MicroRNA-124 suppresses the migration and invasion of osteosarcoma cells via targeting ROR2-mediated non-canonical Wnt signaling

Retraction in: /10.3892/or.2021.8107

  • Authors:
    • Can Zhang
    • Yihe Hu
    • Jun Wan
    • Hongbo He
  • View Affiliations

  • Published online on: August 10, 2015     https://doi.org/10.3892/or.2015.4186
  • Pages: 2195-2201
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRs) have been implicated in tumorigenesis through inhibition of the expression of their target genes at post-transcriptional levels. miR-124 has been found to be downregulated in many malignant tumors including osteosarcoma (OS). However, the detailed mechanism of miR-124 in the regulation of OS malignant phenotypes remains largely unclear. Here we aimed to explore the role of miR-124 in mediating OS cell migration and invasion, as well as the underlying regulatory mechanisms. Real-time RT-PCR data showed that miR-124 was frequently downregulated in OS cell lines compared to normal human osteoblast cells. We further conducted bioinformatic analysis and a luciferase reporter assay, and identified receptor tyrosine kinase-like orphan receptor 2 (ROR2) as a novel target of miR-124. Furthermore, we found that ROR2 was significantly upregulated in OS cell lines compared to normal human osteoblast cells, and miR-124 negatively mediated the protein level of ROR2 in U-2OS and Saos-2 cells. Moreover, transfection with miR-124 mimics significantly suppressed migration and invasion in the U-2OS and Saos-2 cells, while overexpression of ROR2 in the miR-124-transfected OS cells reversed the inhibitory effect of miR-124 upregulation on OS cell migration and invasion. In addition, we found that overexpression of miR-124 significantly suppressed the activity of non-canonical Wnt signaling, downstream of ROR2. Based on these findings, we suggest that miR-124 may inhibit OS metastasis, partly at least, via targeting ROR2 and thus suppressing the activity of ROR2-mediated non-canonical Wnt signaling.

Introduction

Osteosarcoma (OS) is the most common mesenchymal sarcoma in bone, mainly arising from the metaphysis of the long bones (1). Although great efforts have been made to improve OS diagnosis and therapy, the 5-year survival rate of patients with OS is only ~30%, and ~80% of OS patients eventually develop metastasis after surgical resection (1). Aberrant downregulation of tumor-suppressors has been found to play a crucial role in the development and progression of OS (2). Accordingly, exploration of potential targets seems to be promising for the treatment of OS.

MicroRNAs (miRs) are a type of short non-coding RNA, that can bind directly to the 3′-untranslational region (UTR) of their target mRNAs, eventually leading to inhibition of gene expression at post-transcriptional levels (3). To date, miRs have been demonstrated to play key roles in various human cancers. Moreover, many miRs have been implicated in the development and progression of OS, such as miR-101, miR-126, miR-143, miR-194 and miR-217 (49). Among these miRs, miR-124 was recently found to be frequently downregulated in OS tissues and to act as a tumor-suppressor in OS via targeting Rac1. As one miR can target many genes associated with tumorigenesis, the functions of other targets of miR-124 in OS have yet to be investigated.

Receptor tyrosine kinase-like orphan receptor 2 (ROR2) belongs to the receptor tyrosine kinase (RTK) family, which plays an important role in the regulation of cell proliferation, apoptosis, differentiation, adhesion and migration (1012). It has been reported that ROR2 plays an important role in cartilage and growth plate development (13), and mutations in the ROR2 gene lead to the autosomal recessive form of Robinow syndrome (14). Moreover, ROR2 has been demonstrated to be associated with OS severity, and to enhance OS cell migration and invasion via activation of Wnt5a-mediated non-canonical Wnt signaling (1517). However, the relationship between ROR2 and miR-124 in OS cells has never been studied.

Accordingly, we aimed to reveal the regulatory mechanisms of miR-124 in the regulation of malignant phenotypes in OS cells involving ROR2 and its mediated non-canonical Wnt signaling.

Materials and methods

Cell culture

OS cell lines, Saos-2, U-2OS, HOS and MG-63, and normal osteoblast cell line NHOst, were obtained from the American Type Culture Collection (ATCC; Rockville, MD, USA). Cells were cultured in Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum (FBS) (both from Life Technologies, Carlsbad, CA, USA) at 37°C in a humidified incubator containing 5% CO2.

