miR-154 targeting ZEB2 in hepatocellular carcinoma functions as a potential tumor suppressor

Retraction in: /10.3892/or.2021.8113

  • Authors:
    • Xiaoli Pang
    • Kexin Huang
    • Qianqian Zhang
    • Yujiao Zhang
    • Junqi Niu
  • View Affiliations

  • Published online on: October 1, 2015     https://doi.org/10.3892/or.2015.4321
  • Pages: 3272-3279
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA-154 (miR-154) has been identified as a tumor suppressor in several types of human cancers; however, its clinical significance and function in human hepatocellular carcinoma (HCC) remain unclear. The aim of the present study was to analyze the clinical significance and cellular function of miR-154 in HCC patients. The data showed that miR-154 expression was consistently lower in HCC tissues and cell lines compared to that in matched tumor-adjacent tissues and a normal hepatic cell line, and its expression was negatively correlated with tumor differentiation (P<0.01), TNM stage (P<0.01) and lymph node metastasis (P<0.01). Restoration of miR-154 expression in HepG2 cells inhibited cell proliferation, migration and invasion, and induced apoptosis and cell arrest at the G1 phase in vitro, as well as suppressed tumor growth in a nude mouse model. Using a luciferase assay, we identified that miR-154 was able to target the 3'-untranslated region (3'UTR) of ZEB2 mRNA. Then, we revealed that miR-154 was able to reduce ZEB2 expression at the levels of mRNA and protein using qRT-PCR and western blot analysis. Notably, restoration of expression of ZEB2 weakened miR-154-mediated suppression of tumor progression. In conclusion, these results indicate that miR-154 functions as a tumor suppressor in HCC by suppressing ZEB2, suggesting that miR-154 may serve as a potential target for HCC.

Introduction

Hepatocellular carcinoma (HCC) ranks as the fifth most common cancer worldwide (1), and the second most common cause of cancer-related mortality in China (2). Despite the fact that treatments against intrahepatic HCC have almost been standardized, the 5-year overall survival for HCC is less than 30% due to therapeutic resistance and metastasis (3). Although accumulating evidence has demonstrated diverse genetic alterations in HCC, the highly complex molecular mechanisms underlying HCC carcinogenesis and progression remain obscure (4). Therefore, it is necessary to understand the underlying molecular mechanisms involved in HCC progression, which can contribute to the identification of novel markers for HCC, improvement in therapeutic strategies, and effective disease management in the clinic.

MicroRNAs (miRs) are a class of short (~22 nucleotides in length), endogenous, single stranded, RNA molecules that regulate target gene expression at the post-transcriptional level by directly binding to the 3′-untranslated regions (3′UTRs) of target messenger RNAs (mRNAs) (5,6). It has been shown that miRNAs are involved in a variety of cellular processes including proliferation, development, differentiation and tumorigenesis (5,7,8). Since approximately half of all human miRNAs are located in cancer-associated genomic regions; miRNAs can function as either oncogenes or tumor suppressors according to the roles of their target genes (911). Growing evidence suggests that miRNAs play important roles in HCC, including differentiation, proliferation, apoptosis, cell cycle, migration and invasion (12,13), suggesting that miRNAs act not only as diagnostic and prognostic markers but also as potential therapeutic targets of HCC.

miR-154, located on human chromosome 14q32, is a very conservative miRNA cluster in mammalians (14). Recently several studies have demonstrated that miR-154 acts as a tumor suppressor in prostate (15), breast (16), non-small lung (17), colorectal (18) and thyroid cancer (19). In regards to HCC, one study showed that miR-154 expression was downregulated in HCC tissues, and that miR-154 inhibited tumor cell malignant potential and the G1/S phase transition in cancer cells by targeting CCND2 (15). However, the correlation between miR-154 dysregulation and the clinicopathological characteristics of HCC, and its role and underlying molecular mechanism remain poorly understand.

