MicroRNA-92a contributes to tumor growth of human hepatocellular carcinoma by targeting FBXW7

Corrigendum in: /10.3892/or.2023.8596

  • Authors:
    • Wei Yang
    • Changwei Dou
    • Yufeng Wang
    • Yuli Jia
    • Chao Li
    • Xin Zheng
    • Kangsheng Tu
  • View Affiliations

  • Published online on: August 19, 2015     https://doi.org/10.3892/or.2015.4210
  • Pages: 2576-2584
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Deregulation of microRNA-92a (miR-92a) has been reported in several human cancers and is associated with prognosis of patients. However, the clinical significance of miR-92a and the underlying mechanisms involved in hepatocarcinogenesis remain to be determined. The aim of the present study was to determine the role of miR-92a in hepatocellular carcinoma (HCC). The results showed that the expression of miR-92a was upregulated in HCC tissues as compared with matched tumor-adjacent tissues. A high expression of miR-92a was observed in HCC cell lines as compared with a non-transformed hepatic cell line. The gain- and loss-of-function studies revealed that miR-92a significantly promoted proliferation and cell cycle transition from G1 to S phase, and inhibited apoptosis of HCC cell in vitro. In tumor‑bearing nude mice, the downregulation of miR-92a suppressed tumor growth of HCC in vivo. miR-92a was inversely correlated with F-box and WD repeat domain-containing 7 (FBXW7) expression in HCC tissues. Furthermore, miR-92a negatively regulated FBXW7 abundance in HCC cells. In the present study, FBXW7 was identified as a direct target of miR-92a. Notably, alterations of FBXW7 expression abrogated the effects of miR-92a on HCC cell proliferation, cell cycle and apoptosis. Clinical association analysis revealed that a high expression of miR-92a was correlated with poor prognostic characteristics of HCC. Notably, the high expression of miR-92a conferred a reduced 5-year overall survival (OS) and recurrence-free survival (RFS) of HCC patients. The multivariate Cox regression analysis demonstrated that miR-92a expression was an independent prognostic marker for predicting survival of HCC patients. In conclusion, the results of the present study suggested that miR-92a promotes the tumor growth of HCC by targeting FBXW7 and may serve as a novel prognostic biomarker and therapeutic target for HCC.

Introduction

MicroRNAs (miRNAs) are a group of endogenous small non-coding RNA molecules, which regulate protein-coding gene expression by interacting with complementary sites within the 3′-untranslated region (UTR) of target mRNAs and targeting mRNAs for cleavage or translational repression (1). Mounting evidence has indicated that miRNAs are involved in tissue morphogenesis, cell processes such as proliferation, cell cycle and apoptosis and major signaling pathways (24). Previous studies have identified the critical role of miRNAs in human cancers and suggest that deregulation of miRNAs is involved in tumor development and progression by modulating the expression of oncogenes or tumor suppressors (5).

miR-92a, which belongs to the miR-17-92 cluster, plays a critical role in the progression of lung cancer (6), esophageal squamous cell carcinoma (7), colorectal (8), breast (9), ovarian (10) and cervical cancer (11). Previous findings show that miR-92a functions as a regulator of cell proliferation, apoptosis and invasion in human cancers (1113). miR-92a promotes lung cancer cell invasion by targeting reversion-inducing-cysteine-rich protein with Kazal motifs (RECK) and a high expression of miR-92a is associated with the poor survival rate of lung cancer patients (6,13). Plasma miR-92a is a promising novel biomarker for the early detection of colorectal cancer and its overexpression is also a prognostic marker for predicting the poor overall and disease-free survival rate of patients (8,14). However, the expression of miR-92a is downregulated in ovarian and breast cancer (9,10). Upregulation of miR-92a inhibits ovarian cancer cell adhesion, invasion and proliferation by suppressing integrin α5 expression (10). Downregulation of miR-92a is associated with aggressive breast cancer features and increased tumor macrophage infiltration (9). Thus, the functional significance of miR-92a in cancer initiation and development seems to be cancer-type specific. Previously, miR-92a was found to be significantly upregulated in hepatocellular carcinoma (HCC) samples and was also identified as hepatitis B virus (HBV)-specific (15,16). The proliferation of HCC-derived cell lines was enhanced by the upregulation of miR-92a and inhibited by the downregulation of miR-92a (16). However, the clinical significance of miR-92a and the underlying mechanisms involved in the development of HCC remain to be investigated.

