MicroRNA‑708 inhibits the proliferation and invasion of osteosarcoma cells by directly targeting ZEB1

Retraction in: /10.3892/mmr.2023.12936

  • Authors:
    • Jun He
    • Deng Xiang
    • Yanshui Lin
  • View Affiliations

  • Published online on: March 6, 2019     https://doi.org/10.3892/mmr.2019.10013
  • Pages: 3948-3954
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Numerous microRNAs (miRNAs) have been identified as aberrantly expressed in osteosarcoma (OS). miRNAs serve important roles in the pathogenesis of OS as oncogenes or tumor suppressors. Recent studies revealed that miR‑708‑5p (miR‑708) was dysregulated in various types of human cancer; however, its roles and underlying molecular mechanisms in OS remain unknown. Therefore, the present study aimed to determine miR‑708 expression in OS, investigate the roles of miR‑708 in the progression of OS and reveal the potential mechanisms involved. It was demonstrated using reverse transcription‑polymerase chain reaction that miR‑708 was downregulated in OS tissues and cell lines. Cell Counting Kit‑8 and Transwell assays revealed that miR‑708 overexpression suppressed the proliferation and invasion of OS cells in vitro. Additionally, zinc finger E‑box binding homeobox 1 (ZEB1) was validated as a direct target gene of miR‑708 in OS cells. ZEB1 was upregulated in OS tissues; elevated ZEB1 expression was negatively correlated with the levels of miR‑708 expression. Rescue experiments indicated that ZEB1 reintroduction significantly counteracted the inhibitory effects of miR‑708 overexpression on the proliferation and invasion of OS cells. The findings may improve understanding of the roles of miR‑708 in the development of OS, and suggest that miR‑708 may be a potential novel therapeutic target in the treatment of patients with this disease.

Introduction

Osteosarcoma (OS), which originates from primitive transformed cells, is the most common type of primary bone tumor (1). Additionally, OS is the most common type of childhood cancer, accounting for ~2.4% of all malignant tumors reported in pediatric patients (2). OS occurs more frequently in the metaphysis of long bones of the extremities (3). Major advancements in therapeutic approaches have been made in previous decades, including surgical resection, chemotherapy and radiotherapy; however, the treatment outcomes of patients with OS remains poor, particularly those with metastasis or recurrence (4). A number of factors, including alterations of oncogenes or tumor suppressors and environmental radiation, have been associated with the pathogenesis of OS; however, the fundamental mechanisms underlying the formation and progression of OS remain unclear (5,6). Thus, improved understanding of the mechanisms associated with the progression of OS is important for the development of potential therapeutic methods.

MicroRNAs (miRNAs/miRs) refer to a group of evolutionarily conserved, noncoding short (20–23 nucleotides) RNAs (7). miRNAs regulate gene expression by directly interacting with ‘seed sequences’ within the 3′-untranslated regions (3′-UTRs) of target genes, thereby inhibiting translational activity and destabilizing mRNAs (8). Each miRNA modulates numerous genes, suggesting that miRNAs are one of the largest families of gene regulators (9). Increasing evidence suggests that various miRNAs are dysregulated in the majority of human cancers, and that their aberrant expression is required in maintaining the aggressive behaviors of cancer cells (1012). A number of miRNAs are aberrantly expressed in OS, including miR-203 (13), miR-208b (14), miR-448 (15) and miR-635 (16). Dysregulated miRNAs were reported to be involved in various pathological processes, including the proliferation, cell cycle, apoptosis, autophagy, migration, invasion and metastasis of tumor cells (1719). Therefore, miRNAs may represent potential biomarkers for the diagnosis, prognosis and treatment of OS.

Previous studies reported miR-708-5p (miR-708) as dysregulated in various human cancer types, including hepatocellular carcinoma (20,21), gastric cancer (22), melanoma (23) and renal cancer (24). Furthermore, the expression of miR-708 is downregulated in OS (25); however, its functions and underlying molecular mechanisms in OS remain unknown. Therefore, the aims of the present study were to determine the levels of miR-708 expression in OS tissues and cell lines. Additionally, the roles and potential mechanisms of miR-708 in the progression of OS were investigated.

