Next Article in Journal
Impact of Eukaryotic Translation Initiation Factors on Breast Cancer: Still Much to Investigate
Next Article in Special Issue
circ2GO: A Database Linking Circular RNAs to Gene Function
Previous Article in Journal
High-Throughput Sequencing of Gastric Cancer Patients: Unravelling Genetic Predispositions Towards an Early-Onset Subtype
Previous Article in Special Issue
The Expression, Functions and Mechanisms of Circular RNAs in Gynecological Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Biological Role of Sponge Circular RNAs in Gastric Cancer: Main Players or Coadjuvants?

by
Adenilson Leão Pereira
1,2,
Leandro Magalhães
3,
Rafael Pompeu Pantoja
3,
Gilderlanio Araújo
3,
Ândrea Ribeiro-dos-Santos
2,3 and
Amanda Ferreira Vidal
3,*
1
Faculty of Medicine, Federal University of Pará, Altamira 68371-163, Brazil
2
Research Center on Oncology, Graduate Program of Oncology and Medical Science, Federal University of Pará, Belém 66073-000, Brazil
3
Laboratory of Human and Medical Genetics, Institute of Biological Sciences, Graduate Program of Genetics and Molecular Biology, Federal University of Pará, Belém 66075-110, Brazil
*
Author to whom correspondence should be addressed.
Cancers 2020, 12(7), 1982; https://doi.org/10.3390/cancers12071982
Submission received: 12 May 2020 / Revised: 16 June 2020 / Accepted: 17 June 2020 / Published: 21 July 2020
(This article belongs to the Special Issue Circular RNAs in Cancer)

Abstract

:
Circular RNAs (circRNAs) are a new class of long noncoding RNAs able to perform multiple functions, including sponging microRNAs (miRNAs) and RNA-Binding Proteins (RBPs). They play an important role in gastric carcinogenesis, but its involvement during gastric cancer (GC) development and progression are not well understood. We gathered miRNA and/or RBPs sponge circRNAs present in GC, and accessed their biological roles through functional enrichment of their target genes or ligand RBPs. We identified 54 sponge circRNAs in GC that are able to sponge 51 miRNAs and 103 RBPs. Then, we evaluated their host gene expression using The Cancer Genome Atlas (TCGA) database and observed that COL1A2 is the most overexpressed gene, which may be due to circHIPK3/miR-29b-c/COL1A2 axis dysregulation. We identified 27 GC-related pathways that may be affected mainly by circPVT1, circHIPK3 and circNF1. Our results indicate that circHIPK3/miR-107/BDNF/LIN28 axis may mediate chemoresistance in GC, and that circPVT1, circHIPK3, circNF1, ciRS-7 and circ_0000096 appear to be involved in gastrointestinal cancer development. Lastly, circHIPK3, circNRIP1 and circSMARCA5 were identified in different ethnic populations and may be ubiquitous modulators of gastric carcinogenesis. Overall, the studied sponge circRNAs are part of a complex RBP-circRNA-miRNA-mRNA interaction network, and are involved in the establishment, chemoresistance and progression of GC.

1. Introduction

Gastric cancer (GC) is the fifth most frequently diagnosed cancer and the third leading cancer-related deaths worldwide [1]. It is an aggressive disease, whose five-year survival rate is high in some countries like Japan (60%) and South Korea (69%); however, in regions such as South and North America and Europe, this rate does not exceed 30% [2].
Genomic studies have uncovered key changes that result in the development of GC and, the most studied and characterized epigenetic alteration is the tumor-associated DNA methylation profile [3,4]. However, noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) [5] and circular RNAs (circRNAs) [6], are also dysregulated during this disease onset and progression.
CircRNAs are a new class of long noncoding RNAs that have their 5′ and 3′ ends covalently joined [7,8]. Current knowledge of circRNAs suggests that they can perform multiple functions, such as: sponging miRNAs, transcription and splicing modulators and RNA-Binding Proteins (RBPs, e.g., AGO2 and RNA Pol II). They can also act as templates for protein translation and mediate immune responses [7,8]. Dysregulation in their role as miRNA-sponge has been reported in several types of gastrointestinal malignancies, including hepatocellular carcinoma [9,10,11], pancreatic [10,12], colorectal [10,11,13] and gastric cancer [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69].
A large number of studies have demonstrated the role of circRNAs as miRNA-sponges in GC [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69]. For example, it has been shown that alterations in circLARP4/miR-424/LATS1 [57], circRNA_100269/miR-630 [59], circPVT1/miR-125b/E2F2 [14] and circ_0000096/miR-224-5p/miR-200a [58] axes can stimulate proliferation, migration and invasion of GC cells. They were also correlated with staging, poor survival and recurrence of this type of tumor.
More recently, it has been found that circHIPK3/miR-107/BDNF imbalance leads to proliferation and migration of GC cells, and circHIPK3 has been suggested as a potential prognostic biomarker in GC [17]. Additionally, circBANP/let-7a axis imbalance can lead to tumor progression by affecting FZD5/Wnt/ß-catenin signaling [54].
circRNAs can also sponge RBPs in GC and can negatively affect the activity of the anchored protein or favor the expression and/or activity of others. For instance, circ-DONSON has been shown to interact directly with proteins from the NURF chromatin remodeling complex to regulate the transcription of SOX4 oncogene and promote gastric tumorigenesis [70]. On the other hand, circFAT1(e2) is able to bind directly to Y-box binding protein-1 (YBX1) and block tumor-promoting abilities of GC cells [69]. Although studies have demonstrated a relationship between sponge circRNAs and GC, their biological role is still not well understood, and a broader and more integrated scenario of these molecules in gastric carcinogenesis remains to be outlined, being important for future translational applications.
In this study, we gathered experimentally validated sponge circRNAs in GC, in order to understand and to have a broader view of their significance during the development of this type of tumor. Hence, we evaluated the biological roles of these circRNAs, considering their associated RBPs and/or the target genes of their sponged miRNAs, and thus, identified their involvement in GC-related pathways. Our results indicate that the sponge circRNAs in GC are part of a complex RBP-circRNA-miRNA-mRNA interaction network, and may be involved in the establishment, chemoresistance and progression of the tumor.

2. Results

Our search identified 56 papers reporting 54 dysregulated sponge circRNAs in GC, of which 38 were upregulated and 16 were downregulated (Table 1). We explored our results by analyzing them in three different ways (Figure 1). Firstly, we investigated the expression and potential roles of the sponge circRNAs’ host genes; then, we analyzed the sponge circRNAs’ target miRNAs and estimated their roles by drawing circRNA-miRNA-mRNA interaction networks; lastly, we focused at the sponged RBPs and their biological roles.

2.1. Host Genes Expression Profile in GC

We used GC samples from The Cancer Genome Atlas (TCGA) to access the expression of the sponge circRNAs’ host genes. We found 38 differentially expressed host genes in GC (37 upregulated and one downregulated) when compared to the adjacent tissue (Table S1). Ten genes (COL1A2, FAT1, NF1, FN1, NOTCH1, OSBPL10, PDSS1, PTK2, RANGAP1 and SKA3) presented |fold change|>2, being COL1A2, PVT1, RBMS3 and SERPINB5 the ones that showed expression levels higher than three-fold in GC (Table S1). In general, circRNAs and their host genes presented similar expression patterns.

2.2. Dysregulated Sponge Circular RNAs in GC

The 54 circRNAs are able to sponge 51 different miRNAs (Table 1). These studies validated their findings using sensitive and reliable methodologies, such as RT-qPCR, Western blot, cell and/or luciferase assay report.
In general, we observed 46 circRNAs sponging a single microRNA and eight circRNAs regulating at least two microRNAs simultaneously (Table 1). On the other hand, 10 miRNAs can be sponged by more than one circRNA (miR-107 (circHIPK3 and circ-ZFR); miR-124-3p (circPVT1 and circHIPK3); miR-149 (circ_NRIP1 and hsa_circ_0017728); miR-637 (circ_ERBB2 and circ-NOTCH1); miR-130a-3p (circGRAMD1B and circ-ZFR); miR-182-5p (circ-SFMBT2 and circFN1); miR-1256 (circHECTD1 and circ-DCAF6); miR-503 (circ_ERBB2 and hsa_circ_0000267); miR-145 (circ-PRMT5, circDUSP16 and circMAN2B2); miR-375 (hsa_circ_0008035, circ-SERPINE2 and circUBA1); Table 1).

2.3. Functional Enrichment of the Sponged miRNAs’ Target Genes

To identify the experimentally validated target genes of the 51 sponged miRNAs, we submitted them to the miRTargetLink Human online tool. The query showed that of the 51 miRNAs, 39 miRNAs can regulate 966 experimentally validated genes (Table S2). Of these, 159 genes can be regulated by more than one miRNA and BCL2, CDK6, SP1, IGF1R, VEGFA, CCND2, HMGA2, EGFR, CDKN1A, MCL1 and PTEN genes can be regulated by more than five miRNAs each (Table S3). Twelve miRNAs (miR-214-5p, miR-367-5p, miR-369-3p, miR-449c-5p, miR-503-3p, miR-526b, miR-877-3p, miR-1227-5p, miR-1231, miR-1304, miR-1306-3p and miR-4319) did not show experimentally validated target genes according to miRTargetLink Human tool (Table S2).
Functional enrichment analysis showed that these genes are involved in the regulation of 165 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (Table S4). Of these, we highlight 27 biological pathways that are closely related to the development of GC (e.g., bacterial invasion of epithelial cells, cell cycle, Epstein–Barr virus infection, epithelial cell signaling in Helicobacter pylori infection, etc.; Figure 2).
By performing differential expression analysis of the 966 miRNAs’ target genes using the TCGA database, we found 45 DE genes (p ≤ 0.05 and |fold change| ≥1.5, Table S5). These 45 genes may have a greater impact on gastric cancer and should be further explored. However, we would like to emphasize that an integrative analysis would become more accurate using the same cohort to access mRNA, miRNA and circRNA global expression. By this approach, the identification of hub ncRNAs and genes involved in gastric carcinogenesis would be more precise.

