MicroRNA‑601 serves as a potential tumor suppressor in hepatocellular carcinoma by directly targeting PIK3R3

Retraction in: /10.3892/mmr.2022.12923

  • Authors:
    • Yanan Song
    • Saifei He
    • Juhua Zhuang
    • Guoyu Wang
    • Jing Ni
    • Suiliang Zhang
    • Ying Ye
    • Wei Xia
  • View Affiliations

  • Published online on: January 15, 2019     https://doi.org/10.3892/mmr.2019.9857
  • Pages: 2431-2439
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Recently, microRNAs (miRNAs) have been acknowledged as important regulators of hepatocarcinogenesis and tumor progression. Therefore, identifying the underlying molecular mechanisms of miRNAs in the occurrence and development of hepatocellular carcinoma (HCC) may be important for understanding the pathogenesis of HCC and aid the identification of potential therapeutic strategies. In the present study, miRNA (miR)‑601 was significantly downregulated in HCC tissues and cell lines; low miR‑601 expression was strongly associated with tumor, node and metastasis staging and lymph node metastasis of patients with HCC. In addition, the overexpression of miR‑601 expression significantly inhibited the proliferation and invasion of HCC cells. Regarding the underlying mechanism, phosphoinositide‑3‑kinase regulatory subunit 3 (PIK3R3) was predicted to be a direct target of miR‑601 in HCC cells. Furthermore, restoration of PIK3R3 expression in these cells counteracted the inhibitory effects of miR‑601 on cell proliferation and invasion in HCC. Notably, miR‑601 overexpression inhibited the protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway in HCC via the regulation of PIK3R3. Collectively, these results demonstrated that miR‑601 may inhibit the progression of HCC by directly targeting PIK3R3 and regulating the AKT/mTOR signaling pathway. Therefore, miR‑601 may be an effective therapeutic target for the treatment of patients with HCC.

Introduction

Hepatocellular carcinoma (HCC), accounting for >90% of all cases of primary liver cancer, ranks as the sixth most common type of cancer and the second leading cause of cancer-associated mortality worldwide (1). There are >1 million novel diagnosed HCC cases and ~1 million incidences of mortality that occur annually worldwide due to HCC (2,3). Exposure to aflatoxin B1 and infection with hepatitis viruses B or C have been identified as two primary risk factors for HCC (4). With no notable symptoms at the early stages of disease, ~80% patients with HCC are diagnosed at an advanced stage (5). As of yet, surgical hepatectomy and liver transplantation remain the predominant treatments for patients with HCC (6). Significant improvements in surgical techniques and perioperative management have been made; however, the outcomes of treatment for patients with HCC remain unsatisfactory with a 5-year survival rate of <5% (7). Recurrence, and intrahepatic and extrahepatic metastases are factors primarily responsible for the poor prognosis of patients with HCC (8). Consequently, comprehensive understanding of the particular mechanisms underlying the initiation and progression of HCC is important for the identification of novel therapeutic strategies.

MicroRNAs (miRNAs) are a series of highly conserved, non-protein-coding, short RNA molecules, comprising 19–23 nucleotides (9). miRNAs negatively modulate protein expression by imperfect complementary sequence pairing to the 3′-untranslated regions (UTRs) of their target genes, resulting in the degradation of target mRNA or suppression of translation (10). More than one-half of miRNAs have been targeted to cancer-associated genomic regions, and this phenomenon suggests that dysregulated miRNAs may be involved in tumorigenesis and tumor development (11). Alterations in the expression of miRNAs have been recently identified in various types of human cancer, including HCC (12), gastric (13), lung (14) and colorectal cancer (15). miRNAs have been associated with malignancy by serving as oncogenes or tumor suppressors (1618); miRNAs are involved in the regulation of numerous pathological behaviors, including cell proliferation, apoptosis, invasion, metastasis, stem cell biology and epithelial-mesenchymal transition (1618). Therefore, further investigation on the aberrantly expressed miRNAs and their roles in HCC may contribute to the identification of diagnostic biomarkers and therapeutic targets for patients with this aggressive malignancy.

The dysregulation of miRNA (miR)-601 has been observed in colorectal (19), breast (20) and pancreatic cancer (21); however, to the best of our knowledge, the association between miR-601 and HCC has not yet been investigated. In the present study, the expression levels and clinical value of miR-601 in HCC were determined; the functional roles and underlying mechanism of miR-601 in HCC were additionally examined.