Materials and methods

Tissue specimens

Paired OS tissues and adjacent normal tissues were collected from 29 patients (17 males and 12 females; age range, 24–61 years) with OS who underwent surgical resection at The First Affiliated Hospital of Chengdu Medical College (Chengdu, China) between January 2015 and May 2017. Patients that had received preoperative chemotherapy and radiotherapy were not included in the study. Patients that had been treated with chemotherapy or radiotherapy before surgery were excluded from the present study. Tissues specimens were stored in liquid nitrogen prior to subsequent experimentation. Written informed consent was provided by all patients enrolled, and the present study was approved by the Ethics Committee of The First Affiliated Hospital of Chengdu Medical College.

Cell culture

The human osteoblast cell line hFOB1.19 and three human OS cell lines (MG-63, U2OS and HOS) were acquired from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in Dulbecco's Modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin, all obtained from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA). All cells were maintained at 37°C in a 5% CO2 humidified atmosphere.

Transfection

miR-708 mimics and negative control miRNA mimics (miR-NC) were acquired from Wuhan GeneCreate Biological Engineering Co., Ltd. (Wuhan, China). The miR-708 mimics sequence was 5′-AAGGAGCUUACAAUCUAGCUGGG-3′ and the miR-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. The zinc finger E-box binding homeobox 1 (ZEB1) overexpression plasmid pcDNA3.1-ZEB1 and control empty plasmid pcDNA3.1 were chemically synthesized by Amspring (Changsha, China). The restriction sites were HindIII and XhoI. Cells were plated into 6-well plates with an initial density of 50–60% confluence. Cell transfection was conducted using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. The concentration of plasmid and miRNAs used for transfection was 100 pmol and 4 µg, respectively. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Transwell assay was performed out at 48 h post-transfection. Cell Counting Kit-8 (CCK-8) assay and western blot analysis was performed 24 h and 72 h respectively after incubation at 37°C with 5% CO2.

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

Extraction of total RNA from tissues or cells was performed using TRIzol® reagent (Thermo Fisher Scientific, Inc.). To determine miR-708 expression, single-strand complementary DNA (cDNA) was reverse-transcribed using the TaqMan™ MicroRNA Reverse Transcription kit according to the manufacturer's protocols, and the synthesized cDNA was then subjected to qPCR using the TaqMan MicroRNA PCR kit (kits were obtained from Applied Biosystems; Thermo Fisher Scientific, Inc.). The cycling conditions for reverse transcription were: 16°C for 30 min, 42°C for 30 min and 85°C for 5 min. The cycling conditions for qPCR were as follows: 50°C for 2 min, 95°C for 10 min; 40 cycles of denaturation at 95°C for 15 sec; and annealing/extension at 60°C for 60 sec. To detect ZEB1 mRNA expression, RT was performed with the PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China) according to the manufacturer's protocols. The cycling conditions for reverse transcription were as follows: 37°C for 15 min and 85°C for 5 sec. qPCR was subsequently conducted using the SYBR® Premix Ex Taq™ kit (Takara Biotechnology Co., Ltd.). The temperature protocols for qPCR were as follows: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. The relative levels of miR-708 and ZEB1 mRNA expression were analyzed using the 2−ΔΔCq method (26) and normalized to U6 small nuclear RNA and GAPDH, respectively. All reaction was performed on ABI Prism 7500 Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The primers were designed as follows: miR-708, 5′-CGGCGGAAGGAGCTTACAATCTA-3′ (forward) and 5′-GTGCAGGGTCCGAGG-3′ (reverse); U6, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ (forward) and 5′-CGCTTCACGAATTTGCGTGTCAT-3′ (reverse); ZEB1 forward, 5′-AAGTGGCGGTAGATGGTA-3′ and reverse, 5′-TTGTAGCGACTGGATTTT-3′; and GAPDH, 5′-TGCACCACCAACTGCTTAGC-3′ (forward) and 5′-GGCATGCACTGTGGTCATGAG-3′ (reverse). Each sample was analyzed in triplicate and repeated three times.

