MicroRNA‑744 inhibits migration and invasion of hepatocellular carcinoma cells by targeting SOX12

Retraction in: /10.3892/or.2022.8364

  • Authors:
    • Wei Zhang
    • Kai Liu
    • Songyang Liu
    • Bai Ji
    • Yahui Liu
  • View Affiliations

  • Published online on: October 8, 2018     https://doi.org/10.3892/or.2018.6774
  • Pages: 3585-3592
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA‑744 (miR‑744) reportedly plays an oncogenic or tumor‑suppressive role in different human malignancies. Although a previous study demonstrated that miR‑744 significantly inhibits hepatocellular carcinoma (HCC) proliferation in vitro, the role of miR‑744 in the migration and invasion of HCC cells remains largely unknown. The present study investigated the significance of miR‑744 in HCC tissues and cell lines and evaluated its role and underlying mechanism of action in HCC cells. miR‑744 expression was detected in HCC tissues and cell lines by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). The effect of miR‑744 on cell proliferation, migration and invasion was determined using Cell Counting Kit‑8, wound‑healing and Matrigel invasion assays, respectively. Targeting gene of miR‑744 in HCC cells was determined by luciferase reporter assay, RT‑qPCR and western blotting. It was revealed that miR‑744 was poorly expressed in HCC tissues compared with adjacent normal tissues (P<0.05), and that decreased miR‑744 levels were significantly associated with the tumor node metastasis stage and lymph node metastasis. Additionally, restoration of miR‑744 in HCC cells significantly suppressed their proliferation, migration, and invasion. Furthermore, sex determining region Y‑box 12 (SOX12) was identified as a functional target of miR‑744 in HCC cells, and its expression was demonstrated to be upregulated in HCC tissues and inversely correlated with the expression of miR‑744. Notably, overexpression of SOX12 antagonized the suppressive effect of miR‑744 on HCC cell migration and invasion. These findings suggested that miR‑744 inhibited migration and invasion by targeting SOX12, and that it may represent a therapeutic target for the treatment of metastatic HCC.

Introduction

Hepatocellular carcinoma (HCC) is the second most common cause of cancer-associated mortality worldwide and is characterized by its highly invasive, migratory, and proliferative capability (1). Although surgical resection combined with radiation, chemotherapy, and biological-therapy options are available, patients with HCC have very poor prognoses, with a low 5-year survival rate (2,3). Therefore, elucidation of the molecular mechanisms underlying HCC initiation and development are required to identify new diagnostic markers and therapeutic strategies.

A growing number of studies report that microRNAs (miRNAs) are involved in tumor development and progression and act by regulating target genes involved in cell proliferation, cell cycle, apoptosis, migration, angiogenesis, and epithelial-mesenchymal transition (EMT) (46). Numerous miRNAs have been implicated in HCC progression and function as either oncogenes or tumor-suppressor genes (7,8).

MiR-744 is a cancer-associated miRNA that exhibits a crucial role in tumor evolution and progression in multiple cancers (914). Previous studies have reported that miR-744 is downregulated in HCC (15,16), and that overexpression of miR-744 inhibits cell proliferation by suppressing c-Myc expression (16). However, the contribution of miR-744 dysregulation to HCC migration and invasion remains unclear. The present study measured the expression of miR-744 in HCC tissues and cell lines and evaluated its clinical significance. The effect of miR-744 on HCC cell migration and invasion was also investigated and a potential target of miR-744 identified through bioinformatics analysis, coupled with a luciferase assay, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting.

Materials and methods

Tissue samples

Primary HCC tissues and adjacent normal liver tissues were obtained from 48 patients (mean age, 53.5±4.1 years, age range, 41.2–76.8 years; male, 21 cases and female 27 cases) with HCC at the Department of Hepatopancreatobiliary Surgery, the First Hospital of Jilin University (Jilin, China) between July 2015 and August 2016. The histopathological diagnosis of HCC was confirmed after the operation by specialists at the Department of Pathology at the First Hospital of Jilin University based on the criteria defined by the World Health Organization (17). All tissue samples from the surgical procedure were immediately snap frozen in liquid nitrogen and stored at −80°C until RNA extraction. No patients received radiotherapy, chemotherapy, or other therapy prior to surgical intervention. This study was approved by the Ethics Committee of the First Hospital of Jilin University, and written informed consent was obtained from all patients whose biological samples were used in the study.