Materials and methods

Patients and tissue samples

The present study was approved by the Ethics Committee of The Seventh People's Hospital (Shanghai, China). Written informed consent was obtained from all patients prior to enrolment. A total of 56 paired HCC tissues and matched adjacent normal tissues were collected from patients undergoing surgery at The Seventh People's Hospital from March 2014 to December 2016. Tumor Node Metastasis (TNM) system (22) was used for classification of characteristics. None of the patients had undergone chemotherapy, radiotherapy or other treatments prior to surgery. All patients were divided into either low (n=28) or high (n=28) miR-601 groups based on the median levels of miR-601 expression in HCC tissues (1.02). All tissue specimens were quickly frozen following collection and stored in liquid nitrogen.

Cell lines

A total of two human HCC cell lines (Hep3B and Huh7) and an immortalized normal human liver epithelial cell (L-O2) were acquired from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China). All cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum (FBS; both from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and 100 U/ml of penicillin and 100 µg/ml of streptomycin (both from Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). The cells were subsequently cultured at 37°C in a humidified atmosphere containing 5% CO2.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RT-qPCR was performed to detect phosphoinositide-3-kinase regulatory subunit 3 (PIK3R3) mRNA and miR-601 expression. Total RNA was extracted from tissue specimens or cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. For the detection of PIK3R3 mRNA expression, cDNA was synthesized from total RNA using TaqMan RT reagent (Applied Biosystems; Thermo Fisher Scientific, Inc.). A SYBR®-Green PCR Master Mix kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) was subsequently used to perform qPCR with GAPDH as an internal control. qPCR was performed with the following cycling conditions: 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 1 min. For the quantification of miR-601 expression, a TaqMan MicroRNA Assay kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) was employed to produce cDNA. The temperature protocol for reverse transcription was as follows: 16°C for 30 min, 42°C for 30 min and 85°C for 5 min. Subsequently, qPCR was conducted with a TaqMan MicroRNA assay (Applied Biosystems; Thermo Fisher Scientific, Inc.). qPCR was performed with the following cycling conditions: 50°C for 2 min, 95°C for 10 min; 40 cycles of denaturation at 95°C for 15 sec; and annealing/extension at 60°C for 60 sec. U6 small nuclear RNA was used as an internal control for the normalization of miR-601 expression. RT-qPCR was conducted using the Applied Biosystems 7500 Real-time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). Relative gene expression was quantified using the 2−∆∆Cq method (23). The sequences of primers were as follows: PIK3R3, 5′-CTTGCTCTGTGGTGGCCGAT-3′ (forward) and 5′-GACGTTGAGGGAGTCGTTGT-3′ (reverse); and GAPDH, 5′-CGGAGTCAACGGATTTGGTCGTAT-3′ (forward) and 5′-AGCCTTCTCCATGGTGGTGAAGAC-3′ (reverse); miR-601, 5′-GCTCTCCCAACCCTTGT-3′ (forward) and 5′-CUCCUCCAACAAUCCUAGACCATT-3′ (reverse); U6, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ (forward) and 5′-CGCTTCACGAATTTGCGTGTCAT-3′ (reverse).

Transfection assay

miR-601 mimics and negative control miRNA mimics (miR-NC) were provided by Shanghai GenePharma Co., Ltd. (Shanghai, China). The miR-601 mimics sequence was 5′-UGGUCUAGGAUUGUUGGAGGAG-3′ and the miR-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT−3′. A PIK3R3 overexpression plasmid pcDNA3.1-PIK3R3 and empty control pcDNA3.1 plasmid were designed and chemically produced by GeneCopoeia, Inc. (Rockville, MD, USA). For transfection assays, Hep3B and Huh7 cells were plated into 6-well plates at a density of 4×105 cells/well. Cells were transfected with miRNA (100 pmol) or plasmid (4 µg) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol; total RNA and protein were extracted at 48 and 72 h after transfection, respectively. Transfection efficiency was evaluated with RT-qPCR and western blot analysis.

