Open Access

MicroRNA‑101 has a suppressive role in osteosarcoma cells through the targeting of c‑FOS

Retraction in: /10.3892/etm.2021.10410

  • Authors:
    • Zili Wang
    • Rongzhen He
    • Hansong Xia
    • Yu Wei
    • Song Wu
  • View Affiliations

  • Published online on: February 17, 2016     https://doi.org/10.3892/etm.2016.3085
  • Pages: 1293-1299
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRs) have been implicated in the development and progression of osteosarcoma (OS). However, the underlying mechanism of miR‑101 in regulating of the proliferation, migration and invasion of OS cells remains to be elucidated. In the present study, reverse transcription‑quantitative polymerase chain reaction data revealed that miR‑101 was frequently downregulated in the tissue samples of 12 patients with OS compared with their matched adjacent non‑tumor tissues. Furthermore, miR‑101 was significantly downregulated in three common OS cell lines, Saos‑2, MG63 and U2OS, compared with the human osteoblast cell line, hFOB1.19 (P<0.01). A luciferase reporter assay was also performed and identified c‑FOS as a novel target of miR‑101 in U2OS cells; overexpression of miR‑101 significantly suppressed the protein expression levels of c‑FOS, while knockdown of miR‑101 significantly enhanced the formers' expression levels in U2OS cells (P<0.01). Independent inhibition of c‑FOS and overexpression of miR‑101 expression levels significantly suppressed U2OS cell proliferation, migration and invasion (P<0.01). However, overexpression of c‑FOS reversed the inhibitory effect of miR‑101 upregulation on proliferation, migration and invasion of U2OS cells, suggesting that miR‑101 acts as a tumor suppressor in OS cells via targeting of c‑FOS. Thus, we propose that the miR‑101/c‑FOS axis may be a potential therapeutic target for OS.

Introduction

Osteosarcoma (OS) is the most prevalent malignant cancer of the bone, typically arising in the metaphysis of long bones, and is predominately observed in adolescents and young adults (1). The relative 5-year survival rate of young-onset osteosarcoma is 61.6% globally (2). Pain and swelling in the affected bone are the most common symptom of OS. Despite surgical resection (which is the standard treatment strategy) combined with chemotherapy and radiotherapy, the overall survival rate of patients with OS still remains poor (1). It has been demonstrated that the deregulation of certain oncogenes or tumor suppressors is strongly associated with OS. Therefore, continued investigations for potential therapeutic targets in the aforementioned areas may reduce mortality associated with OS (3).

MicroRNAs (miRs/miRNAs), a class of non-coding RNAs 18–25 nucleotides in length, have been demonstrated to inhibit the protein expression levels of their target genes via direct binding to the 3′-untranslated region (UTR) of their target mRNAs (4). Through mediating the expression of target genes associated with tumorigenesis, miRs participate in the development and progression of human cancer, including OS. For example, miR-199a-3p is downregulated in OS and regulates cell proliferation and migration (5). Additionally, miR-218 was revealed to inhibit OS cell migration and invasion by targeting T-cell lymphoma invasion and metastasis 1, and matrix metalloproteinases 2 and 9 (6).

Deregulation of miR-101 has been demonstrated to be involved in multiple types of human malignances (7,8). For example, the downregulation of miR-101 in gastric cancer is associated with cyclooxygenase-2 (Cox-2) overexpression and tumor growth (9). Recently, miR-101 has been implicated in OS. For example, Chang et al reported that miR-101 blocked the autophagy of OS cells and thus enhanced OS cell chemosensitivity (10). Another study has revealed that miR-101 inhibited the metastasis of OS cells by targeting enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) (11). In addition, miR-101 was revealed to inhibit proliferation and induce the apoptosis of OS cells by targeting mechanistic target of rapamycin (mTOR) (12). The aforementioned findings suggest that miR-101 has a tumor suppressive role in OS. However, the underlying mechanism of miR-101 in regulating the proliferation, migration and invasion of OS cells remains largely unclear. In addition, as miR has various target genes (4), other targets of miR-101 may also be involved in the effect of miR-101 on the malignant phenotypes of OS cells.