Therefore, in the present study, we firstly analyzed the association of miR-154 expression with clinicopathological features of HCC patients. We next investigated the functional role of miR-154 and the potential mechanism in the regulation of HCC proliferation, apoptosis, cell cycle arrest, migration and invasion, and tumor growth of xenografts in vivo. The present study contributes to the elucidation of the specific roles and the underlying mechanisms of miR-154 in HCC.

Materials and methods

Clinical specimens

Primary HCC specimens and their matched normal specimens were obtained from patients undergoing hepatic resection at the Department of Hepatopancreatobiliary Surgery, The First Hospital, Jilin University (Changchun, China), and were snap-frozen in liquid nitrogen and stored at −80°C until use. None of the patients received radiotherapy, chemotherapy, or other anticancer treatment prior to surgery. All patients provided written informed consent according to the protocol approved by the Ethics Committee of Jilin University.

Cell culture

Four human HCC cell lines (Huh-7, SMMC7721, HepG2 and HCCLM3) and a normal human hepatocyte cell line HL-7702, were obtained from the Shanghai Institute for Biological Sciences (Shanghai, China). All cell lines were cultured in RPMI-1640 medium containing 100 U/ml penicillin, 100 mg/ml streptomycin and 10% fetal calf serum (FCS) (all from Gibco, Grand Island, NY, USA) at 37°C in a humidified chamber supplemented with 5% CO2.

Real-time quantitative RT-PCR analysis

Total RNA was extracted from the clinical specimens and cultured cells with TRIzol reagent (Invitrogen Corp., Carlsbad, CA, USA) according to the manufacturer's instructions. The purity and concentration of RNA were measured by using a dual-beam ultraviolet spectrophotometer (Eppendorf, Hamburg, Germany). Ten nanograms of total RNA was transcribed into cDNAs using a TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems, Foster City, CA, USA) according to the manufacturer's instructions. To detect miR-154 expression, real-time PCR was performed with a TaqMan MicroRNA Assay kit on the ABI 7900 Fast system (both from Applied Biosystems). U6 small nuclear RNA was used as an internal control.

To measure ZEB2 mRNA expression, qRT-PCR was performed with SYBR Green Premix Ex Taq (Takara) on ABI 7900 Fast system. The primer sequences of ZEB2 and GAPDH used were: ZEB2 sense, 5′-AGGAGCAGGTAATCG-3′ and antisense, 5′-TGGGCACTCGTAAGG-3′; GAPDH sense, 5′-GAAGGTGAAGGTCGGAGTC-3′ and antisense, 5′-GAAGATGGTGATGGGATTTC-3′. GAPDH was used as an internal control. All reactions were run in triplicate, and fold changes in gene expression were calculated by the 2−ΔΔCt method.

Transfection with miRNA mimics, ZEB2 vectors and siRNA

miR-154 mimics and corresponding controls (miR-NC) were purchased from Ribobio Co. (Guangzhou, China). The plasmids carrying ZEB2 (pCDNA3-ZEB2) were a gift from Dr Lin (Jilin University, Changchun, China). These molecular products were transiently transfected into HepG2 cells using Lipofectamine 2000 reagent (Invitrogen) according to the manufacturer's protocol. Transfection efficiencies were evaluated in every experiment at 48 h after transfection.

MTT assay

The cell proliferation was determined by the MTT assay. Briefly, 5×103 transfected cells were seeded into 96-well culture plate, and cultured for 24, 48 and 72 h, respectively. Then, 20 µl of MTT (5 mg/ml; Sigma-Aldrich, St. Louis, MO, USA) was added, and cultured for 4 h at 37°C. Then, the medium was removed and the residue was dissolved in 150 µl DMSO (Sigma-Aldrich). The absorbance of each well was read at 570 nm with a microplate reader.

Colony forming assay

Transfected cells were digested, and a single-cell suspension was prepared. Then, the cells (1,000 cells/well) were added to 6-well plates followed by incubation under a standard condition for 24 h. Non-adherent cells were removed. After culturing for 10–14 days, the cells were stained with 0.5% crystal violet for 30 min. The percentage of colony formation was calculated by adjusting the control (miR-NC group) to 100%.

