Open Access

circMTO1 sponges microRNA‑219a‑5p to enhance gallbladder cancer progression via the TGF‑β/Smad and EGFR pathways

  • Authors:
    • Pingfan Wang
    • Chenggang Zhou
    • Donghai Li
    • Dongsheng Zhang
    • Long Wei
    • Ying Deng
  • View Affiliations

  • Published online on: May 27, 2021     https://doi.org/10.3892/ol.2021.12824
  • Article Number: 563
  • Copyright: © Wang 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

Circular mitochondrial translation optimization 1 homologue (circMTO1) has been reported to regulate the tumorigenesis of different types of cancer; however, the role of circMTO1 in gallbladder cancer (GBC) remains unknown. The present study aimed to identify the potential miRNAs and target genes of circMTO1 during GBC progression, and clarify the regulatory mechanism between circMTO1 and miRNAs or target genes. The present study performed MTT and Transwell assays, and Annexin V staining to assess cell viability, migration and apoptosis, respectively. In addition, a lymphatic vessel formation assay was performed to assess tube formation of human dermal lymphatic endothelial cells (HDLECs), and GBC‑SD and NOZ cells. The results demonstrated that circMTO1 knockdown significantly attenuated the viability and migration of GBC cells and tube formation of HDLECs, and promoted apoptosis, indicating a tumor‑promoting role of circMTO1. In addition, transfection with microRNA (miRNA/miR)‑219a‑5p inhibitor rescued short hairpin RNA‑circMTO1‑inhibited tumorigenesis of GBC cells, suggesting that miR‑219a‑5p acts as a downstream effector for circMTO1. Mechanistically, transfection with miR‑219a‑5p mimic suppressed the expression levels of Smad2/4 and epidermal growth factor receptor. Analysis of The Cancer Genome Atlas datasets revealed that circMTO1 expression was associated with overall survival and the stage of patients with GBC. Taken together, the results of the present study provide novel insight for the role of circMTO1‑induced GBC tumorigenesis via regulation of miR‑219a‑5p expression.

Introduction

Gallbladder cancer (GBC) is considered the most common bile tract cancer (1), and patients with advanced disease have a 5-year survival rate of <5% (2). Given that there are no obvious symptoms and relevant biomarkers for GBC, patients are commonly diagnosed at advanced stages (3,4). Chemotherapy, radiotherapy and surgical resection remain the most common treatment strategies for GBC (5). Despite advancements in treatment approaches, the overall survival rate of patients with GBC remains low (5). Thus, it is important to determine the molecular mechanisms underlying GBC tumorigenesis.

Circular RNAs (circRNAs) are derived from endogenous non-coding RNAs with a closed loop structure (6). circRNAs have been reported to play important roles in different types of cancer by binding to microRNAs (miRNAs/miRs) (7). Circular mitochondrial translation optimization 1 homologue (circMTO1) inhibits progression of gastric cancer (8), colorectal cancer (9), bladder cancer (10) and glioblastoma (11). However, the role of circMTO1 in GBC remains unknown. Recently, Wang et al (12) demonstrated that circMTO1 acts as a biomarker in GBC. Shi et al (13) reported that overexpression of circMTO1 suppresses cell apoptosis via flow cytometry. Thus, it is important to determine the molecular mechanism underlying circMTO1-induced GBC tumorigenesis.

miRNAs are small non-coding RNAs of 20–22 nucleotides in length, which regulate gene expression at the post-transcriptional level (14). Notably, miRNAs are involved in tumor progression (15,16). Previous studies have reported that miR-219a-5p inhibits cancer cell proliferation and migration (17,18). In addition, miR-219a-5p can sensitize non-small cell lung cancer cells to cisplatin by regulating fibroblast growth factor 9 expression (17). miR-219a-5p promotes irradiation-induced apoptosis of non-small cell lung cancer cells (19). Furthermore, miR-219a-5p can also induce neuronal apoptosis (20). Based on these findings, it was hypothesized that miR-219a-5p acts as a potential effector for circMTO1 in GBC.

The present study aimed to determine the underlying molecular mechanism during GBC progression. In addition, rationales for diagnosing and treating GBC via the circMTO1-targeted approach are discussed.

Materials and methods

Cell culture

Human GBC-SD cells were purchased from Kunming Cell Bank, while NOZ cells were purchased from the Japanese Collection of Research Bioresources Cell Bank. Cells were maintained in DMEM supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (all purchased from Gibco; Thermo Fisher Scientific, Inc.), at 37°C with 5% CO2, according to the manufacturer's instructions. Human dermal lymphatic endothelial cells (HDLECs; PromoCell GmbH) and maintained in endothelial cell growth medium (PromoCell GmbH), at 37°C with 5% CO2.