The present study aim was to determine the role of miR-92a in HCC. The results showed that the expression of miR-92a was upregulated in HCC tissues. miR-92a promoted proliferation, cell cycle and apoptosis resistance in vitro. Moreover, the downregulation of miR-92a inhibited the tumor growth of HCC in vivo. Notably, F-box and WD repeat domain-containing 7 (FBXW7) was identified as a direct target of miR-92a. A high expression of miR-92a was associated with poor clinicopathological characteristics and the reduced survival of HCC patients. The results showed a new role for miR-92a in prediction of prognosis and promoting tumor growth of human HCC.

Materials and methods

Clinical samples

HCC and matched normal tumor-adjacent tissues were obtained from 106 patients including 94 males and 12 females, who underwent resection of their primary HCC in the Department of Hepatobiliary Surgery at the First Affiliated Hospital of Xi'an Jiaotong University (Shaanxi, China) from January, 2006 to December, 2009, with a median follow-up period of 38,7 months. None of the patients received preoperative chemo- or radiotherapy. The stage of cancer was determined according to the cancer staging system published in 2010 by the Union for International Cancer Control (UICC). The demographic features and clinicopathological data are shown in Table I. Samples were used after informed consent was obtained. The Xi'an Jiaotong University Ethics Committee approved all the protocols according to the Declaration of Helsinki (as revised in Tokyo 2004).

Table I

Clinicopathological correlation of miR-92a expression in HCC.

Table I

Clinicopathological correlation of miR-92a expression in HCC.

Clinicopathological characteristicsmiR-92a
P-value
High expression (n=53)Low expression (n=53)
Age (years)
 ≤5020220.691
 >503331
Gender
 Male46480.540
 Female75
HBsAg
 No3110.022a
 Yes5042
Serum AFP level (ng/ml)
 ≤2014180.397
 >203935
Tumor size (cm)
 ≤518300.019a
 >53523
No. of tumor nodules
 143450.605
 ≥2108
Cirrhosis
 Absent7170.020a
 Present4636
Venous infiltration
 Absent24270.560
 Present2926
Edmondson-Steiner grading
 I+II33470.002a
 III+IV206
TNM tumor stage
 I+II34450.014a
 III+IV198

a Statistically significant. HCC, hepatocellular carcinoma; HBV, hepatitis B virus; AFP, α-fetoprotein; TNM, tumor-node-metastasis.

Reverse transcription-quantitative PCR (RT-qPCR)

qPCR primer against mature miRNA hsa-miR-92a-3p (HmiRQP0832) and Homo sapiens snRNA U6 qPCR Primer (HmiRQP9001) were purchased from Genecopoeia (Guangzhou, China). The PCR amplification for the quantification of the miR-92a and U6 was performed using the TaqMan miRNA Reverse Transcription kit (Applied Biosystems, Foster City, CA, USA) and TaqMan Human miRNA Assay kit (Applied Biosystems). The relative expression of miR-92a was shown as the fold difference relative to U6.

The PCR amplification for the quantification of the FBXW7 and GAPDH mRNAs was performed using an ABI PRISM 7300 Sequence Detection system (Applied Biosystems) and a SYBR® Premix Ex Taq™ II (Perfect Real Time) kit (Takara Bio, Shiga, Japan), as previously reported (17).

Cell lines and transfection

The human immortalized normal hepatic cell line LO2 and five HCC cell lines (HepG2, Hep3B, Huh7, SMMC-7721 and Bel-7402) were purchased from the Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China. The cells were maintained in Dulbecco's modified Eagle's medium (DMEM; Gibco, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (Gibco) with 100 U/ml penicillin and 100 µg/ml streptomycin (Sigma, St. Louis, MO, USA) at 37°C with 5% CO2.

miRNA vectors, including miR-92a expression vector (HmiR0204-MR04), the control vector for miR-92a (CmiR0001-MR04 and miR-control), miR-92a inhibitor (HmiR-AN0832-AM04 and anti-miR-92a) and the negative control for the miR-92a inhibitor (CmiR-AN0001-AM04 and anti-miR-NC), were purchased from Genecopoeia. Retroviral vectors pMMP-FBXW7 were generated by inserting the FBXW7 cDNA into pMMP. Retrovirus packaging and transduction were performed as previously described (18). The targeted sequences for FBXW7 siRNA sense, 5′-GGA GUA UGG UCA UCA CAA Att-3′ and antisense, 5′-UUU GUG AUG ACC AUA CUC Cac-3′ or a non-specific duplex oligonucleotide as a negative control were produced by Sangon Biotech Co., Ltd. (Shanghai, China). The cells were transfected with the vectors mentioned above using Lipofectamine 2000 according to the manufacturer's instructions (Invitrogen, Carlsbad, CA, USA).