Accordingly, the present study aimed to explore the molecular mechanism involving miR-101 in regulating the proliferation, migration and invasion of OS cells.

Materials and methods

Reagents and kits

RPM-1640 medium, fetal bovine serum (FBS), Lipofectamine 2000, TRIzol reagent (Thermo Fisher Scientific, Inc. (Waltham, MA, USA), MTT and TaqMan miRNA Reverse Transcription kit were purchased from Thermo Fisher Scientific, Inc. A miRNA Q-PCR Detection kit was purchased from GeneCopoeia, Inc. (All-in-One™ miRNA qPCR kit; Rockville, MD, USA). Scrambled miR mimics, miR-101 mimics (accession no. MIMAT0000099) and miR-101 inhibitors were purchased from GeneChem Co., Ltd. (Shanghai, China). c-FOS small interfering (si)RNA and c-FOS plasmid were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Mouse anti-human c-FOS monoclonal antibody (cat. no. ab184666), mouse anti-human GAPDH monoclonal antibody (cat. no. ab9484) and rabbit anti-mouse polyclonal secondary antibody (cat. no. ab6728) were purchased from Abcam (Cambridge, MA, USA). Enhanced chemiluminescence (ECL) kit and polyvinylidene difluoride (PVDF) membrane were purchased from Pierce Biotechnology, Inc. (Rockford, IL, USA). Transwell chambers and Matrigel were purchased from BD Biosciences (Franklin Lakes, NJ, USA).

Tissue specimen collection

The present study was approved by the Ethics Committee of Central South University, Changsha, China. Primary OS samples (n=12) and their normal matched adjacent tissues were collected from Xiangya Hospital of Central South University between March 2010 and April 2012. The inclusion criteria stated that patients (male, n=7 and female, n=6; age 26–50 years) had not received radiation therapy or chemotherapy prior to surgical resection. Written informed consent was obtained from all patients. Tissue samples were immediately snap-frozen in liquid nitrogen following surgical removal and stored at −80°C.

Cell culture

Human OS cell lines, Saos-2, MG63 and U2OS, and a human osteoblast cell line, hFOB1.19, were obtained from the American Type Culture Collection (Manassas, VA, USA). Cells were plated in 6-well plates and cultured to 100% confluence in RPMI-1640 medium supplemented with 10% FBS at 37°C in a humidified incubator containing 5% CO2.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay

An RT-qPCR assay was used to determine the expression levels of miR-101. Total RNA was extracted using TRIzol reagent, according to the manufacturer's protocol. miRNA Reverse Transcription kit was used to convert RNA into cDNA. RT-qPCR was then performed by using miRNA Q-PCR Detection kit on an ABI 7500 thermocycler (Thermo Fisher Scientific, Inc.). The conditions were 50°C for 2 min, 95°C for 10 min, and 40 cycles of denaturation at 95°C for 15 sec and annealing/elongation at 60°C for 60 sec. U6 was used as an internal reference. The relative expression was analyzed by the 2−ΔΔCq method (13).

Western blot analysis

Western blotting was performed to detect the protein expression levels of c-FOS. In brief, tissues and cells were solubilized in cold radioimmunoprecipitation lysis buffer (Beyotime Institute of Biotechnology, Shanghai, China). Proteins were separated with 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (Beyotime Institute of Biotechnology) at 60 V for 30 min and transferred onto PVDF membranes, which were then incubated with phosphate-buffered saline (PBS; Beyotime Institute of Biotechnology) containing 5% milk (Mengniu, Beijing, China) overnight at 4°C. The PVDF membrane was subsequently incubated with mouse anti-c-FOS monoclonal antibody (1:100) and mouse anti-GAPDH monoclonal antibody (1:100) at room temperature for 3 h, followed by rabbit anti-mouse secondary antibodies (1:10,000) at room temperature for 1 h. An ECL kit was then used to perform chemiluminescent detection. The relative protein expression was analyzed using Image Pro Plus software (version 6.0; Media Cybernetics, Rockville, MD, USA) and data are represented as the density ratio compared with GAPDH.