2.4. Biological Pathways and the Potential Roles of the Sponge circRNAs in GC

Of the 966 target genes used during functional enrichment analysis, 271 are involved in 27 biological pathways directly associated with GC (Figure 1; Table S6). circPVT1, circHIPK3 and circNF1 may interfere and regulate the expression of the largest number of genes associated with these pathways (107, 103 and 43 genes, respectively; Figure S1). Other circRNAs, such as circLARP4, hsa_circ_0008035, circ_0000096, circ-ZFR, ciRS-7, circ-SFMBT2, hsa_circ_0027599, ciRS-133 and circ-EIF4G3, can regulate the expression of more than 10 genes each, suggesting they have an important role in GC (Table S6). circDLST, circOSBPL10, circNHSL1, circHECTD1, circRHOBTB3, hsa_circ_0001368 and circFAT1(e2) regulate one gene each (Table S6).
According to the KEGG database, the GC pathway (hsa05226) is composed of 147 main genes, of which 53 can be regulated by 24 miRNAs evaluated in this study (Tables S4 and S7). Through the mRNA-miRNA-circRNA interaction, we identified 23 sponge circRNAs that can regulate these miRNAs and may affect the expression of their target genes directly involved in the gastric cancer pathway (Table S7). circNF1, circHIPK3, circPVT1 and circ_0000096 are the ones that can regulate the highest number of genes present in this pathway. Meanwhile, hsa_circ_100269, circ-CEP85L, circSMARCA5, circRHOBTB3, circRBMS3, circOSBPL10 and circAKT3 regulate one gene each (Table S7). BCL2 gene can be regulated by the greatest number of miRNAs (10 target miRNAs) and, consequently, the gene associated with the highest number of sponge circRNAs (13 circRNAs, Table S7).

2.5. Biological Pathways and the Potential Roles of the Sponge circRNAs in Other Cancers

Among the 165 biological pathways, we also observed that the circRNA-miRNA-mRNA interaction may be involved in 16 different types of tumors, of which four are gastrointestinal tumors (Figure 3; Table S8). So, we evaluated the types of tumors in which the target genes could be associated (consequently, the sponge circRNAs and their target miRNAs too). We identified that these 16 different types of tumors can be affected by the dysregulated expression of 140 sponged miRNAs’ target genes (Figure 3; Table S8). We also noticed that 51 of them are shared by at least two different tumors of the gastrointestinal tract and can be regulated by 25 miRNAs and 28 sponge circRNAs (Table S8). circHIPK3, circNF1, circ_0000096 and circPVT1 can regulate the highest number of genes (≥11 genes each); while circAKT3, circ-CEP85L, circOSBPL10, circRHOBTB3, circSMARCA5 and hsa_circ_100269 regulate one gene each (Table S8). Twenty-five genes were associated with the four types of gastrointestinal tumors, and can be regulated by 18 miRNAs and 17 circRNAs sponge (Table S8). Of these, CDKN1A, EGFR, SMAD4 and TP53 genes can be regulated by four to five miRNAs each, and by six to eight sponge circRNAs each one (Table S8).

2.6. Functional and Enrichment Analysis of RBPs

We also analyzed the existence of RBP sites in the sponge circRNAs. We observed that 13 of the 54 studied circRNAs have RBP anchoring sites (circHIPK3, circPVT1, circFAT1(e2), circAKT3, ciRS-7, circ_SPECC1, circSMARCA5, hsa_circ_0000467, hsa_circ_0000267, hsa_circ_0000673, hsa_circ_0001368, circ_0000096 and hsa_circ_0000993), and that together, they are able to bind to 103 different proteins (Table S9). circFAT1(e2) and hsa_circ_0000673 have the highest number of RBP-binding sites (78 and 76 sites, respectively), while ciRS-7 has 11 sites (Table S9). IGF2BP2 and MOV10 are able to bind to all analyzed circRNAs (Table S10). We highlight LIN28A and LIN28B, which have binding sites in 11 circRNAs (Table S10) and were previously related to the GC [71,72]. Functional enrichment analysis of these proteins revealed that they are involved in 27 different Reactome pathways, and that they have a strong association with RNA metabolism and processing (Figure 4).