Cell cycle, proliferation and detection of apoptosis

Flow cytometry was performed using the fluorescence-activated cell sorting (FACS) Calibur and Cell Quest software (both from Becton-Dickinson, San Jose, CA, USA). For the proliferation assay, HCC cells transfected with different vectors were seeded in 96-well plates at a density of 5×103 cells/well for 24 h and assessed using Cell Proliferation ELISA, BrdU kit (5-bromodeoxyuridine) (chemiluminescent) (Roche, Indianapolis, IN, USA). For cell cycle analysis, the cells were seeded in 6-well plates at 2×105/well. Forty-eight hours after transfection, the cells were fixed in 70% ethanol at 4°C for 24 h and stained with 50 µg/ml propidium iodide (Keygen, Nanjing, China). An Annexin-V-Fluos Staining kit (Roche) was used to analyze apoptosis levels, as previously described (17).

Western blot analysis

The primary antibodies used in the immunoblotting assays were: FBXW7 (WH0055294M2; Sigma) and GAPDH (G8140; US Biological, Swampscott, MA, USA). Horseradish peroxidase (HRP)-conjugated sheep anti-mouse secondary antibodies (Bio-Rad, Hercules, CA, USA) were used at a 1:1,000–1:5,000 dilution and detected using a Western Blotting Luminol Reagent (sc-2048; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), as described in a previous study (18).

Immunohistochemical staining

Immunohistochemistry was performed on paraformaldehyde-fixed paraffin sections. FBXW7 (WH0055294M2; Sigma) antibody was used in immunohistochemistry using a streptavidin peroxidase-conjugated (SP-IHC) method. Immunohistochemistry was performed as previously reported (19). The percentage of positive tumor cells was graded as: 0, <10%; 1, 10–30%; 2, 31–50%; 3, >50%.

Luciferase reporter assay

The predicted 3′-UTR sequence of FBXW7 that interacted with miR-92a, together with a corresponding mutated sequence within the predicted target sites, were created and inserted into the pRL-TK control vector (Promega, Madison, WI, USA). SMMC-7721 cells that were seeded in a 96-well plate were transfected with 120 ng miR-92a expression vector, miR-92a inhibitor, control vector or negative control. Cells were co-transfected with 30 ng of the wild-type or mutant 3′-UTR of FBXW7 mRNA. Transfections were performed using 0.45 µl of Fugene (Promega). Fourty eight hours after transfection, the cells were collected and measured according to the manufacturer's instructions (Dual-Luciferase Assay System; Promega). The pRL-TK expressing Renilla luciferase was cotransfected as an internal control to correct the differences in transfection and harvest efficiencies (20).

In vivo experiments

Four-to-six week-old female BALB/c nude mice (Centre of Laboratory Animals, The Medical College of Xi'an Jiaotong University, Xi'an, China) were used to establish the nude mouse xenograft model. SMMC-7721 (5×106) cells that were transfected with anti-miR-92a or -miR-NC vectors were mixed in 150 µl of Matrigel and were inoculated subcutaneously into the flank of nude mice. Tumor growth curves were generated as previously described (18). Animal protocols were approved by the Institutional Animal Care and Use Committee of Xi'an Jiaotong University.

Statistical analysis

Data are presented as the mean ± SEM from at least three independent replicates. Where appropriate, a Pearson's Chi-squared test, a Kaplan-Meier plot, a log-rank test, the multi-variant Cox regression analysis, a Spearman's rank correlation coefficient and a two-tailed Student's t-test were used with the SPSS statistical package for Windows Version 13 (SPSS, Chicago, IL, USA) or GraphPad Prism 5 software (GraphPad Software, Inc, San Diego, CA, USA). P≤0.05 was considered significant.