Cell cycle distribution and analysis of cell apoptosis

Cell cycle distribution and apoptosis were examined by flow cytometry at 48 h post-transfection. For the cell cycle assay, 2×104 transfected cells were fixed in 10 µl ice-cold ethanol for at least 2 h. and then washed twice in PBS and incubated with 500 µl RNase (0.25 mg/ml) at 37°C for 30 min. The cells were pelleted by centrifugation at 1,000 × g for 5 min, resuspended in 50 µg/ml propidium iodide (PI; KeyGen, Nanjing, China), and incubated at 4°C for 30 min in the dark. The PI signal was examined using a FACSCalibur flow cytometer (BD Biosciences, Mansfield, MA, USA).

The cell apoptosis assay was performed using cell staining with PI and FITC-labeled Annexin V (KeyGen) to detect phosphatidylserine externalization as an endpoint indicator of early apoptosis, according to the manufacturer's instructions.

Wound-healing assay

Transfected cells were seeded at 2.0×104 cells/well in 6-well culture plates. After the cells had grown to confluency, the confluent monolayer in each well was scratched using a 10-µl pipette tip in order to evaluate cell migration by testing the capability of cells to migrate into the wounded area. The cells were photographed at 0 and 24 h; the scratch area was measured using Image J software (National Institutes of Health, Bethesda, MD, USA).

Cell migration and invasion assays

Invasion assays were carried out in modified Boyden chambers (BD Biosciences, San Jose, CA, USA) with 8-mm pore filter inserts in 24-well plates. Twenty-four hours after transfection, 2.0×104 transfected cells suspended in serum-free RPMI-1640 medium were added to the upper chamber coated with Matrigel (BD Biosciences) and incubated at 37°C for 4 h, allowing it to solidify. RPMI-1640 containing 10% FCS was added to the lower chambers as a chemoattractant. After a 24-h culture at 37°C, the non-filtered cells were gently removed with a cotton swab. Filtered cells located on the lower side of the chamber were stained with 2% crystal violet, imaged, and counted with a microscope (Olympus, Tokyo, China). All experiments were performed in triplicate.

Vector construction and luciferase reporter assay

A human ZEB2 3′UTR was amplified from cDNAs of the HCC cell line by RT-PCR, and cloned into the downstream of the firefly luciferase gene of the psiCHECK-2 vector (Promega, Madison, WI, USA). For mutagenesis of the miR-154-binding site, a QuickChange Site-Directed Mutagenesis kit (Agilent Technologies, Palo Alto, CA, USA) was used according to the manufacturer's instructions.

For the luciferase activity assay, HepG2 cells were co-transfected with wild-type (WT) or mutant (Mut) 3′UTR of psiCHECK-2-ZEB2 and miR-154 or miR-NC. The cells were incubated for 48 h, and dual-luciferase activities were assessed using the Dual-Luciferase Reporter Assay system (Promega), according to the manufacturer's instructions. Renilla luciferase activity was normalized to firefly luciferase activity.

Western blot analysis

Tissue samples and cells were harvested and homogenized with RIPA lysis buffer (Beyotime, Beijing, China) according to the manufacturer's instructions. Equal amounts of proteins (30 µg each lane) were separated by 10% SDS-PAGE and transferred to PVDF membranes (Millipore, Billerica, MA, USA). The membranes were incubated overnight at 4°C with the following primary antibodies: anti-ZEB2, anti-E-cadherin, anti-N-cadherin, anti-vimentin, and anti-human GAPDH (all from Cell Signaling Technology, USA). Then the blots were incubated with the corresponding HRP-labeled secondary antibody for 1 h at room temperature. Proteins on the membranes were detected using the ECL detection system (Pierce, Cambridge, MA, USA).