Cell lines and transfection

The three HCC cell lines (SMMC-7721, Hep3B and Huh-7) and the human hepatic cell line (LO2) were purchased from the cell bank of the Chinese Academy of Sciences (Shanghai, China), grown in Dulbecco's modified Eagle's medium (DMEM; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; HyClone; GE Healthcare Life Sciences, Logan, UT, USA), and incubated at 37°C in a humidified atmosphere containing 5% CO2.

The miR-744 mimic (5′-GACAACGGTGAUUGGAGUUGGA-3′) and the appropriate negative control mimic (miR-NC, 5′-GUCCTUGCUCGAGCGAGGUGA-3′) were obtained from Guangzhou RiboBio Co., Ltd. (Guangzhou, China). The sex determining region Y-box 12 (SOX12)-overexpression vector (pCDNA3.1-SOX12) and blank vector (pCDNA3.1) were a kind gift from Tao Jiang (Jilin University, Jilin, China). SMMC-7721 cells were transiently transfected with one of the aforementioned mimics or the overexpression vector using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Transfection efficiency was determined in every experiment at 24 h post-transfection.

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA from tissue samples and cultured cells was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. cDNA was reverse transcribed from 2 µg total RNA using a PrimeScript first-strand cDNA synthesis kit (Takara Biotechnology Co., Ltd., Dalian China) according to the manufacturer protocol. cDNA was amplified using SYBR Premix ExTaq (Takara Biotechnology Co., Ltd.) using the ABI 7900 fast system (Applied Biosystems; Thermo Fisher Scientific, Inc.). Primer sequences for miR-744 and U6 (RiboBio Co., Ltd.) were as follows: miR-744, 5′-CTGTTGCCACTAACCTCAACCT-3′ (sense) and 5′-GCGAGCACAGAATTAATACGAC-3′ (anti-sense); U6, 5′-TGCGGGTGCTCGCTTCGGCAGC-3′ (sense) and 5′-CCAGTGCAGGGTCCGAGGT-3′ (anti-sense). U6 was used as an internal control. The sequences of the SOX12 and GAPDH primers were as previously described (18). The following PCR conditions were used: Denaturation at 94°C for 3 min, followed by 40 cycles of amplification (denaturation at 94°C for 10 sec, annealing at 60°C for 20 sec and extension at 72°C for 20 sec). Relative gene expression was calculated according to the 2−ΔΔCq method (19) following normalization against U6 for miR-744 or GAPDH for SOX12.

Cell proliferation

Cell proliferation was assessed using Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Kumamoto, Japan). Briefly, transfected cells were seeded in 96-well plates at a density of 5×103 cells/well and cultured for 24 to 72 h. At the indicated time points (24, 48 and 72 h), 20 µl CCK-8 solution was added to each well and incubated for an additional for 4 h. Absorbance was measured at a wavelength of 450 nm using an enzyme-linked immunosorbent assay reader (Thermo Labsystems, Helsinki, Finland).

Cell migration and invasion

The migratory ability of the transfected cells was analyzed by a wound-healing assay. Briefly, transfected cells were cultured in 6-well plates (5×104 cells/well) and grown to 100% confluency. Subsequently, an artificial homogenous wound was scratched into the monolayer using a sterile plastic micropipette tip, followed by culture for 24 h in serum-free medium. The spread of the wound was observed and photographed using an inverted microscope (Olympus Corporation, Tokyo, Japan) to directly assess the level of migration.

Transwell insert chambers (Corning Inc., Corning, NY, USA) were used to determine cell invasion ability. Briefly, 1×105 transfected cells were seeded into each well of the upper chamber of the Matrigel-coated inserts in serum-free medium, and DMEM supplemented with 10% FBS was added to the lower chamber to serve as a chemoattractant. After incubation for 24 h at 37°C in a 5% CO2 atmosphere, cells that had migrated to the lower surface of the filter were fixed with 4% paraformaldehyde and stained with 1% crystal violet for 30 min at 37°C. After washing three times with phosphate-buffered saline, cells were imaged and counted in five random fields using a light microscope (Olympus Corporation).