3. Discussion

Although genomic big data have found key genetic modifications that are capable of leading to GC onset [3,4], studies have elegantly shown that a complex, dysregulated and poorly understood regulatory network that involves ncRNAs, mRNAs and RBPs can induce gastric tumorigenesis.
In this study, we gathered circRNAs that can sponge miRNAs and RBPs, and observed that many of them may break homeostasis and disturb biological pathways in GC. An overview of our analyses and results is presented in Figure 1. For instance, the circHIPK3/miR-124/miR-29b/COL1A1/COL4A1/CDK6 [16], ciRS-7/miR-7/PTEN/PI3K/AKT [18] and other signaling pathways (Table S11) may promote the onset, establishment and progression of the tumor by modulating the cell cycle progression, tumor growth, proliferation, apoptosis, invasion, migration, survival and radiosensitivity, stemness, epithelial–mesenchymal transition (EMT) and metastasis of malignant gastric cells. In addition, dysregulation of these axes has an important clinical impact during the course of the disease, since they are associated with poor prognosis, overall and disease-free survival and clinicopathological characteristics (Table S11).
COL1A2 was the circRNAs’ host gene with the highest fold change in GC (Table S1). This gene was positively related to clinicopathological characteristics such as tumor size and depth of invasion in GC [73]. Interestingly, circHIPK3 upregulates COL1A1 and COL4A1 expression by sponging miR-29b and miR-124, promoting GC progression [16]. Meanwhile, COL1A2 has been shown to be a direct target of miR-29b and miR-29c [74,75], which are members of the miR-29 family (miR-29a, 29b and 29c) and share a common seed sequence [76]. Since miR-29b and miR-29c are known to be downregulated in GC [77,78], it is possible that the overexpression of COL1A2 observed in gastric tumor tissue is maintained in part by the disturbance in circHIPK3/miR-29b-c/COL1A2 axis, leading to gastric cancer progression.
Gene functional enrichment in biological pathways indicates that circRNA/miRNA/mRNA axes can directly affect several pathways (Figure 2) that are known to be dysregulated during gastric carcinogenesis [79,80,81]. We highlight circPVT1, circHIPK3 and circNF1 that have the potential to regulate the expression of the highest number of target genes related to these pathways, suggesting that they are critical modulators in gastric adenocarcinoma. Therefore, these findings indicate that the sponge circRNAs may modulate a greater number of pathways in GC than have been associated so far.
Among the pathways shown in Figure 2, we highlight PI3K/Akt signaling pathway because it has the highest number of sponged miRNAs’ target genes and can be regulated by 28 miRNAs and 33 circRNAs (Tables S4–S6). This pathway can be modulated by different epigenetic mechanisms, such as noncoding RNAs, DNA methylation and chromatin remodeling systems [82,83,84]. Here, we highlight BCL2, CDK4, CDK6, CDKN1A, EGFR, ERBB2, FGFR1, IGF1R, ITGB1, MYC, NRAS, KRAS, PHLPP1, PIK3CA, PTEN, TP53 and VEGFA, genes that are potentially the most affected since they are modulated by the highest number of circRNA/miRNA axes (Table S6). In addition, AKT1 (circPVT1/miR-125b-5p), AKT2 (circHIPK3/miR-29b-3p) and AKT3 (cirNF1/miR-16-5p) may also be under the regulatory influence of these axes (Table S6). Interestingly, many of these genes have been found to be associated with this pathway [82,85] and promoting gastric carcinogenesis [3,4].
PI3K/Akt signaling pathway regulates metabolism, survival, cell growth, motility and angiogenesis in several tumors [86,87]. In this pathway, there is the recruitment of phosphorylated AKT (pAkt) and PDK1 (a kinase that phosphorylates Akt protein) by PIP3 action (class IA PI3Ks), caused by the downregulation of PTEN, a negative regulator of PIP3 and PI3K [79,83]. AKT is responsible for activating growth, proliferation and survival signaling [79]. Interestingly, we have found several circRNA-miRNA interactions that are capable of affecting the activity or expression of genes (e.g., PTEN, PI3K, AKT, p21 and PDK1) that are directly involved in PI3K/Akt pathway (Table 2). For instance, circGRAMD1B/miR-130a-3p axis negatively regulates PTEN and p21, allowing higher proliferation, migration and invasion rates of GC cells [62]. Curiously, p21 is a cyclin/cdk inhibitor protein directly associated with the PI3K/Akt pathway [88]. We also highlight the tumor suppressor miR-375 that decreases PDK1 activity in PI3K/Akt signaling [79]. We observed that this miRNA is sponged by three different circRNAs (Table 2), suggesting that they can modulate the miR-375/PDK1/Akt axis to activate cell growth, survival and motility in GC (Table 2, Figure 5A). Therefore, the circRNAs we found may potentially promote gastric carcinogenesis by affecting mainly the PI3K/Akt pathway (Figure 5A).
Regarding the gastric cancer pathway (hsa05226), we noticed that it can be more affected by circNF1, circHIPK3, circPVT1 and circ_0000096, since they can regulate many target genes (Table S7). These circRNAs were also the most critical for other types of gastrointestinal tumors (Table S8). Surprisingly, circHIPK3 and circPVT1 have already been found to be dysregulated in hepatocellular carcinoma, colorectal and pancreatic cancers [9,10,11,12,13]. ciR-7 is another circRNA that is dysregulated in these four types of tumors [11]. Interestingly, circNF1, circHIPK3 and circPVT1 are involved in the 27 GC-related pathways, in the GC pathway itself (hsa05226) and in other cancer-associated pathways, including gastrointestinal ones (Figure 1). Therefore, we suggest that circHIPK3, circPVT1, ciR-7, circNF1 and circ_0000096 may be involved in gastrointestinal carcinogenesis. However, it is necessary to experimentally validate whether circNF1 and circ_0000096 are also dysregulated in hepatocellular carcinoma, colorectal and pancreatic cancer.
circRNAs can act as an anchoring point for RBPs and this interaction can block these binding protein’s activity [69]. We found that circAKT3, circFAT1(e2), circHIPK3, circPVT1, circSMARCA5, circ_SPECC1, circ_0000096, hsa_circ_0000993, hsa_circ_0000467, hsa_circ_0001368 and hsa_circ_0000673 may bind to LIN28A and LIN28B. High Lin28 protein levels seem to mediate the chemoresistance to drugs such as oxaliplatin, paclitaxel, doxorubicin and fluorouracil in GC by regulating miR-107 expression [71,72,89]. Interestingly, only four circRNAs were overexpressed (circAKT3, circHIPK3, circPVT1 and hsa_circ_0000467) in GC, while the remaining seven were downregulated (Table 1), suggesting that the low expression of these circRNAs may allow the activity of LIN28 in GC to favor chemoresistance.
Although LIN28 can inhibit the maturation of miR-107, this miRNA can also negatively regulate LIN28 [89]. LIN28 is capable of blocking the maturation of a subset of miRNA precursors (i.e., pri- and pre-miRNA) such as let-7, miR-107, miR-143 and miR-200c [90,91]. circHIPK3 also has an inhibitory effect on miR-107 expression by sponging it, which allows brain-derived neurotrophic factor (BDNF) activity [17]. Interestingly, it was observed that BDNF is a positive regulator and increases the activity of LIN28 to act in the regulation of specific miRNAs [91]. In this context, we believe that LIN28 suppresses miR-107 at the beginning of its biogenesis, while circHIPK3 reinforces the repression of this miRNA (those that eventually deviated from the repression via LIN28) in its mature state and enhances LIN28 activity (Figure 5B). Therefore, circHIPK3/miR-107/BDNF/LIN28 axis is an alternative mechanism proposed for LIN28-mediated chemoresistance in GC (Figure 5B) and requires experimental validation.
Lastly, of the 54 circRNAs identified in this study, all were tested in the Asian/Chinese population. However, we observed that circHIPK3, circNRIP1 and circSMARCA5 were also identified in the Brazilian population [6]. The Brazilian population is highly substructured and has a strong genetic mixture, due to the genetic contribution of different parental populations, such as European, African, Asian and Amerindian ones [92,93]. It is known that the genetic/ethnic contribution may influence the expression of many genes in response to diseases and/or infections [94,95]. Although the ethnic factor contributes to the modulation of the expression of some genes, in the case of these three circRNAs this apparently does not occur. Thus, we suggest that these three circRNAs may be key ubiquitous modulators of gastric carcinogenesis.
The sponge function of the circRNAs has been debated since most of them harbor only a few miRNA-binding sites and effective target repression may often require many binding sites [96]. However, the growing number of experimental studies have demonstrated that the circRNAs are able to successfully repress miRNAs by sponging them (Table S11). Our network-based approach provided a valuable and easy way of integrating heterogeneous data, which allowed better recognition of a regulatory module involving RBP-circRNA-miRNA-mRNA interactions in gastric cancer.
In general, comprehensive bioinformatics analysis of regulatory networks has pointed out potential interactions and new biological roles between small and long noncoding RNAs associated with complex diseases, such as gastric and colorectal cancers [97,98,99]. We understand that there are inherent limitations in predictive computational analysis; however, in this study, these limitations were minimized by only using experimentally tested and validated data, in order to avoid biased results. This integrative study, using tested biological interactions and computational analysis, offers a broader picture of the epigenetic complexity that involves RBP-ncRNA-mRNA in gastric tumorigenesis.

4. Materials and Methods

4.1. Search for the Sponge circRNAs

Sponge circRNAs were extracted from previous studies selected from PubMed and/or Google Scholar repository databases. Only original research articles published until March 2020 were selected. Keywords that relate and/or associate sponge circRNA, miRNA and gastric cancer were used in the search. In both repositories, we used the following entries: “circRNA sponge and Gastric Cancer”, “circRNA sponge in Gastric Cancer” and “circRNA-miRNA in Gastric Cancer”. Only studies that conducted experimental validation (e.g., RT-qPCR, Western blot, cell and/or luciferase reporter assay) of the analyzed sponge circRNA were included in the analyzes. Studies based only on in silico predictions of circRNA-miRNA interaction were excluded. Restrictions by year of publication or language were not applied.

4.2. Expression Profile of the Sponge circRNAs’ Host Genes

To analyze the differential expression of sponge circRNAs’ host genes, we used the “UALCAN: Analyze, Integrate, Discovery” online tool [100] (http://ualcan.path.uab.edu/index.html). This tool is connected to The Cancer Genome Atlas (TCGA) database and provides OMICS expression data of 415 gastric cancer and 34 controls (adjacent to gastric cancer) samples in transcripts per million (TPM).

4.3. Target Gene Identification and Enrichment Analysis

Target genes of the sponged miRNAs were analyzed using miRTargetLink Human online tool [101] (https://ccb-web.cs.uni-saarland.de/mirtargetlink/). This tool is connected to the miRTarBase database (release 7.0) [102] (http://miRTarBase.cuhk.edu.cn/), which provides information on experimentally validated interaction between the miRNA and its target gene. We only considered target genes that were validated with “strong evidence” (e.g., RT-qPCR, Western blot, cell and/or luciferase reporter assay).
Functional enrichment analysis of the sponged miRNAs’ target genes was performed using Kyoto Encyclopedia of Genes and Genomes (KEGG) and/or Reactome pathways in STRING online tool (v.11.0). In this analysis, we considered the total number of target genes of each miRNA. Graphs were made in R statistical package (Release 3.6.0; https://www.r-project.org/) [103,104].

4.4. Target RBPs and Functional Analysis

Target RBPs of the sponge circRNAs were accessed using the starBase (v. 2.0) online tool (http://starbase.sysu.edu.cn/starbase2/index.php), which provides large-scale CLIP-Seq data of circRNA-RBP interaction. Its functional analysis was performed using Reactome pathways in STRING online tool (v.11.0). Graphs were made in R statistical package (Release 3.6.0; https://www.r-project.org/) [103,104].

5. Conclusions

The functional roles of the sponge circRNAs in the pathophysiology of GC remains poorly understood. However, in this study, we pointed to new directions for understanding this context. Overall, the 54 sponge circRNAs studied are capable of interfering in 27 GC-related biological pathways by modulating the interaction of 51 different miRNAs and their target genes, with a more pronounced effect on PI3K/Akt signaling pathway. COL1A2 was the host gene with the highest fold change and circHIPK3/miR-29b-c/COL1A2 axis may be the one that maintains high levels of this gene in GC, favoring the severe clinicopathological characteristics. Thirteen circRNAs may interact with 103 RBPs that are mostly associated with RNA metabolism. Among them, Lin28 stands out for being previously associated with GC. In this context, circHIPK3 may control the balance between LIN28/miR-107 to favor the activity of Lin28 in a negative feedback loop, promoting chemoresistance of GC cells. circHIPK3, circNRIP1 and circSMARCA5 are involved with GC in different ethnic populations and may be ubiquitous regulators of this type of tumor. Finally, circHIPK3, circPVT1, ciR-7, circNF1 and circ_0000096 are potentially promoting gastrointestinal tumors. Therefore, the sponge circRNAs are favoring gastric carcinogenesis by affecting complex mRNA-miRNA-circRNA-RBP interaction networks, and future studies are required to validate their potential as therapeutic targets for this type of tumor. In this context, the identified sponge circRNAs appear to play a central role in the background of gastric tumors and may be considered major players in tumorigenesis.

