miR-132 inhibits cell proliferation, invasion and migration of hepatocellular carcinoma by targeting PIK3R3

Retraction in: /10.3892/ijo.2021.5238

  • Authors:
    • Kai Liu
    • Xingliang Li
    • Yuchen Cao
    • Yuanyuan Ge
    • Jianmeng Wang
    • Bo Shi
  • View Affiliations

  • Published online on: August 4, 2015     https://doi.org/10.3892/ijo.2015.3112
  • Pages: 1585-1593
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA-132 (miR‑132) has been reported to play a tumor suppressive role in different human malignancies. However, its role and underling mechanism in hepatocellular carcinoma (HCC) remains poorly defined due to lack of target gene information. In the present study, we demonstrated that the mean level of miR‑132 in hepatocellular carcinoma (HCC) tissues was significantly lower than that in matched tumor-adjacent tissues, and its expression negatively correlated with tumor differentiation (P<0.01), TNM stage (P<0.01) and lymph node metastasis (P<0.01). Similarly, the expression of miR‑132 was obviously reduced in HCC cell lines as compared with a normal hepatic cell line. Ectopic expression of miR‑132 inhibited cell proliferation, colony formation, migration and invasion, and induced apoptosis in HepG2 cells. In vivo studies showed that miR‑132 inhibited tumor growth of HCC and decreased tumor volume and weight. In addition, phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) was identified as a direct target of miR‑132 by a luciferase reporter assay. Western blot and qRT-PCR analysis indicated that PIK3R3 was significantly downregulated by miR‑132 in HCC cells. miR‑132 expression inversely correlated with PIK3R3 mRNA expression in clinical HCC tissues. Investigations into possible mechanisms revealed that miR‑132 inactivated the AKT/mTOR signaling pathway, which may contribute to inhibition of proliferation, migration, and invasion of HCC. These findings suggested that miR‑132 may serve as a potential target in the treatment of human HCC.

Introduction

Hepatocellular carcinoma (HCC) is one of the most commonly occurring cancers worldwide and is a major cause of cancer-related mortality, resulting in ~0.7 million deaths each year (1). Despite the use of multimodal treatments such as surgical resection, liver transplantation, chemotherapy and radiotherapy or a combination of these options (2), a 5-year overall survival for HCC is <30% due to its high recurrence and metastasis rate (3). Therefore, it is urgent to understand the underlying molecular mechanisms involved in HCC for developing cancer prevention strategies and possible guiding disease management in the clinic.

MicroRNAs (miRNA) are an abundant class of endogenous, highly conserved, small non-coding 18–25 nucleotide RNA molecules that have been identified as key negative regulators of gene expression by binding to the 3′-untranslated regions (UTR) of the corresponding target mRNAs of protein-coding genes, thereby resulting in target mRNA degradation or the inhibition of mRNA translation (4,5) Accumulating evidence indicates that miRNAs are often deregulated in various cancers, and have a role in diverse biological processes, such as proliferation, apoptosis, migration, invasion and tumorigenesis (68). Many miRNAs have been demonstrated to function as oncogenes by repressing tumor suppressors, or tumor suppressors by negatively regulating oncogenes (9). These findings suggested that miRNAs play important roles in tumor progression, development and metastasis, therefore, miRNAs are consider as potential novel targets for various cancers therapy.

miR-132, arising from the miR-212/132 cluster (10), is reported as dysregulated in several malignancies and the function of miR-132 is complicated because it can be oncogenic in endocrine pancreatic tumors (11), squamous cell carcinoma of the tongue (12), breast cancer (13), and colorectal carcinoma (14,15), or a tumor suppressor in osteosarcoma (16), prostate cancer (17), non-small lung cancer (18), and ovarian cancer (19). It has been reported that miR-132 was significantly down-regulated in HCC tissue compared to adjacent non-cancerous hepatic tissues (20,21). However, the mechanism of miR-132 in HCC development is not very clear due to poor target gene information.

In the present study, we identify that miR-132 is down-regulated in HCC tissue and cell lines and can suppress cell proliferation, colony formation, migration and invasion, and induce apoptosis in HCC cells, as well as suppress tumor growth in a nude mouse model. We also showed that PIK3R3 is a new target gene of miR-132. Furthermore, we showed that miR-132 might function as a tumor suppressor by directly targeting the PIK3R3 and regulating the AKT/mTOR pathway.