In vivo nude mouse tumorigenesis assay

Sixteen BALB/C nude male mice (20–25 g) (5-weeks old) purchased from the Experimental Animal Center of Changchun Institute for Biological Sciences (Changchun, China), were maintained under pathogen-free (SPF) conditions. All animal experiments were 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-154 mimic or the miR-NC were injected subcutaneously into the right rear flank of each mouse (8 per group) to establish an HCC xenograft model. Tumor volume was calculated using the formula: Volume (mm3) = 1/2 width2 × length. All mice were sacrificed, and the tissues were removed and weighed 5 weeks after the injection. Partial liver tissues were snap-frozen for RNA and protein extraction.

Statistical analysis

Data are expressed as the mean ± standard deviation (SD) from at least three independent experiments. Data were imaged with GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA). Two-tailed Student's t-test or ANOVA was used to analyze differences. P<0.05 was considered to indicate a statistically significant difference.

Results

Downregulation of miR-154 in human HCC tissues and cell lines

To investigate the potential role of miR-154 in HCC carcinogenesis, we assessed the miR-154 expression in a panel of human HCC cell lines by real-time quantitative RT-PCR (qRT-PCR). The result showed that the expression level of miR-154 was downregulated in the HCC cell lines compared with that noted in the normal hepatic HL-7702 cell line (Fig. 1A). Additionally, the expression level of miR-154 in the HepG2 cell line was lowest, thus, we selected it for subsequent study. Consistent with the results from the cell lines, the miR-154 level in the HCC tissues was significantly lower than that in the corresponding non-cancerous tissues (Fig. 1B).

The associations of miR-154 expression with various clinicopathological parameters of HCC tissues were also analyzed (Table I). The patients were divided into two groups according to their miR-154 expression levels, using the median (0.467) of miR-154 expression in all 50 patients as a cut-off: high miR-154 expression group (n=24) and low miR-154 expression group (n=26). It was found that expression of miR-154 was significantly downregulated in HCC patients with poorly differentiated tumors (P<0.01), positive lymph node metastasis (P<0.01) and advanced TNM stage (P<0.01). No significant difference was observed between miR-154 expression and patient age and gender. These results suggest that miR-154 may be involve in the initiation and development of HCC.

Table I

Correlation between miR-154 expression and the clinicopathological features of the HCC cases.

Table I

Correlation between miR-154 expression and the clinicopathological features of the HCC cases.

VariablesNo. of casesmiR-154 expression
P-value
Low n (%)High n (%)
Age (years)0.845
 <552211 (50.0)11 (50.0)
 ≥552815 (53.5)13 (46.5)
Gender0.775
 Male2714 (51.9)13 (48.1)
 Female2312 (52.3)11 (47.7)
TNM stage<0.01
 T1-T2329 (28.1)23 (72.9)
 T3-T41817 (94.4)1 (5.6)
Differentiation<0.01
 Well/moderate297 (24.1)22 (75.9)
 Poor2119 (90.5)2 (9.5)
Lymph node<0.01
metastasis
 No339 (27.3)24 (72.7)
 Yes1717 (100)0 (0)
miR-154 inhibits the proliferation and colony formation and induces the cell apoptosis of HCC cells in vitro

To investigate the cellular function of miR-154 in HCC, HepG2 cells were transfected with the miR-154 mimic or miR-NC, and miR-154 expression was examined by qRT-PCR. The expression of miR-154 was markedly elevated in the miR-154 mimictransfected cells compared with the miR-NC-transfected cells (Fig. 2A). Cell proliferation and colony formation were then measured using MTT and colony formation assays. Our results showed that restoration of miR-154 led to a significant decrease in cell proliferation (Fig. 2B) and colony formation (Fig. 2C) of the HepG2 cells. As proliferation is directly linked to cell cycle distribution, the effect of miR-154 on cell cycle progression was analyzed. As expected, the percentage of S phase cells was reduced, while the percentage of G1 phase cells was increased in the HepG2 cells upon transfection with the miR-154 mimic (Fig. 2D). Next, we used flow cytometry to test the role of miR-154 in apoptosis. Our results showed that restoration of miR-22 significantly induced cell apoptosis in the HCC cells (Fig. 2E). Thus, restoration of miR-154 expression inhibited cell growth in HCC by inhibiting cell proliferation and inducing cell apoptosis.