Supplementary Materials

The following are available online at https://www.mdpi.com/2072-6694/12/7/1982/s1, Figure S1: Most important circRNAs to 27 GC-related pathways, Table S1: Host genes expression in GC from TCGA database obtained by UALCAM online tool, Table S2: Sponged miRNAs target genes provided by miRTargetLink Human - Only target genes valued by strong evidence, Table S3: List of miRNAs and target genes, Table S4: Functional analysis of sponged miRNAs’ target genes, Table S5: Genes differentially expressed in GC, Table S6: Intersection among the lists of genes of the 27 GC-related KEGG pathways and the sponged miRNAs’ target genes, Table S7: Genes involved in gastric cancer pathway (hsa05226) and their associated mIRNAs and sponge circRNAs, Table S8: Intersection between the lists of genes of the 16 cancer KEGG pathways and the sponged miRNAs’ target genes, Table S9: List of the target RBPs, Table S10: Target RBPs and their sponge circRNAs, Table S11: Clinical importance and main effects of circRNA-miRNA-mRNA interaction in GC.

Author Contributions

Conceptualization, A.L.P., Â.R.-d.-S. and A.F.V.; formal analysis, A.L.P. and R.P.P.; funding acquisition, Â.R.-d.-S. and A.F.V.; writing—original draft, A.L.P.; writing—review and editing, A.L.P., L.M., G.A., Â.R.-d.-S. and A.F.V. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by Rede de Pesquisa em Genômica Populacional Humana (CAPES/Biologia Computacional: No. 3381/2013/CAPES) and PROPESP/UFPA. A.L.P. was supported by CAPES/BRASIL Post-Doctoral fellowship (Edital No.51/2013/Biologia Computacional; Proc. No. 88887.313819/2019-00/CAPES) in Oncology and Medical Sciences - UFPA. Â.R.S. was supported by CNPq/Productivity: 304413/2015-1.