Cell Counting Kit-8 (CCK-8) assay

The CCK-8 assay was performed to investigate the proliferation of OS cells. A total of 2,000 transfected cells were seeded into 96-well plates and incubated at 37°C in a 5% CO2 humidified atmosphere. Following incubation for 0, 24, 48 and 72 h, 10 µl of CCK-8 reagent (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was added to each well, prior to incubation at 37°C under 5% CO2 for an additional 2 h. The absorbance value at 450 nm of each well was measured using a SpectraMax Microplate® Spectrophotometer (Molecular Devices, LLC, Sunnyvale, CA, USA).

Transwell assay

To determine the invasive ability of OS cells, transfected cells from each group were harvested and resuspended in DMEM without FBS. A total of 1×105 cells were inoculated in the upper chamber of 24-well Transwell inserts (8-µm pore size; Costar; Corning Incorporated, Corning, NY, USA) coated with Matrigel (BD Biosciences, San Jose, CA, USA). DMEM (500 µl) with 20% FBS (Gibco; Thermo Fisher Scientific,) was inserted into the lower chamber to serve as a chemoattractant. Following incubation at 37°C with 5% CO2 for 24 h, the cells on the upper surface of the membrane were carefully removed using a cotton swab. The invasive cells were fixed with 4% paraformaldehyde at 37°C for 30 min, stained with 0.5% crystal violet at 37°C for 30 min, washed with PBS and then air dried. The number of invasive cells was counted in five randomly selected fields under an inverted microscope (magnification, ×200; Olympus IX83; Olympus Corporation, Tokyo, Japan).

Bioinformatics analysis

The putative targets of miR-708 were predicted using TargetScan (release 7.2; http://www.targetscan.org/) and microRNA.org (August 2010 release; www.microRNA.org). The bioinformatics analysis indicated that ZEB1 may be a potential downstream target of miR-708.

Luciferase reporter assay

The wild-type (wt) or mutant (mut) 3′-UTR of ZEB1 was amplified by Shanghai GenePharma Co., Ltd. (Shanghai, China), and was sub-cloned into a pMIR-Report plasmid (Promega Corporation, Madison, WI, USA). The recombined plasmids were named pMIR-ZEB1-3′-UTR wt and pMIR-ZEB1-3′-UTR mut, respectively. Cells were seeded into 24-well plates, and co-transfected with miR-708 mimics or miR-NC, and pMIR-ZEB1-3′-UTR wt or pMIR-ZEB1-3′-UTR mut, using Lipofectamine 2000 according to the manufacturer's protocols. Transfected cells were collected 48 h following incubation at 37°C with 5% CO2 and analyzed using the Dual-Luciferase® Reporter assay system (Promega Corporation). Firefly luciferase activity was normalized to Renilla luciferase activity.

Western blot analysis

Radioimmunoprecipitation assay lysis buffer (Thermo Fisher Scientific, Inc.) was used to isolate total protein from tissues or cells, and protein concentration was determined using a bicinchoninic acid assay (Thermo Fisher Scientific, Inc.). Equal amounts of proteins (30 µg per lane) were separated via 10% SDS-PAGE and subsequently transferred to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). Following blocking at room temperature with 5% fat-free milk in TBS-0.1% Tween-20 (TBST) for 2 h, the membranes were incubated at 4°C overnight with mouse anti-human ZEB1 antibody (1:1,000; ab181451, Abcam, Cambridge, UK) and mouse anti-human GAPDH antibody (1:1,000; ab8245, Abcam). The membranes were then washed three times with TBST and incubated with goat anti-mouse horseradish peroxidase-conjugated secondary antibody (1:5,000; ab6789, Abcam) for 2 h at room temperature. Protein signals were visualized using an enhanced chemiluminescence system (EMD Millipore). Quantity One software version 4.62 (Bio-Rad Laboratories, Inc., Hercules, CA, USA) was used for the densitometry.