Transfection

Lipofectamine 2000 was used to perform cell transfection. For miR-101 and c-FOS function analysis, U2OS cells were transfected with scrambled miR mimics, miR-101 mimics, miR-101 inhibitor, c-FOS siRNA or c-FOS plasmid, respectively, according to the manufacturer's protocol. After incubation at 37°C and 5% CO2 for 6 h, the transfection medium was replaced by RPMI-1640 medium supplemented with 10% FBS. Following this, U2OS cells were cultured for 24 h prior to the subsequent assays.

Dual luciferase reporter assay

The predicted seed sequence of miR-101 in the c-FOS 3′-UTR, in addition to a mutant seed sequence in the c-FOS 3′-UTR, were cloned downstream of the luciferase gene, driven by the cytomegalovirus promoter, generating vectors containing wild-type of c-FOS 3′-UTR (Luc-c-FOS vector) or mutant type of c-FOS 3′UTR (Luc-mutant C-FOS vector), respectively. U2OS cells were co-transfected with miR-101 mimics or miR-101 scramble mimics and Luc-c-FOS vector or Luc-mutant C-FOS vector, respectively. Luciferase activity was determined following transfection for 24 h using a Bio-Tek ELX-800 Absorbance Microplate reader (Biotek Instruments, Inc., Winooski, VT, USA).

MTT assay

An MTT assay was performed to measure cell proliferation. At 48 h post-transfection, the transfection medium was replaced with 100 µl fresh serum-free RPMI-1640 medium containing 0.5 g/l MTT. Following incubation at 37°C for 4 h, the MTT medium was removed by aspiration and 50 µl dimethylsulfoxide was added to each well. Following incubation at 37°C for 10 min, the optical density was measured at 570 nm using a Bio-Tek ELX-800 Absorbance Microplate reader (Biotek Instruments, Inc.).

Cell migration assay

A wound healing assay was used to determine cell migration. Cells were cultured to full confluence and a wound of ~1 mm width was made with a plastic scriber. Cells were then washed with PBS and cultured at 37°C with 5% CO2 for 48 h. Finally, cells in each group were fixed in 1% paraformaldehyde (Beyotime Institute of Biotechnology) and observed under a microscope (CX23; Olympus Corporation, Tokyo, Japan).

Cell invasion assay

A cell invasion assay was performed using Transwell chambers pre-coated with Matrigel. Cell suspension containing 5×105 cells/ml was prepared in serum-free media and 300 µl of the cell suspension was added into the upper chamber. Then, 500 µl RPMI-1640 in 10% FBS was added into the lower chamber. Cells were incubated for 24 h at 37°C in a humidified incubator containing 5% CO2. Following this, a cotton-tipped swab was used to carefully eliminate the cells that did not migrate or invade through the pores. The filters were fixed in 90% alcohol, stained with crystal violet (Beyotime Institute of Biotechnology) and observed under an inverted microscope (CX23; Olympus Corporation). Following this, 0.5 g/l MTT was added and incubated at 37°C for 4 h. Then, the medium containing MTT was removed and 50 µl dimethylsulfoxide was added to each well. Following incubation at 37°C for 10 min, the optical density was measured at 570 nm using a Bio-Tek ELX-800 Absorbance Microplate reader (Biotek Instruments, Inc.).