Bioinformatics, miRNA-target identification and luciferase assay

TargetScan (http://www.targetscan.org/vert_71/), miRanda (http://www.miranda-im.org/), and miRDB (http://www.mirdb.org/) were used to predict potential miR-744 targets. A luciferase assay was performed to validate prediction of SOX12 as a target of miR-744. Briefly, the 3′ untranslated region (UTR) of SOX12 containing a potential binding site for miR-744 (position 1386–1392) was synthesized and inserted into a luciferase-reporter vector (psiCHECK2; Promega Corporation, Madison, WI, USA) and designated as WT-SOX12. A mutant version of the SOX12 3′ UTR was constructed using the QuikChange XL site-directed mutagenesis kit (Agilent Technologies, Santa Clara, CA, USA) and designated as MUT-SOX12. For the luciferase assay, SMMC-7721 cells were cultured to 70–80% confluence in 24-well plates and co-transfected with an miR-744 mimic or miR-NC and WT-SOX12 or MUT-SOX12 reporter plasmids using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer protocol. At 48 h post-transfection, firefly luciferase and Renilla luciferase were visualized using a dual-luciferase reporter assay (Promega Corporation) according to the manufacturer's protocol, and Renilla luciferase activity was normalized against that of firefly luciferase.

Western blot analysis

Total protein extraction, sodium dodecyl sulfate polyacrylamide gel electrophoresis, and western blot analyses were performed as previously described (20). The membranes were probed with primary antibodies overnight at 4°C as follows: Mouse monoclonal antihuman SOX12 (1:1,000; cat. no. Ab54371; Abcam, Cambridge, MA, USA), mouse monoclonal antihuman Twist (1:1,000; cat. no. sc-81417; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) and mouse monoclonal antihuman GAPDH (1:5,000; cat no sc-365062; Santa Cruz Biotechnology, Inc.). Subsequently, the membranes were incubated with polyclonal goat antimouse horseradish peroxidaseconjugated immunogloblin G (1:10,000; cat. no sc-2005; Santa Cruz Biotechnology, Inc.) for 2 h at room temperature. GAPDH was used as an internal control. Protein bands were observed using an enhanced chemiluminescence reagent (ECL; GE Healthcare, Chicaogo, IL, USA). Gray analysis was performed using software Gel-Pro Analyzer 4 (United States Biochemical, Cleveland, OH, USA).

Statistical analysis

All statistical analyses were performed using SPSS software, version 19.0 (IBM SPSS, Armonk, NY, USA). All data are presented as the mean ± standard deviation from at least three independent experiments with similar results. Continuous data were compared using the Student's two-tailed t-test or one-way analysis of variance with post hocTukey's tests. Correlations between miR-744 expression and SOX12 expression were evaluated by Pearson's correlation analysis. In all cases, P<0.05 was considered to indicate a statistically significant difference.

Results

MiR-744 levels are decreased in HCC tissues and cell lines

MiR-744 expression in 48 paired HCC tissues and corresponding adjacent non-tumor tissues was examined by RT-qPCR, revealing that HCC tissues exhibited lower miR-744 expression compared with adjacent normal liver tissues (Fig. 1A). It was also revealed that decreased miR-744 was associated with advanced TNM stage and lymph node metastasis (Fig. 1B and C). MiR-744 expression in three HCC cell lines was examined, and RT-qPCR results revealed that miR-744 levels in three human HCC cell lines (SMMC-7721, Hep3B, and Huh-7) were significantly downregulated compared with normal human hepatocytes (LO2) (Fig. 1D). These data suggested that miR-744 was involved in HCC carcinogenesis.

MiR-744 inhibits HCC proliferation

To investigate the biological roles of miR-744 in HCC proliferation, miR-744 mimic or miR-NC was transiently transfected into human SMMC-7721 cells exhibiting low endogenous levels of miR-744 (Fig. 1D). It was observed that transfection of the miR-744 mimic restored miR-744 expression in SMMC-7721 cells (Fig. 2A), and the CCK-8 assay demonstrated that miR-744 overexpression in SMMC-7721 cells significantly decreased proliferation (Fig. 2B). Consistent with these results, miR-744 overexpression significantly decreased the expression of Ki-67 a proliferation marker, in SMMC7721 cells (Fig. 2C). These results suggested that miR-744 inhibited HCC cell proliferation.