Materials and methods

Human tissues and cell lines

Forty paired human HCC tissues and their adjacent non-cancerous liver tissues were collected from patients undergoing hepatic resection in the Department of Thoracic Surgery, The First Hospital, Jilin University (Changchun, Jilin Province) between July 2009 and September 2014. None of the patients received radiotherapy or chemotherapy before surgery. The specimens were immediately frozen in liquid nitrogen and then stored at −80°C until use. All HCC specimens were confirmed by pathological examination. Prior informed consent was obtained, and the study protocol was approved by the Ethics Committee of Jilin University (Changchun, China).

Human HCC cell lines (Huh-7, SMMC7721, HepG2 and HCCLM3), and normal human heaptocyte cell line HL-7702, were purchased from Shanghai Institute for Biological Sciences (Shanghai, China) and the American Type Culture Collection. All cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM, Gibco, USA) supplemented with 10% fetal bovine serum (FBS, Hyclone, USA) at 37°C in a humidified chamber supplemented with 5% CO2.

Cell transfection

miR-132 mimic and corresponding miRNA negative control (miR-control) were designed and synthesized by GenePharma Co., Ltd. (Shanghai, China), and were transfected into HepG2 cells using Lipofectamine 2000 reagent (Invitrogen) according to the manufacturer's instructions at final concentration of 100 nM. Transfection efficiency was evaluated in every experiment by qRT-PCR 48 h after transfection.

RNA isolation and qRT-PCR assays

miRNA was extracted from human tissue samples and cultured cells using mirVana miRNA Isolation kit (Ambion, Carlsbad, CA, USA) according to the manufacturer's instructions. The expression level of miR-132 was separately quantified with specific primers and probes using TaqMan miRNA assays (Applied Biosystems, Carlsbad, CA, USA) according to the manufacturer's instructions, and normalized by U6 small nuclear RNA.

Total RNA was extracted from human tissue samples and cultured cells using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's protocol and were quantified with Nanodrop 2000 (Thermo Fisher Scientific, Waltham, MA, USA). First-strand cDNA was synthesized using the PrimeScript RT Reagent kit (Takara, Dalian, Japan). qRT-PCR was performed with SYBR Green premix Ex Taq (Takara) on ABI 7900 Fast system (Applied Biosystems). The primer sequences used were as follows: PIK3R3 (sense, 5′-CTTGCTCTGTGGTGGCCGAT-3′ and antisense, 5′-GAC GTTGAGGGAGTCGTTGT-3′); GAPDH (sense, 5′-GAAGGT GAAGGTCGGAGTC-3′ and antisense, 5′-GAAGATGGTG ATGGGATTTC-3′) was used as an internal control. All reactions were run in triplicate and the fold changes of genes were calculated by the 2−ΔΔCt method.

Cell proliferation and colony formation assay

Cell proliferation was assessed by CCK-8 assay (Cell Counting Kit-8, Dojindo, Kumamoto, Japan). In brief, 5×103 cells/well were seeded in 96-well plates, then transfected with miR-132 mimic and corresponding negative control (miR-control), the cell proliferation was detected at 24, 48, 72 and 96 h after transfection using Cell Counting Kit-8 (Dojindo) at 45 nm according to the manufacturer's instructions.

For colony formation, transfected HepG2 cells were resuspended and seeded onto 6-well plates at a density of 1,000 cells/well and cultured for 14 days, and then stained with 0.5% crystal violet for 30 min. The percentage colony formation was calculated by adjusting control (miR-control group) to 100%.

Cell cycle and apoptosis assay

Cell cycle and apoptosis were examined by flow cytometry at 48 h posttransfection. For the cell cycle assay, the transfected cells were collected and fixed in 70% ethanol at 4°C for 16 h and then stained with propidium iodide (PI, Sigma, USA) at 4°C for 30 min in the dark. Cell apoptosis assay was performed by using phycoerythrin (PE)-Annexin V apoptosis detection kit (BD Pharmingen, San Jose, CA, USA). The apoptotic rate and cycle distribution were measured by using a FACSCalibur flow cytometer (BD Biosciences, Mansfield, MA, USA), and analyzed using CellQuest software (BD Biosciences).

Wound-healing assays

For cell motility assay, 5×104 transfected cells were seeded in 6-well plates to near confluence. A linear wound was carefully made by a sterile pipette tip across the confluent cell monolayer, and the cell debris was removed by washing with PBS and incubated with DMEM (1% FBS). The wounded monolayers were then photographed at 0 and 24 after wounding.