Results

Elevated expression of miR-92a is observed in HCC tissues and cells

One hundred and six pairs of HCC tissues and matched tumor-adjacent tissues were tested for miR-92a using RT-qPCR. The results showed that the mean level of miR-92a expression in HCC tissues was significantly higher than that in the non-tumor tissues (P<0.001, Fig. 1A). We then analyzed miR-92a expression in a non-transformed hepatic cell line (LO2) and a panel of HCC cell lines (HepG2, Huh7, Hep3B, SMCC-7721 and Bel-7402). As expected, the relative expression of miR-92a was obviously upregulated in the HCC cell lines as compared with that in the non-transformed LO2 hepatic cell line (P<0.05, Fig. 1B). The results suggested that an elevated expression of miR-92a may facilitate hepatocarcinogenesis.

Promotive effects of miR-92a on proliferation, cell cycle and apoptosis of HCC cells

To identify the biological function of miR-92a in HCC, we transduced an miR-92a expression plasmid or anti-miR-92a vector into human HCC cell lines with different endogenous expression levels of miR-92a. As measured by RT-qPCR, the expression of miR-92a was significantly altered by the corresponding vector in HCC cells (P<0.05, respectively, Fig. 2A). BrdU incorporation assays were performed to determine the effect of altering miR-92a levels on HCC cell proliferation. We found that the downregulation of miR-92a led to a significant reduction of cell proliferation in SMMC-7721 cells (P<0.05, Fig. 2B). Furthermore, as determined by flow cytometric analysis, the downregulation of miR-92a resulted in G1 phase arrest in SMMC-7721 cells (P<0.05, Fig. 2C). Otherwise, the percentage of apoptotic SMMC-7721 cells was significantly increased following the reduction of miR-92a (P<0.001, Fig. 2D). By contrast, the upregulation of miR-92a promoted the proliferation, cell cycle transition from G1 to S phase and apoptosis resistance in Hep3B cells (P<0.05, respectively, Fig. 2B–D). Notably, the tumor growth curve revealed that knockdown of miR-92a significantly retarded the tumor growth of human HCC in subcutaneous nude mouse models (P<0.001, Fig. 3). These results demonstrated that miR-92a regulates the proliferation, cell cycle and apoptosis of HCC cells.

FBXW7 is a direct downstream target of miR-92a

A search for the candidate target genes of miR-92a was conducted using publically available databases TargetScan 6.2 (http://www.targetscan.org/) and miRanda (microrna.org and miRbase). Of note, the complementary sequence of miR-92a was identified in the 3′-UTR of FBXW7 mRNA, which was selected for subsequent studies (Fig. 4A). One hundred and six samples of HCC tissues were subjected to RT-qPCR and immunostaining for FBXW7 expression. The expression levels of FBXW7 mRNA and protein in the miR-92a high-expressing tumors were significantly lower than those in the miR-92a low-expressing tumors (P<0.05, respectively, Fig. 4B and C). Notably, an obvious inverse correlation between mRNA levels of miR-92a and FBXW7 (r= 0.745, P<0.001) and between miR-92a mRNA and FBXW7 protein (r=−0.632, P<0.001) was revealed by Spearman's correlation analysis in HCC tissues. Furthermore, the expression levels of FBXW7 mRNA and protein were significantly increased by the downregulation of miR-92a in SMMC-7721 cells (P<0.05, respectively, Fig. 5A and B). By contrast, the overexpression of miR-92a markedly reduced the mRNA and protein levels of FBXW7 in Hep3B cells (P<0.05, respectively, Fig. 5A and B). In addition, the upregulation of miR-92a prominently inhibited the luciferase activity of FBXW7 containing a wild-type (wt) 3′-UTR but did not suppress the activity of FBXW7 with a mutant (mt) 3′-UTR (P<0.001, Fig. 5C). Suppression of miR-92a by anti-miR-92a increased the luciferase activity of wt FBXW7 3′-UTR (P<0.001, Fig. 5C). However, with the mt FBXW7 3′-UTR constructs, there was no relative increase in activity. Collectively, these results strongly suggested that FBXW7 is a downstream target of miR-92a in HCC.