Cell Counting Kit-8 (CCK-8) assay

Following 24 h post-transfection, a total of 2,000 transfected cells were plated into each well of a 96-well plate with five replicates. Subsequent to incubation at 37°C with 5% CO2 for 0, 24, 48 and 72 h, a CCK-8 assay was conducted to assess the cellular proliferative ability of transfected cells. CCK-8 reagent (10 µl; Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was added into each well and the plates were incubated at 37°C with 5% CO2 for an additional 2 h. The optical density was detected at 450 nm with a Spectramax M5 microplate reader (Molecular Devices LLC, Sunnyvale, CA, USA).

Invasion assay

Matrigel-coated 24-well Transwell Boyden chambers (BD Biosciences, Franklin Lakes, NJ, USA) were used to quantify the invasive ability of the HCC cell lines. A total of 1×105 cells in FBS-free DMEM were inoculated into the upper Boyden chambers. The lower Boyden chambers were filled with 600 µl DMEM containing 20% FBS. Following culture for 24 h at 37°C, the non-invasive cells and Matrigel were gently removed with cotton swabs. The invaded cells were fixed in 100% methanol at room temperature for 20 min and stained with 0.1% crystal violet at room temperature for 20 min. Following washing with PBS, the invaded cells were counted under an inverted light microscope (magnification, ×200; Olympus Corporation, Tokyo, Japan) in five predetermined fields.

Bioinformatics analysis

The potential targets of miR-601 were predicted using a publicly available databases: TargetScan version 7.1 (http://www.targetscan.org/) and miRDB (http://mirdb.org/).

Luciferase reporter assay

PIK3R3 was predicted to be a principal target of miR-601 in the present study. The ability of miR-601 to interact with the 3′-UTR of PIK3R3 was experimentally evaluated with a luciferase reporter assay. The wild-type (Wt) 3′-UTR of PIK3R3 was predicted to bind to miR-601. The Wt and a corresponding mutation (Mut) in the miR-601 binding sites of the 3′UTR was designed and produced by Shanghai GenePharma Co., Ltd., and inserted into the pGL3 plasmid (Ambion; Thermo fisher Scientific, Inc.) which were denoted as pGL3-PIK3R3-3′-UTR Wt and pGL3-PIK3R3-3′-UTR Mut, respectively. Cells were inoculated into 24-well plates at a density of 8×104 cells/well and transfected with miR-601 mimics or miR-NC in combination with one of the two reporter plasmids, using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). After 48 h of incubation, luciferase activities were measured using a Dual-Luciferase Reporter Assay System (Promega Corporation, Madison, WI, USA). Firefly luciferase activity was normalized to Renilla luciferase.

Western blot analysis

Total protein of the transfected cells was isolated using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China). The concentration of total protein was determined using a bicinchoninic acid assay kit (Beyotime Institute of Biotechnology). Equal amounts of protein (30 µg) were separated by 10% SDS-PAGE and subsequently transferred to polyvinylidene difluoride membranes (Beyotime Institute of Biotechnology). The membranes were blocked at room temperature with TBS containing 0.1% Tween-20 (TBST) and 5% skimmed milk for 2 h, and incubated with primary antibodies at 4°C overnight. Following three washes with TBST, the membranes were incubated with a corresponding horseradish peroxidase-conjugated secondary antibody (1:5,000; cat. nos. sc-2004 and sc-2005; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at room temperature for 2 h. Finally, a Pierce™ Enhanced Chemiluminescent Western Blotting Substrate (Pierce™; Thermo Fisher Scientific, Inc.) was used to visualize band signals. The primary antibodies used in the present study included: Mouse anti-human monoclonal PIK3R3 antibody (1:1,000; cat. no. sc-376615; Santa Cruz Biotechnology, Inc.), mouse anti-human monoclonal protein kinase B (AKT) antibody (1:1,000; cat. no. sc-81434; Santa Cruz Biotechnology, Inc.), mouse anti-human monoclonal phosphorylated (p)-AKT antibody (1:1,000; cat. no. sc-81433; Santa Cruz Biotechnology, Inc.), rabbit anti-human monoclonal p-mammalian mechanistic target of rapamycin (mTOR) Ser 2481 antibody (1:1,000; cat. no. ab137133; Abcam, Cambridge, UK), rabbit anti-human monoclonal mTOR antibody (1:1,000; cat. no. ab134903; Abcam) and rabbit anti-human monoclonal GAPDH antibody (1:1,000; cat. no. ab128915; Abcam). Quantity One software version 4.62 (Bio-Rad Laboratories, Inc., Hercules, CA, USA) was used to quantify the protein expression.