Cell invasion assay

The invasive ability of HCC cells was determined using 24-well transwell chambers coated with Matrigel (BD Biosciences). Chambers include upper and lower culture compartments that are separated by polycarbonate membranes with 8-μm pores (Costar, Cambridge, MA, USA). The bottom chamber was filled with DMEM medium containing 10% FBS as a chemoattractant. Transfected cells in serum-free medium were seeded at 2×105 in the top chamber and incubated at 37°C in a humidified incubator containing 5% CO2 for 48 h. Cells that migrated to the underside of the membrane were stained with 0.1% crystal violet, imaged, and counted with a microscope (Olympus, Tokyo, China). All experiments were performed in triplicate.

Prediction of putative targets

To predict the putative targets of miR-132, the following online software was applied: TargetScan (http://www.targetscan.org/), miRanda (http://www.microrna.org/), and miRWalk (http://www.umm.uni-heidelberg.de/apps/zmf/mirwalk/).

Plasmid construction and luciferase reporter assay

The wild-type 3′-UTR segment of PIK3R3, which contained a putative binding site for miR-132, was amplified from normal human genomic DNA by PCR and cloned downstream of the luciferase gene in pGL3-control vector (Promega, Madison, WI, USA) at the NheI and XhoI sites. A mutant 3′-UTR of PIK3R3 containing a mutation in the complementary seed region of miR-132 was synthesized by and cloned into pGL3-control vector. For luciferase assay, 1×105 HepG2 cells were seeded in 24-well plates and allowed to settle for 24 h. Cells were cotransfected with 100 ng wild-type or mutant reporter plasmid, 100 nM miR-132 mimics or miR-NC, and 20 ng pRL-TK renilla plasmid (Promega) using Lipofectamine 2000 according to the manufacturer's instructions. At 48 h post-transfection, both firefly and Renilla luciferase activities were determined using a Dual-Luciferase Reporter system (Promega).

Western blot analysis

Tissue sample and cells were collected and homogenized with RIPA lysis buffer (Beyotime, Beijing, China) according to the manufacturer's instructions. Total protein concentration was detected using a bicinchoninic acid (BCA) protein assay kit (Boster, China). Equal amounts of protein lysates (30 μg each lane) was separated by sodium 10% dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and then electrotransferred to PVDF membranes (Millipore, Bedford, MA, USA). The membranes were blocked with TBST containing 5% non-fat dry milk for 2 h at room temperature, and incubated with primary antibody overnight at 4°C. Then the membrane was incubated with the corresponding horseradish peroxidase (HRP)-conjugated secondary antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA) for 2 h at room temperature. Proteins were visualized with ECL-chemiluminescent kit (ECL-plus, Thermo Scientific). The primary antibodies were: anti-PIK3R3, anti-mTOR, anti-phosphor-mTOR (ser2481), anti-AKT, anti-phosphor-AKT (ser473), anti-GAPDH. All antibodies were purchased from Santa Cruz Biotechnology. GAPDH was used as the internal control.

In vivo nude mouse tumorigenesis assay

Twenty of five-week-old BALB/C-nu/nu nude male mice were obtained from the Experimental Animal Center of Changchun Institute for Biological Sciences (Changchun, China), and maintained in the pathogen-free (SPF) conditions. All animal experiments and their maintenance was performed according to the guidelines approved by the Animal Ethics Committee of Jilin University (Changchun, China).

For the in vivo tumor assay, 2×106 HepG2 cells stably expressing the miR-132 mimic or the miR-control were injected subcutaneously into the right rear flank of each mouse (10 per group) to establish a HCC xenograft model. Tumor volumes were measured every week using calipers two major axes after treatment, and calculated according to the formula: V = 0.5 × L (length) × W2 (width). After 5 weeks, all the mice were sacrificed and the tissue were removed and weighted. Part of the tumor tissue was collected for analysis of the expression of PIK3R3 using western blot analysis.

Statistical analysis

All data are expressed as means ± standard deviation (SD) from three independent experiments. Data were imaged with GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA). The differences between groups were analyzed using Student's t-test. The relationship between miR-132 expression level and clinical and pathological variables was analysed using Pearson's χ2 test. All the analyses were performed with the SPSS software (version 13.0) (IBM Corp., New York, NY, USA). P-values <0.05 were considered statistically significant.

Results

miR-132 is downregulated in human HCC cell lines and tissues

To examine levels of miR-132 in HCC, and used qRT-PCR to quantify the miR-132 expression in four HCC cell lines and 40 pairs of HCC tissues and their corresponding non-cancerous liver tissues. The results showed that the expression of miR-132 was markedly downregulated in the human HCC cell lines Huh-7, SMMC7721, HCCLM3, and HepG2 compared with the normal hepatic cell line HL-7702 (Fig. 1A). Additionally, the expression level of miR-132 in the HepG2 cells was the lowest, thus, we selected this cell line for further study. Consistent with the results from cell lines, miR-132 levels were significantly lower in HCC tissues compared with their matched normal liver tissues (Fig. 1B), indicating that miR-132 was downregulated in HCC.