Statistical analysis

SPSS software (version 17.0; SPSS, Inc. Chicago, IL, USA) was used to perform statistical analysis. All data are expressed as mean ± standard deviation. Differences were analyzed using one-way analysis of variance. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-101 is downregulated in OS tissues and cell lines

To reveal the role of miR-101 in OS, RT-qPCR was performed to examine the expression levels of miR-101 in the tissue samples of 12 patients with OS, with adjacent normal tissues from the respective patients used as controls. As revealed in Fig. 1A, miR-101 was frequently downregulated in OS tissues compared with their adjacent non-tumor tissues. Following this, the expression levels of miR-101 in several common OS cell lines, Saos-2, MG63 and U2OS, were investigated. The human osteoblast cell line hFOB1.19 was used as a control. The data of the present study revealed that miR-101 is significantly downregulated in OS cell lines compared with the human osteoblast cell line, hFOB1.19 (P<0.01; Fig. 1B). In addition, U2OS cells displayed the most significant decrease in miR-101 expression levels. Therefore, U2OS cells were used in the subsequent investigations in the present study.

c-FOS is a target gene of miR-101 in U2OS cells

c-FOS was hypothesized to be a potential target of miR-101 (14). To verify whether miR-101 was able to directly bind to its seed sequences in the c-FOS 3′-UTR in U2OS cells, Luc-c-FOS and Luc-mutant C-FOS vectors containing the wild-type and mutant binding sequences of miR-101 within the 3′-UTR of c-FOS mRNA were generated, respectively (Fig. 2A). A luciferase reporter assay revealed that the luciferase activity was significantly reduced in U2OS cells co-transfected with Luc-c-FOS vector and miR-101 mimics compared with the control group (P<0.01). However, the luciferase activity revealed no significant difference in U2OS cells co-transfected with Luc-mutant C-FOS vector and miR-101 mimics when compared with the control group (Fig. 2B). The data indicates that miR-101 is able to directly bind to its seed sequences in the c-FOS 3′-UTR in U2OS cells. Therefore, c-FOS was identified as a target of miR-101 in U2OS cells.

miR-101 negatively regulates the protein expression levels of c-FOS in U2OS cells

As miR negatively regulates the expression levels of its targets at the post-transcriptional level, the effect of miR-101 upregulation or downregulation on the protein expression levels of c-FOS in U2OS cells was further investigated. U2OS cells were transfected with miR-101 mimics or miR-101 inhibitor and the miR-101 level in each group was then detected. RT-qPCR data indicated that transfection with miR-101 mimics led to a significant increase in miR-101 expression levels, while transfection with miR-101 inhibitor significantly suppressed the miR-101 level in U2OS cells compared with the control (P<0.01; Fig. 3A). The protein levels of c-FOS in each group were then determined by western blot analysis. As demonstrated in Fig. 3B, upregulation of miR-101 significantly decreased the protein expression level of c-FOS (P<0.01), while downregulation of miR-101 with miR-101 inhibitor significantly increased the protein expression level of c-FOS in U2OS cells (P<0.01) compared with the control, indicating that miR-101 negatively regulates the protein expression levels of c-FOS in U2OS cells.

miR-101 suppresses U2OS cell proliferation through targeting c-FOS

To further explore the roles of miR-101 and c-FOS in the regulation of OS cell proliferation, as well as to reveal whether c-FOS is involved in miR-101-mediated OS cell proliferation, U2OS cells were transfected with miR-101 mimics or c-FOS siRNA, or co-transfected with miR-101 mimics and c-FOS plasmid. Subsequently, the protein expression levels of c-FOS were detected in each group. As displayed in Fig. 4A, transfection with miR-101 mimics or c-FOS siRNA led to significant reduction in c-FOS protein expression levels compared with the control group (P<0.01). However, the protein expression levels of c-FOS were higher in U2OS cells co-transfected with miR-101 mimics and c-FOS plasmid, compared with cells transfected with miR-101 mimics alone, suggesting that transfection with c-FOS plasmid reversed the inhibitory effect of miR-101 overexpression on c-FOS protein expression (Fig. 4A). Following this, it was revealed that the upregulation of miR-101 or the knockdown of c-FOS were able to significantly suppress the proliferation of U2OS cells compared with the control (P<0.01; Fig. 4B). However, the suppressive effect of miR-101 upregulation on U2OS cell proliferation was markedly reversed by overexpression of c-FOS (Fig. 4B). The aforementioned findings suggest that miR-101 is able to inhibit U2OS cell proliferation, to a certain extent, via targeting c-FOS.