Statistical analysis

All statistical analyses were conducted using SPSS version 18.0 (SPSS, Inc., Chicago, IL, USA). The data are presented as the mean ± standard deviation and were compared using two-tailed Student's t-test or one-way analysis of variance followed by a Student-Newman-Keuls post-hoc test. The association between miR-601 expression and the various clinicopathological features of patients of HCC was evaluated using a χ2 test. Each assay was repeated at least three times. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-601 expression is downregulated in human HCC tissues and cell lines

The expression of miR-601 in 56 pairs of HCC tissues and matched adjacent normal tissues was detected to investigate the expression profile of miR-601 in HCC. RT-qPCR analysis demonstrated that miR-601 was expressed at significantly lower levels in HCC tissues compared with the matched adjacent normal tissues (P<0.05; Fig. 1A). Additionally, miR-601 expression was measured in two human HCC cell lines, including Hep3B and Huh7. Compared with the immortalized normal human liver epithelial cell (L-O2), the expression levels of miR-601 were significantly decreased in the two human HCC cell lines (P<0.05; Fig. 1B). These results implied that miR-601 downregulation may be associated with the development and progression of HCC.

Association between miR-601 expression and the clinicopathological features of HCC

To evaluate the association between miR-601 and the clinicopathological features of patients with HCC, all patients were divided into either low (n=28) or high (n=28) miR-601 groups based on the median levels of miR-601 expression. Statistical analyses identified that decreased miR-601 expression levels were significantly associated with TNM stage (P=0.007) and lymph node metastasis (P=0.016); however, no significant association was observed with other clinicopathological factors, including age (P=0.589), sex (P=0.408), tumor size (P=0.284) and differentiation (P=0.101; Table I). These results suggested that the downregulation of miR-601 may serve important roles in the progression of HCC.

Table I.

Association between miR-601 expression and the clinicopathological factors of patients with hepatocellular carcinoma.

Table I.

Association between miR-601 expression and the clinicopathological factors of patients with hepatocellular carcinoma.

miR-601 expression

Clinicopathological factorsLowHighP-value
Age 0.589
  <55 years1113
  ≥55 years1715
Sex 0.408
  Female912
  Male1916
Tumor size 0.284
  <5 cm1317
  ≥5 cm1511
Differentiation 0.101
  Well and moderate814
  Poor2014
TNM stage 0.007
  I–II717
  III–IV2111
Lymph node metastasis 0.016
  Negative918
  Positive1910

[i] miR, microRNA; TNM, tumor, node and metastasis.

Upregulation of miR-601 expression may inhibit HCC malignancy in vitro

To demonstrate the role of miR-601 in the progression of HCC, Hep3B and Huh7 cells were transfected with miR-601 mimics to ectopically express miR-601; the expression levels of miR-601 were significantly higher compared with the miR-NC-transfected cells (P<0.05; Fig. 2A). Functionally, the effects of miR-601 overexpression on HCC cell proliferative ability were analyzed by CCK-8 assays. As presented in Fig. 2B and C, the upregulation of miR-601 significantly decreased the proliferative ability of Hep3B and Huh7 cells compared with cells transfected with miR-NC at 48 and 72 h (P<0.05). In addition, invasion assays were performed to demonstrate the effects of miR-601 on HCC cell invasion. The results demonstrated that miR-601 upregulation notably decreased the invasive abilities of Hep3B and Huh7 cells; the number of invaded cells was significantly decreased compared with cells transfected with miR-NC (P<0.05; Fig. 2D). Therefore, miR-601 may exhibit tumor-suppressive effects in HCC.