Acknowledgments

We thank to Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Conselho Nacional de Desenvolvimento (CNPq) and Pró-Reitoria de Pesquisa e Pós-Graduação da Universidade Federal do Pará (PROPESP-UFPA).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Allemani, C.; Matsuda, T.; Di-Carlo, V.; Harewood, R.; Matz, M.; Nikšić, M.; Bonaventure, A.; Valkov, M.; Johnson, C.J.; CONCORD Working Group; et al. Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): Analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet 2018, 391, 1023–1075. [Google Scholar] [CrossRef] [Green Version]
  3. Bass, A.; Thorsson, V.; Shmulevich, I.; Reynolds, S.M.; Miller, M.; Bernard, B.; Hinoue, T.; Laird, P.W.; Curtis, C.; Shen, H.; et al. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar]
  4. Cristescu, R.; Lee, J.; Nebozhyn, M.; Kim, K.M.; Ting, J.C.; Wong, S.S.; Liu, J.; Yue, Y.G.; Wang, J.; Yu, K.; et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat. Med. 2015, 21, 449–456. [Google Scholar] [CrossRef] [PubMed]
  5. Pereira, A.; Moreira, F.; Vinasco-Sandoval, T.; Cunha, A.; Vidal, A.; Ribeiro-dos-Santos, A.M.; Pinto, P.; Magalhães, L.; Assumpção, M.; Demachki, S.; et al. miRNome Reveals New Insights Into the Molecular Biology of Field Cancerization in Gastric Cancer. Front. Genet. 2019, 10, 592. [Google Scholar] [CrossRef]
  6. Vidal, A.F.; Ribeiro-dos-Santos, A.M.; Vinasco-Sandoval, T.; Magalhães, L.; Pinto, P.; Anaissi, A.K.M.; Demachki, S.; de Assumpção, P.P.; Santos, S.E.B.; Ribeiro-dos-Santos, Â. The comprehensive expression analysis of circular RNAs in gastric cancer and its association with field cancerization. Sci. Rep. 2017, 7, 14551. [Google Scholar] [CrossRef] [Green Version]
  7. Li, X.; Yang, L.; Chen, L.L. The Biogenesis, Functions, and Challenges of Circular RNAs. Mol. Cell 2018, 71, 428–442. [Google Scholar] [CrossRef] [Green Version]
  8. Ng, W.L.; Mohd Mohidin, T.B.; Shukla, K. Functional role of circular RNAs in cancer development and progression. RNA Biol. 2018, 15, 995–1005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Chen, G.; Shi, Y.; Liu, M.; Sun, J. circHIPK3 regulates cell proliferation and migration by sponging miR-124 and regulating AQP3 expression in hepatocellular carcinoma. Cell Death Dis. 2018, 9, 175. [Google Scholar] [CrossRef] [PubMed]
  10. Naeli, P.; Pourhanifeh, M.H.; Karimzadeh, M.R.; Shabaninejad, Z.; Movahedpour, A.; Tarrahimofrad, H.; Mirzaei, H.R.; Bafrani, H.H.; Savardashtaki, A.; Mirzaei, H.; et al. Circular RNAs and gastrointestinal cancers: Epigenetic regulators with a prognostic and therapeutic role. Crit. Rev. Oncol. Hematol. 2020, 145, 102854. [Google Scholar] [CrossRef] [PubMed]
  11. Derderian, C.; Orunmuyi, A.T.; Olapade-Olaopa, E.O.; Ogunwobi, O.O. PVT1 Signaling Is a Mediator of Cancer Progression. Front. Oncol. 2019, 9, 502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Liu, Y.; Xia, L.; Dong, L.; Wang, J.; Xiao, Q.; Yu, X.; Zhu, H. CircHIPK3 Promotes Gemcitabine (GEM) Resistance in Pancreatic Cancer Cells by Sponging miR-330-5p and Targets RASSF1. Cancer Manag. Res. 2020, 12, 12921–12929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zeng, K.; Chen, X.; Xu, M.; Liu, X.; Hu, X.; Xu, T.; Sun, H.; Pan, Y.; He, B.; Wang, S. CircHIPK3 promotes colorectal cancer growth and metastasis by sponging miR-7. Cell Death Dis. 2018, 9, 417. [Google Scholar] [CrossRef]
  14. Chen, J.; Li, Y.; Zheng, Q.; Bao, C.; He, J.; Chen, B.; Lyu, D.; Zheng, B.; Xu, Y.; Long, Z.; et al. Circular RNA profile identifies circPVT1 as a proliferative factor and prognostic marker in gastric cancer. Cancer Lett. 2017, 388, 208–219. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, Y.Y.; Zhang, L.Y.; Du, W.Z. Circular RNA circ-PVT1 contributes to paclitaxel resistance of gastric cancer cells through the regulation of ZEB1 expression by sponging miR-124-3p. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef]
  16. Cheng, J.; Zhuo, H.; Xu, M.; Wang, L.; Xu, H.; Peng, J.; Hou, J.; Lin, L.; Cai, J. Regulatory network of circRNA-miRNA-mRNA contributes to the histological classification and disease progression in gastric cancer. J. Transl. Med. 2018, 16, 216. [Google Scholar] [CrossRef] [Green Version]
  17. Wei, J.; Xu, H.; Wei, W.; Wang, Z.; Zhang, Q.; De, W.; Shu, Y. circHIPK3 Promotes Cell Proliferation and Migration of Gastric Cancer by Sponging miR-107 and Regulating BDNF Expression. Onco Targ. Ther. 2020, 13, 1613–1624. [Google Scholar] [CrossRef] [Green Version]
  18. Pan, H.; Li, T.; Jiang, Y.; Pan, C.; Ding, Y.; Huang, Z.; Yu, H.; Kong, D. Overexpression of circular RNA ciRS-7 abrogates the tumor suppressive effect of miR-7 on gastric cancer via PTEN/PI3K/AKT signaling pathway. J. Cell. Biochem. 2018, 119, 440–446. [Google Scholar] [CrossRef]
  19. Li, G.; Xue, M.; Yang, F.; Jin, Y.; Fan, Y.; Li, W. CircRBMS3 promotes gastric cancer tumorigenesis by regulating miR-153-SNAI1 axis. J. Cell. Physiol. 2019, 234, 3020–3028. [Google Scholar] [CrossRef]
  20. Li, H.; Yao, G.; Feng, B.; Lu, X.; Fan, Y. Circ_0056618 and CXCR4 act as competing endogenous in gastric cancer by regulating miR-206. J. Cell. Biochem. 2018, 119, 9543–9551. [Google Scholar] [CrossRef]
  21. Wang, L.; Shen, J.; Jiang, Y. Circ_0027599/PHDLA1 suppresses gastric cancer progression by sponging miR-101-3p.1. Cell. Biosci. 2018, 8, 58. [Google Scholar] [CrossRef]
  22. Li, X.; He, M.; Guo, J.; Cao, T. Upregulation of circular RNA circ-ERBB2 predicts unfavorable prognosis and facilitates the progression of gastric cancer via miR-503/CACUL1 and miR-637/MMP-19 signaling. Biochem. Biophys. Res. Commun. 2019, 511, 926–930. [Google Scholar] [CrossRef] [PubMed]
  23. Cai, X.; Nie, J.; Chen, L.; Yu, F. Circ_0000267 promotes gastric cancer progression via sponging MiR-503-5p and regulating HMGA2 expression. Mol. Genet. Genomic. Med. 2020, 8, e1093. [Google Scholar] [CrossRef] [Green Version]
  24. Guan, E.C.; Xu, X.G.; Xue, F.X. circ-NOTCH1 acts as a sponge of miR-637 and affects the expression of its target gene Apelin to regulate gastric câncer cell growth. Biochem. Cell. Biol. 2020, 98, 164–170. [Google Scholar] [CrossRef] [PubMed]
  25. Zhao, X.; Zhong, Q.; Cheng, X.; Wang, S.; Wu, R.; Leng, X.; Shao, L. miR-449c-5p availability is antagonized by circ-NOTCH1 for MYC-induced NOTCH1 upregulation as well as tumor metastasis and stemness in gastric cancer. J. Cell. Biochem. 2020, 1–12. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, H.; Wang, X.; Huang, H.; Wang, Y.; Zhang, F.; Wang, S. Hsa_circ_0067997 promotes the progression of gastric cancer by inhibition of miR-515-5p and activation of X chromosome-linked inhibitor of apoptosis (XIAP). Artif. Cells Nanomed. Biotechnol. 2019, 1, 308–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zhang, X.; Wang, S.; Wang, H.; Cao, J.; Huang, X.; Chen, Z.; Xu, P.; Sun, G.; Xu, J.; Lv, J.; et al. Circular RNA circNRIP1 acts as a microRNA-149-5p sponge to promote gastric cancer progression via the AKT1/mTOR pathway. Mol. Cancer 2019, 18, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Yang, C.; Deng, S. Hsa_circ_0017728 as an oncogene in gastric cancer by sponging miR-149 and modulating the IL-6/STAT3 pathway. Arch. Med. Sci. 2019. [Google Scholar] [CrossRef] [Green Version]
  29. Xue, M.; Li, G.; Fang, X.; Wang, L.; Jin, Y.; Zhou, Q. hsa_circ_0081143 promotes cisplatin resistance in gastric cancer by targeting miR-646/CDK6 pathway. Cancer Cell. Int. 2019, 19, 25. [Google Scholar] [CrossRef] [Green Version]
  30. Wang, Z.; Ma, K.; Pitts, S.; Cheng, Y.; Liu, X.; Ke, X.; Kovaka, S.; Ashktorab, H.; Smoot, D.T.; Schatz, M.; et al. Novel circular RNA NF1 acts as a molecular sponge, promoting gastric cancer by absorbing miR-16. Endocr. Relat. Cancer 2018. [Google Scholar] [CrossRef]
  31. Zhang, J.; Hou, L.; Liang, R.; Chen, X.; Zhang, R.; Chen, W.; Zhu, J. CircDLST promotes the tumorigenesis and metastasis of gastric cancer by sponging miR-502-5p and activating the NRAS/MEK1/ERK1/2 signaling. Mol. Cancer 2019, 18, 80. [Google Scholar] [CrossRef] [PubMed]
  32. Ouyang, Y.; Li, Y.; Huang, Y.; Li, X.; Zhu, Y.; Long, Y.; Wang, Y.; Guo, X.; Gong, K. CircRNA circPDSS1 promotes the gastric cancer progression by sponging miR-186-5p and modulating NEK2. J. Cell. Physiol. 2019, 234, 10458–10469. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, H.; Zhu, L.; Bai, M.; Liu, Y.; Zhan, Y.; Deng, T.; Yang, H.; Sun, W.; Wang, X.; Zhu, K.; et al. Exosomal circRNA derived from gastric tumor promotes white adipose browning by targeting the miR-133/PRDM16 pathway. Int. J. Cancer 2019, 144, 2501–2515. [Google Scholar] [CrossRef]