Generating stable cell lines

To generate stable cell lines, 1 µg pLKO.1-short hairpin (sh)RNAs (Invitrogen; Thermo Fisher Scientific, Inc.), packaging vectors 0.5 µg pCMV–VSVG (Addgene, Inc.) and 0.5 µg pCMV-PAX2 (Addgene, Inc.) were introduced into 293T cells to generate 2nd lentiviruses. Lentiviruses were collected from the medium after 36–48 h of transfection and maintained in fresh media. We used lentiviruses (MOI=5) to infect into GBC-SD and NOZ cells for 24 h. Puromycin (2 µg/ml) was used to obtain pooled resistant cells after 24 h and 1 µg/ml of puromycin were used for maintenance. After 1 week, subsequent experiments were performed. The following sequences were used: sh-negative control (NC), 5′-AUCAGCCAAUCGGUCAACCUUC-3′; sh-circMTO1-1, 5′-GUGGGGUUGUUUUGGGUCAGA-3′; and sh-circMTO1-2, 5′-GUUGUUUUGGGUCAGAUGUCA-3′.

Cell transfection

miRNAs (50 µM; Shanghai GenePharma Co., Ltd.) were transfected into GBC-SD and NOZ cells using Lipofectamine® (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Following incubation for 48 h at 37°C, the cells were collected for subsequent analysis. The following sequences were used: NC mimic, 5′-CAUUCAUCCAUCAAUCGGGCAGGCCUUUAAGCUAACAUGGAA-3′; miR-219a-5p mimic, 5′-UGAUUGUCCAAACGCAAUUCUAAUUGCGUUUGGACAAUCAUU-3′; and NC inhibitor, 5′-UUCAGGCAAUCCAAAUGCAGG-3′; miR-219a-5p inhibitor, 5′-AGAAUUGCGUUUGGACAAUCA-3′.

StarBase and TargetScan prediction

The StarBase 2.0 online software (http://starbase.sysu.edu.cn/starbase2) created by Sun Yat-sen University was used to predict the potential interacting miRNAs. In addition, TargetScan v7.2 (http://www.targetscan.org) created by MIT was used to predict protein-coding genes as potential targets.

MTT assay

GBC-SD and NOZ cells were seeded into 96-well plates at a density of ~5,000 cells/well. MTT reagent was added to each well and incubated for 4 h at 37°C with 5% CO2. Following the MTT incubation, the purple formazan crystals were dissolved using 150 µl DMSO for 10 min and viability was subsequently analyzed at a wavelength of 490 nm.

Migration assay

GBC cells (1×105 cells) were suspended in 200 µl of DMEM media (Gibco; Thermo Fisher Scientific, Inc.). In this assay, the upper chambers had 8-µm-pore membranes and incubated for 4–6 h at 37°C. DMEM media (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 20% FBS was added to the lower chambers and incubated for 24 h at 37°C. Migratory cells were stained with 0.005% crystal violet dye for 2 h at room temperature and observed under an invert light microscope (magnification, ×200).

Immunofluorescence

GBC cells were fixed with 4% paraformaldehyde for 10 min at room temperature. Triton was subsequently used to permeabilize the cell membranes. Annexin V-FITC (eBioscience; Thermo Fisher Scientific, Inc.) was used to probe the signal, and cell nuclei were stained with DAPI at room temperature for 30 min and observed under a fluorescence microscope (magnification, ×200).

Western blotting

GBC cells were lysed using RIPA buffer and total proteins were extracted. The Rapid Gold BCA method (Thermo Fisher Scientific, Inc.) was used to determine protein concentrations, according to the manufacturer's protocol and ~50 µg total protein/lane was separated via SDS-PAGE on a 10% gel. The separated proteins were subsequently transferred onto PVDF membranes and blocked with 5% non-fat milk at room temperature for 1 h. The membranes were incubated with primary antibodies against Smad2 (cat. no. 5339), Smad4 (cat. no. 46535), epidermal growth factor receptor (EGFR; cat. no. 2085), cleaved caspase-3 (cat. no. 25546-1-AP) and GAPDH (cat. no. 8884) overnight at 4°C (all 1:1,000 and purchased from Cell Signaling Technology, Inc.). Following the primary incubation, membranes were incubated with anti-rabbit HRP-conjugated IgG (cat. no. 7074) and anti-mouse HRP-conjugated IgG (cat. no. 7076) secondary antibodies at room temperature for 1 h (both 1:2,000 and purchased from Cell Signaling Technology, Inc.). Protein bands were visualized using ECL reagents (Thermo Fisher Scientific, Inc.) and Bio-Rad gel imaging machine (Bio-Rad Laboratories, Inc.).

The Cancer Genome Atlas (TCGA) analysis

The gene expression data (RNAseq, NGS) of GBC were downloaded from TCGA dataset (36 tumor tissues and nine normal tissues, http://tcga.xenahubs.net). The present study did not have any inclusion or exclusion criteria. circMTO1 expression was analyzed using a two-tailed unpaired Student's t-test. Clinical characteristics of patients with gallbladder cancer were presented in Table I.