PIK3R3 is a direct target gene of miR-601 in HCC cells

To elucidate the mechanisms associated with the tumor-suppressive roles of miR-601 in HCC, the potential targets of miR-601 were investigated using bioinformatics analysis. A conserved principal putative miR-601 binding site was predicted at the 3′-UTR of PIK3R3 (Fig. 3A). PIK3R3, a member of the PI3K family, was identified to be overexpressed in HCC and was closely associated with the development of HCC (24,25); therefore, PIK3R3 was selected for further analysis. In the present study, luciferase reporter assays were conducted to evaluate whether the 3′-UTR of PIK3R3 may be directly targeted by miR-601 in HCC cells. pGL3-PIK3R3-3′-UTR Wt or pGL3-PIK3R3-3′-UTR Mut was transfected into Hep3B and Huh7 cells, in addition to miR-601 mimics or miR-NC. miR-601 overexpression significantly decreased the luciferase activities of cell transfected with pGL3-PIK3R3-3′-UTR Wt compared with the control groups (P<0.05). However, ectopic miR-601 expression did not affect the luciferase activities of the plasmid possessing the PIK3R3 3′-UTR Mut in Hep3B and Huh7 cells (Fig. 3B). In addition, RT-qPCR and western blot analysis demonstrated that upregulation of miR-601 expression significantly decreased PIK3R3 mRNA (P<0.05; Fig. 3C) and protein (P<0.05; Fig. 3D) expression levels compared with the control. Collectively, these results demonstrated that PIK3R3 may be a direct target of miR-601 in HCC cells.

Upregulated PIK3R3 reverses the inhibitory effects of miR-601 in HCC cells

To further clarify whether PIK3R3 mediates decreases in miR-601-induced cell proliferation and invasion in HCC, PIK3R3 expression was restored in miR-601 mimics-transfected Hep3B and Huh7 cells by transfection with a PIK3R3 overexpression plasmid, pcDNA3.1-PIK3R3. Western blot analysis demonstrated that decreased PIK3R3 expression levels induced by miR-601 overexpression were restored in Hep3B and Huh7 cells following cotransfection with pcDNA3.1-PIK3R3 (P<0.05; Fig. 4A). Furthermore, CCK-8 and invasion assays demonstrated that restored PIK3R3 expression levels significantly eliminated the suppressive effects of miR-601 overexpression on proliferation (P<0.05; Fig. 4B) and invasion (P<0.05; Fig. 4C) of Hep3B and Huh7 cells compared with cells transfected with miR-601 mimics and pcDNA3.1. These results suggested that miR-601 may suppress cell proliferation and invasion of HCC, at least in part, by directly inhibiting PIK3R3 expression.

miR-601 inactivates the AKT/mTOR signaling pathway in HCC cells

Recent studies demonstrated that PIK3R3 is involved in the regulation of the AKT/mTOR signaling pathway (2528). Therefore, the present study investigated whether miR-601 may affect the AKT/mTOR signaling pathway in HCC cells. Hep3B and Huh7 cells were transfected with miR-601 mimics in combination with pcDNA3.1 or pcDNA3.1-PIK3R3. Following transfection for 72 h, western blot analysis was performed to detect p-AKT, AKT, p-mTOR and mTOR expression. As presented in Fig. 5, upregulated expression of miR-601 notably suppressed p-AKT and p-mTOR expression compared with the control and the cotransfected PIK3R3 overexpression groups; however, the expression levels of total AKT and mTOR appeared to be unaltered in Hep3B and Huh7 cells. Additionally, p-AKT and p-mTOR expression was recovered in Hep3B and Huh7 cells following cotransfection with pcDNA3.1-PIK3R3. These results suggested that miR-601 may inactivate the AKT/mTOR signaling pathway in HCC cells by regulating PIK3R3.

Discussion

Recently, miRNAs have been acknowledged as important regulators during hepatocarcinogenesis and tumor progression (2931). Therefore, identifying the underlying molecular mechanisms of miRNAs in the occurrence and development of HCC may improve the current understanding of the associated pathogenesis and aid the identification of potential therapeutic strategies for the treatment of HCC. The results of the RT-qPCR analysis in the present study demonstrated that miR-601 expression was significantly downregulated in HCC tissues and cell lines. Decreased miR-601 expression was additionally associated with TNM staging and lymph node metastasis. Upregulation of miR-601 expression decreased the proliferation and invasion of HCC cells. In addition, PIK3R3 was identified as a novel target of miR-601 in HCC cells. Furthermore, recovered PIK3R3 expression eliminated the suppressive effects of miR-601 overexpression in HCC cells in the present study. Additionally, the upregulation of miR-601 inactivated the AKT/mTOR signaling pathway in HCC cells via the inhibition of PIK3R3. Therefore, miR-601 may be considered a promising therapeutic target for patients with HCC.