MiR-744 inhibits HCC migration and invasion

To clarify the role of miR-744 in HCC metastasis, the present study analyzed the effects of miR-744 on the migration and invasion of HCC cells. Using a wound-healing assay, a significantly decreased migration of SMMC-7721 cells transfected with the miR-744 mimic was observed compared with cells transfected with miR-NC (Fig. 3A). Similarly, an in vitro Transwell-invasion assay indicated that miR-744 overexpression significantly inhibited the HCC cell invasion (Fig. 3B). These results suggested that miR-744 suppressed HCC metastasis.

SOX12 is a direct target of miR-744 in HCC cells

To explore the mechanism of miR-744 activity in HCC progression, we used three algorithms (TargetScan, miRanda, and miRDB) to search for candidate targets of miR-744, finding that the SOX12 3′ UTR matched the miR-744 seed sequence (Fig. 4A). To verify whether SOX12 was a direct target of miR-744 in HCC cells, a luciferase-reporter assay was performed in SMMC-7721 cells, revealing that miR-744 overexpression clearly inhibited the luciferase activity of the WT-SOX12 3′UTR, whereas it had no influence on that of the MUT-SOX12 3′UTR (Fig. 4B). Furthermore, miR-744 overexpression in SMMC-7721 cells markedly suppressed SOX12 mRNA and protein levels (Fig. 4C and D) and decreased levels of Twist, a protein associated with downstream SOX12 activity (Fig. 4C and D). These results suggested SOX12 as a target of miR-744 in HCC cells.

MiR-744 expression is inversely correlated with that of SOX12 in HCC tissues

To further investigate the relationship between miR-744 and SOX12 ex vivo, SOX12 mRNA levels were examined in HCC tissues and adjacent normal tissues. It was demonstrated that SOX12 mRNA levels were significantly upregulated in HCC tissues compared with those in adjacent normal tissues (Fig. 5A). Additionally, it was demonstrated that SOX12 mRNA levels were inversely correlated with miR-744 expression levels in HCC tissues (R2 =0.450, P<0.0001; Fig. 5B).

SOX12 overexpression partially rescues cells from the biological effects of miR-744 induction in HCC cells

To investigate whether miR-744 functions by regulating SOX12, the present study restored SOX12 expression by transfecting a SOX12-overexpression plasmid (pCDNA3.1-SOX12) into miR-744-overexpressing SMMC-7721 cells (Fig. 6A). The results demonstrated that restoration of SOX12 partially abrogated the effect of miR-744-overexpressing SMMC-7721 cells (Fig. 6B-D). The present study detected SOX12 expression levels in SMMC-7721 cells following transfection with pcDNA3.1-SOX12 or blank vector pCDNA3.1 by western blotting, to establish whether transfection had been successful. It was revealed that transfection with pcDNA3.1-SOX12 increased SOX12 expression compared with pcDNA3.1 (Fig. 7).

Discussion

Multiple miRNAs are involved in HCC tumorigenesis and development (21,22). In the present study, it was demonstrated that miR-744 was downregulated in HCC tissues and cell lines compared with levels in adjacent normal tissues and hepatocytes, which was consistent with previous results (15,16). Furthermore, it was demonstrated that decreased miR-744 levels were associated with TNM stage and lymph node metastasis, and that restoration of miR-744 suppressed the proliferation, migration, and invasion of HCC cells, which suggested that miR-744 plays a fundamental role in HCC progression.

Previous studies have reported that miR-744 is frequently dysregulated and functions as a tumor suppressor in breast cancer (12) and cervical cancer (14). Conversely miR-744 is highly expressed in head and neck cancer (23), non-small lung cancer (24), pancreatic (10) and prostate cancer (13), and nasopharyngeal carcinoma (9,11) and functions as an oncogene in these cancer types. The contradictory effects of miR-744 in various tumors indicate that miR-744 may exhibit different biological functions in different types of cancer. Previous studies suggest that miR-744 expression is decreased in HCC tissues (15,16), and that miR-744 overexpression inhibits cell proliferation by targeting c-Myc (16). However, the role of miR-744 in HCC cell migration and invasion remains largely unknown. In the present study, it was demonstrated that miR-744 significantly suppressed the proliferation, migration, and invasion of HCC cells, further supporting the function of miR-744 as a tumor suppressor in HCC.