Statistical analysis

Data were expressed as the mean ± standard deviation, and were analyzed using SPSS version 20.0 (IBM Corp., Armonk, NY, USA). Differences between groups were investigated using Student's t-tests (two groups) or one-way analyses of variance followed by a Tukey's post-hoc test (>2 groups). Associations between miR-708 and ZEB1 mRNA expression were determined by Spearman's correlation analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of miR-708 is reduced in OS tissues and cell lines

To determine the expression profile of miR-708 in OS, total RNA was extracted from 29 paired OS tissues and adjacent normal tissues and then subjected to RT-qPCR. It was revealed that miR-708 expression was significantly downregulated in OS tissues compared with in adjacent normal tissues (P<0.05; Fig. 1A). Levels of miR-708 expression were subsequently measured in three human OS cell lines, namely, MG-63, U2OS and HOS. RT-qPCR analysis demonstrated that each OS cell line exhibited significantly reduced levels of miR-708 expression than the normal human osteoblast cell line, hFOB1.19 (P<0.05; Fig. 1B). These observations suggest that miR-708 may serve important roles in the carcinogenesis and progression of OS.

miR-708 upregulation suppresses the proliferation and invasion of OS cells

To clarify the potential functional roles of miR-708 in the development of OS, mi-708 mimics or miR-NC was transfected into MG-63 and U2OS cells, which exhibited markedly reduced miR-708 expression compared with HOS cells (Fig. 1B). The expression levels of miR-708 were significantly increased in MG-63 and U2OS cells following transfection with miR-708 mimics (P<0.05; Fig. 2A). A CCK-8 assay was performed to determine the effects of miR-708 overexpression on the proliferation of OS cells. MG-63 and U2OS cells transfected with miR-708 mimics exhibited a significant decrease in proliferative ability compared with cells transfected with miR-NC (P<0.05; Fig. 2B). The results of the Transwell assay indicated that ectopic miR-708 expression significantly inhibited the invasion of MG-63 and U2OS cells compared with the control (P<0.05; Fig. 2C). The results suggest that miR-708 may serve a tumor-suppressor role by reducing the proliferation and invasion of OS cells.

ZEB1 is a direct target gene of miR-708 in OS

To investigate the mechanisms by which miR-708 regulates OS proliferation and invasion, bioinformatic analysis was performed to determine putative targets of miR-708. ZEB1 was revealed to be a candidate target of miR-708 (Fig. 3A); this prediction was evaluated via luciferase reporter assay. miR-708 mimics or miR-NC, along with reporter plasmids containing the wt or mut 3′-UTR of ZEB1, were co-transfected into MG-63 and U2OS cells. Overexpression of miR-708 significantly reduced the luciferase activity of the plasmid containing the wt 3′-UTR of ZEB1 in MG-63 and U2OS cells (P<0.05; Fig. 3B); however, luciferase activity was markedly unaffected following transfection with miR-708 mimics when the binding sequence for miR-708 in the 3′-UTR of ZEB1 was mutated. Via RT-qPCR and western blot analysis, it was demonstrated that the expression levels of ZEB1 mRNA (P<0.05; Fig. 3C) and protein (P<0.05; Fig. 3D) were decreased as a result of miR-708 overexpression. Collectively, the results indicated that ZEB1 is a direct target gene of miR-708 in OS.

Upregulation of ZEB1 is negatively associated with miR-708 expression in OS tissues

As miR-708 was reported to exhibit reduced expression in OS and directly target ZEB1 by binding to its 3′-UTR, it was investigated as to whether ZEB1 expression was inversely correlated with the expression levels of miR-708 in OS tissues. RT-qPCR analysis revealed that ZEB1 mRNA was significantly overexpressed in OS tissues compared with in adjacent normal tissues (P<0.05; Fig. 4A). Furthermore, Spearman's correlation analysis demonstrated that the expression levels of ZEB1 mRNA were inversely correlated with miR-708 expression in OS tissues (r=−0.5390, P=0.0026; Fig. 4B). The results suggested that upregulation of ZEB1 in OS tissues may be associated with the downregulation of miR-708.