miR-154 inhibits migration, invasion and epithelial-mesenchymal transition (EMT) in HCC cells

As it has been shown that miR-154 expression is associated with lymph node metastasis in patients suffering from HCC, we aimed to ascertain whether miR-154 affects cell migration and invasion in HepG2 cells transfected with the miR-154 mimic or miR-NC by wound healing and Transwell chamber assays. Consistent with the clinical data, restoration of miR-154 significantly decreased migration and invasion capacities of the HepG2 cells (P<0.05; Fig. 3A and B).

Since EMT is closely related to cancer cell metastasis ability, we next examined EMT markers in HCC cells transfected with the miR-154 mimic or miR-NC by western blot analysis. We found that overexpression of miR-154 led to increased expression of E-cadherin and decreased expression of N-cadherin and vimentin (Fig. 3C), suggesting that miR-154 inhibits EMT.

ZEB2 is a target of miR-154

To understand the molecular mechanism of miR-154 action in HCC, we searched for miR-154 targets by using TargetScan, miRanda, and miRWalk algorithms. Among the common predicted targets of miR-154, ZEB2 was selected since ZEB2 has been reported to play an important role in cell proliferation and invasion in various types of cancers including HCC (20). Using prediction tools, we predicted that ZEB2 may be a target of miR-154 (Fig. 4A). To further confirm that miR-154 directly targets ZEB2, luciferase reporter assays were performed. Our results showed that HepG2 cells transfected with miR-154 significantly decreased wild-type ZEB2-3′UTR reporter activity compared with the cells co-transfected with miR-NC (P<0.01), while miR-154 had no inhibitory effect on mutant ZEB2-3′UTR reporter activity (Fig. 4B). To further validate the association between miR-154 and ZEB2, we detected endogenous ZEB2 expression in the HepG2 cells transfected with the miR-154 mimic or miR-NC. Our results showed that the miR-154 mimic markedly decreased the expression levels of ZEB2 mRNA (Fig. 4C) and protein (Fig. 4D). Taken together, these data indicate that ZEB2 is a target of miR-154.

Overexpression of ZEB2 abrogates the inhibitory effects of miR-154 on cell proliferation, migration and invasion in HCC cells

To determine whether the role of miR-154 in HCC is mediated by ZEB2, HepG2 cells were transfected with the pCDNA3.0-ZEB2 plasmid or the vector. Western blot assay confirmed that the pCDNA3.0-ZEB2 plasmid significantly increased ZEB2 expression in the HepG2 cells (Fig. 5A). HepG2 cells were co-transfected with miR-154 and pcDNA3-ZEB2, or the blank pCDNA3 vector, and then cell proliferation, migration and invasion were assessed at the indicated times. MTT assay showed that overexpression of ZEB2 markedly reversed the tumor-suppressive effect of miR-154 on cell proliferation (Fig. 5B). Wound-healing migration and invasion assays demonstrated that overexpression of ZEB2 reversed the inhibitory effects of miR-154 on HCC cell migration and invasion (Fig. 5C and D). These data suggest that miR-154 acts as a tumor suppressor in HCC by targeting ZEB2.

miR-154 suppresses tumor growth in a mouse xenograft model

To evaluate the effect of miR-154 in HCC in vivo, we examined the potential effect of miR-154 on tumorigenesis using a HepG2 xenograft model. Tumors grew slower in the HepG2/miR-154 group compared with the rate in the HepG2/miR-NC group (Fig. 6A). A significant decrease in tumor size (Fig. 6B) and weight (Fig. 6C) was observed in the HepG2/miR-154 group compared to the HepG2/miR-NC group after the mice were sacrificed. Furthermore, we also determined ZEB2 expression in the tumor tissues by western blot analysis. ZEB2 expression was markedly decreased in the HepG2/miR-154 group compared to the level in the HepG2/miR-NC group (Fig. 6D). These data indicate that miR-154 suppresses tumor growth of HCC in vivo by targeting ZEB2.