  34. Zhang, L.; Song, X.; Chen, X.; Wang, Q.; Zheng, X.; Wu, C.; Jiang, J. Circular RNA CircCACTIN Promotes Gastric Cancer Progression by Sponging MiR-331-3p and Regulating TGFBR1 Expression. Int. J. Biol. Sci. 2019, 15, 1091–1103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Huang, S.; Zhang, X.; Guan, B.; Sun, P.; Hong, C.T.; Peng, J.; Tang, S.; Yang, J. A novel circular RNA hsa_circ_0008035 contributes to gastric cancer tumorigenesis through targeting the miR-375/YBX1 axis. Am. J. Transl. Res. 2019, 11, 2455–2462. [Google Scholar] [PubMed]
  36. Liu, J.; Song, S.; Lin, S.; Zhang, M.; Du, Y.; Zhang, D.; Xu, W.; Wang, H. Circ-SERPINE2 promotes the development of gastric carcinoma by sponging miR-375 and modulating YWHAZ. Cell Prolif. 2019, 52, e12648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Pan, H.; Pan, J.; Chen, P.; Gao, J.; Guo, D.; Yang, Z.; Ji, L.; Lv, H.; Guo, Y.; Xu, D. Circular RNA circUBA1 promotes gastric cancer proliferation and metastasis by acting as a competitive endogenous RNA through sponging miR-375 and regulating TEAD4. Cancer Lett. 2020, 19. [Google Scholar] [CrossRef]
  38. Lai, Z.; Yang, Y.; Wang, C.; Yang, W.; Yan, Y.; Wang, Z.; Xu, J.; Jiang, K. Circular RNA 0047905 acts as a sponge for microRNA4516 and microRNA1227-5p, initiating gastric cancer progression. Cell Cycle 2019, 18, 1560–1572. [Google Scholar] [CrossRef]
  39. Wang, Q.; Wang, T.; Hu, Y.; Jiang, W.; Lu, C.; Zheng, W.; Zhang, W.; Chen, Z.; Cao, H. Circ-EIF4G3 promotes the development of gastric cancer by sponging miR-335. Pathol. Res. Pract. 2019, 215, 152507. [Google Scholar] [CrossRef]
  40. Wang, S.; Zhang, X.; Li, Z.; Wang, W.; Li, B.; Huang, X.; Sun, G.; Xu, J.; Li, Q.; Xu, Z.; et al. Circular RNA profile identifies circOSBPL10 as an oncogenic factor and prognostic marker in gastric cancer. Oncogene 2019, 38, 6985–7001. [Google Scholar] [CrossRef]
  41. Huang, X.; Li, Z.; Zhang, Q.; Wang, W.; Li, B.; Wang, L.; Xu, Z.; Zeng, A.; Zhang, X.; Zhang, X.; et al. Circular RNA AKT3 upregulates PIK3R1 to enhance cisplatin resistance in gastric cancer via miR-198 suppression. Mol. Cancer 2019, 18, 71. [Google Scholar] [CrossRef]
  42. Zhu, Z.; Rong, Z.; Luo, Z.; Yu, Z.; Zhang, J.; Qiu, Z.; Huang, C. Circular RNA circNHSL1 promotes gastric cancer progression through the miR-1306-3p/SIX1/vimentin axis. Mol. Cancer 2019, 18, 126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Cai, J.; Chen, Z.; Wang, J.; Wang, J.; Chen, X.; Liang, L.; Huang, M.; Zhang, Z.; Zuo, X. circHECTD1 facilitates glutaminolysis to promote gastric cancer progression by targeting miR-1256 and activating β-catenin/c-Myc signaling. Cell Death Dis. 2019, 10, 576. [Google Scholar] [CrossRef] [PubMed]
  44. Wu, L.; Liu, D.; Yang, Y. Enhanced expression of circular RNA circ-DCAF6 predicts adverse prognosis and promotes cell progression via sponging miR-1231 and miR-1256 in gastric cancer. Exp. Mol. Pathol. 2019, 110, 104273. [Google Scholar] [CrossRef] [PubMed]
  45. Du, W.; Li, D.; Guo, X.; Li, P.; Li, X.; Tong, S.; Tong, J.; Kuang, L.; Liang, D. Circ-PRMT5 promotes gastric cancer progression by sponging miR-145 and miR-1304 to upregulate MYC. Artif. Cells Nanomed. Biotechnol. 2019, 47, 4120–4130. [Google Scholar] [CrossRef]
  46. Zhang, Z.; Wang, C.; Zhang, Y.; Yu, S.; Zhao, G.; Xu, J. CircDUSP16 promotes the tumorigenesis and invasion of gastric cancer by sponging miR-145-5p. Gastric Cancer 2020, 23, 437–448. [Google Scholar] [CrossRef] [Green Version]
  47. Sun, B.; Sun, H.; Wang, Q.; Wang, X.; Quan, J.; Dong, D.; Lun, Y. Circular RNA circMAN2B2 promotes growth and migration of gastric cancer cells by down-regulation of miR-145. J. Clin. Lab. Anal. 2020, 5, e23215. [Google Scholar] [CrossRef]
  48. Sun, H.; Xi, P.; Sun, Z.; Wang, Q.; Zhu, B.; Zhou, J.; Jin, H.; Zheng, W.; Tang, W.; Cao, H.; et al. Circ-SFMBT2 promotes the proliferation of gastric cancer cells through sponging miR-182-5p to enhance CREB1 expression. Cancer Manag. Res. 2018, 10, 5725–5734. [Google Scholar] [CrossRef] [Green Version]
  49. Huang, X.X.; Zhang, Q.; Hu, H.; Jin, Y.; Zeng, A.L.; Xia, Y.B.; Xu, L. A novel circular RNA circFN1 enhances cisplatin resistance in gastric cancer via sponging miR-182-5p. J. Cell. Biochem. 2020, 1–12. [Google Scholar] [CrossRef]
  50. Mo, W.L.; Jiang, J.T.; Zhang, L.; Lu, Q.C.; Li, J.; Gu, W.D.; Cheng, Y.; Wang, H.T. Circular RNA hsa_circ_0000467 Promotes the Development of Gastric Cancer by Competitively Binding to MicroRNA miR-326-3p. Biomed. Res. Int. 2020, 2020, 4030826. [Google Scholar] [CrossRef]
  51. Liu, Y.; Yao, K.; Zhang, K.; Wang, J.; Daic, Q.; Wangd, R. Circular RNA PTK2 modifies the progression and radiosensitivity in gastric cancer via miR-369-3p/ZEB1 axis. RSC Adv. 2020, 10, 1711. [Google Scholar] [CrossRef] [Green Version]
  52. Lu, J.; Wang, Y.H.; Yoon, C.; Huang, X.Y.; Xu, Y.; Xie, J.W.; Wang, J.B.; Lin, J.X.; Chen, Q.Y.; Cao, L.L.; et al. Circular RNA circ-RanGAP1 regulates VEGFA expression by targeting miR-877-3p to facilitate gastric cancer invasion and metastasis. Cancer Lett. 2020, 471, 38–48. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, Z.; Wu, H.; Chen, Z.; Li, G.; Liu, B. Circular RNA ATXN7 promotes the development of gastric cancer through sponging miR-4319 and regulating ENTPD4. Cancer Cell Int. 2020, 20, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Xun, J.; Wang, C.; Yao, J.; Gao, B.; Zhang, L. CircBANP acts as a sponge of let-7a to promote gastric cancer progression via the FZD5/Wnt/β-catenin pathway. RSC Adv. 2020, 10, 7221–7231. [Google Scholar] [CrossRef] [Green Version]
  55. Lu, J.; Wang, Y.H.; Huang, X.Y.; Xie, J.W.; Wang, J.B.; Lin, J.X.; Chen, Q.Y.; Cao, L.L.; Huang, C.M.; Zheng, C.H.; et al. circ-CEP85L suppresses the proliferation and invasion of gastric cancer by regulating NFKBIA expression via miR-942-5p. J. Cell Physiol. 2020, 1–13. [Google Scholar] [CrossRef] [PubMed]
  56. Deng, G.; Mou, T.; He, J.; Chen, D.; Lv, D.; Liu, H.; Yu, J.; Wang, S.; Li, G. Circular RNA circRHOBTB3 acts as a sponge for miR-654-3p inhibiting gastric cancer growth. J. Exp. Clin. Cancer Res. 2020, 39, 1. [Google Scholar] [CrossRef]
  57. Zhang, J.; Liu, H.; Hou, L.; Wang, G.; Zhang, R.; Huang, Y.; Chen, X.; Zhu, J. Circular RNA_LARP4 inhibits cell proliferation and invasion of gastric cancer by sponging miR-424-5p and regulating LATS1 expression. Mol. Cancer 2017, 16, 151. [Google Scholar] [CrossRef]
  58. Li, P.; Chen, H.; Chen, S.; Mo, X.; Li, T.; Xiao, B.; Yu, R.; Guo, J. Circular RNA 0000096 affects cell growth and migration in gastric cancer. Br. J. Cancer 2017, 116, 626–633. [Google Scholar] [CrossRef] [Green Version]
  59. Zhang, Y.; Liu, H.; Li, W.; Yu, J.; Li, J.; Shen, Z.; Ye, G.; Qi, X.; Li, G. CircRNA_100269 is downregulated in gastric cancer and suppresses tumor cell growth by targeting miR-630. Aging 2017, 9, 1585–1594. [Google Scholar] [CrossRef] [Green Version]
  60. Chang, P.; Wang, F.; Li, Y. Hsa_circ_0000673 is down-regulated in gastric cancer and inhibits the proliferation and invasion of tumor cells by targetting miR-532-5p. Biosci. Rep. 2018, 38. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, T.; Liu, S.; Xu, Y.; Shu, R.; Wang, F.; Chen, C.; Zeng, Y.; Luo, H. Circular RNA-ZFR inhibited cell proliferation and promoted apoptosis in gastric cancer by sponging miR-130a/miR-107 and modulating PTEN. Cancer Res. Treat. 2018, 50, 1396–1417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Dai, X.; Guo, X.; Liu, J.; Cheng, A.; Peng, X.; Zha, L.; Wang, Z. Circular RNA circGRAMD1B inhibits gastric cancer progression by sponging miR-130a-3p and regulating PTEN and p21 expression. Aging 2019, 11, 9689–9708. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, H.; Liu, Y.; Bian, Z.; Zhang, J.; Zhang, R.; Chen, X.; Huang, Y.; Wang, Y.; Zhu, J. Circular RNA YAP1 inhibits the proliferation and invasion of gastric cancer cells by regulating the miR-367-5p/p27Kip1 axis. Mol. Cancer 2018, 17, 151. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, L.H.; Wang, L.P.; Ma, X.Q. Circ_SPECC1 enhances the inhibition of miR-526b on downstream KDM4A/YAP1 pathway to regulate the growth and invasion of gastric câncer cells. Biochem. Biophys. Res. Commun. 2019, 517, 253–259. [Google Scholar] [CrossRef]
  65. Zhong, S.; Wang, J.; Hou, J.; Zhang, Q.; Xu, H.; Hu, J.; Zhao, J.; Feng, J. Circular RNA hsa_circ_0000993 inhibits metastasis of gastric cancer cells. Epigenomics 2018, 10, 1301–1313. [Google Scholar] [CrossRef]