miR-101 inhibits the migration and invasion of U2OS cells via targeting c-FOS

The effects of miR-101 and c-FOS on the migration and invasion of U2OS cells were investigated. Similar to the cell proliferation data, it was identified that miR-101 overexpression or c-FOS knockdown significantly inhibited U2OS cell migration and invasion (P<0.01; Fig. 5A and B, respectively). However, the suppressive effect of miR-101 overexpression on U2OS cell migration and invasion was reversed by the upregulation of c-FOS. The aforementioned data suggest that miR-101 inhibits U2OS cell migration and invasion, to a certain extent, via targeting c-FOS.

Discussion

miR-101 has been demonstrated to act as a tumor suppressor in multiple types of human cancer (1517). However, the underlying mechanism of miR-101 in the regulation of OS cell proliferation, migration and invasion remains largely unclear. In the present study, it was revealed that miR-101 is significantly downregulated in OS tissues and cell lines. Subsequently, c-FOS was identified as a direct target of miR-101 and it was revealed that the expression of c-FOS was negatively regulated by miR-101 at the post-transcriptional level in U2OS cells. Furthermore, the current results indicate that miR-101 inhibits the proliferation, migration and invasion of U2OS cells, to a certain extent, via direct suppression of the protein expression levels of c-FOS.

The tumor suppressive role of miR-101 has been demonstrated in multiple types of human cancer, including OS. Zhang et al (18) revealed that the expression levels of miR-101 significantly decreased in hepatocellular carcinoma (HCC), and its downregulation was associated with tumor aggressiveness and poor prognosis. Furthermore, the aforementioned study demonstrated that overexpression of miR-101 significantly inhibited the proliferation and tumorigenicity of HCC cells by targeting SRY-box 9 (19). Lei et al (16) reported that miR-101 inhibited migration and invasion, and enhanced the cisplatin chemosensitivity, of bladder cancer cells via inhibition of vascular endothelial growth factor C. In addition, Guo et al (19) identified that miR-101 suppressed the epithelial-to-mesenchymal transition in ovarian carcinoma cells by targeting zinc finger E-Box binding homeobox 1 and 2. Recently, the role of miR-101 in OS has been proposed in several studies (1012). Lin et al (12) demonstrated that overexpression of miR-101 inhibited the proliferation and promoted the apoptosis of Saos-2 cells in an mTOR-dependent manner. Zhang et al (11) revealed that overexpression of miR-101 suppressed EZH2 expression levels, and significantly inhibited the migration and invasion of OS cells, while knockdown of EZH2 also suppressed OS cell migration and invasion, suggesting that miR-101 inhibits the metastasis of OS cells by downregulation of EZH2 expression levels. In the present study, it was demonstrated that miR-101 suppressed the proliferation, migration and invasion of OS cells. Chang et al (10) previously reported the effect of miR-101 on the chemoresistance of OS cells. The aforementioned study indicated that treatment with doxorubicin induced a significantly high level of autophagy-characteristic acidic vesicular organelles and autophagy-related protein expression levels in U2OS cells, which were significantly inhibited by miR-101 overexpression. Furthermore, the autophagy blockage by miR-101 sensitized the U2OS cells to doxorubicin treatment, suggesting that miR-101 blocks autophagy during chemotherapy in OS cells and enhances chemosensitivity (10).