Real-time RT-PCR assay

Total RNA was extracted by using the miRNA isolation kit (Life Technologies) according to the manufacturer's instructions. For detection of miR expression, the miRNA reverse transcription kit (Life Technologies) was used to convert 10 ng of total RNA into cDNA, according to the manufacturer's instructions. Real-time PCR was then performed using an miRNA Q-PCR detection kit (GeneCopoeia, Rockville, MD, USA) on Applied Biosystems 7500 Real-Time PCR system. The U6 gene was used as an internal reference. The primers were: miR-124 forward, TCGGCAGGTAAGGCACGCGGTG and reverse, TCAACT GGTGTCGTGGAGTCGGC; and U6 forward, CTCGCTTC GGCAGCACATATACT and reverse, ACGCTTCACGAATT TGCGTGTC. Expression of mRNA was detected using the standard SYBR-Green RT-PCR kit (Life Technologies) following the manufacturer's instructions. GAPDH was used as an internal reference. The primers were: ROR2 forward, TCCGAACGACCCTTTAGGAC and reverse, TTTAGCCAC CGCACGTTAGG; and GAPDH forward, GGAGCGAGATC CCTCCAAAAT and reverse, GCCATCACGC CACAGTTTC. The PCR cycling conditions used were: 94°C for 3 min followed by 40 cycles of 94°C for 30 sec, 56°C for 30 sec and 72°C for 30 sec. The relative expression was analyzed by the 2−ΔΔCt method.

Western blotting

Cells were lysed with ice-cold lysis buffer (50 mM Tris-HCl, pH 6.8, 100 mM 2-ME, 2% w/v SDS, 10% glycerol). After centrifugation at 20,000 × g for 10 min at 4°C, proteins in the supernatants were quantified and separated with 10% SDS-PAGE. Then, proteins were transferred onto a polyvinylidene difluoride (PVDF) membrane (Amersham Biosciences, Buckinghamshire, UK), which was then incubated with PBS containing 5% milk overnight at 4°C. The PVDF membrane was then incubated with rabbit anti-ROR2 monoclonal antibody (1:100) or the rabbit anti-GAPDH monoclonal antibody (1:200) (both from Abcam, Cambridge, UK) at room temperature for 3 h, respectively, and then with HRP-linked goat anti-rabbit secondary antibody (Abcam) at room temperature for 1 h. SuperSignal West Pico chemiluminescent substrate kit (Pierce, Rockford, IL, USA) was then used to detect signals according to the manufacturer's instructions. The relative protein expression was analyzed by Image-Pro Plus 6.0 software, represented as the density ratio vs. GAPDH.

Transfection

Lipofectamine 2000 (Life Technologies) was used to perform cell transfection following the manufacturer's instructions. For functional analysis, Saos-2 and U-2OS cells were transfected with scramble miR and miR-124 mimics (both from Life Technologies), or co-transfected with miR-124 mimics and ROR2 plasmid (Santa Cruz Biotechnology), respectively.

Bioinformatic predication

We screened the target genes of miR-124 using TargetScan (http://www.targetscan.org/index.html).

Luciferase reporter assay

Total cDNA from the cells was used to amplify the 3′-UTR of ROR2, which was then cloned into the pMir-REPORT vector (Life Technologies). Mutations were introduced within the potential seed sequences of the 3′UTR of ROR2 using the QuikChange site-directed mutagenesis kit (Stratagene), changing the seed sequences GUGCCUU into GUAAAUU. Using Lipofectamine 2000, Saos-2 and U-2OS cells were transfected with miR-124 mimics, as well as the pMir-REPORT vectors containing the wild-type or mutant-type of ROR2 3′-UTR, respectively. The pRL-SV40 vector (Promega, Madison, WI, USA) carrying the Renilla luciferase gene was used as an internal control. Luciferase activity was determined after 48 h using the Dual-Glo substrate system and LD400 luminometer (Beckman Coulter, Brea, CA, USA). Data are presented as the ratio of Renilla luciferase to firefly luciferase.

Wound-healing assay

A wound-healing assay was performed to evaluate the cell migratory capacity of OS cells in each group. Briefly, OS cells were cultured to full confluence. Wounds of ~1 mm width were created with a plastic scriber, and cells were washed and incubated in a serum-free medium. Twenty-four hours after wounding, cells were incubated in a medium containing 10% FBS. After 36-h of culture, cells were fixed and observed under a microscope.