Alterations of FBXW7 levels influence the effects of miR-92a on HCC cells

To confirm that FBXW7 is a functional target of miR-92a, FBXW7 was knocked down by a specific siRNA in miR-92a-suppressive SMMC-7721 cells (P<0.05, Fig. 6A). We found that cell proliferation was significantly increased by FBXW7 knockdown (P<0.05, Fig. 6B). Furthermore, FBXW7 knockdown markedly rescued the downregulation of miR-92a-induced cell cycle arrest and apoptosis (P<0.05, respectively, Fig. 6C and D). Similarly, FBXW7 overexpression inhibited cell proliferation and promoted G1 phase arrest and apoptosis in miR-92a-overexpressing Hep3B cells (P<0.05, respectively, Fig. 6A–D). The results showed that FBXW7 is a downstream mediator of miR-92a in HCC.

Clinical significance of miR-92a expression in HCC cases

The expression of miR-92a was considered as low (n=53) or high (n=53) in accordance with the cut-off value, which was defined as the median of the cohort. The association between clinicopathological characteristics and miR-92a expression was subsequently analyzed. As shown in Table I, a high expression of miR-92a was evidently associated with HBV infection (P=0.022), large tumor size (P=0.019), cirrhosis (P=0.020), high Edmondson-Steiner grading (P=0.002) and advanced tumor-node-metastasis (TNM) tumor stage (P=0.014). Furthermore, the prognostic value of miR-92a was analyzed by Kaplan-Meier estimation. A high expression of miR-92a conferred a poor overall survival (OS) and recurrence-free survival (RFS) of HCC patients (P=0.001, respectively, Fig. 7). The multi-variant Cox regression analysis revealed that miR-92a expression was an independent prognostic marker for predicting 5-year OS and RFS in HCC patients (P=0.026 and P=0.042, respectively, Table II). These results showed that miR-92a is a potent biomarker for predicting prognosis of HCC patients.

Table II

Multivariate Cox regression analysis of 5-year OS and RFS of 106 HCC patients.

Table II

Multivariate Cox regression analysis of 5-year OS and RFS of 106 HCC patients.

VariablesOS
RFS
HR95% CIP-valueHR95% CIP-value
Age0.8530.512–1.440.5230.8690.558–1.3620.552
Gender1.3730.587–3.1920.4540.7810.424–1.4790.455
HBV0.9340.396–2.2130.8690.9780.463–2.1560.996
No. of tumor nodules1.7610.966–3.1920.0631.6841.002–2.8490.051
Tumor size3.5982.041–6.352<0.001a2.5711.613–4.076<0.001a
Venous infiltration4.0422.362–6.931<0.001a3.5312.159–5.782<0.001a
Serum AFP level1.5540.872–2.7550.1371.4630.883–2.4200.140
Cirrhosis1.0080.561–1.7980.9641.0530.631–1.7530.851
Edmondson-Steiner grading1.2410.702–2.1860.4501.1320.678–1.8880.639
TNM tumor stage2.8011.348–5.8140.006a2.1981.030–4.6730.032a
miR-92a expression2.2831.104–4.7170.026a3.7061.079–5.1550.042a

a Statistically significant. OS, overall survival; RFS, recurrence-free survival; HBV, hepatitis B virus; AFP, α-fetoprotein; TNM, tumor-node-metastasis; HR, hazard ratio; CI, confidence interval.

Discussion

In previous studies, the deregulation of miR-92a has been studied in various human types of cancer and it is a prognostic marker for predicting survival of patients (6,7,9,11,14,16). miR-92a has been found to regulate HCC cell proliferation (16), facilitate G1/S transition (11,21), mediate apoptosis (21) and promote in vivo tumor growth (22). The clinical significance of miR-92a and the molecular mechanisms by which miR-92a promotes HCC development remain unclear. In the present study, we identified that the mean level of miR-92a expression was obviously increased in HCC tissues vs. matched tumor-adjacent tissues. Elevated expression of miR-92a was observed in the HCC cell lines as compared with the normal hepatic cell line. The role of miR-92a deregulation in tumor growth was examined via in vitro and in vivo experiments. We found that the downregulation of miR-92a inhibited HCC cell proliferation and induced G1 phase arrest and apoptosis in vitro and suppressed in vivo tumor growth. By contrast, the overexpression of miR-92a promoted proliferation and cell cycle transition to S phase and inhibited apoptosis in vitro. The results suggested that miR-92a is a novel tumor-suppressive miRNA that plays a critical role in the regulation of tumor growth in HCC.