In addition, the relationship between the miR-132 expression levels and the clinicopathological characteristics of the HCC patients was investigated and summarized in Table I. The results showed that no statistically significant correlations were observed between the miR-132 expression and age and gender (Table I). However, miR-132 expression correlated with tumor differentiation (P<0.01), TNM stage (P<0.01) and lymph node metastasis (P<0.01) (Table I) were observed. These results suggested that miR-132 might be involved in HCC initiation and progression.

Table I

Association of miR-132 expression with clinicopathological factors of 40 HCC patients.

Table I

Association of miR-132 expression with clinicopathological factors of 40 HCC patients.

VariablesNo. of casesRelative miR-132 expression (miR-132/U6)P-value
Age (years)P>0.05
 <50180.421±0.621
 ≥50220.416±0.537
GenderP>0.05
 Male240.419±0.734
 Female160.416±0.644
TNM stageP<0.01
 I–II280.368±0.579
 III–IV120.535±0.612
DifferentiationP<0.01
 Well/moderate300.371±0.746
 Poor100.559±0.438
Lymph node metastasisP<0.01
 No260.336±0.695
 Yes140.570±0.521
miR-132 inhibits proliferation and colony formation of HCC cells in vitro

Based on the decreased expression of miR-132 in HCC tissues we assumed miR-132 to be a tumor suppressor. To test the miR-132 in HCC growth, miR-132 mimic and miR-control were transfected into HepG2 cells. We found that transfection of miR-132 mimics restored miR-132 expression level in HepG2 cells (Fig. 2A). Then we investigated the effects of miR-132 restoration on proliferation in HepG2 cells by CCK-8 assay. As shown in Fig. 2B, miR-132 could significantly inhibit the proliferation of HepG2 cells in vitro. To assess the role of miR-132 in cancer cell growth, colony formation was performed in HepG2 cells transfected with miR-132 mimic or miR-control. Compared with miR-control group, the numbers of HepG2 colonies were reduced significantly by restoration of miR-132 (P<0.05, Fig. 2C). Taken together, the results indicate that miR-132 inhibits the proliferation and colony formation of HCC cells in vitro.

miR-132 induces the cell cycle and apoptosis of HCC cells in vitro

To determine whether miR-132 regulates the cell cycle, flow cytometry was performed. The results indicate that cell cycle was arrested in G1 phase, with 71.36% of miR-132 transfected cells in G0/G1 versus 58.32% of miR-control transfected cells (Fig. 3A). Next, the ability of miR-132 to induce apoptosis in HepG2 cell lines was evaluated by co-staining with Annexin V and propidium iodide (PI). The results demonstrated that miR-132 could significantly induce apoptosis in HepG2 cells compared with the miR-control (Fig. 3B). These results suggested that that miR-132 induces apoptosis and cell arrest at G0/G1 stage of HCC cells in vitro.

miR-132 inhibits the migration and invasion of HCC cells in vitro

We tested the role of miR-132 in migration and invasion of HCC cells. Wound healing assay showed that the ectopic expression of miR-132 HepG2 cells significantly inhibited cell migration, compared to the control group (Fig. 4A). Additionally, we performed the Boyden chamber assay to investigate the effect of miR-132 on cell invasion. As shown in Fig. 4B, overexpression of miR-132 significantly decreased the invasion capacity of HepG2 cells.

miR-132 targets PIK3R3 and inhibits its expression and AKT/mTOR signaling pathway activation

To understand the molecular mechanism of miR-132 action in HCC, we searched for miR-132 targets by using TargetScan, miRanda, and miRWalk algorithms. Phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) was predicted as a potential target of miR-132. To further confirm whether PIK3R3 was a direct target of miR-132, a human PIK3R3 3′-UTR fragment containing the binding sites of miR-132 or the mutant sites (Fig. 5A) were cloned into the pGL3 vector, then along with miR-132 mimic or miR-control were cotransfected into HepG2 cells. At 48 h posttransfection, luciferase activities were measured. It was found that restoration of miR-132 obviously suppressed the luciferase activity of wild-type PIK3R3 site, but the activity of the mutant PIK3R3 site was not affected (Fig. 5B), suggesting that PIK3R3 is directly targeted by miR-132. Then, qRT-PCR and western blot analysis was performed to measure PIK3R3 on mRNA (Fig. 5C) and protein level (Fig. 5D) in HepG2 cells transfected with miR-132 mimic. The result showed that PIK3R3 mRNA and protein levels decreased in miR-132-transfected HCC cells.