Cell invasion assay

Cells in each group were starved in serum-free medium for 24 h, and then resuspended in serum-free medium. The cell suspension was added into the upper chamber, while the lower chamber was filled with base medium containing 10% FBS. After 24-h of incubation, cells attached to the bottom were stained with crystal violet for 20 min, and then washed and dried in air. Invasive cells were observed under a microscope.

Statistical analysis

Data are expressed as mean ± standard deviation from at least 3 separate experiments. The differences were analyzed using one-way analysis of variance (ANOVA). SPSS 18.0 software was used to perform statistical analysis. A p-value <0.05 were considered to indicate a statistically significant result.

Results

miR-124 is frequently downregulated in OS cells

Real-time RT-PCR was used to detect the expression of miR-124 in OS cell lines, U-2OS, HOS, Saos-2 and MG-63, and in the normal osteoblast cell line NHOst. As shown in Fig. 1, the expression of miR-124 was decreased in the OS cell lines compared with the normal osteoblast NHOst cells, suggesting that downregulation of miR-124 may be associated with OS development.

Overexpression of miR-124 inhibits migration and invasion of OS cells

We further studied the role of miR-124 in the regulation of OS cell migration and invasion. As shown in Fig. 2A, the miR-124 level was significantly increased after transfection with the miR-124 mimics in the U-2OS and Saos-2 cells, compared to the control group. We then determined the cell migration by conducting a wound healing assay. The migratory capacity of the OS cells overexpressing miR-124 was significantly decreased compared to the control group, suggesting that miR-124 plays an inhibitory role in OS cell migration (Fig. 2B). After that, we performed a Transwell assay to determine the cell invasive capacity in each group. As shown in Fig. 2C, overexpression of miR-124 significantly suppressed the invasive capacity of Saos-2 and U-2OS cells compared to the control group, indicating that miR-124 plays a suppressive role in mediating OS cell invasion.

Identification of ROR2 as a target gene of miR-124

According to bioinformatic analysis, ROR2 is a putative target gene of miR-124 (Fig. 3A). However, whether miR-124 directly targets ROR2 has never been previously reported. Therefore, we conducted a luciferase reporter assay to clarify whether miR-124 can bind directly to their seed sequences in the ROR2 3′-UTR. Luciferase activity was significantly decreased in the U-20S and Saos-2 cells co-transfected with the wild-type (WT) ROR2 3′-UTR and miR-124 mimics, but showed no difference in U-20S and Saos-2 cells co-transfected with the mutant-type (MUT) ROR2 3′-UTR and miR-124 mimics, when compared with that in the control group (Fig. 3B), indicating that ROR2 is a direct target gene of miR-124.

ROR2 is significantly upregulated in OS cell lines and is negatively regulated by miR-124

We then detected the protein level of ROR2 in OS cell lines, U-2OS, HOS, Saos-2 and MG-63, and in the normal osteoblast cell line NHOst. As shown in Fig. 4A and B, the mRNA and protein levels of ROR2 were significantly increased in the OS cell lines compared to the NHOst cells. As miRs generally play suppressive roles in the regulation of their target expression at post-transcriptional levels, we further determined the effect of miR-124 overexpression or inhibition on the protein level of ROR2 in OS cells. Saos-2 and U-2OS cells were transfected with miR-124 mimics or miR-124 inhibitor, respectively. Transfection with miR-124 inhibitor significantly downregulated the level of miR-124 compared to the control group (Fig. 4C). As shown in Fig. 4D, western blot data demonstrated that overexpression of miR-124 inhibited the protein level of ROR2 in the OS cells when compared to the control group, while inhibition of miR-124 increased the protein level of ROR2 in OS cells, indicating that miR-124 negatively mediates the protein expression of ROR2 in OS cells.

Overexpression of ROR2 reverses the inhibitory effect of miR-124 upregulation on OS cell migration and invasion by activation of non-canonical Wnt signaling

To further clarify whether ROR2 acts as a downstream effector in miR-124-mediated migration and invasion of OS cells, we transfected OS cells with miR-124 mimics, or co-transfected them with miR-124 mimics and the ROR2 plasmid, and then determined the migratory and invasive capacities of the OS cells in each group. As shown in Fig. 5A and B, OS cells co-transfected with miR-124 mimics and ROR2 plasmid showed higher migratory and invasive capacities, when compared with the OS cells transfected with miR-124 mimics, suggesting that ROR2 is involved in miR-124-mediated migration and invasion of OS cells. To further confirm our data, we conducted a western blot assay to determine the protein level of ROR2 in each group, and found that transfection with the ROR2 plasmid reversed the inhibitory effect of miR-124 overexpression on ROR2 protein levels in the OS cells (Fig. 5C).