Although miR-744 has been indicated to inhibit cell proliferation by targeting c-Myc, the mechanism of miR-744 in HCC progression remains largely unclear. To investigate the underlying mechanisms by which miR-744 exerts its biological effects on HCC cell migration and invasion, it is necessary to identify its targets. The present study used three algorithms (TargetScan, miRanda, and miRDB) to identify SOX12, a member of the Sox (SRY-related HMG-box) family of transcription factors (25), as a candidate target for further investigation based on its biological function. Previous studies report that SOX12 is upregulated in HCC tissues (26,27) and functions as an oncogene associated with HCC progression (26,27). Additionally, SOX12 induces the EMT process by regulating E-cadherin and Twist expression (18,26). In the present study, SOX12 was identified as a direct target of miR-744 by luciferase-reporter assay, RT-qPCR, and western blot analysis. Furthermore, an inverse correlation between miR-744 expression and SOX12 mRNA levels in HCC tissues was observed. It was confirmed that restoration of SOX12 expression partially abrogated the functional effect of miR-744 on HCC cell proliferation, migration, and invasion. These data provided reliable evidence suggesting that miR-744 exerts an inhibitory effect on HCC progression, at least in part, by inhibiting SOX12 expression and translation.

In conclusion, the findings revealed that miR-744 levels were lower in HCC tissues and cell lines, and low levels of miR-744 were associated with TNM stage and lymph node metastasis. The results also revealed that restoring miR-744 inhibited HCC cell proliferation, migration, and invasion by repressing SOX12 expression. Therefore, targeting miR-744/SOX12 interactions or rescuing miR-744 expression may represent a novel therapeutic strategy for treating HCC patients.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Research Fund of the Education Department of Jilin Province (grant no. 2017000032JC).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

WZ and YL conceived the experiments; WZ, KL and SL performed the experiments; BJ and SL analyzed the data; WZ and YL wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of the First Hospital of Jilin University, and written informed consent was obtained from all patients whose biological samples were used in the study.

Patient consent for publication

Written informed consent was obtained from all patients.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Venook AP, Papandreou C, Furuse J and de Guevara LL: The incidence and epidemiology of hepatocellular carcinoma: A global and regional perspective. Oncologist. 15 Suppl 4:S5–S13. 2010. View Article : Google Scholar

3 

Lau WY and Lai EC: Hepatocellular carcinoma: Current management and recent advances. Hepatobiliary Pancreat Dis Int. 7:237–257. 2008.PubMed/NCBI

4 

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

5 

Pillai RS: MicroRNA function: Multiple mechanisms for a tiny RNA? RNA. 11:1753–1761. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Hayes J, Peruzzi PP and Lawler S: MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol Med. 20:460–469. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Mao B and Wang G: MicroRNAs involved with hepatocellular carcinoma (Review). Oncol Rep. 34:2811–2820. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Giordano S and Columbano A: MicroRNAs: New tools for diagnosis, prognosis, and therapy in hepatocellular carcinoma? Hepatology. 57:840–847. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Yu Q, Zhang F, Du Z and Xiang Y: Up-regulation of serum miR-744 predicts poor prognosis in patients with nasopharyngeal carcinoma. Int J Clin Exp Med. 8:13296–13302. 2015.PubMed/NCBI

10 

Zhou W, Li Y, Gou S, Xiong J, Wu H, Wang C, Yan H and Liu T: MiR-744 increases tumorigenicity of pancreatic cancer by activating Wnt/beta-catenin pathway. Oncotarget. 6:37557–37569. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Fang Y, Zhu X, Wang J, Li N, Li D, Sakib N, Sha Z and Song W: MiR-744 functions as a proto-oncogene in nasopharyngeal carcinoma progression and metastasis via transcriptional control of ARHGAP5. Oncotarget. 6:13164–13175. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Vislovukh A, Kratassiouk G, Porto E, Gralievska N, Beldiman C, Pinna G, El'skaya A, Harel-Bellan A, Negrutskii B and Groisman I: Proto-oncogenic isoform A2 of eukaryotic translation elongation factor eEF1 is a target of miR-663 and miR-744. Br J Cancer. 108:2304–2311. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Guan H, Liu C, Fang F, Huang Y, Tao T, Ling Z, You Z, Han X, Chen S, Xu B and Chen M: MicroRNA-744 promotes prostate cancer progression through aberrantly activating Wnt/beta-catenin signaling. Oncotarget. 8:14693–14707. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Chen XF and Liu Y: MicroRNA-744 inhibited cervical cancer growth and progression through apoptosis induction by regulating Bcl-2. Biomed Pharmacother. 81:379–387. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Tan YL, Bai ZG, Zou WL, Ma XM, Wang TT, Guo W, Liu J, Li JS, Jie Yin, Zang YJ, et al: miR-744 is a potential prognostic marker in patients with hepatocellular carcinoma. Clin Res Hepatol Gastroenterol. 39:359–365. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Lin F, Ding R, Zheng S, Xing D, Hong W, Zhou Z and Shen J: Decrease expression of microRNA-744 promotes cell proliferation by targeting c-Myc in human hepatocellular carcinoma. Cancer Cell Int. 14:582014. View Article : Google Scholar : PubMed/NCBI