Enhanced expression of ZEB1 reverses the suppressive effects of miR-708 in OS cells

A series of rescue experiments was performed to determine the effects of ZEB1 on the potential tumor-suppressive roles of miR-708 in OS cells. The ZEB1 overexpression plasmid pcDNA3.1-ZEB1 was utilized to recover ZEB1 protein expression in MG-63 and U2OS cells (P<0.05; Fig. 5A). CCK-8 and Transwell assays revealed that the rescued expression of ZEB1 reversed the effects of miR-708 overexpression on the proliferative (P<0.05; Fig. 5B and C) and invasive (P<0.05; Fig. 5D) abilities of MG-63 and U2OS cells. The results indicated that ZEB1, as a direct target of miR-708, is involved in the inhibitory effects of miR-708 on the proliferation and invasion of OS cells.

Discussion

Numerous miRNAs have been identified to be aberrantly expressed in OS, and serve important roles in the genesis and progression of OS via oncogenic or tumor suppressor activities (2729). Therefore, improved understanding of the dysregulated expression of miRNAs in OS may provide novel insight regarding the diagnosis and treatment of patients with OS. In the present study, data from RT-qPCR analysis revealed that the expression levels of miR-708 were downregulated in OS tissues and cell lines. Additionally, miR-708 overexpression attenuated the proliferation and invasion of OS cells in vitro. A significant inverse correlation between the expression of miR-708 and ZEB1 mRNA was reported in OS tissues. Furthermore, a series of rescue experiments demonstrated that ZEB1 was a direct target of miR-708 in OS cells, and that restored ZEB1 expression significantly eliminated the miR-708-induced suppression of the proliferation and invasion of OS cells. These findings provide novel evidence of the tumor suppressor roles of miR-708 in the progression of OS via targeting of ZEB1, suggesting that this miRNA may serve as a potential therapeutic target in the treatment of patients with OS.

The expression of miR-708 has been investigated in various human malignancies. For instance, the expression levels of miR-708 are decreased in hepatocellular carcinoma tissues and cell lines (20,21). Reduced miR-708 expression is significantly associated with Edmondson-Steiner grading and tumor-node-metastasis (TNM) stage (20,21). miR-708 is downregulated in gastric cancer, and decreased expression of miR-708 is associated with lymphatic metastasis, invasive depth and TNM stage (22). Furthermore, miR-708 is downregulated in melanoma (23), renal cancer (24) and glioblastoma (30). Conversely, miR-708 was reported to be overexpressed in colorectal cancer (31), lung adenocarcinoma (32) and bladder cancer (33). These opposing observations indicate that the expression status of miR-708 exhibits tissue specificity in malignant tumors. Therefore, miR-708 may serve as a promising biomarker for the detection of specific types of tumor.

miR-708 exhibits tumor suppressor activity in numerous types of human cancer; for example, miR-708 upregulation suppresses the proliferation and motility of hepatocellular carcinoma cells via negative regulation of mothers against decapentaplegic homolog family member 3 (20,21). Exogenous miR-708 expression inhibits the proliferative and invasive abilities of gastric cancer in vitro by directly targeting Notch homolog 1 (22). In melanoma, miR-708 overexpression suppresses the proliferation and epithelial-mesenchymal transition of cells, and promotes apoptosis via targeting lymphoid enhancer-binding factor 1 and regulating the Wnt signaling pathway (23). In renal cancer, miR-708 targets ZEB2 and Polycomb complex protein B lymphoma Mo-MLV insertion region 1 homolog to suppress the growth and metastasis, induce apoptosis and improve sensitivity to anti-cancer drugs of cells in vitro, and inhibit tumor growth in vivo (24). In glioblastoma, miR-708 upregulation suppresses the proliferation and invasion, and induces the apoptosis of cells via the regulation of various genes, including protein kinase B, cyclin D1, matrix metalloproteinase-2, Enhancer of zeste homolog 2, poly (adenosine 5′-diphosphate-ribose) polymerase 1 and B-cell lymphoma 2 (30). Conversely, miR-708 serves oncogenic roles in lung adenocarcinoma (31), bladder cancer (33) and acute lymphoblastic leukemia (34). These findings indicate that miR-708 may serve as a potential therapeutic target in the treatment of patients with these specific types of cancer.