Dysregulation of miR-601 is a frequent molecular event in numerous types of human cancer, and has been identified to be closely associated with the biological mechanism of tumor formation and development (1921). miR-601 was downregulated in the plasma of patients with colorectal cancer compared with healthy controls (19). Receiver operating characteristic curve analysis identified the value of miR-601 in the diagnosis of patients with colorectal cancer of an advanced stage (19). In breast cancer, expression levels of miR-601 were decreased in tumor tissues and cell lines. Low miR-601 expression levels were significantly correlated with distant metastasis and poor distant metastasis-free survival in breast cancer (20). Additionally, miR-601 expression levels were negatively correlated with the metastatic potential of breast cancer cells (20); functionally, miR-601 targets protein tyrosine phosphatase type IVA 1 to inhibit breast cancer cell growth and metastasis (20). Cao et al (21) observed that miR-601 is downregulated in pancreatic cancer, which was closely associated with metastasis. Ectopic miR-601 expression decreased cell proliferation and migration in pancreatic cancer via the inhibition of Sirtuin 1 (21). These results suggested that miR-601 may have the potential as a diagnostic and prognostic marker, in addition to a therapeutic target for the treatment of certain types of human cancer.

Identifying the targets of miR-601 in HCC is important for understanding the mechanisms underlying the effects of miR-601 on the onset and development of HCC. Detection of these targets may contribute to the development of effective therapeutic targets for patients with HCC. In the present study, PIK3R3 was identified as a direct target gene of miR-601 in HCC. PIK3R3 was observed to be overexpressed in various types of human cancer, including ovarian cancer (32), colorectal cancer (33), glioma (34) and lung cancer (35). A number of previous studies demonstrated that PIK3R3 may be involved in a variety of cellular processes, including cell growth, apoptosis, migration, invasion, metastasis and epithelial-mesenchymal transition (28,36,37). Hence, targeting PIK3R3 may represent as a novel therapeutic method for cancer treatment.

PIK3R3 expression levels were additionally increased in HCC tissues and cell lines (23,24). This upregulation has been closely associated with the occurrence and development of HCC by affecting cell growth, apoptosis, migration and invasion (24,25). The PI3K/AKT/mTOR signaling pathway serves important roles in the malignant transformation of numerous human cancer types and their subsequent rapid growth, apoptosis and metastasis (3840). PIK3R3, a member of the PI3K family, was observed to be negatively regulated by miRNAs and induced the inactivation of the PI3K/AKT/mTOR signaling pathway in a number of types of human cancer, including HCC (24,34,35). miR-132 was identified to target PIK3R3, deactivating the AKT/mTOR signaling pathway, and decreasing cell growth and motility in HCC (25). Xu et al (35) demonstrated that miR-7 overexpression suppressed TLR9 signaling-enhanced cell growth and metastasis in lung cancer by directly targeting PIK3R3 and affecting the PI3K/AKT signaling pathway. Zhu et al (34) observed that miR-365 inhibited the progression of glioma by downregulating PIK3R3 and regulating the AKT/mTOR signaling pathway. Collectively, the miR-601/PIK3R3 signaling pathway requires further investigation as a potential therapeutic target for the treatment of patients with HCC. However, the present study also has several limitations. First, the correlation between miR-601 and the prognosis of HCC patients was not examined. Second, the effects of miR-601 in the growth and metastasis of HCC cells in vivo were not explored. Finally, miR-601 inhibitor was not used to illustrate the effects of miR-601 downregulation in HCC cell proliferation and invasion. These limitations will be resolved in further investigations.

In conclusion, miR-601 was downregulated in HCC tissues and cell lines, and this dysregulation was significantly associated with TNM staging and lymph node metastasis. miR-601 overexpression inhibited cell proliferation and invasion of HCC, possibly by directly targeting PIK3R3 and regulating the AKT/mTOR signaling pathway. The results of the present study suggested that miR-601 may be considered a therapeutic target for the treatment of patients with HCC.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the Natural Science Foundation of China (grant no. 81703791), the Key Disciplines Group Construction Project of Pudong Health and Family Planning Commission of Shanghai (grant no. PWZxk2017-06), Shanghai Municipal Commission of Health and Family Planning (grant no. 20174Y0044), the Science and Technology Development Fund of Shanghai Pudong New Area (grant no. PKJ2017-Y13) and the Talents Training Program of the Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine (grant nos. XX2017-06 and XX2017-04).