As ROR2 is an important receptor of non-canonical Wnt signaling, which plays a key role in the regulation of cell motility and is closely associated with tumor metastasis, we further determined the activity of non-canonical Wnt signaling in each group. As shown in Fig. 5D, we demonstrated that overexpression of miR-124 significantly inhibited the activity of c-JNK, the pivotal member in the non-canonical Wnt signaling pathway. However, restoration of ROR2 rescued the inhibitory effect of miR-124 overexpression on the activity of c-JNK. Therefore, we suggest that miR-124 can inhibit OS cell migration and invasion, partly at least, via targeting ROR2 and thus suppressing the activity of ROR2-mediated non-canonical Wnt signaling.

Discussion

Tumor cell proliferation, migration and invasion play key roles in the development and progression of OS (6). It has been demonstrated that miR-124 generally acts as a tumor-suppressor in human cancers (1820). However, the detailed molecular mechanism of miR-124 in the regulation of OS progression remains largely unknown. In the present study, we found that miR-124 was frequently downregulated in OS cell lines compared to normal osteoblast cells, and showed suppressive effects on the proliferation, migration and invasion of OS cells. Further investigation identified ROR2 as a novel target of miR-124, and was negatively mediated by miR-124 in the OS cells.

Moreover, deregulation of miRs has been found to be associated with the development and progression of various human cancers including OS (2,21). He et al found that miR-23a functions as a tumor-suppressor in OS via inhibition of OS cell proliferation, migration, and invasion (22). Zhang et al showed that miR-451 expression is associated with the prognosis of OS patients, and it inhibits OS cell growth and invasion by targeting CXCL16 (23). In addition, overexpression of miR-101 inhibited OS cell proliferation and promoted cell apoptosis via inhibition of mTOR (24). In the present study, we found that miR-124 played a suppressive role in mediating OS cell migration and invasion. In fact, similar findings were also reported in other types of human cancers. For example, Liang et al reported that miR-124 could inhibit the invasive and metastatic potential of breast cancer, probably by inhibition of the epithelial to mesenchymal transition (18). More recently, the suppressive role of miR-124 in OS was revealed. Han et al found that miR-124 downregulation occurred more frequently in OS tissues at an advanced clinical stage, with positive distant metastasis and a poor response to neoadjuvant chemotherapy, and low miR-124 expression was identified as an unfavorable prognostic factor for overall survival (25). Furthermore, they demonstrated that transfection of a miR-124 mimic into MG-63 cells was able to reduce cell proliferation, invasion and migration, and promote cell apoptosis (25). Geng et al reported that expression of miR-124 was significantly downregulated in OS tissues and cell lines, compared with that in adjacent tissues (26). Moreover, the expression of miR-124 in the metastatic OS tissues was lower than that in non-metastatic tissues, suggesting that miR-124 is associated with OS metastasis, consistent with our findings. It was also found that miR-124 could inhibit OS cell proliferation, migration and invasion, partly at least, by targeting Rac1 (26). As one miR targets many target genes, our study aimed to reveal novel targets of miR-124 in OS, and found that ROR2 is a target of miR-124, which is involved in miR-124-mediated OS cell migration and invasion.

ROR2 is a type I transmembrane protein that belongs to the ROR subfamily of cell surface receptors (27). ROR2 is involved in the development of bone by promoting osteoblast differentiation (28,29). Recently, the oncogenic role of ROR2 in OS has been demonstrated. Lu et al investigated the expression of non-canonical Wnt ligand Wnt5a and its receptor ROR2 in OS. They found that the expression levels of Wnt5a and ROR2 were significantly higher in OS samples than in osteochondroma, and the expression of Wnt5a and ROR2 was correlated to Enneking surgical stage and tumor metastasis. This suggested that Wnt5a and ROR2 play a coordinated role in the occurrence and progression of OS (15). Through interaction with Wnt5a, ROR2 promotes OS cell invasion by upregulation of MMP13 (17,30). Wnt5b was also found to be a putative ROR2 ligand, and the physiological interaction of Wnt5b and ROR2 could enhance OS cell migration (31). Moreover, ROR2 was also found to be involved in OS cell motility by mediating Snail-mediated epithelial-mesenchymal transition (16). In the present study, we found that overexpression of ROR2 reversed the inhibitory effect of miR-124 upregulation on OS cell migration and invasion by upregulation of non-canonical Wnt signaling activity.

