axis. Biochem Biophys Res Commun. 512:29–33. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Mukasa A, Ueki K, Ge X, Ishikawa S, Ide T, Fujimaki T, Nishikawa R, Asai A, Kirino T and Aburatani H: Selective expression of a subset of neuronal genes in oligodendroglioma with chromosome 1p loss. Brain Pathol. 14:34–42. 2010. View Article : Google Scholar

33 

Kashima Y, Miki T, Shibasaki T, Ozaki N, Miyazaki M, Yano H and Seino S: Critical role of cAMP-GEFII-Rim2 complex in incretin-potentiated insulin secretion. J Biol Chem. 276:46046–46053. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Fujimoto K, Shibasaki T, Yokoi N, Kashima Y, Matsumoto M, Sasaki T, Tajima N, Iwanaga T and Seino S: Piccolo, a Ca2+ sensor in pancreatic beta-cells. Involvement of cAMP-GEFII.Rim2. Piccolo complex in cAMP-dependent exocytosis. J Biol Chem. 277:50497–50502. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Sani I and Albanese A: Endocrine long-term follow-up of children with neurofibromatosis type 1 and optic pathway glioma. Horm Res Paediatr. 87:178–188. 2017. View Article : Google Scholar

36 

Masanori A, Takayoshi T, Koji M, Yumi A, Ken-Ichi S and Hironobu S: Prevalence of obesity, hyperlipemia and insulin resistance in children with suprasellar brain tumors. Clin Pediatr Endocrinol. 16:1–9. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Anjom-Shoae J, Shayanfar M, Mohammad-Shirazi M, Sadeghi O, Sharifi G, Siassi F and Esmaillzadeh A: Dietary insulin index and insulin load in relation to glioma: Findings from a case-control study. Nutr Neurosci. 29:1–9. 2019. View Article : Google Scholar

38 

Gong Y, Ma Y, Sinyuk M, Loganathan S, Thompson RC, Sarkaria JN, Chen W, Lathia JD, Mobley BC, Clark SW and Wang J: Insulin-mediated signaling promotes proliferation and survival of glioblastoma through Akt activation. Neuro Oncol. 18:48–57. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Yang BC, Wang YS, Wang CH, Lin HH, Tang MJ and Yang TL: Transient apoptosis elicited by insulin in serum-starved glioma cells involves Fas/Fas-L and Bcl-2. Cell Biol Int. 23:533–540. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Jia G, Tang Y, Deng G, Fang D, Xie J, Yan L and Chen Z: miR-590-5p promotes liver cancer growth and chemotherapy resistance through directly targeting FOXO1. Am J Transl Res. 11:2181–2193. 2019.PubMed/NCBI

41 

Kim CW, Oh ET, Kim JM, Park JS, Lee DH, Lee JS, Kim KK and Park HJ: Hypoxia-induced microRNA-590-5p promotes colorectal cancer progression by modulating matrix metalloproteinase activity. Cancer Lett. 416:31–41. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Shen B, Yu S, Zhang Y, Yuan Y, Li X, Zhong J and Feng J: miR-590-5p regulates gastric cancer cell growth and chemosensitivity through RECK and the AKT/ERK pathway. Onco Targets Ther. 9:6009–6019. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Chu Y, Ouyang Y, Wang F, Zheng A, Bai L, Han L, Chen Y and Wang H: MicroRNA-590 promotes cervical cancer cell growth and invasion by targeting CHL1. J Cell Biochem. 115:847–853. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Jiang X, Xiang G, Wang Y, Zhang L, Yang X, Cao L, Peng H, Xue P and Chen D: MicroRNA-590-5p regulates proliferation and invasion in human hepatocellular carcinoma cells by targeting TGF-β RII. Mol Cells. 33:545–551. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Chen L, Wang W, Zhu S, Jin X, Wang J, Zhu J and Zhou Y: MicroRNA-590-3p enhances the radioresistance in glioblastoma cells by targeting LRIG1. Exp Ther Med. 14:1818–1824. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Li Y, Chen D, Jin L, Liu J, Li Y, Su Z, Qi Z, Shi M, Jiang Z, Yang S, et al: Oncogenic microRNA-142-3p is associated with cellular migration, proliferation and apoptosis in renal cell carcinoma. Oncol Lett. 11:1235–12341. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Qi X, Li J, Zhou C, Lv C and Tian M: miR-142-3p suppresses SOCS6 expression and promotes cell proliferation in nasopharyngeal carcinoma. Cell Physiol Biochem. 36:1743–1752. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Gao X, Xu W, Lu T, Zhou J, Ge X and Hua D: MicroRNA-142-3p promotes cellular invasion of colorectal cancer cells by activation of RAC1. Technol Cancer Res Treat. 17:15330338187905082018. View Article : Google Scholar : PubMed/NCBI