Table I.

Clinical characteristics of patients with gallbladder cancer (n=36).

Table I.

Clinical characteristics of patients with gallbladder cancer (n=36).

CharacteristicPatients, n
Age, years
  ≤6017
  >6019
Sex
  Male20
  Female16
Cholecystolithiasis
  Absent28
  Present8
Diabetes
  Absent12
  Present24
Jaundice
  Absent21
  Present15
Pathological type
  Adenocarcinoma36
  Adenosquamous carcinoma0
  Papillocarcinoma0
Degree of differentiation
  Poor22
  Moderate-well14
T stage
  T110
  T29
  T417
Patient sorting criterion

The patients were sorted into high and low expression groups according to the median circMTO1 RNA expression (cut-off value=2.8) in patients with GBC tumors. A total of 18 patients were classified into the high circMTO1 expression group and 18 into the low circMTO1 expression group.

Tube formation assay

GFR Matrigel was mixed with PBS (1:5) and coated in 24-well plates at 37°C for 4–5 h. HDLECs were subsequently seeded into the plates (12 wells) at a density of ~5,000 cells/well, and the tubes were observed under an inverted light microscope (magnification, ×200).

Reverse transcription-quantitative (RT-q)PCR

Total RNA was extracted from GBC cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Total RNA (1 µg) was reverse transcribed into cDNA using the PrimeScript RT reagent kit (Takara Bio, Inc.) at 37°C for 15 min and 85°C for 1 min. qPCR was subsequently performed to measure relative gene expression levels, using the 2−ΔΔCq method (21). SYBR Green dye (Roche Applied Science) was used for qPCR. The following thermocycling conditions were used: Initial denaturation at 95°C for 5 min (step 1), 95°C for 30 sec (step 2), 60°C for 30 sec (step 3), 72°C for 30 sec (step 4), 40 cycles (step 2–4). β-actin was used as the reference gene for circMTO1, Smad2, Smad4 and EGFR. U6 was used as the reference gene for miR-219a-5p. The following primer sequences were used for qPCR: circMTO1 forward, 5′-GCCTGAACACACTGGGAAAT-3′ and reverse, 5′-CACAGATGCGAGAACACAGG-3′; Smad2 forward, 5′-CTTTGTGCAGAGCCCCAATT-3′ and reverse, 5′-CTTGTTACCGTCTGCCTTCG-3′; Smad4 forward, 5′-TCCAGCCTCCCATTTCCAAT-3′ and reverse, 5′-ACCTTGCTCTCTCAATGGCT-3′; EGFR forward, 5′-AGGTGAAAACAGCTGCAAGG-3′ and reverse, 5′-AGGTGATGTTCATGGCCTGA-3′; miR-219a-5p forward, 5′-CTCCTGATTGTCCAAAC-3′ and reverse, 5′-CGCTCGAGGTTTGGGG-3′; β-actin forward, 5′-TGGCATCCACGAAAC TAC CT-3′ and reverse, 5′-TCTCCTTCTGCATCCTGTCG-3′; and U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′.

Statistical analysis

Statistical analysis was performed using GraphPad Prism 8.0 software (GraphPad Software, Inc.). Unpaired Student's t-test was used to compare differences between two groups, while one-way ANOVA followed by Tukey's post hoc test was used to compare differences between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

circMTO1 knockdown impairs GBC tumorigenesis

To determine the role of circMTO1 in GBC progression, circMTO1 expression was suppressed using shRNAs (sh-circMTO1-1 and sh-circMTO1-2). RT-qPCR analysis was performed to detect circMTO1 expression. The results demonstrated that circMTO1 expression significantly decreased (~70-80%) in circMTO1 knockdown cells compared with sh-NC cells (Fig. 1A) (GBC-SD, sh-circMTO1-1 vs. sh-NC, P=0.0002; sh-circMTO1-2 vs. sh-NC, P=0.0003; NOZ, sh-circMTO1-1 vs. sh-NC, P=0.0005; sh-circMTO1-2 vs. sh-NC, P=0.0006).