In conclusion, the present study demonstrates that miR-124 can inhibit the migratory and invasive capacities of OS cells via targeting ROR2 as well as its downstream non-canonical Wnt signaling. Based on these findings, we suggest that miR-124/ROR2 may become a promising therapeutic target for the treatment of OS.

References

1 

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

2 

Liang W, Gao B, Fu P, Xu S, Qian Y and Fu Q: The miRNAs in the pathgenesis of osteosarcoma. Front Biosci (Landmark Ed). 18:788–794. 2013. View Article : Google Scholar

3 

Moss EG: MicroRNAs: Hidden in the genome. Curr Biol. 12:R138–R140. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Shen L, Wang P, Yang J and Li X: MicroRNA-217 regulates WASF3 expression and suppresses tumor growth and metastasis in osteosarcoma. PLoS One. 9:e1091382014. View Article : Google Scholar : PubMed/NCBI

5 

Wang Q, Cai J, Wang J, Xiong C and Zhao J: miR-143 inhibits EGFR-signaling-dependent osteosarcoma invasion. Tumour Biol. 35:12743–12748. 2014. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Zhang K, Zhang Y, Ren K, Zhao G, Yan K and Ma B: MicroRNA-101 inhibits the metastasis of osteosarcoma cells by downregulation of EZH2 expression. Oncol Rep. 32:2143–2149. 2014.PubMed/NCBI

8 

Han K, Zhao T, Chen X, Bian N, Yang T, Ma Q, Cai C, Fan Q, Zhou Y and Ma B: microRNA-194 suppresses osteosarcoma cell proliferation and metastasis in vitro and in vivo by targeting CDH2 and IGF1R. Int J Oncol. 45:1437–1449. 2014.PubMed/NCBI

9 

Yang J and Zhang W: New molecular insights into osteosarcoma targeted therapy. Curr Opin Oncol. 25:398–406. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Sundaram MV: Canonical RTK-Ras-ERK signaling and related alternative pathways. Jul 11–2013, pp. 1–38. View Article : Google Scholar

11 

Jiménez G, Shvartsman SY and Paroush Z: The Capicua repressor - a general sensor of RTK signaling in development and disease. J Cell Sci. 125:1383–1391. 2012. View Article : Google Scholar

12 

Batchu SN and Korshunov VA: Novel tyrosine kinase signaling pathways: Implications in vascular remodeling. Curr Opin Nephrol Hypertens. 21:122–127. 2012. View Article : Google Scholar : PubMed/NCBI

13 

DeChiara TM, Kimble RB, Poueymirou WT, Rojas J, Masiakowski P, Valenzuela DM and Yancopoulos GD: Ror2, encoding a receptor-like tyrosine kinase, is required for cartilage and growth plate development. Nat Genet. 24:271–274. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Mehawej C, Chouery E, Maalouf D, Baujat G, Le Merrer M, Cormier-Daire V and Mégarbané A: Identification of a novel causative mutation in the ROR2 gene in a Lebanese family with a mild form of recessive Robinow syndrome. Eur J Med Genet. 55:103–108. 2012. View Article : Google Scholar

15 

Lu BJ, Wang YQ, Wei XJ, Rong LQ, Wei D, Yan CM, Wang DJ and Sun JY: Expression of WNT-5a and ROR2 correlates with disease severity in osteosarcoma. Mol Med Rep. 5:1033–1036. 2012.PubMed/NCBI

16 

Ren D, Minami Y and Nishita M: Critical role of Wnt5a-Ror2 signaling in motility and invasiveness of carcinoma cells following Snail-mediated epithelial-mesenchymal transition. Genes Cells. 16:304–315. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Enomoto M, Hayakawa S, Itsukushima S, Ren DY, Matsuo M, Tamada K, Oneyama C, Okada M, Takumi T, Nishita M, et al: Autonomous regulation of osteosarcoma cell invasiveness by Wnt5a/Ror2 signaling. Oncogene. 28:3197–3208. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Liang YJ, Wang QY, Zhou CX, Yin QQ, He M, Yu XT, Cao DX, Chen GQ, He JR and Zhao Q: miR-124 targets Slug to regulate epithelial-mesenchymal transition and metastasis of breast cancer. Carcinogenesis. 34:713–722. 2013. View Article : Google Scholar :