Discussion

In recent years, emerging evidence has demonstrated that miRNAs play critical roles in the initiation, promotion and progression of human cancers by regulating target gene expression (79); therefore, they can be classified as oncomiRs or tumor-suppressive miRNAs (10,11). Recently, a number of miRNAs related to cell proliferation, migration and invasion in HCC have been identified. For example, overexpression of miR-222 was found to promote cell proliferation, migration and invasion, and decrease cell apoptosis, as well as enhance the resistance of HCC cells to sorafenib through activation of the PI3K/AKT signaling pathway (21). miR-494 was found to promote cell proliferation, migration and invasion, and increase sorafenib resistance in HCC by targeting PTEN (22). miR-26a suppressed the recruitment of macrophages by down-regulating macrophage colony-stimulating factor expression through the PI3K/Akt pathway in HCC (23). miR-211 was found to inhibit the proliferation and invasion of HCC cells by targeting special AT-rich sequence-binding protein 2 (SATB2) (24). Data from the present study demonstrated that miR-154 inhibited proliferation, colony formation, migration and invasion, and decreased cell apoptosis of HCC cells, and suppressed tumor growth in a nude mouse model.

As a tumor suppressor, miR-154 is upregulated in different types of human cancers, such as prostate (15), breast (16), non-small lung (17), colorectal (18) and thyroid cancer (19). Accumulating evidence suggests that miR-154 plays important roles in cancer development, progression, metastasis, and may act as an effective biomarker or therapeutic target for cancer prognosis and treatment (1519). In regards to HCC, one study revealed that miR-154 is downregulated in HCC tissues, and that overexpression of miR-154 suppressed tumor cell malignancy and the G1/S phase transition in HCC cells by targeting CCND2 (15). However, the mechanisms involved in tumorigenesis are so complex, that the effects of miR-154 on HCC development and progression remain largely unclear. Data from the present study showed that the expression of miR-154 was decreased in HCC tissues and cell lines, and the expression level was significantly associated with tumor differentiation, TNM stage and lymph node metastasis. Restoration of miR-154 in HCC cells suppressed tumor growth and metastasis of HCC in vitro and in vivo by targeting ZEB2.

Zinc finger E-box binding homeobox 2 (ZEB2), a member of the δEF-1 family of two-handed zinc-finger factors, was originally identified in a transforming growth factor-β/bone morphogenetic protein (TGF-β/BMP) signaling pathway by its binding to the MH2 domain of receptor-activated Smads (25). ZEB2 has been reported to be upregulated in various types of cancers including HCC (26), and to play a critical regulatory role in promoting EMT and tumor progression (21). Of note, it has been reported that ZEB2 induces EMT in HCC by suppressing E-cadherin or inducing vimentin expression, which facilitates the metastasis of cancer cells (27,28). In the present study, luciferase reporter assays detected the direct binding of miR-154 to the 3′UTR of ZEB1/2 transcripts. qRT-PCR and western blot analysis further confirmed that overexpression of miR-154 inhibited ZEB2 expression at the mRNA and protein levels. Furthermore, overexpression of ZEB2 weakened miR-154-mediated suppression of tumor progression, suggesting that miR-154 functions as a tumor suppressor in HCC, at least in part, by targeting ZEB2.