17 

Edge SB and Compton CC: The American joint committee on cancer: The 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 17:1471–1474. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Ding H, Quan H, Yan W and Han J: Silencing of SOX12 by shRNA suppresses migration, invasion and proliferation of breast cancer cells. Biosci Rep. 36:e003892016. View Article : Google Scholar :

19 

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

20 

Liu Y, Zhang W, Liu S, Liu K, Ji B and Wang Y: miR-365 targets ADAM10 and suppresses the cell growth and metastasis of hepatocellular carcinoma. Oncol Rep. 37:1857–1864. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Huang JT, Liu SM, Ma H, Yang Y, Zhang X, Sun H, Zhang X, Xu J and Wang J: Systematic review and meta-analysis: Circulating miRNAs for diagnosis of hepatocellular carcinoma. J Cell Physiol. 231:328–335. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Khare S, Zhang Q and Ibdah JA: Epigenetics of hepatocellular carcinoma: Role of microRNA. World J Gastroenterol. 19:5439–5445. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Nurul-Syakima AM, Yoke-Kqueen C, Sabariah AR, Shiran MS, Singh A and Learn-Han L: Differential microRNA expression and identification of putative miRNA targets and pathways in head and neck cancers. Int J Mol Med. 28:327–336. 2011.PubMed/NCBI

24 

Sha Z, Zhu X, Li N, Li Y and Li D: Proto-oncogenic miR-744 is upregulated by transcription factor c-Jun via a promoter activation mechanism. Oncotarget. 7:64977–64986. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Hoser M, Potzner MR, Koch JM, Bosl MR, Wegner M and Sock E: Sox12 deletion in the mouse reveals nonreciprocal redundancy with the related Sox4 and Sox11 transcription factors. Mol Cell Biol. 28:4675–4687. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Huang W, Chen Z, Shang X, Tian D, Wang D, Wu K, Fan D and Xia L: Sox12, a direct target of FoxQ1, promotes hepatocellular carcinoma metastasis through up-regulating Twist1 and FGFBP1. Hepatology. 61:1920–1933. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Jiang T, Guan LY, Ye YS, Liu HY and Li R: MiR-874 inhibits metastasis and epithelial-mesenchymal transition in hepatocellular carcinoma by targeting SOX12. Am J Cancer Res. 7:1310–1321. 2017.PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 40 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
Zhang W, Liu K, Liu S, Ji B and Liu Y: MicroRNA‑744 inhibits migration and invasion of hepatocellular carcinoma cells by targeting SOX12 Retraction in /10.3892/or.2022.8364. Oncol Rep 40: 3585-3592, 2018
APA
Zhang, W., Liu, K., Liu, S., Ji, B., & Liu, Y. (2018). MicroRNA‑744 inhibits migration and invasion of hepatocellular carcinoma cells by targeting SOX12 Retraction in /10.3892/or.2022.8364. Oncology Reports, 40, 3585-3592. https://doi.org/10.3892/or.2018.6774
MLA
Zhang, W., Liu, K., Liu, S., Ji, B., Liu, Y."MicroRNA‑744 inhibits migration and invasion of hepatocellular carcinoma cells by targeting SOX12 Retraction in /10.3892/or.2022.8364". Oncology Reports 40.6 (2018): 3585-3592.
Chicago
Zhang, W., Liu, K., Liu, S., Ji, B., Liu, Y."MicroRNA‑744 inhibits migration and invasion of hepatocellular carcinoma cells by targeting SOX12 Retraction in /10.3892/or.2022.8364". Oncology Reports 40, no. 6 (2018): 3585-3592. https://doi.org/10.3892/or.2018.6774