Identification of the direct target genes of miR-708 is important for understanding its functional roles in the initiation and progression of OS, and may aid the development of effective therapeutic strategies. Therefore, the molecular mechanisms underlying the tumor suppressive roles of miR-708 in OS cells were investigated in the present study. ZEB1 was validated as a direct target of miR-708 in OS cells. ZEB1, located on the short arm of human chromosome 10, is overexpressed in various human malignancies, including hepatocellular carcinoma (35), and thyroid (36), colorectal (37), lung (38) and gastric cancers (39). Its expression is also reduced in OS tissues and cell lines. ZEB1 expression is significantly correlated with the lung metastasis of patients with OS (40). Dysregulation of ZEB1 is associated with the aggressive behaviors of OS cells via regulation of numerous pathological processes, including cell proliferation, migration, invasion, metastasis and chemoresistance (4144). Thus, miR-708-based therapy targeted against ZEB1 expression may serve as an effective strategy in the treatment of patients with OS.

In conclusion, it was demonstrated that miR-708 expression was downregulated in OS tissues and cell lines, and that upregulation suppressed the proliferation and invasion of OS cells. The tumor suppressor roles of miR-708 in OS may involve the negative regulation of ZEB1. These findings indicate that the downregulation of miR-708 may serve important roles in the development of OS; thus, miR-708 may be a potential therapeutic target in the treatment of patients with this disease. As the sample size of the present study was small, receiver operating curve analysis should be conducted to determine the sensitivity and specificity of miR-708 as a diagnostic biomarker for patients with OS. Additionally, an RNA immunoprecipitation assay, a technique to verify the binding of ZEB1 and miR-708, was not conducted in the present study. These limitations of the present study should be resolved in future experiments.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

YL made substantial contributions to the design of the present study. JH, DX and YL performed functional experiments. All authors have read and approved the final draft.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of The First Affiliated Hospital of Chengdu Medical College, and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of The First Affiliated Hospital of Chengdu Medical College. Written informed consent was obtained from all patients for the use of their clinical tissues.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ta HT, Dass CR, Choong PF and Dunstan DE: Osteosarcoma treatment: State of the art. Cancer Metastasis Rev. 28:247–263. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Ottaviani G and Jaffe N: The epidemiology of osteosarcoma. Cancer Treat Res. 152:3–13. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Bramer JA, van Linge JH, Grimer RJ and Scholten RJ: Prognostic factors in localized extremity osteosarcoma: A systematic review. Eur J Surg Oncol. 35:1030–1036. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Luetke A, Meyers PA, Lewis I and Juergens H: Osteosarcoma treatment-where do we stand? A state of the art review. Cancer Treat Rev. 40:523–532. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Zhou W, Hao M, Du X, Chen K, Wang G and Yang J: Advances in targeted therapy for osteosarcoma. Discov Med. 17:301–307. 2014.PubMed/NCBI

6 

Tan ML, Choong PF and Dass CR: Osteosarcoma: Conventional treatment vs. gene therapy. Cancer Biol Ther. 8:106–117. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Sun K and Lai EC: Adult-specific functions of animal microRNAs. Nat Rev Genet. 14:535–548. 2013. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

Behm-Ansmant I, Rehwinkel J and Izaurralde E: MicroRNAs silence gene expression by repressing protein expression and/or by promoting mRNA decay. Cold Spring Harb Symp Quant Biol. 71:523–530. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Cai Y, Yu X, Hu S and Yu J: A brief review on the mechanisms of miRNA regulation. Genomics Proteomics Bioinformatics. 7:147–154. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Rupaimoole R and Slack FJ: MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 16:203–222. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Bracken CP, Scott HS and Goodall GJ: A network-biology perspective of microRNA function and dysfunction in cancer. Nat Rev Genet. 17:719–732. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Lin W, Zhu X, Yang S, Chen X, Wang L, Huang Z, Ding Y, Huang L and Lv C: MicroRNA-203 inhibits proliferation and invasion, and promotes apoptosis of osteosarcoma cells by targeting Runt-related transcription factor 2. Biomed Pharmacother. 91:1075–1084. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Jiang Z, Jiang C, Yu C and Fang J: MicroRNA-208b inhibits human osteosarcoma progression by targeting ROR2. Tumour Biol. 39:10104283177057512017. View Article : Google Scholar : PubMed/NCBI