As an activator protein-1 transcription factor, c-FOS has been revealed to be associated with specific enzymes involved in the synthesis of phospholipids at the endoplasmic reticulum and may activate their synthesis to accompany genomic decisions regarding growth (20). In addition, c-FOS has been reported to have an oncogenic role in OS (21). Overexpression of c-FOS has been demonstrated to increase the recombination frequency in human OS cells (22) and c-FOS may induce OS formation in transgenic mice via cooperating with c-jun (23). Gamberi et al (21) reported that c-myc and c-FOS were upregulated in relapsed OS, and overexpression of both in the same tumor was strongly associated with the development of metastases, suggesting that c-myc and c-FOS are involved in the growth and spread of OS; thus, synchronous overexpression of c-myc and c-FOS is a significant indicator of the metastatic potential of primary OS. In the present study, c-FOS was identified as a novel target of miR-101 in OS cells, and it was determined that miR-101 inhibited OS cell proliferation, migration and invasion via targeting c-FOS, suggesting that miR-101 may have a role in the regulation of OS growth and metastasis via targeting c-FOS.

The association between miR-101 and c-FOS has also been identified in other types of cancer. Konno et al (14) demonstrated that miR-101 was able to target EZH2, myeloid cell leukemia 1 (MCL-1) and c-FOS to suppress proliferation, invasion and stem cell-like phenotypes of aggressive endometrial cancer cells. Liang et al (24) reported that miR-101 inhibited the G1 to S phase transition of cervical cancer cells by targeting c-FOS. Furthermore, overexpression of miR-101 may also suppress cellular proliferation, migration and invasion of gastric cancer cells by targeting EZH2, Cox-2, induced MCL-1 and c-FOS. Therefore, the miR-101/c-FOS axis may be a common regulatory mechanism in several types of cancer, highlighting its potential value in cancer therapy.

In conclusion, the present study suggests that miR-101 has a suppressive role in the regulation of OS cell proliferation, migration and invasion, to a certain extent, via targeting c-FOS. Accordingly, miR-101 and c-FOS may serve as potential therapeutic targets for OS.

Acknowledgements

The present study was supported by the Natural Science Foundation of Hunan Province, Hunan, China (grant no. 13JJ2013).

References

1 

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

2 

Mirabello L, Troisi RJ and Savage SA: Osteosarcoma incidence and survival rates from 1973 to 2004: Data from the Surveillance, Epidemiology, and End Results Program. Cancer. 115:1531–1543. 2009. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2:e3632004. View Article : Google Scholar : PubMed/NCBI

5 

Duan Z, Choy E, Harmon D, Liu X, Susa M, Mankin H and Hornicek F: MicroRNA-199a-3p is downregulated in human osteosarcoma and regulates cell proliferation and migration. Mol Cancer Ther. 10:1337–1345. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Jin J, Cai L, Liu ZM and Zhou XS: miRNA-218 inhibits osteosarcoma cell migration and invasion by down-regulating of TIAM1, MMP2 and MMP9. Asian Pac J Cancer Prev. 14:3681–3684. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Wang HJ, Ruan HJ, He XJ, Ma YY, Jiang XT, Xia YJ, Ye ZY and Tao HQ: MicroRNA-101 is down-regulated in gastric cancer and involved in cell migration and invasion. Eur J Cancer. 46:2295–2303. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Zhang JG, Guo JF, Liu DL, Liu Q and Wang JJ: MicroRNA-101 exerts tumor-suppressive functions in non-small cell lung cancer through directly targeting enhancer of zeste homolog 2. J Thorac Oncol. 6:671–678. 2011. View Article : Google Scholar : PubMed/NCBI

9 

He XP, Shao Y, Li XL, Xu W, Chen GS, Sun HH, Xu HC, Xu X, Tang D, Zheng XF, et al: Downregulation of miR-101 in gastric cancer correlates with cyclooxygenase-2 overexpression and tumor growth. FEBS J. 279:4201–4212. 2012. View Article : Google Scholar : PubMed/NCBI