circMTO1 has been reported to promote tumorigenesis of cervical cancer (22). The MTT assay was performed to assess the viability of GBC-SD and NOZ cells transfected with sh-NC, sh-circMTO1-1 and sh-circMTO1-2. The results demonstrated that cells transfected with sh-circMTO1-1 or sh-circMTO1-2 exhibited evidently lower viability compared with sh-NC cells (Fig. 1B) (GBC-SD, sh-circMTO1-1 vs. sh-NC, P=0.0003; sh-circMTO1-2 vs. sh-NC, P=0.0004; NOZ, sh-circMTO1-1 vs. sh-NC, P=0.0002; sh-circMTO1-2 vs. sh-NC, P=0.0007). Annexin V staining was performed to assess apoptosis of GBC-SD and NOZ cells transfected with sh-NC, sh-circMTO1-1 or sh-circMTO1-2. The results indicated that circMTO1 knockdown increased the apoptotic rate of cells (Fig. 1C and D) (GBC-SD, sh-circMTO1-1 vs. sh-NC, P=0.0005; sh-circMTO1-2 vs. sh-NC, P=0.0063; NOZ, sh-circMTO1-1 vs. sh-NC, P=0.0008; sh-circMTO1-2 vs. sh-NC, P=0.036). To confirm, western blot analysis was performed to detect cleaved caspase-3 protein expression in GBC-SD and NOZ cells transfected with sh-NC, sh-circMTO1-1 or sh-circMTO1-2. The results demonstrated that cleaved caspase-3 protein expression was upregulated in circMTO1 knockdown cells (Fig. 1E). In addition, the effect of circMTO1 on tube formation of HDLECs was investigated. The results indicated that circMTO1 knockdown attenuated tube formation of HDLECs (Fig. 1F and G) (GBC-SD, sh-circMTO1-1 vs. sh-NC, P=0.0004; sh-circMTO1-2 vs. sh-NC, P=0.0003; NOZ, sh-circMTO1-1 vs. sh-NC, P=0.0007; sh-circMTO1-2 vs. sh-NC, P=0.0005). The results of the Transwell assay demonstrated that circMTO1 knockdown decreased the number of migratory GBC-SD cells (Fig. 1H). Taken together, these results suggest that circMTO1 acts as an oncogene in GBC progression.

miR-219a-5p inhibitor partially restores sh-circMTO1-attenuated GBC tumorigenesis

The StarBase database was used to determine the potential molecular mechanism underlying circMTO1-induced tumorigenesis of GBC. Analysis revealed that miR-219a-5p is an interacting molecule for circMTO1 in GBC (Fig. 2A). RT-qPCR analysis demonstrated that miR-219a-5p expression was substantially regulated in GBC-SD or NOZ cells transfected with NC mimic, miR-219a-5p mimic, NC inhibitor or miR-219a-5p inhibitor (Fig. 2B) (GBC-SD, miR-219a-5p mimic vs. NC mimic, P=0.0004; miR-219a-5p inhibitor vs. NC inhibitor, P=0.0005; NOZ, miR-219a-5p mimic vs. NC mimic, P=0.0003; miR-219a-5p inhibitor vs. NC inhibitor, P=0.0003).

Figure 2.

miR-219a-5p inhibitor partially restores sh-circMTO1-attenuated GBC tumorigenesis. (A) Bioinformatics analysis revealed that miR-219-5p is a potential target of circMTO1. (B) RT-qPCR analysis was performed to detect miR-219a-5p expression in GBC-SD and NOZ cells transfected with NC mimic, miR-219a-5p mimic, NC inhibitor or miR-219a-5p inhibitor. (C) RT-qPCR analysis was performed to detect miR-219a-5p expression in GBC-SD and NOZ cells transfected with sh-NC, sh-circMTO1-1 or sh-circMTO1-1 + miR-219a-5p inhibitor. (D) The MTT assay was performed to assess the viability of GBC-SD and NOZ cells transfected with sh-NC, sh-circMTO1-1 or sh-circMTO1-1 + miR-219a-5p inhibitor. (E) Western blot analysis was performed to detect cleaved caspase-3 protein expression in GBC-SD and NOZ cells transfected with sh-NC, sh-circMTO1-1 or sh-circMTO1-1 + miR-219a-5p inhibitor. GAPDH was used as the internal control. (F and G) Annexin V staining was performed to assess the apoptosis of GBC-SD and NOZ cells transfected with sh-NC, sh-circMTO1-1 or sh-circMTO1-1 + miR-219a-5p inhibitor. (H and I) The lymphatic vessel formation assay was performed to assess tube formation of human dermal lymphatic endothelial, GBC-SD and NOZ cells transfected with sh-NC, sh-circMTO1-1 or sh-circMTO1-1 + miR-219a-5p inhibitor (scale bar, 2 µm). (J) The Transwell assay was performed to assess the migratory ability of NOZ cells transfected with sh-NC, sh-circMTO1-1 or sh-circMTO1-1 + miR-219a-5p inhibitor. *P<0.05; **P<0.01; ***P<0.001. miR, microRNA; sh, short hairpin; circMTO1, circular mitochondrial translation optimization 1 homologue; GBC, gallbladder cancer; RT-qPCR, reverse transcription-quantitative PCR; NC, negative control.