It has been shown that PIK3R3 played key roles in survival, proliferation, and motility of the tumors because PIK3R3 activated AKT/mTOR signaling pathways (22,23). Here, we investigated whether miR-132 affected activation of AKT/mTOR pathways. The AKT, p-AKT, mTOR and p-mTOR expression were determined in HepG2 cells by western blot analysis after transfected with miR-132 mimic or miR-control. The result of western blot analysis showed that overexpression of miR-132 significantly inhibited the phosphorylation of AKT and mTOR, whereas total AKT and mTOR expression did not change (Fig. 5D). These results indicate that miR-132 can bind directly to PIK3R3 and inhibits the expression of PIK3R3 and AKT/mTOR signaling pathway activation in HCC cells.

miR-132 is inversely correlated with PIK3R3 expression in HCC

To determine whether the inverse relationship between miR-132 and PIK3R3 expression observed in vitro also occurred in vivo, we examined expression of PIK3R3 in HCC specimens and corresponding non-cancerous tissue by qRT-PCR. The results showed that PIK3R3 mRNA levels were higher in HCC tissues than in paired non-cancerous tissues (Fig. 6A) and were negatively correlated with miR-132 (Fig. 6B; r=−0.578, P<0.001).

miR-132 suppresses tumor growth in nude mice by inhibiting PIK3R3

The preceding in vitro studies indicated that miR-132 could suppress HCC cell growth and metastasis in vitro. Thus, we further investigated whether miR-132 could affect HCC tumor growth in vivo. The stably transfected human HepG2 cells (HepG2/miR-132 or HepG2/miR-control) were implanted subcutaneously into nude mice to allow tumor formation. At five weeks post-injection, the mice were sacrificed, and tumor tissue was isolated. We found that the xenograft tumor was significantly smaller in the miR-132 group compared with those in the miR-control group (Fig. 7A), and that the volume (Fig. 7B) and weight (Fig. 7C) also were decreased in miR-132 group, indicating that miR-132 overexpression suppresses HCC tumor growth in vivo. We also detected the PIK3R3 expression in tumor xenograft tumors by western blot analysis. We found that PIK3R3 protein expression was downregulated in xenograft tumors of the miR-132 group (Fig. 7D). These results suggested that miR-132 suppresses HCC tumor growth in nude mice by inhibiting PIK3R3.

Discussion

Hepatocellular carcinoma (HCC) is a primary lethal neoplasm of the liver and the third leading cause of cancer-related deaths worldwide, and the burden of this devastating cancer is expected to increase further in the coming years (1). However, its underlying molecular mechanism remains largely unknown. In recent years, a larger number of microRNAs (miRNAs) have been reported to be involved in the initiation and progression of HCC. For example, miR-222 can promote cell proliferation, migration and invasion, and decrease cell apoptosis, as well as enhance the resistance of HCC cells to sorafenib through activating the PI3K/AKT signaling pathway (24). MicroRNA-188-5p suppresses tumor cell proliferation and metastasis by directly targeting fibroblast growth factor 5 (FGF5) in hepatocellular carcinoma (25).