15 

Wu X, Yan L, Liu Y, Xian W, Wang L and Ding X: MicroRNA-448 suppresses osteosarcoma cell proliferation and invasion through targeting EPHA7. PLoS One. 12:e01755532017. View Article : Google Scholar : PubMed/NCBI

16 

Tian L, Guo Z, Wang H and Liu X: MicroRNA-635 inhibits the malignancy of osteosarcoma by inducing apoptosis. Mol Med Rep. 16:4829–4834. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Kim YH, Goh TS, Lee CS, Oh SO, Kim JI, Jeung SH and Pak K: Prognostic value of microRNAs in osteosarcoma: A meta-analysis. Oncotarget. 8:8726–8737. 2017.PubMed/NCBI

18 

Kushlinskii NE, Fridman MV and Braga EA: Molecular mechanisms and microRNAs in osteosarcoma pathogenesis. Biochemistry (Mosc). 81:315–328. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Sampson VB, Yoo S, Kumar A, Vetter NS and Kolb EA: MicroRNAs and potential targets in osteosarcoma: Review. Front Pediatr. 3:692015. View Article : Google Scholar : PubMed/NCBI

20 

Li Q, Li S, Wu Y and Gao F: miRNA-708 functions as a tumour suppressor in hepatocellular carcinoma by targeting SMAD3. Oncol Lett. 14:2552–2558. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Galun D, Basaric D, Zuvela M, Bulajic P, Bogdanovic A, Bidzic N and Milicevic M: Hepatocellular carcinoma: From clinical practice to evidence-based treatment protocols. World J Hepatol. 7:2274–2291. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Li X, Zhong X, Pan X and Ji Y: Tumor suppressive microRNA-708 targets Notch1 to suppress cell proliferation and invasion in gastric cancer. Oncol Res. 2018.

23 

Song XF, Wang QH and Huo R: Effects of microRNA-708 on epithelial-mesenchymal transition, cell proliferation and apoptosis in melanoma cells by targeting LEF1 through the Wnt signaling pathway. Pathol Oncol Res. 25:377–389. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Kim EA, Kim SW, Nam J, Sung EG, Song IH, Kim JY, Kwon TK and Lee TJ: Inhibition of c-FLIPL expression by miRNA-708 increases the sensitivity of renal cancer cells to anti-cancer drugs. Oncotarget. 7:31832–31846. 2016.PubMed/NCBI

25 

Delsin LEA, Roberto GM, Fedatto PF, Engel EE, Scrideli CA, Tone LG and Brassesco MS: Downregulated adhesion-associated microRNAs as prognostic predictors in childhood osteosarcoma. Pathol Oncol Res. 25:11–20. 2019. View Article : Google Scholar : PubMed/NCBI

26 

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

27 

Liu D, Zhang C, Li X, Zhang H, Pang Q and Wan A: MicroRNA-567 inhibits cell proliferation, migration and invasion by targeting FGF5 in osteosarcoma. EXCLI J. 17:102–112. 2018.PubMed/NCBI