The function of miR-219a-5p in circMTO1-regulated GBC phenotypes was investigated. RT-qPCR analysis demonstrated that miR-219a-5p expression was upregulated following circMTO1 knockdown (Fig. 2C) (GBC-SD, sh-circMTO1-1 vs. sh-NC, P=0.0002; sh-circMTO1-1 + miR-219a-5p inhibitor vs. sh-circMTO1-1, P=0.0006; NOZ, sh-circMTO1-1 vs. sh-NC, P=0.0067; sh-circMTO1-1 + miR-219a-5p inhibitor vs. sh-circMTO1-1, P=0.028). The results of the MTT assay demonstrated that transfection with miR-219a-5p inhibitor rescued the viability of GBC-SD or NOZ cells attenuated by circMTO1 knockdown (Fig. 2D) (GBC-SD, sh-circMTO1-1 vs. sh-NC, P=0.0005; sh-circMTO1-1 + miR-219a-5p inhibitor vs. sh-circMTO1-1, P=0.0036; NOZ, sh-circMTO1-1 vs. sh-NC, P=0.0007; sh-circMTO1-1 + miR-219a-5p inhibitor vs. sh-circMTO1-1, P=0.0083). Western blot analysis revealed that cleaved caspase-3 protein expression was upregulated in circMTO1 knockdown cells, the effects of which were reversed following transfection with miR-219a-5p inhibitor (Fig. 2E). In addition, Annexin V staining demonstrated that transfection with miR-219a-5p inhibitor decreased the apoptotic rate of GBC-SD or NOZ cells enhanced by circMTO1 knockdown (Fig. 2F and G) (GBC-SD, sh-circMTO1-1 vs. sh-NC, P=0.0072; sh-circMTO1-1 + miR-219a-5p inhibitor vs. sh-circMTO1-1, P=0.032; NOZ, sh-circMTO1-1 vs. sh-NC, P=0.0007; sh-circMTO1-1 + miR-219a-5p inhibitor vs. sh-circMTO1-1, P=0.014). Notably, transfection with miR-219a-5p inhibitor enhanced sh-circMTO1-modulated tube formation of HDLECs (Fig. 2H and I) (GBC-SD, sh-circMTO1-1 vs. sh-NC, P=0.0002; sh-circMTO1-1 + miR-219a-5p inhibitor vs. sh-circMTO1-1, P=0.0003; NOZ, sh-circMTO1-1 vs. sh-NC, P=0.0006; sh-circMTO1-1 + miR-219a-5p inhibitor vs. sh-circMTO1-1, P=0.0003). The results of the Transwell assay indicated that circMTO1 knockdown decreased migration of NOZ cells, which was reversed following transfection with miR-219a-5p inhibitor (Fig. 2J). Collectively, these results suggest that miR-219a-5p serves as a downstream effector for circMTO1 in GBC cells.

miR-219a-5p regulates the TGF-β/Smad pathway and EGFR expression

To further investigate the detailed downstream pathway responsible for miR-219a-5p-associated GBC progression, TargetScan software was used to detect Smad2/4 and EGFR expression. RT-qPCR analysis demonstrated that transfection with miR-219a-5p mimic inhibited the expression levels of Smad2, Smad4 and EGFR (Fig. 3A-C) (Fig. 3A, GBC-SD, miR-219a-5p mimic vs. NC mimic, P=0.0003; NOZ, miR-219a-5p mimic vs. NC mimic, P=0.0005. Fig. 3B, GBC-SD, miR-219a-5p mimic vs. NC mimic, P=0.0004; NOZ, miR-219a-5p mimic vs. NC mimic, P=0.0008. Fig. 3C, GBC-SD, miR-219a-5p mimic vs. NC mimic, P=0.0005; NOZ, miR-219a-5p mimic vs. NC mimic, P=0.0004). Similarly, western blot analysis demonstrated that transfection with miR-219a-5p mimic decreased the protein expression levels of Smad2, Smad4 and EGFR (Fig. 3D-G) (Fig. 3E, GBC-SD, miR-219a-5p mimic vs. NC mimic, P=0.0002; NOZ, miR-219a-5p mimic vs. NC mimic, P=0.0003. Fig. 3F, GBC-SD, miR-219a-5p mimic vs. NC mimic, P=0.0004; NOZ, miR-219a-5p mimic vs. NC mimic, P=0.0007. Fig. 3G, GBC-SD, miR-219a-5p mimic vs. NC mimic, P=0.0006; NOZ, miR-219a-5p mimic vs. NC mimic, P=0.0005). Taken together, these results suggest that miR-219a-5p modulates the TGF-β/Smad pathway and EGFR expression.

circMTO1 is closely associated with GBC progression

To further determine whether circMTO1 plays a role in GBC, GBC datasets were downloaded from TCGA database. As expected, the results demonstrated that circMTO1 expression was higher in patients with GBC compared with healthy individuals (P=0.018; Fig. 4A). Notably, patients with low circMTO1 expression had prolonged overall survival times than those with high circMTO1 expression (P=0.0069; Fig. 4B). In addition, patients with high circMTO1 expression were associated with advanced disease (stage II vs. stage I; P=0.024; stage IV vs. stage II, P=0.035; Fig. 4C). Finally, we proposed the working model for circMTO1-induced GBC progression via miR-219a-5p (Fig. 4D). Collectively, these results suggest that circMTO1 is closely associated with GBC progression.