MicroRNA-129-5p inhibits hepatocellular carcinoma cell metastasis and invasion via targeting v-ets erythroblastosis virus E26 oncogene homolog 1 (ETS1) (26). miR-211 repressed proliferation and invasion in HCC cells by targeting special AT-rich sequence-binding protein-2 (SATB2) (27). Here, we found that miR-132 expression level was downregulated in HCC tissues and cell lines, and its expression negatively correlated with tumor differentiation, TNM stage and lymph node metastasis. miR-132 was able to inhibit HCC cell growth and metastasis by directly targeting PIK3R3 and inhibiting activation of the Akt/mTOR signaling pathway. These findings suggest that miR-132 might be a novel tumor suppressor miRNA. miR-132, located in an intergenic region on human chromosome 17, is a highly conserved miRNA (28). miR-132 has been reported to be an oncogene in endocrine pancreatic tumors (11), squamous cell carcinoma of the tongue (12), breast cancer (13), and colorectal carcinoma (14,15). However, miR-132 also has been found to be a tumor suppressor in a series of cancers, such as prostate cancer (17), and ductal carcinoma in situ (DCIS) of the breast cancer (29), osteosarcoma (16), non-small lung cancer (18) and ovarian cancer (19). For HCC, a microarray report showed that miR-132 is downregulated in human HCC (30). Another study also demonstrated that miR-132 expression was more frequently downregulated in HBV-related HCC tissues than in adjacent non-cancerous hepatic tissues and had a significant inverse correlation with HBx expression in HBV-related HCCs (21). Consistent with these results, our results showed that miR-132 expression was downregualted in HCC tissue and cell lines. In addition, recently two studies showed that miR-132 inhibited HCC cell proliferation and colony formation and induced cell apoptosis (20,21). However, the role of miR-132 in HCC is not well known due to the limitation of target gene information. To our knowledge, miR-132 target gene in HCC has not been confirmed until now. In this study, we used the prediction algorithms of TargetScan, miRWalk and miRanda to predict that miR-132 can target the 3′-UTR of PIK3R3. Luciferase activities further confirmed PIK3R3 is a direct target gene of miR-132. Overexpression of miR-132 in HCC cells decreased both mRNA and protein level of PIK3R3 in HCC cells. Our results also showed that miR-132 was negatively correlated with PIK3R3 mRNA in HCC tissues. Our results demonstrated that miR-132 can suppress HCC migration and invasion, and tumor growth in vivo. These findings linked to previous studies suggested that miR-132 functions as a tumor suppressor in HCC, and plays a crucial role in HCC progression.

PIK3R3 is a member of the phosphatidylinositol 3-kinase (PI3K) family, which plays a key regulatory role in cell proliferation, cell differentiation, tumor angiogenesis, and tumor growth by the unique N-terminal domain of PIK3R3 specifically binding to cell growth key proteins, including retinoblastoma protein (Rb) and proliferating cell nuclear antigen (PCNA) (3133). It has been reported that PIK3R3 functioned as an oncogene, involved in cancer development and progression in several types of cancers, such as liver cancer (23), ovarian cancer (34), gastric cancer (35), lung cancer (36), colorectal cancer (37), and breast cancer (38). In addition, studies have showed that PIK3R3 could regulate the AKT/mTOR signaling pathway (22,23). The data from the present study showed that overexpression of miR-132 in HCC cells inhibited pAkt and pmTOR expression, suggesting that miR-132 inhibited HCC growth and metastasis by targeting PIK3R3 through regulating AKT/mTOR signaling pathway to some extent.

In conclusion, our study provides evidence that the expression of miR-132 was decreased in HCC tissue and cell lines, and its expression level was significantly associated with tumor differentiated, TNM stage and lymph node metastasis. miR-132 can inhibit cell proliferation, colony formation, migration and invasion, and induced cell apoptosis, and cell cycle at G0/G1 stage in vitro, as well as suppressed tumor growth in vivo. Moreover, PIK3R3 was identified as a crucial target gene of miR-132. Overexpression of miR-132 inhibited PIK3R3 expression and AKT/mTOR signaling pathway activation. These findings clearly showed that miR-132 functions as a tumor suppressor in HCC, suggesting that miR-132 could be a potential target for the treatment of HCC.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Livraghi T, Makisalo H and Line PD: Treatment options in hepatocellular carcinoma today. Scand J Surg. 100:22–29. 2011.PubMed/NCBI

3 

Thorgeirsson SS and Grisham JW: Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet. 31:339–346. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Malan-Müller S, Hemmings SM and Seedat S: Big effects of small RNAs: A review of microRNAs in anxiety. Mol Neurobiol. 47:726–739. 2013. View Article : Google Scholar :