19 

Furuta M, Kozaki KI, Tanaka S, Arii S, Imoto I and Inazawa J: miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis. 31:766–776. 2010. View Article : Google Scholar

20 

An L, Liu Y, Wu A and Guan Y: microRNA-124 inhibits migration and invasion by down-regulating ROCK1 in glioma. PLoS One. 8:e694782013. View Article : Google Scholar : PubMed/NCBI

21 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

22 

He Y, Meng C, Shao Z, Wang H and Yang S: miR-23a functions as a tumor suppressor in osteosarcoma. Cell Physiol Biochem. 34:1485–1496. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Zhang F, Huang W, Sheng M and Liu T: miR-451 inhibits cell growth and invasion by targeting CXCL16 and is associated with prognosis of osteosarcoma patients. Tumour Biol. 36:2041–2048. 2015. View Article : Google Scholar

24 

Lin S, Shao NN, Fan L, Ma XC, Pu FF and Shao ZW: Effect of microRNA-101 on proliferation and apoptosis of human osteosarcoma cells by targeting mTOR. J Huazhong Univ Sci Technolog Med Sci. 34:889–895. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Han G, Wang Y, Bi W, Jia J and Wang W: MicroRNA-124 functions as a tumor suppressor and indicates prognosis in human osteosarcoma. Exp Ther Med. 9:679–684. 2015.PubMed/NCBI

26 

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

27 

Stricker S and Mundlos S: FGF and ROR2 receptor tyrosine kinase signaling in human skeletal development. Curr Top Dev Biol. 97:179–206. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Liu Y, Bhat RA, Seestaller-Wehr LM, Fukayama S, Mangine A, Moran RA, Komm BS, Bodine PV and Billiard J: The orphan receptor tyrosine kinase Ror2 promotes osteoblast differentiation and enhances ex vivo bone formation. Mol Endocrinol. 21:376–387. 2007. View Article : Google Scholar

29 

Liu Y, Ross JF, Bodine PV and Billiard J: Homodimerization of Ror2 tyrosine kinase receptor induces 14-3-3(beta) phosphorylation and promotes osteoblast differentiation and bone formation. Mol Endocrinol. 21:3050–3061. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Yamagata K, Li X, Ikegaki S, Oneyama C, Okada M, Nishita M and Minami Y: Dissection of Wnt5a-Ror2 signaling leading to matrix metalloproteinase (MMP-13) expression. J Biol Chem. 287:1588–1599. 2012. View Article : Google Scholar :

31 

Morioka K, Tanikawa C, Ochi K, Daigo Y, Katagiri T, Kawano H, Kawaguchi H, Myoui A, Yoshikawa H, Naka N, et al: Orphan receptor tyrosine kinase ROR2 as a potential therapeutic target for osteosarcoma. Cancer Sci. 100:1227–1233. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 34 Issue 4

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 C, Hu Y, Wan J and He H: MicroRNA-124 suppresses the migration and invasion of osteosarcoma cells via targeting ROR2-mediated non-canonical Wnt signaling Retraction in /10.3892/or.2021.8107. Oncol Rep 34: 2195-2201, 2015
APA
Zhang, C., Hu, Y., Wan, J., & He, H. (2015). MicroRNA-124 suppresses the migration and invasion of osteosarcoma cells via targeting ROR2-mediated non-canonical Wnt signaling Retraction in /10.3892/or.2021.8107. Oncology Reports, 34, 2195-2201. https://doi.org/10.3892/or.2015.4186
MLA
Zhang, C., Hu, Y., Wan, J., He, H."MicroRNA-124 suppresses the migration and invasion of osteosarcoma cells via targeting ROR2-mediated non-canonical Wnt signaling Retraction in /10.3892/or.2021.8107". Oncology Reports 34.4 (2015): 2195-2201.
Chicago
Zhang, C., Hu, Y., Wan, J., He, H."MicroRNA-124 suppresses the migration and invasion of osteosarcoma cells via targeting ROR2-mediated non-canonical Wnt signaling Retraction in /10.3892/or.2021.8107". Oncology Reports 34, no. 4 (2015): 2195-2201. https://doi.org/10.3892/or.2015.4186