10 

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. 7947562014.PubMed/NCBI

11 

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

12 

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

13 

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

14 

Konno Y, Dong P, Xiong Y, Suzuki F, Lu J, Cai M, Watari H, Mitamura T, Hosaka M, Hanley SJ, et al: MicroRNA-101 targets EZH2, MCL-1 and FOS to suppress proliferation, invasion and stem cell-like phenotype of aggressive endometrial cancer cells. Oncotarget. 5:6049–6062. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Frankel LB, Wen J, Lees M, Høyer-Hansen M, Farkas T, Krogh A, Jäättelä M and Lund AH: microRNA-101 is a potent inhibitor of autophagy. EMBO J. 30:4628–4641. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Lei Y, Li B, Tong S, Qi L, Hu X, Cui Y, Li Z, He W, Zu X, Wang Z and Chen M: miR-101 suppresses vascular endothelial growth factor C that inhibits migration and invasion and enhances cisplatin chemosensitivity of bladder cancer cells. PLoS One. 10:e01178092015. View Article : Google Scholar : PubMed/NCBI

17 

Su H, Yang JR, Xu T, Huang J, Xu L, Yuan Y and Zhuang SM: MicroRNA-101, down-regulated in hepatocellular carcinoma, promotes apoptosis and suppresses tumorigenicity. Cancer Res. 69:1135–1142. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Zhang Y, Guo X, Xiong L, Kong X, Xu Y, Liu C, Zou L, Li Z, Zhao J and Lin N: MicroRNA-101 suppresses SOX9-dependent tumorigenicity and promotes favorable prognosis of human hepatocellular carcinoma. FEBS Lett. 586:4362–4370. 2012. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Caputto BL, Gizzi Cardozo AM and Gil GA: c-Fos: An AP-1 transcription factor with an additional cytoplasmic, non-genomic lipid synthesis activation capacity. Biochim Biophys Acta. 1841:1241–1246. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Gamberi G, Benassi MS, Bohling T, Ragazzini P, Molendini L, Sollazzo MR, Pompetti F, Merli M, Magagnoli G, Balladelli A and Picci P: C-myc and c-fos in human osteosarcoma: Prognostic value of mRNA and protein expression. Oncology. 55:556–563. 1998. View Article : Google Scholar : PubMed/NCBI

22 

van den Berg S, Rahmsdorf HJ, Herrlich P and Kaina B: Overexpression of c-fos increases recombination frequency in human osteosarcoma cells. Carcinogenesis. 14:925–928. 1993. View Article : Google Scholar : PubMed/NCBI

23 

Wang ZQ, Liang J, Schellander K, Wagner EF and Grigoriadis AE: c-fos-induced osteosarcoma formation in transgenic mice: Cooperativity with c-jun and the role of endogenous c-fos. Cancer Res. 55:6244–6251. 1995.PubMed/NCBI

24 

Liang X, Liu Y, Zeng L, Yu C, Hu Z, Zhou Q and Yang Z: miR-101 inhibits the G1-to-S phase transition of cervical cancer cells by targeting Fos. Int J Gynecol Cancer. 24:1165–1172. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2016
Volume 11 Issue 4

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
Wang Z, He R, Xia H, Wei Y and Wu S: MicroRNA‑101 has a suppressive role in osteosarcoma cells through the targeting of c‑FOS Retraction in /10.3892/etm.2021.10410. Exp Ther Med 11: 1293-1299, 2016
APA
Wang, Z., He, R., Xia, H., Wei, Y., & Wu, S. (2016). MicroRNA‑101 has a suppressive role in osteosarcoma cells through the targeting of c‑FOS Retraction in /10.3892/etm.2021.10410. Experimental and Therapeutic Medicine, 11, 1293-1299. https://doi.org/10.3892/etm.2016.3085
MLA
Wang, Z., He, R., Xia, H., Wei, Y., Wu, S."MicroRNA‑101 has a suppressive role in osteosarcoma cells through the targeting of c‑FOS Retraction in /10.3892/etm.2021.10410". Experimental and Therapeutic Medicine 11.4 (2016): 1293-1299.
Chicago
Wang, Z., He, R., Xia, H., Wei, Y., Wu, S."MicroRNA‑101 has a suppressive role in osteosarcoma cells through the targeting of c‑FOS Retraction in /10.3892/etm.2021.10410". Experimental and Therapeutic Medicine 11, no. 4 (2016): 1293-1299. https://doi.org/10.3892/etm.2016.3085