6 

Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Almeida MI, Reis RM and Calin GA: MicroRNA history: Discovery, recent applications, and next frontiers. Mutat Res. 717:1–8. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Calin GA and Croce CM: MicroRNA-cancer connection: The beginning of a new tale. Cancer Res. 66:7390–7394. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Manikandan J, Aarthi JJ, Kumar SD and Pushparaj PN: Oncomirs: The potential role of non-coding microRNAs in understanding cancer. Bioinformation. 2:330–334. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Remenyi J, Hunter CJ, Cole C, Ando H, Impey S, Monk CE, Martin KJ, Barton GJ, Hutvagner G and Arthur JS: Regulation of the miR-212/132 locus by MSK1 and CREB in response to neurotrophins. Biochem J. 428:281–291. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, et al: A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 103:2257–2261. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Wong TS, Liu XB, Wong BY, Ng RW, Yuen AP and Wei WI: Mature miR-184 as potential oncogenic microRNA of squamous cell carcinoma of tongue. Clin Cancer Res. 14:2588–2592. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Anand S, Majeti BK, Acevedo LM, Murphy EA, Mukthavaram R, Scheppke L, Huang M, Shields DJ, Lindquist JN, Lapinski PE, et al: MicroRNA-132-mediated loss of p120RasGAP activates the endothelium to facilitate pathological angiogenesis. Nat Med. 16:909–914. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Schetter AJ, Leung SY, Sohn JJ, Zanetti KA, Bowman ED, Yanaihara N, Yuen ST, Chan TL, Kwong DL, Au GK, et al: MicroRNA expression profiles associated with prognosis and therapeutic outcome in colon adenocarcinoma. JAMA. 299:425–436. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Yang L, Belaguli N and Berger DH: MicroRNA and colorectal cancer. World J Surg. 33:638–646. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Yang J, Gao T, Tang J, Cai H, Lin L and Fu S: Loss of microRNA-132 predicts poor prognosis in patients with primary osteosarcoma. Mol Cell Biochem. 381:9–15. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Formosa A, Lena AM, Markert EK, Cortelli S, Miano R, Mauriello A, Croce N, Vandesompele J, Mestdagh P, Finazzi-Agrò E, et al: DNA methylation silences miR-132 in prostate cancer. Oncogene. 32:127–134. 2013. View Article : Google Scholar

18 

You J, Li Y, Fang N, Liu B, Zu L, Chang R, Li X and Zhou Q: MiR-132 suppresses the migration and invasion of lung cancer cells via targeting the EMT regulator ZEB2. PLoS One. 9:e918272014. View Article : Google Scholar : PubMed/NCBI

19 

Chung YW, Bae HS, Song JY, Lee JK, Lee NW, Kim T and Lee KW: Detection of microRNA as novel biomarkers of epithelial ovarian cancer from the serum of ovarian cancer patients. Int J Gynecol Cancer. 23:673–679. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Liu HB, Hua Y and Jin ZX: Effects of MicroRNA-132 transfection on the proliferation and apoptosis of human liver cancer cells in vitro and in vivo. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. 37:30–36. 2015.PubMed/NCBI

21 

Wei X, Tan C, Tang C, Ren G, Xiang T, Qiu Z, Liu R and Wu Z: Epigenetic repression of miR-132 expression by the hepatitis B virus x protein in hepatitis B virus-related hepatocellular carcinoma. Cell Signal. 25:1037–1043. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Yu T, Li J, Yan M, Liu L, Lin H, Zhao F, Sun L, Zhang Y, Cui Y, Zhang F, et al: MicroRNA-193a-3p and -5p suppress the metastasis of human non-small-cell lung cancer by downregulating the ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Oncogene. 34:413–423. 2015. View Article : Google Scholar

23 

Cao G, Dong W, Meng X, Liu H, Liao H and Liu S: MiR-511 inhibits growth and metastasis of human hepatocellular carcinoma cells by targeting PIK3R3. Tumour Biol. Jan 22–2015.(Epub ahead of print). View Article : Google Scholar

24 

Liu K, Liu S, Zhang W, Ji B, Wang Y and Liu Y: miR-222 regulates sorafenib resistance and enhance tumorigenicity in hepatocellular carcinoma. Int J Oncol. 45:1537–1546. 2014.PubMed/NCBI

25 

Fang F, Chang RM, Yu L, Lei X, Xiao S, Yang H and Yang LY: MicroRNA-188-5p suppresses tumor cell proliferation and metastasis by directly targeting FGF5 in hepatocellular carcinoma. J Hepatol. S0168-8278 (15)00331-1. 2015. View Article : Google Scholar

26 

Ma N, Chen F, Shen SL, Chen W, Chen LZ, Su Q, Zhang LJ, Bi J, Zeng WT, Li W, et al: MicroRNA-129-5p inhibits hepatocellular carcinoma cell metastasis and invasion via targeting ETS1. Biochem Biophys Res Commun. 461:618–623. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Jiang G, Cui Y, Yu X, Wu Z, Ding G and Cao L: miR-211 suppresses hepatocellular carcinoma by downregulating SATB2. Oncotarget. 6:9457–9466. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Nudelman AS, DiRocco DP, Lambert TJ, Garelick MG, Le J, Nathanson NM and Storm DR: Neuronal activity rapidly induces transcription of the CREB-regulated microRNA-132, in vivo. Hippocampus. 20:492–498. 2010.