49 

Lin RJ, Xiao DW, Liao LD, Chen T, Xie ZF, Huang WZ, Wang WS, Jiang TF, Wu BL, Li EM and Xu LY: miR-142-3p as a potential prognostic biomarker for esophageal squamous cell carcinoma. J Surg Oncol. 105:175–182. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Hankey W, Frankel WL and Groden J: Functions of the APC tumor suppressor protein dependent and independent of canonical WNT signaling: Implications for therapeutic targeting. Cancer Metastasis Rev. 37:159–172. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Naseri Z, Oskuee RK, Jaafari MR and Forouzandeh Moghadam M: Exosome-mediated delivery of functionally active miRNA-142-3p inhibitor reduces tumorigenicity of breast cancer in vitro and in vivo. Int J Nanomedicine. 13:7727–7747. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Wu S, Liu W and Zhou L: miR-590-3p regulates osteogenic differentiation of human mesenchymal stem cells by regulating APC gene. Biochem Biophys Res Commun. 478:1582–1587. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Saito-Diaz K, Benchabane H, Tiwari A, Tian A, Li B, Thompson JJ, Hyde AS, Sawyer LM, Jodoin JN, Santos E, et al: APC inhibits ligand-independent Wnt signaling by the clathrin endocytic pathway. Dev Cell. 44:566–581.e8. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Ghosh N, Hossain U, Mandal A and Sil PC: The Wnt signaling pathway: A potential therapeutic target against cancer. Ann N Y Acad Sci. 1443:54–74. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Cole JM, Simmons K and Prosperi JR: Effect of adenomatous polyposis coli loss on tumorigenic potential in pancreatic ductal adenocarcinoma. Cells. 8:10842019. View Article : Google Scholar

56 

Wang Y, Cao C, Fang D and Hu Y: Role of APC-mediated MDR-1/CLCX-1 signaling pathway in ovarian tumors. J Biol Regul Homeost Agents. 32:529–536. 2018.PubMed/NCBI

57 

Zhang Y, Guo L, Li Y, Feng GH, Teng F, Li W and Zhou Q: MicroRNA-494 promotes cancer progression and targets adenomatous polyposis coli in colorectal cancer. Mol Cancer. 17:12018. View Article : Google Scholar : PubMed/NCBI

58 

Li D, Wang Z, Chen Z, Lin L, Wang Y, Sailike D, Luo K, Du G, Xiang X and Jiafu GD: MicroRNA-106a-5p facilitates human glioblastoma cell proliferation and invasion by targeting adenomatosis polyposis coli protein. Biochem Biophys Res Commun. 481:245–250. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 21 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wei B, Wang L and Zhao J: Circular RNA hsa_circ_0005114‑miR‑142‑3p/miR‑590‑5p-adenomatous polyposis coli protein axis as a potential target for treatment of glioma. Oncol Lett 21: 58, 2021
APA
Wei, B., Wang, L., & Zhao, J. (2021). Circular RNA hsa_circ_0005114‑miR‑142‑3p/miR‑590‑5p-adenomatous polyposis coli protein axis as a potential target for treatment of glioma. Oncology Letters, 21, 58. https://doi.org/10.3892/ol.2020.12320
MLA
Wei, B., Wang, L., Zhao, J."Circular RNA hsa_circ_0005114‑miR‑142‑3p/miR‑590‑5p-adenomatous polyposis coli protein axis as a potential target for treatment of glioma". Oncology Letters 21.1 (2021): 58.
Chicago
Wei, B., Wang, L., Zhao, J."Circular RNA hsa_circ_0005114‑miR‑142‑3p/miR‑590‑5p-adenomatous polyposis coli protein axis as a potential target for treatment of glioma". Oncology Letters 21, no. 1 (2021): 58. https://doi.org/10.3892/ol.2020.12320