ZEB2 has been found to be regulated in HCC by several miRNAs. For example, Qiu et al reported that miR-139-5p inhibited EMT, migration and invasion of HCC cells by targeting ZEB1 and ZEB2 (29). Yang et al found that miR-200a suppressed the metastatic potential of side population cells in human HCC by targeting ZEB2 (30). Wu et al found that miR-141 suppressed both the growth and the motility of HCC cells by targeting ZEB2 (31). Here, we showed that miR-154 inhibited HCC proliferation, migration and invasion by targeting ZEB2. Taken together with previous studies, it appears that various miRNAs exert a tumor suppressive effect on HCC by targeting ZEB2

In summary, we demonstrated that the expression of miR-154 was downregulated in HCC tissues and cell lines, and its expression level was significantly associated with tumor differentiation, TNM stage and lymph node metastasis. Restoration of miR-154 in HCC cells inhibited cell proliferation, colony formation, migration and invasion, and induced cell apoptosis in vitro, as well as suppressed the tumor growth of HCC in vivo by targeting ZEB2. These findings suggest that miR-154 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 

He J, Gu D, Wu X, Reynolds K, Duan X, Yao C, Wang J, Chen CS, Chen J, Wildman RP, et al: Major causes of death among men and women in China. N Engl J Med. 353:1124–1134. 2005. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

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

5 

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

6 

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 :

7 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

McManus MT: MicroRNAs and cancer. Semin Cancer Biol. 13:253–258. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Farazi TA, Spitzer JI, Morozov P and Tuschl T: miRNAs in human cancer. J Pathol. 223:102–115. 2011. View Article : Google Scholar :

11 

Garzon R and Marcucci G: Potential of microRNAs for cancer diagnostics, prognostication and therapy. Curr Opin Oncol. 24:655–659. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Li G, Shen Q, Li C, Li D, Chen J and He M: Identification of circulating microRNAs as novel potential biomarkers for hepatocellular carcinoma detection: A systematic review and meta-analysis. Clin Transl Oncol. 7:684–693. 2015. View Article : Google Scholar

13 

Saito Y, Suzuki H, Matsuura M, Sato A, Kasai Y, Yamada K, Saito H and Hibi T: MicroRNAs in hepatobiliary and pancreatic cancers. Front Genet. 2:662011. View Article : Google Scholar

14 

Lin SP, Youngson N, Takada S, Seitz H, Reik W, Paulsen M, Cavaille J and Ferguson-Smith AC: Asymmetric regulation of imprinting on the maternal and paternal chromosomes at the Dlk1-Gtl2 imprinted cluster on mouse chromosome 12. Nat Genet. 35:97–102. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Wang W, Peng B, Wang D, Ma X, Jiang D, Zhao J and Yu L: Human tumor microRNA signatures derived from large-scale oligonucleotide microarray datasets. Int J Cancer. 129:1624–1634. 2011. View Article : Google Scholar

16 

Miranda PJ, Vimalraj S and Selvamurugan N: A feedback expression of microRNA-590 and activating transcription factor-3 in human breast cancer cells. Int J Biol Macromol. 72:145–150. 2015. View Article : Google Scholar

17 

Lin X, Yang Z, Zhang P and Shao G: miR-154 suppresses non-small cell lung cancer growth in vitro and in vivo. Oncol Rep. 33:3053–3060. 2015.PubMed/NCBI

18 

Xin C, Zhang H and Liu Z: miR-154 suppresses colorectal cancer cell growth and motility by targeting TLR2. Mol Cell Biochem. 387:271–277. 2014. View Article : Google Scholar

19 

Mian C, Pennelli G, Fassan M, Balistreri M, Barollo S, Cavedon E, Galuppini F, Pizzi M, Vianello F, Pelizzo MR, et al: MicroRNA profiles in familial and sporadic medullary thyroid carcinoma: Preliminary relationships with RET status and outcome. Thyroid. 22:890–896. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Yang Z, Sun B, Li Y, Zhao X, Zhao X, Gu Q, An J, Dong X, Liu F and Wang Y: ZEB2 promotes vasculogenic mimicry by TGF-β1 induced epithelial-to-mesenchymal transition in hepatocellular carcinoma. Exp Mol Pathol. 98:352–359. 2015. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Liu K, Liu S, Zhang W, Jia B, Tan L, Jin Z and Liu Y: miR-494 promotes cell proliferation, migration and invasion, and increased sorafenib resistance in hepatocellular carcinoma by targeting PTEN. Oncol Rep. 34:1003–1010. 2015.PubMed/NCBI