  66. Li, Q.; Tang, H.; Hu, F.; Qin, C. Circular RNA SMARCA5 inhibits gastric câncer progression through targeting the miR-346/ FBXL2 axis. RSC Adv. 2019, 9, 18277. [Google Scholar] [CrossRef] [Green Version]
  67. Rong, D.; Lu, C.; Zhang, B.; Fu, K.; Zhao, S.; Tang, W.; Cao, H. CircPSMC3 suppresses the proliferation and metastasis of gastric cancer by acting as a competitive endogenous RNA through sponging miR-296-5p. Mol. Cancer 2019, 18, 25. [Google Scholar] [CrossRef] [Green Version]
  68. Lu, J.; Zhang, P.Y.; Li, P.; Xie, J.W.; Wang, J.B.; Lin, J.X.; Chen, Q.Y.; Cao, L.L.; Huang, C.M.; Zheng, C.H. Circular RNA hsa_circ_0001368 suppresses the progression of gastric cancer by regulating miR-6506-5p/FOXO3 axis. Biochem. Biophys. Res. Commun. 2019, 512, 29–33. [Google Scholar] [CrossRef]
  69. Fang, J.; Hong, H.; Xue, X.; Zhu, X.; Jiang, L.; Qin, M.; Liang, H.; Gao, L. A novel circular RNA, circFAT1(e2), inhibits gastric cancer progression by targeting miR-548g in the cytoplasm and interacting with YBX1 in the nucleus. Cancer Lett. 2019, 442, 222–232. [Google Scholar] [CrossRef] [PubMed]
  70. Ding, L.; Zhao, Y.; Dang, S.; Wang, Y.; Li, X.; Yu, X.; Li, Z.; Wei, J.; Liu, M.; Li, G. Circular RNA circ-DONSON facilitates gastric cancer growth and invasion via NURF complex dependent activation of transcription factor SOX4. Mol. Cancer 2019, 18, 45. [Google Scholar] [CrossRef] [PubMed]
  71. Balzeau, J.; Menezes, M.R.; Cao, S.; Hagan, J.P. The LIN28/let-7 Pathway in Cancer. Front. Genet. 2017, 8, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhang, J.; Xu, A.; Miao, C.; Yang, J.; Gu, M.; Song, N. Prognostic value of Lin28A and Lin28B in various human malignancies: A systematic review and meta-analysis. Cancer Cell Int. 2019, 19, 79. [Google Scholar] [CrossRef] [PubMed]
  73. Li, J.; Ding, Y.; Li, A. Identification of COL1A1 and COL1A2 as candidate prognostic factors in gastric cancer. World J. Surg. Oncol. 2016, 14, 297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Sengupta, S.; den Boon, J.Á.; Chen, I.H.; Newton, M.A.; Stanhope, S.A.; Cheng, Y.J.; Chen, C.J.; Hildesheim, A.; Sugden, B.; Ahlquist, P. MicroRNA 29c is down-regulated in nasopharyngeal carcinomas, up-regulating mRNAs encoding extracellular matrix proteins. Proc. Natl. Acad. Sci. USA 2008, 105, 5874–5878. [Google Scholar] [CrossRef] [Green Version]
  75. Moulin, D.; Salone, V.; Koufany, M.; Clément, T.; Behm-Ansmant, I.; Branlant, C.; Charpentier, B.; Jouzeau, J.Y. MicroRNA-29b Contributes to Collagens Imbalance in Human Osteoarthritic and Dedifferentiated Articular Chondrocytes. Biomed. Res. Int. 2017, 2017, 9792512. [Google Scholar] [CrossRef]
  76. Kriegel, A.J.; Liu, Y.; Fang, Y.; Ding, X.; Liang, M. The miR-29 family: Genomics, cell biology, and relevance to renal and cardiovascular injury. Physiol. Genom. 2012, 44, 237–244. [Google Scholar] [CrossRef]
  77. Vidal, A.F.; Cruz, A.M.; Magalhães, L.; Pereira, A.L.; Anaissi, A.K.; Alves, N.C.; Albuquerque, P.J.; Burbano, R.M.; Demachki, S.; Ribeiro-dos-Santos, Â. hsa-miR-29c and hsa-miR-135b differential expression as potential biomarker of gastric carcinogenesis. World J. Gastroenterol. 2016, 22, 2060–2070. [Google Scholar] [CrossRef]
  78. Wang, T.; Hou, J.; Jian, S.; Luo, Q.; Wei, J.; Li, Z.; Wang, X.; Bai, P.; Duan, B.; Xing, J.; et al. miR-29b negatively regulates MMP2 to impact gastric cancer development by suppress gastric cancer cell migration and tumor growth. J. Cancer 2018, 9, 3776–3786. [Google Scholar] [CrossRef] [Green Version]
  79. Zhang, Z.; Li, Z.; Li, Y.; Zang, A. MicroRNA and signaling pathways in gastric cancer. Cancer Gene Ther. 2014, 21, 305–316. [Google Scholar] [CrossRef] [Green Version]
  80. Riquelme, I.; Saavedra, K.; Espinoza, J.A.; Weber, H.; García, P.; Nervi, B.; Garrido, M.; Corvalán, A.H.; Roa, J.C.; Bizama, C. Molecular classification of gastric cancer: Towards a pathway-driven targeted therapy. Oncotarget 2015, 6, 24750–24779. [Google Scholar] [CrossRef] [Green Version]
  81. Rezalotfi, A.; Ahmadian, E.; Aazami, H.; Solgi, G.; Ebrahimi, M. Gastric Cancer Stem Cells Effect on Th17/Treg Balance; A Bench to Beside Perspective. Front. Oncol. 2019, 9, 226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Slattery, M.L.; Mullany, L.E.; Sakoda, L.C.; Wolff, R.K.; Stevens, J.R.; Samowitz, W.S.; Herrick, J.S. The PI3K/AKT Signaling Pathway: Associations of miRNAs With Dysregulated Gene Expression in Colorectal Cancer. Mol. Carcinog. 2018, 57, 243–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Spangle, J.M.; Roberts, T.M.; Zhao, J.J. The Emerging Role of PI3K/AKT-mediated Epigenetic Regulation in Cancer. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 123–131. [Google Scholar] [CrossRef] [PubMed]
  84. Yang, Q.; Jiang, W.; Hou, P. Emerging Role of PI3K/AKT in Tumor-Related Epigenetic Regulation. Semin. Cancer Biol. 2019, 59, 112–124. [Google Scholar] [CrossRef] [PubMed]
  85. Daragmeh, J.; Barriah, W.; Saad, B.; Zaid, H. Analysis of PI3K pathway components in human cancers. Oncol. Lett. 2016, 11, 2913–2918. [Google Scholar] [CrossRef] [Green Version]
  86. Engelman, J.A.; Luo, J.; Cantley, L.C. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat. Rev. Genet. 2006, 7, 606–619. [Google Scholar] [CrossRef]
  87. Carnero, A.; Paramio, J.M. The PTEN/PI3K/AKT Pathway in vivo, Cancer Mouse Models. Front. Oncol. 2014, 4, 252. [Google Scholar] [CrossRef] [Green Version]
  88. Zhang, Y.; Wang, S.; Qian, W.; Ji, D.; Wang, Q.; Zhang, Z.; Wang, S.; Ji, B.; Fu, Z.; Sun, Y. uc.338 targets p21 and cyclin D1 via PI3K/AKT pathway activation to promote cell proliferation in colorectal cancer. Oncol. Rep. 2018, 40, 1119–1128. [Google Scholar] [CrossRef]
  89. Teng, R.; Hu, Y.; Zhou, J.; Seifer, B.; Chen, Y.; Shen, J.; Wang, L. Overexpression of Lin28 Decreases the Chemosensitivity of Gastric Cancer Cells to Oxaliplatin, Paclitaxel, Doxorubicin, and Fluorouracil in Part via microRNA-107. PLoS ONE 2015, 10, e0143716. [Google Scholar] [CrossRef]
  90. Heo, I.; Joo, C.; Kim, Y.-K.; Há, M.; Yoon, M.-J.; Cho, J.; Yeom, K.-H.; Han, J.; Kim, V.N. TUT4 in Concert with Lin28 Suppresses MicroRNA Biogenesis through Pre-MicroRNA Uridylation. Cell 2009, 138, 696–708. [Google Scholar] [CrossRef] [Green Version]
  91. Huang, Y.W.; Ruiz, C.R.; Eyler, E.C.; Lin, K.; Meffert, M.K. Dual regulation of miRNA biogenesis generates target specificity in neurotrophin-induced protein synthesis. Cell 2012, 148, 933–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Santos, N.P.; Ribeiro-Rodrigues, E.M.; Ribeiro-Dos-Santos, A.K.; Pereira, R.; Gusmão, L.; Amorim, A.; Guerreiro, J.F.; Zago, M.A.; Matte, C.; Hutz, M.H.; et al. Assessing individual interethnic admixture and population substructure using a 48-insertion-deletion (INSEL) ancestry-informative marker (AIM) panel. Hum. Mutat. 2010, 31, 184–190. [Google Scholar] [CrossRef] [PubMed]
  93. Andrade, R.B.; Amador, M.A.T.; Cavalcante, G.C.; Leitão, L.P.C.; Fernandes, M.R.; Modesto, A.A.C.; Moreira, F.C.; Khayat, A.S.; Assumpção, P.P.; Ribeiro-dos-Santos, Â.; et al. Estimating Asian contribution to the Brazilian population: A new application of a validated set of 61 ancestry informative markers. G3 2018, 8, 3577–3582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Pinto, P.S.C.; Santos, N.P.; Santos, S.; Ribeiro-dos-Santos, Â. Influence of genetic ancestry on INDEL markers of NFKb1, CASP8, PAR1, IL4 and CYP19A1 genes in leprosy patients. PLoS Negl. Trop. Dis. 2015, 9, e0004050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Nédélec, Y.; Sanz, J.; Baharian, G.; Szpiech, Z.A.; Pacis, A.; Dumaine, A.; Grenier, J.C.; Freiman, A.; Sams, A.J.; Hebert, S.; et al. Genetic Ancestry and Natural Selection Drive Population Differences in Immune Responses to Pathogens. Cell 2016, 167, 657–669. [Google Scholar] [CrossRef]
  96. Stagsted, L.V.W.; Nielsen, K.M.; Daugaard, I.; Hansen, T.B. Noncoding AUG circRNAs constitute an abundant and conserved subclass of circles. Life Sci. Alliance 2019, 2. [Google Scholar] [CrossRef] [Green Version]
  97. Branco, P.R.; de Araújo, G.S.; Barrera, J.; Suarez-Kurtz, G.; Souza, S.J. Uncovering association networks through an eQTL analysis involving human miRNAs and lincRNAs. Sci. Rep. 2018, 8, 15050. [Google Scholar] [CrossRef]
  98. Falzone, L.; Scola, L.; Zanghì, A.; Biondi, A.; Di Cataldo, A.; Libra, M.; Candido, S. Integrated Analysis of Colorectal Cancer microRNA Datasets: Identification of microRNAs Associated With Tumor Development. Aging 2018, 10, 1000–1014. [Google Scholar] [CrossRef]
