miR-154 inhibits migration and invasion of human non-small cell lung cancer by targeting ZEB2

Retraction in: /10.3892/ol.2021.12873

  • Authors:
    • Xingyu Lin
    • Zhiguang Yang
    • Peng Zhang
    • Yunpeng Liu
    • Guoguang Shao
  • View Affiliations

  • Published online on: May 16, 2016     https://doi.org/10.3892/ol.2016.4577
  • Pages: 301-306
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Emerging evidence suggests that microRNAs (miRs) play critical roles in the development and progression of non-small cell lung cancer (NSCLC). In a previous study, the present authors demonstrated that miR-154 acts as a tumor suppressor in NSCLC; however, its underlying molecular mechanism and target in NSCLC remain poorly understood. In the present study, ectopic expression of miR‑154 remarkably suppressed cell migration and invasion in NSCLC cells. Zinc finger E‑box binding homeobox 2 (ZEB2) was identified as a direct target of miR‑154 in NSCLC cells. Furthermore, overexpression of miR‑154 could decrease the expression of ZEB2 at the messenger RNA and protein levels. Ectopic expression of miR‑154 also increased the levels of E‑cadherin, an epithelial marker, and decreased the levels of vimentin, a mesenchymal marker, which contributed to suppress epithelial‑mesenchymal transition (EMT) and to inhibit cell migration and invasion. In addition, downregulation of ZEB2 exerted similar effects to those caused by miR‑154 overexpression on NSCLC cell migration and invasion, while upregulation of ZEB2 could significantly reverse the inhibitory effects on migration and invasion caused by miR‑154 on NSCLC cells. These findings demonstrated that miR‑154 inhibited migration and invasion of NSCLC cells by regulating EMT through targeting ZEB2, suggesting that miR‑154 may be a potential anticancer therapeutic target for NSCLC.

Introduction

Non-small cell lung cancer (NSCLC), which represents ~75–85% of all lung carcinomas, is one of the most common causes of cancer-associated mortalities worldwide (1,2). The majority of NSCLC patients are diagnosed at an advanced stage, presenting with metastatically or locally advanced disease, leading to ~90% of lung cancer patients succumbing to metastasis (3). The metastasis progression of NSCLC involves multi-step genetic events, and the molecular underlying mechanisms have not been documented to date (4). Accumulating evidence has revealed that non-coding small RNAs are involved in NSCLC initiation, progression and metastasis (5,6), which provides novel insights for the treatment of this disease.

MicroRNAs (miRNAs or miRs) are a class of small, non-coding RNAs of ~19–25 nucleotides in length, which regulate gene expression at the post-transcriptional level by interacting with the 3′-untranslated regions (3′-UTRs) of their target messenger (m)RNAs (7,8). Increasing evidence has suggested that miRNAs are involved in a number of biological processes, including development, differentiation, apoptosis, metabolism, immunity and tumor progression (5,79). In addition, a growing body of evidence strongly suggests that the dysregulation or dysfunction of miRNAs may modulate tumor initiation and progression, and may participate in tumor cell invasion and metastasis (5,914). miRNAs may function as oncogenes or tumor suppressors depending on their specific target genes (15).

The miR-154 cluster, which is located in the human imprinted 14q32 domain (mouse chromosome 12F2), has been identified as a tumor suppressor in various types of human cancer, including prostate (16), breast (17), colorectal (18) and thyroid cancer (19). For NSCLC, a previous study by the present authors revealed that miR-154 expression is downregulated in human primary NSCLC tissues and cell lines, and that exogenous miR-154 significantly suppressed NSCLC growth in vitro and in vivo (20), suggesting that miR-154 has potential therapeutic application against NSCLC. However, the molecular mechanisms by which it exerts its functions remain largely unknown. Thus, the identification of novel miR-154 targets would provide novel insights into the molecular mechanism underlying the miR-154-induced inhibition of tumorigenic properties in cancer cells.

The present study aimed to investigate the role and mechanism of miR-154 in the migration and invasion of NSCLC. Overexpression of miR-154 significantly suppressed the migration and invasion abilities of NSCLC cells in vitro. In addition, the epithelial-mesenchymal transition (EMT) regulator zinc finger E-box binding homeobox 2 (ZEB2) was identified as one of the direct target genes of miR-154. miR-154 inhibited cell migration and invasion by regulating EMT through inhibiting the function of ZEB2.

Materials and methods

Cell culture

The human NSCLC cell line A549 was purchased from the Institute of Biochemistry and Cell Biology of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in the presence of 10% heat-inactivated fetal bovine serum (FBS) (HyClone; GE Healthcare Life Sciences, Chalfont, UK) and penicillin (100 U/ml; Sigma-Aldrich, St. Louis, MO, USA) in a humidified 5% (v/v) atmosphere of CO2 at 37°C.

Plasmids and transfection

miR-154 mimic and the corresponding negative control (miR-NC), as well as a small interfering (si)RNA targeting ZEB2 (si-ZEB2) and the corresponding scramble control (si-Scramble), were purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China). The ZEB2 overexpression plasmid (pcDNA3.0-ZEB2) was generated using the following primers: Sense 5′-GGGGTACCATGCGAACTGCCATCTGA-3′ and antisense 5′-TTGCGGCCGCGTGCTTCAAAGAACAGGGTG-3′. The polymerase chain reaction (PCR) fragment was inserted into the pcDNA3.0 vector within the KpnI and NotI restriction sites (Invitrogen; Thermo Fisher Scientific, Inc.). The 3′-UTRs of human wild-type (WT) ZEB2 (pGL3-ZEB2 WT; Promega Corporation, Madison, WI, USA) containing the potential binding sites of miR-154 were amplified and constructed as described by Guan et al (21). Mutations (MUT)in the miR-154 binding-sites module of ZEB2 (pGL3-ZEB2 MUT) were introduced using the QuikChange Site-Directed Mutagenesis kit (Stratagene; Agilent Technologies, Inc., Santa Clara, CA, USA), according to the manufacturer's protocol.

Transfection was performed in A549 cells using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol.

RNA extraction and reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from cells using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. RNA was reverse transcribed into complementary DNA using PrimeScript™ One Step RT-PCR kit (Takara Biotechnology Co., Ltd., Dalian, China), following the manufacturer's protocol. RT-qPCR was performed using a standard SYBR Green PCR kit (Takara Biotechnology Co., Ltd.) on a 7900HT Fast Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The forward and reverse primers for ZEB2 were 5′-AGGAGCAGGTAATCG-3′ and 5′-TGGGCACTCGTAAGG-3, respectively; while those for glyceraldehyde 3-phosphate dehydrogenase (GAPDH) were 5′-GAAGGTGAAGGTCGGAGTC-3′ and 5′-GAAGATGGTGATGGGATTTC-3′, respectively. The reaction volume was 10 µl, and the mixture contained 5 µl SYBR Premix Ex Taq, 1 µl cDNA, 0.2 µl (10 µM) ZEB2 forward primer and ZEB2 reverse primer, or 0.2 µl (10 µM) GAPDH forward primer and GAPDH reverse primer, and 3.6 µl dH2O. The reaction conditions used for mRNA detection were as follows: 95°C for 5 min, followed by 40 cycles of 95°C for 5 sec, 60°C for 20 sec and 72°C for 20 sec. Relative quantification of ZEB2 was presented as the fold-change upon normalization to the GAPDH RNA levels, according to the equation 2-∆∆Cq (22)in the Rotor-Gene 6000 software version 1.7 (Qiagen GmbH, Hilden, Germany).

Cell migration and invasion assays

For the migration assays, A549 cells were harvested 48 h post-transfection, and 5×104 cells in 200 µl serum-free RPMI-1640 medium were seeded into the upper Transwell chamber (pore size, 8 mm; Corning Life Sciences, Tewksbury, MA, USA). For the invasion assays, transfected cells in 200 µl serum-free RPMI-1640 medium were placed into the upper chamber of an insert coated with Matrigel (BD Biosciences, San Jose, CA, USA), according to the manufacturer's protocol. RPMI-1640 medium containing 20% FBS was added to the lower chamber. Following 24 h of incubation, the cells remaining on the upper membrane were removed with cotton swabs, whereas those that had migrated or invaded through the membrane were fixed in 90% ethanol (Sigma-Aldrich) and stained with 0.1% crystal violet (Sigma-Aldrich). The number of cells migrating or invading were photographed and counted at five randomly selected fields under an IX51 inverted microscope (Olympus Corporation, Tokyo, Japan; magnification, ×200). All experiments were independently repeated three times.

Luciferase assay

A549 cells (2×105) were seeded in a 24-well plate for 24 h prior to be co-transfected with pGL3-ZEB2 WT or pGL3-ZEB2 MUT and miR-154 or miR-NC using Lipofectamine 2000. Cells were collected 48 h post-transfection, and Renilla and firefly luciferase activities were assayed with the Dual-Luciferase® Reporter Assay System (Promega Corporation), according to the manufacturer's protocol. The specific activity was expressed as the fold-changes of the experimental group vs. those of the miR-NC group.

Western blotting

Cells were lysed in ice-cold radioimmunoprecipitation assay buffer (Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at 48 h post-transfection. Proteins were quantified using Pierce BCA Protein Assay kit (Thermo Fisher Scientific, Inc.). Equal amounts of protein (30 µg) were separated by 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis (Bio-Rad Laboratories, Inc., Hercules, CA, USA) for 2 h at 120 V, and then transferred to nitrocellulose membranes (Invitrogen; Thermo Fisher Scientific, Inc.). Membranes were blocked in 4% dry milk diluted with Tri-buffered saline Tween-20 (20 mmol/l Tris-HCl, 150 mmol/l NaCl (pH 7.5) and 0.1% Tween-20) at room temperature for 1 h, and immunostained with the following primary antibodies at 4°C overnight: Rabbit monoclonal anti-human ZEB2 (1:1,000 dilution; cat. no. sc-48789; Santa Cruz Biotechnology, Inc.), mouse monoclonal anti-human E cadherin (1:1,000 dilution; cat. no. sc-8426; Santa Cruz Biotechnology, Inc.), rabbit monoclonal anti-human vimentin (1:1,500 dilution; cat. no. sc-5565; Santa Cruz Biotechnology, Inc.) and rabbit monoclonal anti-human GAPDH (1:5,000; cat. no. 2118L; Cell Signaling Technology, Inc., Danvers, MA, USA). Next, membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit or goat anti-mouse secondary antibody (1:5,000 dilution; cat. no. sc-2004; sc-2005; Santa Cruz Biotechnology, Inc.) at room temperature for 2 h. The blots were detected with an enhanced chemiluminescence detection kit (Thermo Fisher Scientific, Inc.) and exposed in a Molecular Imager® ChemiDoc™ XRS system (Bio-Rad Laboratories, Inc.). Protein levels were normalized to those of GAPDH.

Statistical analysis

Statistical significance was determined by Student's t-test using GraphPad Prism version 5 software (GraphPad Software, Inc., La Jolla, CA, USA). Data were expressed as the mean ± standard deviation. All experiments were repeated ≥3 times, and each experiment consisted of triplicate wells. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-154 inhibited migration and invasion of NSCLC cells

To examine the effect of miR-154 on the migration and invasion abilities of NSCLC cells, miR-154 mimic or miR-NC were transfected into A549 cells, and in vitro migration and invasion assays were then performed. Transwell assay demonstrated that miR-154 significantly repressed in vitro the migration and invasion abilities of NSCLC cells (P<0.01; Fig. 1A and B, respectively).

ZEB2 was a target of miR-154 in NSCLC cells

To detect the molecular mechanism by which miR-154 suppresses the metastasis of NSCLC cells, putative target genes of miR-154 in human cells were predicted using the tools miRanda (http://www.microrna.org/), PicTar (http://pictar.mdc-berlin.de/) and TargetScanS version 6.2 (http://www.targetscan.org/). Among the predicted candidates, ZEB2 was selected as a miR-154 target gene, since ZEB2 has been demonstrated to be involved in the development and metastasis of various types of human cancer (23,24). As indicated in Fig. 2A, miR-154 contains one predicted binding site in the 3′-UTR of ZEB2 mRNA. Luciferase activity assay revealed that miR-154 significantly inhibited the luciferase activity of the WT 3′-UTR of ZEB2 (P<0.01), but not that of the MUT 3′-UTR of ZEB2, in A549 cells (Fig. 2B), indicating the direct regulation of miR-154 in the 3′-UTR of ZEB2 mRNA.

Overexpression of miR-154 regulates ZEB2 expression and EMT

To determine whether miR-154 affects the regulation of endogenous ZEB2, endogenous ZEB2 mRNA and protein expression levels were measured in the A549 cells transfected with miR-154 mimic or miR-NC by RT-qPCR and western blotting, respectively. The results demonstrated that overexpression of miR-154 significantly downregulated ZEB2 mRNA and protein levels in A549 cells (P<0.01; Fig. 3A and B, respectively). These findings suggested that ZEB2 is a bona fide target of miR-154. It has been previously demonstrated that ZEB2 is a vital EMT inducer through suppressing E-cadherin expression or inducing vimentin expression in human cancer (24,25). To further confirm that ZEB2 acts as a target of miR-154, the effect of miR-154 on two downstream effectors of ZEB2 was examined by western blotting. As indicated in Fig. 3B, overexpression of miR-154 in A549 cells markedly upregulated the protein expression of E-cadherin, an epithelial marker, and downregulated that of vimentin, a mesenchymal marker, which contributed to suppress EMT and to inhibit cell migration and invasion. Taken together, the present data indicate that miR-154 could directly inhibit ZEB2 expression and regulate EMT in NSCLC cells.

Inhibition of ZEB2 exerted a similar effect to that of miR-154 overexpression

To study the effect of ZEB2 on NSCLC cell migration and invasion, A549 cells were transfected with si-ZEB2 or si-Scramble. RT-qPCR and western blotting confirmed that si-ZEB2 could significantly inhibit ZEB2 expression at the mRNA and protein levels in A549 cells (P<0.01; Fig. 4A and B, respectively). Furthermore, inhibition of ZEB2 had a similar effect to that of miR-154 overexpression, since it significantly repressed the migration and invasion of A549 cells in vitro (P<0.01; Fig. 4C and D, respectively).

ZEB2 overexpression attenuated the suppressive effect of miR-154

The present study further investigated whether overexpression of ZEB2 could reverse the suppressive effect of miR-154 on cell migration and invasion. A549 cells were transfected with plasmid pcDNA3.0-ZEB2 or empty pcDNA3.0 vector, which served as a blank control. RT-qPCR and western blot assays confirmed that pcDNA3.0-ZEB2 could significantly increase ZEB2 expression at the mRNA and protein levels in A549 cells (P<0.01; Fig. 5A and B, respectively). A549 cells were co-transfected with miR-154 and pcDNA3.0-ZEB2 or empty pcDNA3.0 vector for 24 h, and migration and invasion assays were then performed by Transwell assay. The results demonstrated that overexpression of ZEB2 could markedly reverse the suppressive effect of miR-154 on migration and invasion of A549 cells (Fig. 5C and D, respectively).

Discussion

Accumulating evidence has demonstrated that miRNAs could play crucial roles in tumor growth, migration, invasion and angiogenesis in various malignances such as NSCLC (6,26). For instance, Yongchun et al (27) reported that miR-195 could decrease cell proliferation, migration and invasion of NSCLC via the proto-oncogene Myb. You et al (28) observed that miR-132 blocks the migration and invasion of NSCLC cells through targeting the EMT regulator ZEB2. Yu et al (29) reported that upregulation of miR-1 inhibited A549 cell proliferation, migration and invasion by regulating phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha through the phosphoinositide-3-kinase/Akt signaling pathway. Shan et al (30) noticed that ectopic expression of miR-153 significantly inhibited the proliferation, migration and invasion of NSCLC cells in vitro by targeting a disintegrin and metalloproteinase 9. The present results indicated that miR-154 was involved in the migration and invasion of NSCLC.

miR-154, a recently identified miRNA, has been reported to act as a tumor suppressor in a variety of tumors by targeting several oncogenes (15,17,30,31). For example, Zhu et al (16,31) reported that miR-154 inhibited the growth of prostate cancer cells by targeting high-mobility group AT-hook 2 and cyclin D2 (CCND2). Xin et al (18) reported that miR-154 remarkably suppressed cell proliferation, colony formation, migration and invasion in colorectal cancer cells by targeting Toll-like receptor 2. Wang et al (32) reported that overexpression of miR-154 suppressed tumor cell malignance and G1/S transition in hepatocellular cancer cells by targeting CCND2. In the present study, miR-154 could significantly inhibit the migration and invasion of NSCLC cells by targeting ZEB2.

ZEB2/Smad interacting protein 1, as a member of the delta EF-1 family of two-handed zinc finger factors, has been observed to be elevated in various types of human cancer, including NSCLC (23,33). Growing evidence suggests that ZEB2 could induce EMT through suppressing E-cadherin expression or inducing vimentin expression, thus facilitating the metastasis of cancer cells (24,25,34,35). ZEB2 is regulated by several miRNAs, including miR-132 (28), miR-101 (36), miR-141 (37), miR-144 (21) and miR-335 (38). In the present study, ZEB2 was observed to be a target of miR-154 by luciferase assay, and miR-154 could inhibit EMT and decrease the expression of ZEB2 at the mRNA and protein levels. In addition, underexpression of ZEB2 exerted similar effects to those caused by miR-154 on NSCLC cells in terms of migration and invasion, while overexpression of ZEB2 could significantly reverse the inhibitory effects of miR-154 on NSCLC cell migration and invasion. These findings suggested that miR-154 inhibited cell migration and invasion of NSCLC by targeting ZEB2 through inhibiting EMT.

In summary, the present study offers evidence that miR-154 acts as a tumor suppressor and blocks in vitro migration and invasion of NSCLC cells partially through the downregulation of ZEB2, leading to EMT inhibition. The present data provide novel insights into the mechanism responsible for the development of human NSCLC. Therefore, miR-154 could be regarded as a novel therapeutic target for NSCLC.

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 62:10–29. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Ramalingam SS, Owonikoko TK and Khuri FR: Lung cancer: New biological insights and recent therapeutic advances. CA Cancer J Clin. 61:91–112. 2011. View Article : Google Scholar : PubMed/NCBI

3 

de Sánchez Cos J, Sojo González MA, Montero MV, Pérez Calvo MC, Vicente MJ and Valle MH: Non-small cell lung cancer and silent brain metastasis. Survival and prognostic factors. Lung Cancer. 63:140–145. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Mason DP: The role of surgery for locally advanced non-small cell lung cancer. Cleve Clin J Med. 79Electronic. (Suppl 1): eS38–eS41. 2012.PubMed/NCBI

5 

Tutar Y: miRNA and cancer; computational and experimental approaches. Curr Pharm Biotechnol. 15:4292014. View Article : Google Scholar : PubMed/NCBI

6 

Skrzypski M, Dziadziuszko R and Jassem J: MicroRNA in lung cancer diagnostics and treatment. Mutat Res. 717:25–31. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Osman A: MicroRNAs in health and disease - basic science and clinical applications. Clin Lab. 58:393–402. 2012.PubMed/NCBI

8 

Mendell JT and Olson EN: MicroRNAs in stress signaling and human disease. Cell. 148:1172–1187. 2012. View Article : Google Scholar : PubMed/NCBI

9 

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

10 

Chen PS, Su JL and Hung MC: Dysregulation of microRNAs in cancer. J Biomed Sci. 19:902012. View Article : Google Scholar : PubMed/NCBI

11 

Melo SA and Esteller M: Dysregulation of microRNAs in cancer: Playing with fire. FEBS Lett. 585:2087–2099. 2011. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Boeri M, Pastorino U and Sozzi G: Role of microRNAs in lung cancer: MicroRNA signatures in cancer prognosis. Cancer J. 18:268–274. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Guz M, Rivero-Müller A, Okoń E, Stenzel-Bembenek A, Polberg K, Słomka M and Stepulak A: MicroRNAs-role in lung cancer. Dis Markers. 2014:2181692014. View Article : Google Scholar : PubMed/NCBI

15 

Kent OA and Mendell JT: A small piece in the cancer puzzle: MicroRNAs as tumor suppressors and oncogenes. Oncogene. 25:6188–6196. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Zhu C, Li J, Cheng G, Zhou H, Tao L, Cai H, Li P, Cao Q, Ju X, Meng X, et al: miR-154 inhibits EMT by targeting HMGA2 in prostate cancer cells. Mol Cell Biochem. 379:69–75. 2013. View Article : Google Scholar : PubMed/NCBI

17 

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 : 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 : PubMed/NCBI

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 

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

21 

Guan H, Liang W, Xie Z, Li H, Liu J, Liu L, Xiu L and Li Y: Down-regulation of miR-144 promotes thyroid cancer cell invasion by targeting ZEB1 and ZEB2. Endocrine. 48:566–574. 2015. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Peinado H, Olmeda D and Cano A: Snail, Zeb and bHLH factors in tumour progression: An alliance against the epithelial phenotype? Nat Rev Cancer. 7:415–428. 2007. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

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

26 

Boeri M, Sestini S, Fortunato O, et al: Recent advances of microRNA-based molecular diagnostics to reduce false-positive lung cancer imaging. Expert review of molecular diagnostics. 15:801–813. 2015.PubMed/NCBI

27 

Yongchun Z, Linwei T, Xicai W, Lianhua Y, Guangqiang Z, Ming Y, Guanjian L, Yujie L and Yunchao H: MicroRNA-195 inhibits non-small cell lung cancer cell proliferation, migration and invasion by targeting MYB. Cancer Lett. 347:65–74. 2014. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Yu QQ, Wu H, Huang X, Shen H, Shu YQ, Zhang B, Xiang CC, Yu SM, Guo RH and Chen L: MiR-1 targets PIK3CA and inhibits tumorigenic properties of A549 cells. Biomed Pharmacother. 68:155–161. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Shan N, Shen L, Wang J, He D and Duan C: MiR-153 inhibits migration and invasion of human non-small-cell lung cancer by targeting ADAM19. Biochem Biophys Res Commun. 456:385–391. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Zhu C, Shao P, Bao M, Li P, Zhou H, Cai H, Cao Q, Tao L, Meng X, Ju X, et al: miR-154 inhibits prostate cancer cell proliferation by targeting CCND2. Urol Oncol. 32:31.e9–31.e16. 2014. View Article : Google Scholar

32 

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 : PubMed/NCBI

33 

Gemmill RM, Roche J, Potiron VA, Nasarre P, Mitas M, Coldren CD, Helfrich BA, Garrett-Mayer E, Bunn PA and Drabkin HA: ZEB1-responsive genes in non-small cell lung cancer. Cancer Lett. 300:66–78. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Qi S, Song Y, Peng Y, Wang H, Long H, Yu X, Li Z, Fang L, Wu A, Luo W, et al: ZEB2 mediates multiple pathways regulating cell proliferation, migration, invasion, and apoptosis in glioma. PLoS One. 7:e388422012. View Article : Google Scholar : PubMed/NCBI

35 

Chu PY, Hu FW, Yu CC, Tsai LL, Yu CH, Wu BC, Chen YW, Huang PI and Lo WL: Epithelial-mesenchymal transition transcription factor ZEB1/ZEB2 co-expression predicts poor prognosis and maintains tumor-initiating properties in head and neck cancer. Oral Oncol. 49:34–41. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Guo F, Cogdell D, Hu L, Yang D, Sood AK, Xue F and Zhang W: MiR-101 suppresses the epithelial-to-mesenchymal transition by targeting ZEB1 and ZEB2 in ovarian carcinoma. Oncol Rep. 31:2021–2028. 2014.PubMed/NCBI

37 

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

38 

Sun Z, Zhang Z, Liu Z, Qiu B, Liu K and Dong G: MicroRNA-335 inhibits invasion and metastasis of colorectal cancer by targeting ZEB2. Med Oncol. 31:9822014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 12 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin X, Yang Z, Zhang P, Liu Y and Shao G: miR-154 inhibits migration and invasion of human non-small cell lung cancer by targeting ZEB2 Retraction in /10.3892/ol.2021.12873. Oncol Lett 12: 301-306, 2016
APA
Lin, X., Yang, Z., Zhang, P., Liu, Y., & Shao, G. (2016). miR-154 inhibits migration and invasion of human non-small cell lung cancer by targeting ZEB2 Retraction in /10.3892/ol.2021.12873. Oncology Letters, 12, 301-306. https://doi.org/10.3892/ol.2016.4577
MLA
Lin, X., Yang, Z., Zhang, P., Liu, Y., Shao, G."miR-154 inhibits migration and invasion of human non-small cell lung cancer by targeting ZEB2 Retraction in /10.3892/ol.2021.12873". Oncology Letters 12.1 (2016): 301-306.
Chicago
Lin, X., Yang, Z., Zhang, P., Liu, Y., Shao, G."miR-154 inhibits migration and invasion of human non-small cell lung cancer by targeting ZEB2 Retraction in /10.3892/ol.2021.12873". Oncology Letters 12, no. 1 (2016): 301-306. https://doi.org/10.3892/ol.2016.4577