Availability of data and materials

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

Authors' contributions

YY and WX designed this study. YS and SH performed RT-qPCR, CCK-8 assay and luciferase reporter assay. Invasion assay and western blot analysis were performed by JZ, GW, JN and SZ.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of The Seventh People's Hospital (Shanghai, China). Written informed consent was obtained from all patients prior to enrolment.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kew MC: Epidemiology of chronic hepatitis B virus infection, hepatocellular carcinoma, and hepatitis B virus-induced hepatocellular carcinoma. Pathol Biol (Paris). 58:273–277. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Ferlay J, Parkin DM and Steliarova-Foucher E: Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer. 46:765–781. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Nguyen VT, Law MG and Dore GJ: Hepatitis B-related hepatocellular carcinoma: Epidemiological characteristics and disease burden. J Viral Hepat. 16:453–463. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Sun VC and Sarna L: Symptom management in hepatocellular carcinoma. Clin J Oncol Nurs. 12:759–766. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Venook AP, Papandreou C, Furuse J and de Guevara LL: The incidence and epidemiology of hepatocellular carcinoma: A global and regional perspective. Oncologist. 4:5–13. 2010. View Article : Google Scholar

7 

Yang LY, Fang F, Ou DP, Wu W, Zeng ZJ and Wu F: Solitary large hepatocellular carcinoma: A specific subtype of hepatocellular carcinoma with good outcome after hepatic resection. Ann Surg. 249:118–123. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Zhu AX, Duda DG, Sahani DV and Jain RK: HCC and angiogenesis: Possible targets and future directions. Nat Rev Clin Oncol. 8:292–301. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Herranz H and Cohen SM: MicroRNAs and gene regulatory networks: Managing the impact of noise in biological systems. Genes Dev. 24:1339–1344. 2010. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

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

12 

Qu Z, Wu J, Wu J, Ji A, Qiang G, Jiang Y, Jiang C and Ding Y: Exosomal miR-665 as a novel minimally invasive biomarker for hepatocellular carcinoma diagnosis and prognosis. Oncotarget. 8:80666–80678. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Yu L, Chen J, Liu Y, Zhang Z and Duan S: MicroRNA-937 inhibits cell proliferation and metastasis in gastric cancer cells by downregulating FOXL2. Cancer Biomark. 21:105–116. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Li G, Wu F, Yang H, Deng X and Yuan Y: MiR-9-5p promotes cell growth and metastasis in non-small cell lung cancer through the repression of TGFBR2. Biomed Pharmacother. 96:1170–1178. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Wang X and Wu X: The Role of MicroRNA-1207-5p in colorectal cancer. Clin Lab. 63:1875–1882. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Chen CZ: MicroRNAs as oncogenes and tumor suppressors. N Engl J Med. 353:1768–1771. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Kong YW, Ferland-McCollough D, Jackson TJ and Bushell M: MicroRNAs in cancer management. Lancet Oncol. 13:e249–e258. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Wang X, Hu G and Zhou J: Repression of versican expression by microRNA-143. J Biol Chem. 285:23241–23250. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Wang Q, Huang Z, Ni S, Xiao X, Xu Q, Wang L, Huang D, Tan C, Sheng W and Du X: Plasma miR-601 and miR-760 are novel biomarkers for the early detection of colorectal cancer. PLoS One. 7:e443982012. View Article : Google Scholar : PubMed/NCBI

20 

Hu JY, Yi W, Wei X, Zhang MY, Xu R, Zeng LS, Huang ZJ and Chen JS: miR-601 is a prognostic marker and suppresses cell growth and invasion by targeting PTP4A1 in breast cancer. Biomed Pharmacother. 79:247–253. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Cao W, Jin H, Zhang L, Chen X and Qian H: Identification of miR-601 as a novel regulator in the development of pancreatic cancer. Biochem Biophys Res Commun. 483:638–644. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Qiao CX, Zhai XF, Ling CQ, Lang QB, Dong HJ, Liu Q and Li MD: Health-related quality of life evaluated by tumor node metastasis staging system in patients with hepatocellular carcinoma. World J Gastroenterol. 18:2689–2694. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

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. 36:4453–4459. 2015. View Article : Google Scholar : PubMed/NCBI

25 

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. Int J Oncol. 47:1585–1593. 2015. View Article : Google Scholar : PubMed/NCBI