Discussion

The results of the present study confirmed that circMTO1 interacts with miR-219a-5p to promote the TGF-β/Smad signaling pathway and EGFR expression in GBC cells. To the best of our knowledge, the present study was the first to demonstrate that circMTO1 may serve as an oncogene in patients with GBC. The results demonstrated that circMTO1 expression was dysregulated in GBC cells and tumors. In addition, circMTO1 was involved in GBC progression. Based on these findings, circMTO1 may serve as a biomarker or target for diagnosis and treatment of GBC.

circRNAs are generated by back-splicing of 3′ and 5′ splice sites (23). As circRNAs are ubiquitously expressed in several tissues, they play important roles across various biological processes (24,25). Several circRNAs, including circERBB2, circFOXP1 and circHIPK3, have been reported to regulate GBC progression (2628). Consistent with these findings, the results of the present study demonstrated that circMTO1 knockdown decreased cell viability and tube formation of HDLECs, while promoting the apoptosis of GBC cells. Notably, a few studies have reported that overexpression of circMTO1 enhances the apoptosis of gastric cancer cells and hepatoma cells (8,29). These findings oppose the results of the present study, thus it was hypothesized that circMTO1 may exert different roles in different types of cancer.

miRNAs play key roles in different types of human cancer by modulating gene expression levels (16). It has been reported that circRNAs sponge and decoy miRNAs in cancer cells (30). The results of the present study demonstrated that circMTO1 negatively regulated miR-219a-5p expression. Previous studies have verified that miR-92, miR-9 and miR-6893 are downstream effectors in different types of cancer (11,22,31). Thus, the results of the present study broaden the interacting spectrum of circMTO1. The results presented here demonstrated that miR-219a-5p inhibitor attenuated circMTO1 knockdown-induced apoptosis, which is consistent with previous studies (19,20). Long et al (32) reported that miR-219-5p targets B-cell lymphoma 2 (Bcl-2) to inhibit melanoma growth and metastasis. Thus, it was hypothesized that circMTO1 interacts with miR-219-5p to regulate GBC progression by targeting anti-apoptotic genes, such as Bcl-2.

TGF-β signaling plays a key role in tumor progression, including epithelial-to-mesenchymal transition (EMT), via Smads and the MAPK pathway (33). A recent study suggested that TGF-β/Smad signaling promotes GBC metastasis by upregulating the miR-182/CADM1 axis (34). Smads exert their roles via phosphorylation (35). The results of the present study demonstrated that miR-219a-5p decreased Smad2/4 expression levels at the transcriptional level. Thus, detection of Smad 2/4 phosphorylation levels were not necessary and significant. In addition, miR-219a-5p downregulated EGFR expression in GBC cells, which was consistent with a previous study, suggesting that miR-219a-5p regulates EGFR to affect EMT of ovarian cancer cells (36). Notably, Shen et al (37) demonstrated that activation of the EGFR signaling pathway promotes gallbladder cancer invasion and metastasis. The authors reported that PLEK2 interacts with the kinase domain of EGFR and suppresses EGFR ubiquitination by c-CBL, which results in constitutive activation of EGFR signaling and elevated expression of the downstream effector, CCL2.

In conclusion, the results of the present study suggest that circMTO1 contributes to GBC progression via the miR-219a-5p/Smad/EGFR axis. This novel mechanism can be used to develop targeted therapeutic strategies for patients with GBC. However, the present study is not without limitations. First, further studies are required to determine the role of circMTO1 in different models (other GBC cell lines and animal models). Secondly, although the present study demonstrated that miR-219a-5p negatively regulated TGF-β signaling and EGFR, further studies are required to determine whether other downstream targets are involved in the circMTO1/miR-219a-5p axis-modulated GBC progression.

Acknowledgements

Not applicable.

Funding

The present study was supported by Lanzhou Science and Technology Development Guide Project (grant no. 2019-ZD-21).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Authors' contributions

PW, CZ and YD designed the present study and performed the experiments. DL, LW and DZ analyzed the data and prepared the figures. PW and LW drafted the initial manuscript. DL, DZ and LW confirmed the authenticity of all the raw data. PW and YD reviewed and edited the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Shaffer EA: Gallbladder cancer: The basics. Gastroenterol Hepatol (N Y). 4:737–741. 2008.PubMed/NCBI