29 

Li S, Meng H, Zhou F, Zhai L, Zhang L, Gu F, Fan Y, Lang R, Fu L, Gu L, et al: MicroRNA-132 is frequently down-regulated in ductal carcinoma in situ (DCIS) of breast and acts as a tumor suppressor by inhibiting cell proliferation. Pathol Res Pract. 209:179–183. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Gramantieri L, Ferracin M, Fornari F, Veronese A, Sabbioni S, Liu CG, Calin GA, Giovannini C, Ferrazzi E, Grazi GL, et al: Cyclin G1 is a target of miR-122a, a microRNA frequently down-regulated in human hepatocellular carcinoma. Cancer Res. 67:6092–6099. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Xia X, Cheng A, Akinmade D and Hamburger AW: The N-terminal 24 amino acids of the p55 gamma regulatory subunit of phosphoinositide 3-kinase binds Rb and induces cell cycle arrest. Mol Cell Biol. 23:1717–1725. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Wang G, Cao X, Lai S, Luo X, Feng Y, Xia X, Yen PM, Gong J and Hu J: PI3K stimulates DNA synthesis and cell-cycle progression via its p55PIK regulatory subunit interaction with PCNA. Mol Cancer Ther. 12:2100–2109. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Wang G, Chen C, Yang R, Cao X, Lai S, Luo X, Feng Y, Xia X, Gong J and Hu J: p55PIK-PI3K stimulates angiogenesis in colorectal cancer cell by activating NF-κB pathway. Angiogenesis. 16:561–573. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Zhang L, Huang J, Yang N, Greshock J, Liang S, Hasegawa K, Giannakakis A, Poulos N, O'Brien-Jenkins A, Katsaros D, et al: Integrative genomic analysis of phosphatidylinositol 3′-kinase family identifies PIK3R3 as a potential therapeutic target in epithelial ovarian cancer. Clin Cancer Res. 13:5314–5321. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Soroceanu L, Kharbanda S, Chen R, Soriano RH, Aldape K, Misra A, Zha J, Forrest WF, Nigro JM, Modrusan Z, et al: Identification of IGF2 signaling through phosphoinositide-3-kinase regulatory subunit 3 as a growth-promoting axis in glioblastoma. Proc Natl Acad Sci USA. 104:3466–3471. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Xu L, Wen Z, Zhou Y, Liu Z, Li Q, Fei G, Luo J and Ren T: MicroRNA-7-regulated TLR9 signaling-enhanced growth and metastatic potential of human lung cancer cells by altering the phosphoinositide-3-kinase, regulatory subunit 3/Akt pathway. Mol Biol Cell. 24:42–55. 2013. View Article : Google Scholar :

37 

Wang G, Yang X, Li C, Cao X, Luo X and Hu J: PIK3R3 induces epithelial-to-mesenchymal transition and promotes metastasis in colorectal cancer. Mol Cancer Ther. 13:1837–1847. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Klahan S, Wu MS, Hsi E, Huang CC, Hou MF and Chang WC: Computational analysis of mRNA expression profiles identifies the ITG family and PIK3R3 as crucial genes for regulating triple negative breast cancer cell migration. BioMed Res Int. 2014:5365912014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 47 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu K, Li X, Cao Y, Ge Y, Wang J and Shi B: miR-132 inhibits cell proliferation, invasion and migration of hepatocellular carcinoma by targeting PIK3R3 Retraction in /10.3892/ijo.2021.5238. Int J Oncol 47: 1585-1593, 2015
APA
Liu, K., Li, X., Cao, Y., Ge, Y., Wang, J., & Shi, B. (2015). miR-132 inhibits cell proliferation, invasion and migration of hepatocellular carcinoma by targeting PIK3R3 Retraction in /10.3892/ijo.2021.5238. International Journal of Oncology, 47, 1585-1593. https://doi.org/10.3892/ijo.2015.3112
MLA
Liu, K., Li, X., Cao, Y., Ge, Y., Wang, J., Shi, B."miR-132 inhibits cell proliferation, invasion and migration of hepatocellular carcinoma by targeting PIK3R3 Retraction in /10.3892/ijo.2021.5238". International Journal of Oncology 47.4 (2015): 1585-1593.
Chicago
Liu, K., Li, X., Cao, Y., Ge, Y., Wang, J., Shi, B."miR-132 inhibits cell proliferation, invasion and migration of hepatocellular carcinoma by targeting PIK3R3 Retraction in /10.3892/ijo.2021.5238". International Journal of Oncology 47, no. 4 (2015): 1585-1593. https://doi.org/10.3892/ijo.2015.3112