Results of the present study suggest that miR-92a promoted tumor growth, at least in part, by targeting FBXW7. Firstly, miR-92a was inversely correlated with the levels of both FBXW7 mRNA and protein in HCC tissues. Secondly, miR-92a negatively regulated FBXW7 abundance in HCC cells. Thirdly, the complementary sequence of miR-92a was identified in the 3′-UTR of FBXW7 mRNA. Knockdown of miR-92a increased the luciferase reporter activity of wt 3′-UTR but not mt 3′-UTR of FBXW7. Conversely, the overexpression of miR-92a decreased the luciferase activity of wt 3′-UTR but not mt 3′-UTR of FBXW7. The effects of miR-92a alteration on proliferation, cell cycle and apoptosis of HCC cells were also abolished by FBXW7 modulation. In addition, FBXW7 has been reported to be a direct target of miR-92a in cervical cancer (11). Collectively, our results support FBXW7 as a downstream mediator of miR-92a function in HCC.

Our previous findings have shown that FBXW7 is a potent tumor suppressor and inhibits tumor growth of HCC by inducing growth arrest and apoptosis in vitro and in vivo (1719). FBXW7 is a well-known E3 ligase and recognizes target proteins for ubiquitination and degradation. Several oncoproteins have been identified as substrates of FBXW7 including c-Myc (23) and Cyclin E (24). Impaired expression of FBXW7 leads to the accumulation of these oncoproteins, which promote cancer cell proliferation, cell cycle progression and apoptosis resistance. c-Myc protein plays crucial roles in mitogenic signaling and cell growth responses (25) and Cyclin E is a key component of the cell cycle machinery (26). Active Cyclin E-Cdk2 complex is required for G1 and S-phase transition, modulating pRb and thereby activating E2F transcription factors that enable DNA replication (27). It has been reported that c-Myc can induce apoptosis by Caspase-3-dependent and caspase-independent signaling (28). Upregulation of c-Myc and Cyclin E have been reported in HCC and lead to rapid tumor growth in vitro and in vivo (29,30). Thus, upregulation of c-Myc and Cyclin E may be a mechanism by which miR-92a promotes the proliferation and cell cycle transition from G1 to S phase and inhibits apoptosis of HCC. Alternatively, our previous results showed that Yes-associated protein (YAP) is a potential target of FBXW7 and is involved in FBXW7-induced growth arrest and apoptosis of HCC (18). It has been shown that YAP plays critical role in hepatocarcinogenesis (31). This mechanism may account for the effects of miR-92a in HCC.

The role of miR-92a in cancer cell proliferation, cell cycle and apoptosis suggests its potential application as a prognostic biomarker of HCC patients. Furthermore, our previous findings showed that a low expression of FBXW7 was an independent prognostic marker for predicting the survival of HCC patients (18). Thus, we demonstrated, for the first time, that high expression of miR-92a was associated with poor prognostic characteristics including HBV infection, large tumor size, cirrhosis, high Edmondson-Steiner grading and advanced TNM tumor stage. We also confirmed that a high expression of miR-92a was an independent prognostic marker for predicting 5-year OS and RFS of HCC patients.

In conclusion, the results show that the expression of miR-92a is reduced in HCC tissues and cell lines. We confirm that miR-92a is an independent prognostic marker for HCC. In vitro and in vivo studies revealed that miR-92a functions as a novel oncomiRNA by promoting proliferation, cell cycle transition and apoptosis resistance of HCC. Its multiple tumor-promotive effects are mediated by FBXW7. Collectively, the deregulation of miR-92a may play an important role in tumor growth and may be a novel prognostic factor and potential therapeutic target for HCC.

Acknowledgments

The present study was supported by a grant from the National Natural Science Foundation of China (no. 81402039).