2 

Sachs TE, Akintorin O and Tseng J: How should gallbladder cancer be managed? Adv Surg. 52:89–100. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Kakaei F, Beheshtirouy S, Nejatollahi SM, Zarrintan S and Mafi MR: Surgical treatment of gallbladder carcinoma: A critical review. Updates Surg. 67:339–351. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Lazcano-Ponce EC, Miquel JF, Munoz N, Herrero R, Ferrecio C, Wistuba II, Alonso de Ruiz P, Aristi Urista G and Nervi F: Epidemiology and molecular pathology of gallbladder cancer. CA Cancer J Clin. 51:349–364. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Misra S, Chaturvedi A, Misra NC and Sharma ID: Carcinoma of the gallbladder. Lancet Oncol. 4:167–176. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Patop IL and Kadener S: circRNAs in cancer. Curr Opin Genet Dev. 48:121–127. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Guo JU, Agarwal V, Guo H and Bartel DP: Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 15:4092014. View Article : Google Scholar : PubMed/NCBI

8 

Song R, Li Y, Hao W, Yang L, Chen B, Zhao Y, Sun B and Xu F: Circular RNA MTO1 inhibits gastric cancer progression by elevating PAWR via sponging miR-199a-3p. Cell Cycle. 19:3127–3139. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Ge Z, Li LF, Wang CY, Wang Y and Ma WL: circMTO1 inhibits cell proliferation and invasion by regulating Wnt/β-catenin signaling pathway in colorectal cancer. Eur Rev Med Pharmacol Sci. 22:8203–8209. 2018.PubMed/NCBI

10 

Li Y, Wan B, Liu L, Zhou L and Zeng Q: Circular RNA circMTO1 suppresses bladder cancer metastasis by sponging miR-221 and inhibiting epithelial-to-mesenchymal transition. Biochem Biophys Res Commun. 508:991–996. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Zhang X, Zhong B, Zhang W, Wu J and Wang Y: Circular RNA circMTO1 inhibits proliferation of glioblastoma cells via miR-92/WWOX signaling pathway. Med Sci Monit. 25:6454–6461. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Wang X, Lin YK, Lu ZL and Li J: Circular RNA circ-MTO1 serves as a novel potential diagnostic and prognostic biomarker for gallbladder cancer. Eur Rev Med Pharmacol Sci. 24:8359–8366. 2020.PubMed/NCBI

13 

Shi CC, Pan LY, Peng ZY and Li JG: circMTO1 attenuated acute kidney injury through regulating miR-337. Inflammation. 43:1304–1311. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Guo H, Ingolia NT, Weissman JS and Bartel DP: Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 466:835–840. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Braicu C, Zimta AA, Harangus A, Iurca I, Irimie A, Coza O and Berindan-Neagoe I: The function of non-coding RNAs in lung cancer tumorigenesis. Cancers (Basel). 11:6052019. View Article : Google Scholar : PubMed/NCBI

16 

Boufraqech M, Nilubol N, Zhang L, Gara SK, Sadowski SM, Mehta A, He M, Davis S, Dreiling J, Copland JA, et al: miR30a inhibits LOX expression and anaplastic thyroid cancer progression. Cancer Res. 75:367–377. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Rao C, Miao X, Zhao G, Zhang C, Shen H, Dong C and Yang M: miR-219a-5p enhances cisplatin sensitivity of human non-small cell lung cancer by targeting FGF9. Biomed Pharmacother. 114:1086622019. View Article : Google Scholar : PubMed/NCBI

18 

Xu K, Shi J, Mo D, Yang Y, Fu Q and Luo Y: miR-219a-1 inhibits colon cancer cells proliferation and invasion by targeting MEMO1. Cancer Biol Ther. 21:1163–1170. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Wei T, Cheng S, Fu XN and Feng LJ: miR-219a-5p enhances the radiosensitivity of non-small cell lung cancer cells through targeting CD164. Biosci Rep. 40:BSR201927952020. View Article : Google Scholar : PubMed/NCBI

20 

Yan J, Bu X, Li Z, Wu J, Wang C, Li D, Song J and Wang J: Screening the expression of several miRNAs from TaqMan low density array in traumatic brain injury: miR-219a-5p regulates neuronal apoptosis by modulating CCNA2 and CACUL1. J Neurochem. 150:202–217. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Maruyama T, Nishihara K, Umikawa M, Arasaki A, Nakasone T, Nimura F, Matayoshi A, Takei K, Nakachi S, Kariya KI and Yoshimi N: MicroRNA-196a-5p is a potential prognostic marker of delayed lymph node metastasis in early-stage tongue squamous cell carcinoma. Oncol Lett. 15:2349–2363. 2018.PubMed/NCBI

22 

Chen M, Ai G, Zhou J, Mao M, Li H and Guo J: circMTO1 promotes tumorigenesis and chemoresistance of cervical cancer via regulating miR-6893. Biomed Pharmacother. 117:1090642019. View Article : Google Scholar : PubMed/NCBI