23 

Chai ZT, Zhu XD, Ao JY, Wang WQ, Gao DM, Kong J, Zhang N, Zhang YY, Ye BG, Ma DN, et al: microRNA-26a suppresses recruitment of macrophages by down-regulating macrophage colony-stimulating factor expression through the PI3K/Akt pathway in hepatocellular carcinoma. J Hematol Oncol. 8:562015. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Verschueren K, Remacle JE, Collart C, Kraft H, Baker BS, Tylzanowski P, Nelles L, Wuytens G, Su MT, Bodmer R, et al: SIP1, a novel zinc finger/homeodomain repressor, interacts with Smad proteins and binds to 5′-CACCT sequences in candidate target genes. J Biol Chem. 274:20489–20498. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Cai MY, Luo RZ, Chen JW, Pei XQ, Lu JB, Hou JH and Yun JP: Overexpression of ZEB2 in peritumoral liver tissue correlates with favorable survival after curative resection of hepatocellular carcinoma. PLoS One. 7:e328382012. View Article : Google Scholar : PubMed/NCBI

27 

Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, Mareel M, Huylebroeck D and van Roy F: The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell. 7:1267–1278. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Vandewalle C, Comijn J, De Craene B, Vermassen P, Bruyneel E, Andersen H, Tulchinsky E, Van Roy F and Berx G: SIP1/ZEB2 induces EMT by repressing genes of different epithelial cell-cell junctions. Nucleic Acids Res. 33:6566–6578. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Qiu G, Lin Y, Zhang H and Wu D: miR-139-5p inhibits epithelial-mesenchymal transition, migration and invasion of hepatocellular carcinoma cells by targeting ZEB1 and ZEB2. Biochem Biophys Res Commun. 463:315–321. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Yang X, Wang J, Qu S, Zhang H, Ruan B, Gao Y, Ma B, Wang X, Wu N, Li X, et al: MicroRNA-200a suppresses metastatic potential of side population cells in human hepatocellular carcinoma by decreasing ZEB2. Oncotarget. 6:7918–7929. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Wu SM, Ai HW, Zhang DY, Han XQ, Pan Q, Luo FL and Zhang XL: miR-141 targets ZEB2 to suppress HCC progression. Tumour Biol. 35:9993–9997. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2015
Volume 34 Issue 6

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
Pang X, Huang K, Zhang Q, Zhang Y and Niu J: miR-154 targeting ZEB2 in hepatocellular carcinoma functions as a potential tumor suppressor Retraction in /10.3892/or.2021.8113. Oncol Rep 34: 3272-3279, 2015
APA
Pang, X., Huang, K., Zhang, Q., Zhang, Y., & Niu, J. (2015). miR-154 targeting ZEB2 in hepatocellular carcinoma functions as a potential tumor suppressor Retraction in /10.3892/or.2021.8113. Oncology Reports, 34, 3272-3279. https://doi.org/10.3892/or.2015.4321
MLA
Pang, X., Huang, K., Zhang, Q., Zhang, Y., Niu, J."miR-154 targeting ZEB2 in hepatocellular carcinoma functions as a potential tumor suppressor Retraction in /10.3892/or.2021.8113". Oncology Reports 34.6 (2015): 3272-3279.
Chicago
Pang, X., Huang, K., Zhang, Q., Zhang, Y., Niu, J."miR-154 targeting ZEB2 in hepatocellular carcinoma functions as a potential tumor suppressor Retraction in /10.3892/or.2021.8113". Oncology Reports 34, no. 6 (2015): 3272-3279. https://doi.org/10.3892/or.2015.4321