References

1 

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

2 

Kloosterman WP and Plasterk RH: The diverse functions of microRNAs in animal development and disease. Dev Cell. 11:441–450. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Szabo G and Bala S: MicroRNAs in liver disease. Nat Rev Gastroenterol Hepatol. 10:542–552. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Mendell JT: MicroRNAs: Critical regulators of development, cellular physiology and malignancy. Cell Cycle. 4:1179–1184. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Tu K, Zheng X, Dou C, Li C, Yang W, Yao Y and Liu Q: MicroRNA-130b promotes cell aggressiveness by inhibiting peroxisome proliferator-activated receptor gamma in human hepatocellular carcinoma. Int J Mol Sci. 15:20486–20499. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Ranade AR, Cherba D, Sridhar S, Richardson P, Webb C, Paripati A, Bowles B and Weiss GJ: MicroRNA 92a-2*: A biomarker predictive for chemoresistance and prognostic for survival in patients with small cell lung cancer. J Thorac Oncol. 5:1273–1278. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Chen ZL, Zhao XH, Wang JW, Li BZ, Wang Z, Sun J, Tan FW, Ding DP, Xu XH, Zhou F, et al: microRNA-92a promotes lymph node metastasis of human esophageal squamous cell carcinoma via E-cadherin. J Biol Chem. 286:10725–10734. 2011. View Article : Google Scholar :

8 

Wu CW, Ng SS, Dong YJ, Ng SC, Leung WW, Lee CW, Wong YN, Chan FK, Yu J and Sung JJ: Detection of miR-92a and miR-21 in stool samples as potential screening biomarkers for colorectal cancer and polyps. Gut. 61:739–745. 2012. View Article : Google Scholar

9 

Nilsson S, Möller C, Jirström K, Lee A, Busch S, Lamb R and Landberg G: Downregulation of miR-92a is associated with aggressive breast cancer features and increased tumour macrophage infiltration. PLoS One. 7:e360512012. View Article : Google Scholar : PubMed/NCBI

10 

Ohyagi-Hara C, Sawada K, Kamiura S, Tomita Y, Isobe A, Hashimoto K, Kinose Y, Mabuchi S, Hisamatsu T, Takahashi T, et al: miR-92a inhibits peritoneal dissemination of ovarian cancer cells by inhibiting integrin α5 expression. Am J Pathol. 182:1876–1889. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Zhou C, Shen L, Mao L, Wang B, Li Y and Yu H: miR-92a is upregulated in cervical cancer and promotes cell proliferation and invasion by targeting FBXW7. Biochem Biophys Res Commun. 458:63–69. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Ranji N, Sadeghizadeh M, Shokrgozar MA, Bakhshandeh B, Karimipour M, Amanzadeh A and Azadmanesh K: MiR-17-92 cluster: An apoptosis inducer or proliferation enhancer. Mol Cell Biochem. 380:229–238. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Lin HY, Chiang CH and Hung WC: STAT3 upregulates miR-92a to inhibit RECK expression and to promote invasiveness of lung cancer cells. Br J Cancer. 109:731–738. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Ke TW, Wei PL, Yeh KT, Chen WT and Cheng YW: MiR-92a Promotes Cell Metastasis of Colorectal Cancer Through PTEN-Mediated PI3K/AKT Pathway. Ann Surg Oncol. 22:2649–2655. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Li LM, Hu ZB, Zhou ZX, Chen X, Liu FY, Zhang JF, Shen HB, Zhang CY and Zen K: Serum microRNA profiles serve as novel biomarkers for HBV infection and diagnosis of HBV-positive hepatocarcinoma. Cancer Res. 70:9798–9807. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Shigoka M, Tsuchida A, Matsudo T, Nagakawa Y, Saito H, Suzuki Y, Aoki T, Murakami Y, Toyoda H, Kumada T, et al: Deregulation of miR-92a expression is implicated in hepatocellular carcinoma development. Pathol Int. 60:351–357. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Tu K, Zheng X, Zhou Z, Li C, Zhang J, Gao J, Yao Y and Liu Q: Recombinant human adenovirus-p53 injection induced apoptosis in hepatocellular carcinoma cell lines mediated by p53-Fbxw7 pathway, which controls c-Myc and cyclin E. PLoS One. 8:e685742013. View Article : Google Scholar : PubMed/NCBI

18 

Tu K, Yang W, Li C, Zheng X, Lu Z, Guo C, Yao Y and Liu Q: Fbxw7 is an independent prognostic marker and induces apoptosis and growth arrest by regulating YAP abundance in hepatocellular carcinoma. Mol Cancer. 13:1102014. View Article : Google Scholar : PubMed/NCBI