23 

Ashwal-Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, Hanan M, Evantal N, Memczak S, Rajewsky N and Kadener S: circRNA biogenesis competes with pre-mRNA splicing. Mol Cell. 56:55–66. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Li R, Jiang J, Shi H, Qian H, Zhang X and Xu W: circRNA: A rising star in gastric cancer. Cell Mol Life Sci. 77:1661–1680. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Yan Z, Xiao Y, Chen Y and Luo G: Screening and identification of epithelial-to-mesenchymal transition-related circRNA and miRNA in prostate cancer. Patho Res Pract. 216:1527842020. View Article : Google Scholar : PubMed/NCBI

26 

Huang X, He M, Huang S, Lin R, Zhan M, Yang D, Shen H, Xu S, Cheng W, Yu J, et al: Circular RNA circERBB2 promotes gallbladder cancer progression by regulating PA2G4-dependent rDNA transcription. Mol Cancer. 18:1662019. View Article : Google Scholar : PubMed/NCBI

27 

Kai D, Yannian L, Yitian C, Dinghao G, Xin Z and Wu J: Circular RNA HIPK3 promotes gallbladder cancer cell growth by sponging microRNA-124. Biochem Biophys Res Commun. 503:863–869. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Wang S, Zhang Y, Cai Q, Ma M, Jin LY, Weng M, Zhou D, Tang Z, Wang JD and Quan Z: Circular RNA FOXP1 promotes tumor progression and Warburg effect in gallbladder cancer by regulating PKLR expression. Mol Cancer. 18:1452019. View Article : Google Scholar : PubMed/NCBI

29 

Wang J, Tan Q, Wang W and Yu J: Mechanism of the regulatory effect of overexpression of circMTO1 on proliferation and apoptosis of hepatoma cells via miR-9-5p/NOX4 axis. Cancer Manag Res. 12:3915–3925. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Kristensen LS, Hansen TB, Veno MT and Kjems J: Circular RNAs in cancer: Opportunities and challenges in the field. Oncogene. 37:555–565. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Han D, Li J, Wang H, Su X, Hou J, Gu Y, Qian C, Lin Y, Liu X, Huang M, et al: Circular RNA circMTO1 acts as the sponge of microRNA-9 to suppress hepatocellular carcinoma progression. Hepatology. 66:1151–1164. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Long J, Menggen Q, Wuren Q, Shi Q and Pi X: miR-219-5p inhibits the growth and metastasis of malignant melanoma by targeting BCL-2. Biomed Res Int. 2017:90325022017. View Article : Google Scholar : PubMed/NCBI

33 

Bao RF, Shu YJ, Hu YP, Wang XA, Zhang F, Liang HB, Ye YY, Li HF, Xiang SS, Weng H, et al: miR-101 targeting ZFX suppresses tumor proliferation and metastasis by regulating the MAPK/Erk and Smad pathways in gallbladder carcinoma. Oncotarget. 7:22339–22354. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Qiu Y, Luo X, Kan T, Zhang Y, Yu W, Wei Y, Shen N, Yi B and Jiang X: TGF-β upregulates miR-182 expression to promote gallbladder cancer metastasis by targeting CADM1. Mol Biosyst. 10:679–685. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Syed V: TGF-β signaling in cancer. J Cell Biochem. 117:1279–1287. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Wang L, Yu M and Zhao S: lncRNA MEG3 modified epithelial-mesenchymal transition of ovarian cancer cells by sponging miR-219a-5p and regulating EGFR. J Cell Biochem. 120:17709–17722. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Shen H, He M, Lin R, Zhan M, Xu S, Huang X, Xu C, Chen W, Yao Y, Mohan M and Wang J: PLEK2 promotes gallbladder cancer invasion and metastasis through EGFR/CCL2 pathway. J Exp Clin Cancer Res. 38:2472019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2021
Volume 22 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
Wang P, Zhou C, Li D, Zhang D, Wei L and Deng Y: circMTO1 sponges microRNA‑219a‑5p to enhance gallbladder cancer progression via the TGF‑β/Smad and EGFR pathways. Oncol Lett 22: 563, 2021
APA
Wang, P., Zhou, C., Li, D., Zhang, D., Wei, L., & Deng, Y. (2021). circMTO1 sponges microRNA‑219a‑5p to enhance gallbladder cancer progression via the TGF‑β/Smad and EGFR pathways. Oncology Letters, 22, 563. https://doi.org/10.3892/ol.2021.12824
MLA
Wang, P., Zhou, C., Li, D., Zhang, D., Wei, L., Deng, Y."circMTO1 sponges microRNA‑219a‑5p to enhance gallbladder cancer progression via the TGF‑β/Smad and EGFR pathways". Oncology Letters 22.1 (2021): 563.
Chicago
Wang, P., Zhou, C., Li, D., Zhang, D., Wei, L., Deng, Y."circMTO1 sponges microRNA‑219a‑5p to enhance gallbladder cancer progression via the TGF‑β/Smad and EGFR pathways". Oncology Letters 22, no. 1 (2021): 563. https://doi.org/10.3892/ol.2021.12824