26 

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 : PubMed/NCBI

27 

Zhang HD, Sun DW, Mao L, Zhang J, Jiang LH, Li J, Wu Y, Ji H, Chen W, Wang J, et al: MiR-139-5p inhibits the biological function of breast cancer cells by targeting Notch1 and mediates chemosensitivity to docetaxel. Biochem Biophys Res Commun. 465:702–713. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Zhao J, Xu J and Zhang R: MicroRNA-411 inhibits malignant biological behaviours of colorectal cancer cells by directly targeting PIK3R3. Oncol Rep. 39:633–642. 2018.PubMed/NCBI

29 

Long XD, Huang XY, Yao JG, Liao P, Tang YJ, Ma Y and Xia Q: Polymorphisms in the precursor microRNAs and aflatoxin B1-related hepatocellular carcinoma. Mol Carcinog. 55:1060–1072. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Wu XM, Xi ZF, Liao P, Huang HD, Huang XY, Wang C, Ma Y, Xia Q, Yao JG and Long XD: Diagnostic and prognostic potential of serum microRNA-4651 for patients with hepatocellular carcinoma related to aflatoxin B1. Oncotarget. 8:81235–81249. 2017.PubMed/NCBI

31 

Liu L, Cai X, Liu E, Tian X and Tian C: MicroRNA-18a promotes proliferation and metastasis in hepatocellular carcinoma via targeting KLF4. Oncotarget. 8:68263–68269. 2017.PubMed/NCBI

32 

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

33 

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

34 

Zhu Y, Zhao H, Rao M and Xu S: MicroRNA-365 inhibits proliferation, migration and invasion of glioma by targeting PIK3R3. Oncol Rep. 37:2185–2192. 2017. View Article : Google Scholar : PubMed/NCBI

35 

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 : PubMed/NCBI

36 

Sun J and Fu L: IL-6 promotes gastric cancer cell proliferation and EMT through regulating miR-152/PIK3R3 pathway. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 42:1241–1247. 2017.(In Chinese). PubMed/NCBI

37 

Dong L and Hui L: HOTAIR promotes proliferation, migration, and invasion of ovarian cancer SKOV3 cells through regulating PIK3R3. Med Sci Monit. 22:325–331. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Mabuchi S, Kuroda H, Takahashi R and Sasano T: The PI3K/AKT/mTOR pathway as a therapeutic target in ovarian cancer. Gynecol Oncol. 137:173–179. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Asati V, Mahapatra DK and Bharti SK: PI3K/Akt/mTOR and Ras/Raf/MEK/ERK signaling pathways inhibitors as anticancer agents: Structural and pharmacological perspectives. Eur J Med Chem. 109:314–341. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Samarin J, Laketa V, Malz M, Roessler S, Stein I, Horwitz E, Singer S, Dimou E, Cigliano A, Bissinger M, et al: PI3K/AKT/mTOR-dependent stabilization of oncogenic far-upstream element binding proteins in hepatocellular carcinoma cells. Hepatology. 63:813–826. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 19 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song Y, He S, Zhuang J, Wang G, Ni J, Zhang S, Ye Y and Xia W: MicroRNA‑601 serves as a potential tumor suppressor in hepatocellular carcinoma by directly targeting PIK3R3 Retraction in /10.3892/mmr.2022.12923. Mol Med Rep 19: 2431-2439, 2019
APA
Song, Y., He, S., Zhuang, J., Wang, G., Ni, J., Zhang, S. ... Xia, W. (2019). MicroRNA‑601 serves as a potential tumor suppressor in hepatocellular carcinoma by directly targeting PIK3R3 Retraction in /10.3892/mmr.2022.12923. Molecular Medicine Reports, 19, 2431-2439. https://doi.org/10.3892/mmr.2019.9857
MLA
Song, Y., He, S., Zhuang, J., Wang, G., Ni, J., Zhang, S., Ye, Y., Xia, W."MicroRNA‑601 serves as a potential tumor suppressor in hepatocellular carcinoma by directly targeting PIK3R3 Retraction in /10.3892/mmr.2022.12923". Molecular Medicine Reports 19.3 (2019): 2431-2439.
Chicago
Song, Y., He, S., Zhuang, J., Wang, G., Ni, J., Zhang, S., Ye, Y., Xia, W."MicroRNA‑601 serves as a potential tumor suppressor in hepatocellular carcinoma by directly targeting PIK3R3 Retraction in /10.3892/mmr.2022.12923". Molecular Medicine Reports 19, no. 3 (2019): 2431-2439. https://doi.org/10.3892/mmr.2019.9857