19 

Tu K, Zheng X, Zan X, Han S, Yao Y and Liu Q: Evaluation of Fbxw7 expression and its correlation with the expression of c-Myc, cyclin E and p53 in human hepatocellular carcinoma. Hepatol Res. 42:904–910. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Liu Z, Tu K and Liu Q: Effects of microRNA-30a on migration, invasion and prognosis of hepatocellular carcinoma. FEBS Lett. 588:3089–3097. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Lv XB, Zhang X, Deng L, Jiang L, Meng W, Lu Z and Wang X: MiR-92a mediates AZD6244 induced apoptosis and G1-phase arrest of lymphoma cells by targeting Bim. Cell Biol Int. 38:435–443. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Li M, Guan X, Sun Y, Mi J, Shu X, Liu F and Li C: miR-92a family and their target genes in tumorigenesis and metastasis. Exp Cell Res. 323:1–6. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Welcker M, Orian A, Jin J, Grim JE, Harper JW, Eisenman RN and Clurman BE: The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation. Proc Natl Acad Sci USA. 101:9085–9090. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Koepp DM, Schaefer LK, Ye X, Keyomarsi K, Chu C, Harper JW and Elledge SJ: Phosphorylation-dependent ubiquitination of cyclin E by the SCFFbw7 ubiquitin ligase. Science. 294:173–177. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Grandori C, Cowley SM, James LP and Eisenman RN: The Myc/Max/Mad network and the transcriptional control of cell behavior. Annu Rev Cell Dev Biol. 16:653–699. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Hwang HC and Clurman BE: Cyclin E in normal and neoplastic cell cycles. Oncogene. 24:2776–2786. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Ohtsubo M, Theodoras AM, Schumacher J, Roberts JM and Pagano M: Human cyclin E, a nuclear protein essential for the G1-to-S phase transition. Mol Cell Biol. 15:2612–2624. 1995.PubMed/NCBI

28 

Prendergast GC: Mechanisms of apoptosis by c-Myc. Oncogene. 18:2967–2987. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Sargent LM, Zhou X, Keck CL, Sanderson ND, Zimonjic DB, Popescu NC and Thorgeirsson SS: Nonrandom cytogenetic alterations in hepatocellular carcinoma from transgenic mice overexpressing c-Myc and transforming growth factor-alpha in the liver. Am J Pathol. 154:1047–1055. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Li K, Lin SY, Brunicardi FC and Seu P: Use of RNA interference to target cyclin E-overexpressing hepatocellular carcinoma. Cancer Res. 63:3593–3597. 2003.PubMed/NCBI

31 

Xu MZ, Chan SW, Liu AM, Wong KF, Fan ST, Chen J, Poon RT, Zender L, Lowe SW, Hong W, et al: AXL receptor kinase is a mediator of YAP-dependent oncogenic functions in hepatocellular carcinoma. Oncogene. 30:1229–1240. 2011. View Article : Google Scholar

Related Articles

Journal Cover

November-2015
Volume 34 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang W, Dou C, Wang Y, Jia Y, Li C, Zheng X and Tu K: MicroRNA-92a contributes to tumor growth of human hepatocellular carcinoma by targeting FBXW7 Corrigendum in /10.3892/or.2023.8596. Oncol Rep 34: 2576-2584, 2015
APA
Yang, W., Dou, C., Wang, Y., Jia, Y., Li, C., Zheng, X., & Tu, K. (2015). MicroRNA-92a contributes to tumor growth of human hepatocellular carcinoma by targeting FBXW7 Corrigendum in /10.3892/or.2023.8596. Oncology Reports, 34, 2576-2584. https://doi.org/10.3892/or.2015.4210
MLA
Yang, W., Dou, C., Wang, Y., Jia, Y., Li, C., Zheng, X., Tu, K."MicroRNA-92a contributes to tumor growth of human hepatocellular carcinoma by targeting FBXW7 Corrigendum in /10.3892/or.2023.8596". Oncology Reports 34.5 (2015): 2576-2584.
Chicago
Yang, W., Dou, C., Wang, Y., Jia, Y., Li, C., Zheng, X., Tu, K."MicroRNA-92a contributes to tumor growth of human hepatocellular carcinoma by targeting FBXW7 Corrigendum in /10.3892/or.2023.8596". Oncology Reports 34, no. 5 (2015): 2576-2584. https://doi.org/10.3892/or.2015.4210