  99. Qi, D.; Wang, Q.; Wu, M.; Zhang, X. Comprehensive bioinformatics analysis of lncRNAs in gastric cancer. Oncol. Lett. 2019, 17, 1279–1291. [Google Scholar] [CrossRef] [Green Version]
  100. Chandrashekar, D.S.; Bashel, B.; Balasubramanya, S.A.H.; Creighton, C.J.; Rodriguez, I.P.; Chakravarthi, B.V.S.K.; Varambally, S. UALCAN: A portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia 2017, 19, 649–658. [Google Scholar] [CrossRef]
  101. Hamberg, M.; Backes, C.; Fehlmann, T.; Hart, M.; Meder, B.; Meese, E.; Keller, A. MiRTargetLink-miRNAs, Genes and Interaction Networks. Int. J. Mol. Sci. 2016, 17, 564. [Google Scholar] [CrossRef] [PubMed]
  102. Huang, H.Y.; Lin, Y.C.; Li, J.; Huang, K.Y.; Shrestha, S.; Hong, H.C.; Tang, Y.; Chen, Y.G.; Jin, C.N.; Yu, Y.; et al. miRTarBase 2020: Updates to the experimentally validated microRNA-target interaction database. Nucleic Acids Res. 2020, 48, D148–D154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yu, G.; Wang, L.; Han, Y.; He, Q. clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS 2012, 16, 284–287. [Google Scholar] [CrossRef] [PubMed]
  104. Yu, G.; He, Q. ReactomePA: An R/Bioconductor package for reactome pathway analysis and visualization. Mol. BioSyst. 2016, 12, 477–479. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart showing an overview of our study design and results. After selecting the 54 sponge circRNAs, we divided our study by analyzing their host genes (Analysis 1), target miRNAs (Analysis 2) and target RNA-Binding Proteins (RBPs, Analysis 3). Each analysis was performed using different filters.
Figure 1. Flowchart showing an overview of our study design and results. After selecting the 54 sponge circRNAs, we divided our study by analyzing their host genes (Analysis 1), target miRNAs (Analysis 2) and target RNA-Binding Proteins (RBPs, Analysis 3). Each analysis was performed using different filters.
Cancers 12 01982 g001
Figure 2. Gastric cancer (GC)-related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways in which the sponged miRNAs’ target genes are potentially involved.
Figure 2. Gastric cancer (GC)-related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways in which the sponged miRNAs’ target genes are potentially involved.
Cancers 12 01982 g002
Figure 3. Cancer KEGG pathways in which the sponged miRNAs’ target genes are potentially involved. (*) Gastrointestinal tumors.
Figure 3. Cancer KEGG pathways in which the sponged miRNAs’ target genes are potentially involved. (*) Gastrointestinal tumors.
Cancers 12 01982 g003
Figure 4. Reactome pathways for which the sponge circRNAs’ target RBPs are enriched.
Figure 4. Reactome pathways for which the sponge circRNAs’ target RBPs are enriched.
Cancers 12 01982 g004
Figure 5. Graphic representation of signaling pathways that promote gastric cancer. (A) circRNA-miRNA interactions that affect the PI3K/Akt signaling pathway in gastric cancer. (B) circHIPK3/miR-107/Lin28-mediated chemoresistance of gastric cancer cells.
Figure 5. Graphic representation of signaling pathways that promote gastric cancer. (A) circRNA-miRNA interactions that affect the PI3K/Akt signaling pathway in gastric cancer. (B) circHIPK3/miR-107/Lin28-mediated chemoresistance of gastric cancer cells.
Cancers 12 01982 g005
Table 1. Description of the sponge circular RNAs (circRNAs) in gastric cancer and their target microRNAs (miRNAs).
Table 1. Description of the sponge circular RNAs (circRNAs) in gastric cancer and their target microRNAs (miRNAs).
Sponge circRNAExpressionHost GeneExpressionSponged miRNARef.
hsa_circ_0001821 (circPVT1)UPPVT1UP *miR-124-3p; miR-125b-5p[14,15]
hsa_circ_0000284 (circHIPK3)UPHIPK3UP *miR-124-3p; miR-29b-3p; miR-107[16,17]
hsa_circ_0001946 (ciRS-7)UPCDR1DOWN *miR-7-5p[18]
hsa_circ_0064644 (circRBMS3)UPRBMS3DOWN *miR-153[19]
hsa_circ_0056618UPSPOPL, UPUPmiR-206[20]
hsa_circ_0027599DOWNPHLDA1UPmiR-101-3p.1[21]
hsa_circ_0007766 (circ_ERBB2)UPERBB2UPmiR-503; miR-637[22]
hsa_circ_0000267UPFAM53BUPmiR-503-5p[23]
hsa_circ_0089548 (circ-NOTCH1)UPNOTCH1UPmiR-637[24]
hsa_circ_0089547 (circ-NOTCH1)UPNOTCH1UPmiR-449c-5p[25]
hsa_circ_0067997UPFNDC3BUPmiR-515-5p[26]
hsa_circ_0004771 (circ_NRIP1)UPNRIP1UPmiR-149-5p[27]
hsa_circ_0017728UPDHTKD1UPmiR-149[28]
hsa_circ_0081143UPCOL1A2UPmiR-646[29]
hsa_circ_0042881 (circNF1)UPNF1UPmiR-16[30]
hsa_circ_0032627 (circDLST)UPDLSTUPmiR-502-5p[31]
hsa_circ_0093398 (circPDSS1)UPPDSS1UPmiR-186-5p[32]
hsa_circ_0010522 (ciRS-133)UPRAP1GAPDOWN *miR-133[33]
hsa_circ_0092303 (circCACTIN)UPC19orf29UPmiR-331-3p[34]
hsa_circ_0008035UPEXT1UPmiR-375[35]
hsa_circ_0008365 (circ-SERPINE2)UPSERPINE2UPmiR-375[36]
hsa_circ_0090410 (circUBA1)UPUBA1UPmiR-375[37]
hsa_circ_0047905UPSERPINB5UP *miR-4516; miR-1227-5p[38]
hsa_circ_0005075 (circ-EIF4G3)UPEIF4G3UPmiR-335[39]
hsa_circ_0008549 (circOSBPL10)UPOSBPL10UPmiR-136-5p[40]
hsa_circ_0000199 (circAKT3)UPAKT3UP *miR-198[41]
hsa_circ_0006835 (circNHSL1)UPNHSL1UP *miR-1306-3p[42]
hsa_circHECTD1UPHECTD1UP *miR-1256[43]
hsa_circ_0009109 (circ-DCAF6)UPDCAF6UPmiR-1231; miR-1256[44]
hsa_circ_0031250 (circ-PRMT5)UPPRMT5UPmiR-145; miR-1304[45]
hsa_circ_0003855 (circDUSP16)UPDUSP16UPmiR-145-5p[46]
hsa_circ_0069086 (circMAN2B2)UPMAN2B2UPmiR-145[47]
hsa_circ_0017639 (circ-SFMBT2)UPSFMBT2UPmiR-182-5p[48]
hsa_circ_0058147 (circFN1)UPFN1UPmiR-182-5p[49]
hsa_circ_0000467UPSKA3UPmiR-326- 3p[50]
hsa_circ_0003221 (circPTK2)UPPTK2UPmiR-369-3p[51]
hsa_circ_0063526 (circ-RanGAP1)UPRANGAP1UPmiR-877-3p[52]
hsa_circ_0066436 (circATXN7)UPATXN7UPmiR-4319[53]
hsa_circ_0040809 (circBANP)UPBANPUPmiR-let-7a[54]
hsa_circ_0077736 (circ-CEP85L)DOWNCEP85L (C6ORF204)DOWN *miR-942-5p[55]
hsa_circ_00074444 (circRHOBTB3)DOWNRHOBTB3UP *miR-654-3p[56]
hsa_circ_101057 (circLARP4)DOWNLARP4UPmiR-424-5p[57]
hsa_circ_0000096DOWNHIAT1UPmiR-224-5p; miR-200a-3p[58]
hsa_circ_0013048 (hsa_circ_100269)DOWNLPHN2UPmiR-630[59]
hsa_circ_0000673DOWNRSL1D1UPmiR-532-5p[60]
hsa_circ_0072088 (circ-ZFR)DOWNZFRUPmiR-107; miR-130a-3p[61]
hsa_circ_0004798 (circGRAMD1B)DOWNGRAMD1BDOWN *miR-130a-3p[62]
hsa_circ_0002320 (circYAP)DOWNYAP1UPmiR-367-5p[63]
hsa_circ_0000745 (circ_SPECC1)DOWNSPECC1 (CYTSB)UPmiR-526b[64]
hsa_circ_0000993DOWNATL2DOWN *miR-214-5p[65]
hsa_circ_0001445 (circSMARCA5)DOWNSMARCA5UPmiR-346[66]
hsa_circ_0021977 (circPSMC3)DOWNPSMC3UPmiR-296-5p[67]
hsa_circ_0001368DOWNKLHL24UPmiR-6506-5p[68]
hsa_circ_0001461 (circFAT1(e2))DOWNFAT1UPmiR-548g[69]
(*) Statistically not significant. UP: Upregulated; DOWN: Downregulated; Ref.: References.
Table 2. circRNA-miRNA interactions that affect PI3K/Akt signaling pathway in gastric cancer.
Table 2. circRNA-miRNA interactions that affect PI3K/Akt signaling pathway in gastric cancer.
circRNA/miRNA Interaction [Ref.]Effect in Gene and/or Protein [Ref.]Effect in Gastric Cancer [Ref.]
ciRS-7↑/miR-7↓ [18]PTEN↓, PI3K↑ and pAkt↑ [18]Apoptosis inhibition, migration stimulus and poor clinicopathological and survival [18]
circNRIP1↑/miR-149-5p↓ [27]AKT1↑ [27]Cell proliferation, migration and invasion and growth of tumor [27]
circGRAMD1B↓/miR-130a-3p↑ [62]PTEN↓ and p21↓ [62]Cell proliferation, migration and invasion [62]
circRNA0047905↑ [38]AKT1/CREB↑ [38]Cell proliferation and tumor progression [38]
circMAN2B2↑/miR-145↓ [47]pPI3K↑ and pAkt ↑ [47]Cell growth and migration [47]
hsa_circ_0008035↑/miR-375↓ [35]PDK1↑ *Increased of pAkt (Proliferation, migration and invasion) *
circ-SERPINE2↑/miR-375↓ [36]PDK1↑ *
circUBA1↑/miR-375↓ [37]PDK1↑ *
(*) Mechanism proposed in this study. ↑: Upregulated; ↓: Downregulated; Ref.: References.

Share and Cite

MDPI and ACS Style

Pereira, A.L.; Magalhães, L.; Pantoja, R.P.; Araújo, G.; Ribeiro-dos-Santos, Â.; Vidal, A.F. The Biological Role of Sponge Circular RNAs in Gastric Cancer: Main Players or Coadjuvants? Cancers 2020, 12, 1982. https://doi.org/10.3390/cancers12071982

AMA Style

Pereira AL, Magalhães L, Pantoja RP, Araújo G, Ribeiro-dos-Santos Â, Vidal AF. The Biological Role of Sponge Circular RNAs in Gastric Cancer: Main Players or Coadjuvants? Cancers. 2020; 12(7):1982. https://doi.org/10.3390/cancers12071982

Chicago/Turabian Style

Pereira, Adenilson Leão, Leandro Magalhães, Rafael Pompeu Pantoja, Gilderlanio Araújo, Ândrea Ribeiro-dos-Santos, and Amanda Ferreira Vidal. 2020. "The Biological Role of Sponge Circular RNAs in Gastric Cancer: Main Players or Coadjuvants?" Cancers 12, no. 7: 1982. https://doi.org/10.3390/cancers12071982

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop