MicroRNA‑212 suppresses the proliferation and migration of osteosarcoma cells by targeting forkhead box protein A1

Retraction in: /10.3892/etm.2021.10409

  • Authors:
    • Jian Liu
    • Bohua Chen
    • Bin Yue
    • Junde Yang
  • View Affiliations

  • Published online on: November 7, 2016     https://doi.org/10.3892/etm.2016.3880
  • Pages: 4135-4141
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs) are a class of small non-coding RNAs that function as critical gene regulators by targeting the 3' untranslated region (UTR) of mRNA, causing translational repression or mRNA degradation. Deregulation of specific miRNAs, including miR‑212, has been identified in patients with osteosarcoma. However, the underlying mechanism is yet to be fully elucidated. The present study aimed to reveal the regulatory mechanism of miR‑212 in osteosarcoma cell viability and migration. Quantitative polymerase chain reaction data revealed that miR‑212 was significantly downregulated in osteosarcoma tissues compared with normal bone tissues. miR‑212 was also downregulated in osteosarcoma cell lines compared with normal osteoblast cell lines. Overexpression of miR‑212 significantly suppressed the viability and migration of human osteosarcoma MG‑63 and Saos‑2 cell lines. In addition, forkhead box protein A1 (FOXA1), an oncogene in osteosarcoma, was predicted to be a putative target of miR‑212 by bioinformatical analysis. Furthermore, luciferase reporter assay data confirmed that miR‑212 could directly bind to the seed sequences within the 3'UTR of FOXA1 mRNA, and miR‑212 negatively mediated the protein levels of FOXA1 in osteosarcoma MG‑63 and Saos‑2 cells. Moreover, knockdown of FOXA1 also led to a significant decrease in the viability and migration of osteosarcoma MG‑63 and Saos‑2 cells and the expression levels of FOXA1 were significantly upregulated in osteosarcoma tissues and cell lines. These data suggest that miR‑212 inhibits the viability and migration of osteosarcoma cells by targeting FOXA1. Accordingly, miR‑212 may become a potential candidate for osteosarcoma therapy.

Introduction

Osteosarcoma (OS) is the most common mesenchymal sarcoma in bones (1). In recent years, various oncogenes and tumor suppressors have been identified to be associated with the development and progression of osteosarcoma (24). Therefore, investigation into the genes involved in the regulation of osteosarcoma cell viability and migration is crucial for the development of therapeutic targets for osteosarcoma.

Accumulating evidence supports the cancer-associated effects of microRNAs (miRNAs), which are a group of endogenous small RNA containing about 22 nucleotides, which function through the inhibition of the expression of various genes. Bioinformatics algorithms suggest that human miRNA regulates up to 30% of human genes, which represents the majority of genetic pathways (5). Moreover, various miRNA are deregulated in different types of human cancers and are important in promoting or suppressing the development and progression of malignant tumors (6). Therefore, these miRNA act as oncogenes or tumor suppressors in cancer, which may become novel targets for anticancer therapies.

The role of miR-212 has been elucidated in several cancer types. Xu et al (7) identified that the expression of miR-212 was significantly decreased in gastric cancer caused by DNA hypermethylation, suggesting that downregulation of miR-212 may be associated with the development of gastric cancer. On the contrary, miR-212 was demonstrated to be upregulated in pancreatic adenocarcinoma tissues, and overexpression of miR-212 enhanced the proliferation of pancreatic cancer cells, indicating that miR-212 may be an oncogenic miRNA in pancreatic cancer (8). Thus, miR-212 has opposing roles in cancer, and further investigation is required in different cancer types. Recently, Luo et al (9) found that the expression level of miR-212 was markedly reduced in osteosarcoma tissues compared with adjacent normal tissues. Furthermore, they identified that overexpression of miR-212 inhibited cell proliferation and invasion, partly at least, via targeting the sex-determining region Y-box 4 (Sox4) in osteosarcoma cells. These observations suggest that miR-212 is suppressive in osteosarcoma. However, as one miRNA has multiple types of targets, whether other genes are also involved in miR-212-mediated malignant phenotypes of osteosarcoma cells remains unknown.

The present study aimed to examine the expression of miR-212 in osteosarcoma tissues, and elucidate its role in the regulation of osteosarcoma cell viability and migration. miR-212 targets were also studied, which may be involved in this process.

Materials and methods

Tissue

The present study was approved by the Ethics Committee of the Medical School of Qingdao University (Qingdao, China). Osteosarcoma tissues and matched normal non-tumor tissues were obtained from 13 patients with osteosarcoma diagnosed by pathological analysis from the Eighth People's Hospital of Qingdao (Qingdao, China). Written informed consent was obtained from all patients and the characteristics of patients are shown in Table I. Patients did not receive any treatment prior to the surgery. Tissues were stored at −70°C until further use.

Table I.

Clinical characteristics of patients with osteosarcoma.

Table I.

Clinical characteristics of patients with osteosarcoma.

FactorsOsteosarcoma
Number of patients13
Age range, years (mean)27–51 (44.1)
Gender
  Male8
  Female5
Cell culture

Human osteosarcoma cell lines (HOS, Saos-2, U-2OS and MG-63) and the normal osteoblast cell line NHOst were purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS; both Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in a cell incubator containing 5% CO2 at 37°C.

Transfection

miR-212 mimics, negative control miRNA (miR-NC) and FOXA1 siRNA were provided by Sigma-Aldrich (Merck Millipore, Darmstadt, Germany). Cells were seeded in 24-well plates (1×105 cells/well) and transfected using a concentration of 100 nM that was diluted using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Following incubation at 5% CO2 and 37°C for 48 h, the cells were used for further analysis.

Quantitative polymerase chain reaction (qPCR)

Total RNA was isolated using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). miR-212 expression was determined using TaqMan MicroRNA assays (Applied Biosystems; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. U6 (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used as a normalization control for miRNA expression. To detect the mRNA levels of FOXA1, primers for FOXA1 and GAPDH were obtained from Shanghai Shenggong Co., Ltd., (Shanghai, China). The primer sequences were as follows: FOXA1 forward, 5′-GCAATACTCGCCTTACGGCT-3′ and reverse, 5′-TACACACCTTGGTAGTACGCC-3′; and GAPDH forward 5′GGAGCGAGATCCCTCCAAAAT-3′ and reverse 5′-GGCTGTTGTCATACTTCTCATGG-3′. GAPDH was used as a normalization control for gene expression and the qPCR reaction was performed using a Applied Biosystems 7500 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The qPCR reaction mixture contained 0.33 µl cDNA solution, 10 µl of 1X TaqMan universal PCR master mix, 2 µl 1X gene specific primer/probe set and 7.67 µl H2O, at a final reaction volume of 20 µl. The thermal cycling conditions were as follows: 95°C for 10 min, 40 cycles of denaturation at 95°C for 15 sec, and annealing/elongation step at 60°C for 60 sec. The relative fold changes of the miRNA and genes were calculated using the 2−ΔΔCt method (10).

Western blot analysis

Cells were lysed using assay lysis buffer (Beyotime Institute of Biotechnology, Wuhan, China) and the protein (50 µg) was separated by 12% SDS PAGE and subsequently transferred to polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). Rabbit anti-FOXA1 and anti-GAPDH antibodies (both 1:200; cat. nos. ab23738 and ab9485, respectively; both Abcam Cambridge MA, USA) were incubated with the membrane overnight at 4°C. Following washing three times, the membrane was incubated with goat anti-rabbit secondary antibodies (1:10,000; cat. no. ab6721; Abcam) at room temperature for 1 h and visualized using enhanced chemiluminescence (EMD Millipore).

Cell viability assay

Cells were seeded in 96-well plates (1×105 cells/well) and cultured for 24 h. The MTT assay was used to measure cell viability of MG63 and SAOS2 cells transfected with miR 212 mimics and negative control miRNA. Non-transfected cells were used as control group. At 48 h post transfection, the transfection medium in each well was replaced with 100 µl fresh serum free medium containing 0.5 g/l MTT (Sigma-Aldrich; Merck Millipore). Subsequent to incubation at 37°C for 4 h, the MTT medium was removed by aspiration and 50 µl dimethylsulfoxide (Sigma-Aldrich; Merck Millipore) was added to each well. Following incubation at 37°C for a further 10 min, the optical density at 570 nm was measured using the BioTek ELX 800 Absorbance Microplate reader (Biotek, Winooski, VT, USA).

Cell migration assay

Cells (1×105 cells/well) were trypsinized and seeded in the top chamber of Matrigel-coated polyethylene terephthalate membrane (Corning Inc., Steuben County, NY, USA). FBS (10%) was added into the lower chamber and after culturing for 24 h, the cells that did not migrate through the membrane were removed; and the cells that had migrated through the membrane were stained using 0.1% crystal violet for 30 min. Migrated cells were counted under an optical microscope (Olympus Corp., Tokyo, Japan).

Luciferase reporter assay

Targets of miR-212 were analyzed using the TargetScan program (targetscan.org/). In order to confirm whether FOXA1 was a direct target gene of miR-212, a luciferase assay was performed using pMIR-Report vector (Applied Biosystems; Thermo Fisher Scientific, Inc.) containing the wild type (WT) or mutant (MT) FOXA1 3′ untranslated region (UTR). Subsequently, 100 ng WT or MT vector were co-transfected with miR-212 mimics or miR-NC into MG-63 and Saos-2 human osteosarcoma cells. Following transfection for 48 h, the luciferase activity was determined by the Dual-Luciferase Reporter Assay System (Promega Corp., Madison, WI, USA).

Statistical analysis

All of the experiments were performed three times. Values are shown as the mean ± standard deviation. Significant differences among the groups were determined using one-way analysis of variance in SPSS 17.0 (SPSS Inc., Chicago, IL, USA). P<0.05 was used to indicate a statistically significant difference.

Results

miR-212 is downregulated in osteosarcoma

Τhe present study initially examined the expression level of miR-212 in osteosarcoma tissues. qPCR data revealed that miR-212 was frequently downregulated in osteosarcoma tissues compared with their matched normal adjacent bone tissues (P<0.05; Fig. 1A). In addition, miR-212 was also significantly downregulated in several common osteosarcoma cell lines compared with the normal human osteoblast cell line (P<0.05; Fig. 1B). These data indicate that deregulation of miR-212 may be associated with the development of osteosarcoma.

Restoration of miR-212 levels suppresses the viability of osteosarcoma cells

Human osteosarcoma MG-63 and Saos-2 cells were transfected with miR-212 mimics and miR-NC. As shown in Fig. 2A, the miR-212 level was significantly increased in MG-63 cells (P<0.01). Similarly, transfection of miR-212 also significantly upregulated the miR-212 level in Saos-2 cells (P<0.01; Fig. 2B). The MTT assay further determined the cell viability capacity of MG-63 and Saos-2 cells in each group. As shown in Fig. 3A, the viability capacity of MG-63 cells overexpressing miR-212 was significantly decreased compared with the control group (P<0.01). Similarly, overexpression of miR-212 also significantly suppressed the viability of Saso-2 cells compared with the control group (P<0.01; Fig. 3B).

Restoration of miR-212 levels suppresses the migration of osteosarcoma cells

Transwell assay analysis was used to determine the cell migration capacity of MG-63 and Saos-2 cells in each group. The data of the present study demonstrated that the migration capacity of MG-63 cells overexpressing miR-212 was significantly reduced compared with the control group (P<0.01; Fig. 4A). Furthermore, overexpression of miR-212 led to a significant decrease in the migration of Saos-2 cells compared with the control group (P<0.01; Fig. 4B).

miR-212 directly targets the 3′UTR of FOXA1 and negative ly mediates its expression in osteosarcoma cells

Putative targets of miR-212 were identified using TargetScan. As shown in Fig. 5A, a putative 8-mer binding site for miR-212 was indicated in the 3′UTR of FOXA1 mRNA. To further confirm this prediction, a luciferase reporter assay was conducted. As shown in Fig. 5B, transfection with a miR-212 mimic led to a significant decrease in the luciferase activity of MG-63 cells (P<0.01) in the WT group, but no significant change was identified in the MT group compared with the control group. Similar observations were also indicated in Saos-2 cells (Fig. 5C). Therefore, the present data indicates that miR-212 directly targets FOXA1 by interacting with the 3′UTR. As miR-212 negatively mediates the protein levels of their targets, the effects of miR-212 on the protein levels of FOXA1 were investigated further. FOXA1 protein levels were observed to have decreased after overexpression of miR-212 in MG-63 and Saos-2 cells (Fig. 6A and B, respectively).

Knockdown of FOXA1 inhibits the viability and migration of osteosarcoma cells

Human osteosarcoma MG-63 and Saos-2 cells were transfected with FOXA1 siRNA. As shown in Fig. 7A, the FOXA1 level was significantly reduced in MG-63 cells (P<0.01). Similarly, transfection of FOXA1 siRNA also significantly suppressed the FOXA1 level in Saos-2 cells (P<0.01; Fig. 7B). MTT assay further determined the cell viability capacity of MG-63 and Saos-2 cells in each group. As shown in Fig. 8, knockdown of FOXA1 significantly inhibited cell viability compared with the control group (P<0.01). Subsequently, a Transwell assay was conducted to determine the cell migration capacity in each group. Consistent with the effect of miR-212 overexpression, inhibition of FOXA1 expression also significantly suppressed the migration of MG-63 and Saos-2 cells (both P<0.01; Fig. 9A and B, respectively). These data indicate that FOXA1 promotes the regulation of the viability and migration of osteosarcoma cells.

Discussion

miRNA have been demonstrated to directly bind to the 3′UTR of their targets mRNA, through which they inhibit the protein expression of their targets. Deregulation of miRNAs are associated with the development and progression of human cancer. miRNA have been identified to act as tumor suppressors in osteosarcoma. Jiang et al (11) revealed that miR-126 inhibited cell growth, invasion and migration of osteosarcoma cells by downregulating ADAM-9. Geng et al (12) revealed that the expression of miR-124 is significantly downregulated in osteosarcoma tissues and cell lines, compared with adjacent tissues, and overexpression of miR-124 suppressed cell proliferation, migration and invasion, and induced apoptosis in osteosarcoma cells. Furthermore, miR-145 was found to target the vascular endothelial growth factor and inhibit invasion and metastasis of osteosarcoma cells (13). The present study identified that miR-212 was also downregulated in osteosarcoma tissues and cell lines, and acted as a tumor suppressor in osteosarcoma, thus supporting the hypothesis that miRNA function in the tumorigenesis of osteosarcoma.

miR-212 has been found to be important in different cancer types. For instance, miR-212 is downregulated in human gastric cancer suppresses the methyl-CpG-binding protein (14). Zhao et al (15) revealed that miR-212 suppressed the G1/S phase transition of the cell cycle and the epithelial to mesenchymal transition in cervical cancer cells via the inhibition of SMAD2 expression. Moreover, miR-212 was also identified to exert a suppressive effect on SKOV3 ovarian cancer cells by targeting HBEGF (16). These aforementioned observations indicate that miR-212 may have a suppressive role in human cancer. However, miR-212 was also identified to promote the malignance of non-small cell lung cancer cells and target the hedgehog pathway receptor, Patched 1 (17). These observations suggest that miR-212 has a complex role in cancer progression and its exact function is tumor-specific. The present study revealed that miR-212 was downregulated in osteosarcoma, and overexpression of miR-212 significantly inhibited the viability and migration of osteosarcoma cells, suggesting a tumor suppressive role of miR-212 in osteosarcoma. This evidence strongly suggests that miR-212 is involved in the progression of osteosarcoma.

Recently, Luo et al (9) also revealed that miR-212 was downregulated in osteosarcoma tissues compared with adjacent normal tissues. They suggested that the introduction of miR-212 mimics into MG63 and U2OS cells inhibited cell proliferation and invasion, partly at least, by targeting Sox4. In the present study, FOXA1 was identified as a direct target of miR-212, and its expression level was negatively mediated by miR-212 in osteosarcoma cells. Moreover, knockdown of FOXA1 was observed to have similar effects as miR-212 overexpression on osteosarcoma cell viability and migration, suggesting that the role of miR-212 in the regulation of the malignant phenotypes of osteosarcoma cells is via the mediation of FOXA1.

FOXA1, also known as HNF3α, is a member of the FoxA gene family. Genome-wide location analyses indicate that FOXA1 binds to adjacent cis-regulatory domain with estrogen receptor (ER)α or androgen receptor (AR) (18). FOXA1 participates in the recruitment of ERα or AR, which are crucial in the regulation of estrogen and androgen signaling. FOXA1 has been identified to have a promoting role in several types of human cancer. For instance, FOXA1 enhanced the proliferation and migration of prostate cancer cells by modulating EAF2 regulation of AR transcriptional activity (19). Qiu et al (20) revealed that FOXA1 promoted tumor cell proliferation through AR involving the Notch pathway in endometrial cancer. Furthermore, FOXA1 was suggested to be associated with methylation of the tumor suppressor genes promoter, and may be a potential demethylation target for the prevention and treatment of breast cancer (21). However, to the best of our knowledge, no previous study has elucidated the exact role of FOXA1 in osteosarcoma. In the present study, knockdown of FOXA1 suppressed the viability and migration of osteosarcoma cells. Consistent with our observations a recent study also found that miR-212 directly targets FOXA1 (22). Moreover, Dou et al (22) revealed that miR-212 suppresses tumor growth of human hepatocellular carcinoma by targeting FOXA1. Therefore, the miR-212/FOXA1 axis may have similar effects on the malignant phenotypes of different types of human cancer. Future studies should focus on this signaling pathway in other types of cancer.

In conclusion, the present study demonstrated that miR-212 is significantly downregulated in osteosarcoma. In addition, restoration of miR-212 expression suppressed the viability and migration of osteosarcoma cells, partly at least, by the direct inhibition of FOXA1 expression. Therefore, miR-212 may be a potential therapeutic candidate for the treatment of osteosarcoma.

References

1 

Thompson LD: Osteosarcoma. Ear Nose Throat J. 92:288–290. 2013.PubMed/NCBI

2 

Zhang J, Yu XH, Yan YG, Wang C and Wang WJ: PI3K/Akt signaling in osteosarcoma. Clin Chim Acta. 444:182–192. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Chang Z, Huo L, Li K, Wu Y and Hu Z: Blocked autophagy by miR-101 enhances osteosarcoma cell chemosensitivity in vitro. Scientific World Journal. 2014:7947562014. View Article : Google Scholar : PubMed/NCBI

4 

Zhou Y, Huang Z, Wu S, Zang X, Liu M and Shi J: miR-33a is up-regulated in chemoresistant osteosarcoma and promotes osteosarcoma cell resistance to cisplatin by down-regulating TWIST. J Exp Clin Cancer Res. 33:122014. View Article : Google Scholar : PubMed/NCBI

5 

Hwang HW and Mendell JT: MicroRNAs in cell proliferation, cell death, and tumorigenesis. Br J Cancer. 94:776–780. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Croce CM and Calin GA: miRNAs, cancer, and stem cell division. Cell. 122:6–7. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Xu L, Wang F, Xu XF, Mo WH, Xia YJ, Wan R, Wang XP and Guo CY: Down-regulation of miR-212 expression by DNA hypermethylation in human gastric cancer cells. Med Oncol. 28:(Suppl 1). S189–S196. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Park JK, Henry JC, Jiang J, Esau C, Gusev Y, Lerner MR, Postier RG, Brackett DJ and Schmittgen TD: miR-132 and miR-212 are increased in pancreatic cancer and target the retinoblastoma tumor suppressor. Biochem Biophys Res Commun. 406:518–523. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Luo XJ, Tang DG, Gao TL, Zhang YL, Wang M, Quan ZX and Chen J: MicroRNA-212 inhibits osteosarcoma cells proliferation and invasion by down-regulation of Sox4. Cell Physiol Biochem. 34:2180–2188. 2014. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Jiang L, He A, Zhang Q and Tao C: miR-126 inhibits cell growth, invasion, and migration of osteosarcoma cells by downregulating ADAM-9. Tumour Biol. 35:12645–12654. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Geng S, Zhang X, Chen J, Liu X, Zhang H, Xu X, Ma Y, Li B, Zhang Y, Bi Z and Yang C: The tumor suppressor role of miR-124 in osteosarcoma. PLoS One. 9:e915662014. View Article : Google Scholar : PubMed/NCBI

13 

Fan L, Wu Q, Xing X, Wei Y and Shao Z: MicroRNA-145 targets vascular endothelial growth factor and inhibits invasion and metastasis of osteosarcoma cells. Acta Biochim Biophys Sin (Shanghai). 44:407–414. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Wada R, Akiyama Y, Hashimoto Y, Fukamachi H and Yuasa Y: miR-212 is downregulated and suppresses methyl-CpG-binding protein MeCP2 in human gastric cancer. Int J Cancer. 127:1106–1114. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Zhao JL, Zhang L, Guo X, Wang JH, Zhou W, Liu M, Li X and Tang H: miR-212/132 downregulates SMAD2 expression to suppress the G1/S phase transition of the cell cycle and the epithelial to mesenchymal transition in cervical cancer cells. IUBMB Life. 67:380–394. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Wei LQ, Liang HT, Qin DC, Jin HF, Zhao Y and She MC: MiR-212 exerts suppressive effect on SKOV3 ovarian cancer cells through targeting HBEGF. Tumour Biol. 35:12427–12434. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Li Y, Zhang D, Chen C, Ruan Z, Li Y and Huang Y: MicroRNA-212 displays tumor-promoting properties in non-small cell lung cancer cells and targets the hedgehog pathway receptor PTCH1. Mol Biol Cell. 23:1423–1434. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Robinson JL and Carroll JS: FoxA1 is a key mediator of hormonal response in breast and prostate cancer. Front Endocrinol (Lausanne). 3:682012.PubMed/NCBI

19 

Guo W, Keener AL, Jing Y, Cai L, Ai J, Zhang J, Fisher AL, Fu G and Wang Z: FOXA1 modulates EAF2 regulation of AR transcriptional activity, cell proliferation, and migration in prostate cancer cells. Prostate. 75:976–987. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Qiu M, Bao W, Wang J, Yang T, He X, Liao Y and Wan X: FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer. BMC Cancer. 14:782014. View Article : Google Scholar : PubMed/NCBI

21 

Zheng L, Qian B, Tian D, Tang T, Wan S, Wang L, Zhu L and Geng X: FOXA1 positively regulates gene expression by changing gene methylation status in human breast cancer MCF-7 cells. Int J Clin Exp Pathol. 8:96–106. 2015.PubMed/NCBI

22 

Dou C, Wang Y, Li C, Liu Z, Jia Y, Li Q, Yang W, Yao Y, Liu Q and Tu K: MicroRNA-212 suppresses tumor growth of human hepatocellular carcinoma by targeting FOXA1. Oncotarget. 6:13216–13228. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 12 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu J, Chen B, Yue B and Yang J: MicroRNA‑212 suppresses the proliferation and migration of osteosarcoma cells by targeting forkhead box protein A1 Retraction in /10.3892/etm.2021.10409. Exp Ther Med 12: 4135-4141, 2016
APA
Liu, J., Chen, B., Yue, B., & Yang, J. (2016). MicroRNA‑212 suppresses the proliferation and migration of osteosarcoma cells by targeting forkhead box protein A1 Retraction in /10.3892/etm.2021.10409. Experimental and Therapeutic Medicine, 12, 4135-4141. https://doi.org/10.3892/etm.2016.3880
MLA
Liu, J., Chen, B., Yue, B., Yang, J."MicroRNA‑212 suppresses the proliferation and migration of osteosarcoma cells by targeting forkhead box protein A1 Retraction in /10.3892/etm.2021.10409". Experimental and Therapeutic Medicine 12.6 (2016): 4135-4141.
Chicago
Liu, J., Chen, B., Yue, B., Yang, J."MicroRNA‑212 suppresses the proliferation and migration of osteosarcoma cells by targeting forkhead box protein A1 Retraction in /10.3892/etm.2021.10409". Experimental and Therapeutic Medicine 12, no. 6 (2016): 4135-4141. https://doi.org/10.3892/etm.2016.3880