28 

Du L, Chen T, Zhao K and Yang D: miR-30a suppresses osteosarcoma proliferation and metastasis by downregulating MEF2D expression. Onco Targets Ther. 11:2195–2202. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Yao Q, Pei Y, Zhang X and Xie B: microRNA-96 acts as a tumor suppressor gene in human osteosarcoma via target regulation of EZRIN. Life Sci. 203:1–11. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Guo P, Lan J, Ge J, Nie Q, Mao Q and Qiu Y: miR-708 acts as a tumor suppressor in human glioblastoma cells. Oncol Rep. 30:870–876. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Lei SL, Zhao H, Yao HL, Chen Y, Lei ZD, Liu KJ and Yang Q: Regulatory roles of microRNA-708 and microRNA-31 in proliferation, apoptosis and invasion of colorectal cancer cells. Oncol Lett. 8:1768–1774. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Jang JS, Jeon HS, Sun Z, Aubry MC, Tang H, Park CH, Rakhshan F, Schultz DA, Kolbert CP, Lupu R, et al: Increased miR-708 expression in NSCLC and its association with poor survival in lung adenocarcinoma from never smokers. Clin Cancer Res. 18:3658–3667. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Song T, Zhang X, Zhang L, Dong J, Cai W, Gao J and Hong B: miR-708 promotes the development of bladder carcinoma via direct repression of Caspase-2. J Cancer Res Clin Oncol. 139:1189–1198. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Zhang Y, Li H, Cao R, Sun L, Wang Y, Fan S, Zhao Y, Kong D, Cui L, Lin L, et al: Suppression of miR-708 inhibits the Wnt/β-catenin signaling pathway by activating DKK3 in adult B-all. Oncotarget. 8:64114–64128. 2017.PubMed/NCBI

35 

Zhou YM, Cao L, Li B, Zhang RX, Sui CJ, Yin ZF and Yang JM: Clinicopathological significance of ZEB1 protein in patients with hepatocellular carcinoma. Ann Surg Oncol. 19:1700–1706. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Zhang Y, Liu G, Wu S, Jiang F, Xie J and Wang Y: Zinc finger E-box-binding homeobox 1: Its clinical significance and functional role in human thyroid cancer. Onco Targets Ther. 9:1303–1310. 2016.PubMed/NCBI

37 

Li J, Xia L, Zhou Z, Zuo Z, Xu C, Song H and Cai J: MiR-186-5p upregulation inhibits proliferation, metastasis and epithelial-to-mesenchymal transition of colorectal cancer cell by targeting ZEB1. Arch Biochem Biophys. 640:53–60. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Larsen JE, Nathan V, Osborne JK, Farrow RK, Deb D, Sullivan JP, Dospoy PD, Augustyn A, Hight SK, Sato M, et al: ZEB1 drives epithelial-to-mesenchymal transition in lung cancer. J Clin Invest. 126:3219–3235. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Jia B, Liu H, Kong Q and Li B: Overexpression of ZEB1 associated with metastasis and invasion in patients with gastric carcinoma. Mol Cell Biochem. 366:223–229. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Shen A, Zhang Y, Yang H, Xu R and Huang G: Overexpression of ZEB1 relates to metastasis and invasion in osteosarcoma. J Surg Oncol. 105:830–834. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Liu C and Lin J: Long noncoding RNA ZEB1-AS1 acts as an oncogene in osteosarcoma by epigenetically activating ZEB1. Am J Transl Res. 8:4095–4105. 2016.PubMed/NCBI

42 

Xu J, Wang Z, Liao Z, Dai D and Ma X: MicroRNA-150 functions as an antioncogenic regulator in osteosarcoma. Oncol Lett. 14:2483–2490. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Yan H, Zhang B, Fang C and Chen L: miR-340 alleviates chemoresistance of osteosarcoma cells by targeting ZEB1. Anticancer Drugs. 29:440–448. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Wang H, Xing D, Ren D, Feng W, Chen Y, Zhao Z, Xiao Z and Peng Z: MicroRNA643 regulates the expression of ZEB1 and inhibits tumorigenesis in osteosarcoma. Mol Med Rep. 16:5157–5164. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 19 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He J, Xiang D and Lin Y: MicroRNA‑708 inhibits the proliferation and invasion of osteosarcoma cells by directly targeting ZEB1 Retraction in /10.3892/mmr.2023.12936. Mol Med Rep 19: 3948-3954, 2019
APA
He, J., Xiang, D., & Lin, Y. (2019). MicroRNA‑708 inhibits the proliferation and invasion of osteosarcoma cells by directly targeting ZEB1 Retraction in /10.3892/mmr.2023.12936. Molecular Medicine Reports, 19, 3948-3954. https://doi.org/10.3892/mmr.2019.10013
MLA
He, J., Xiang, D., Lin, Y."MicroRNA‑708 inhibits the proliferation and invasion of osteosarcoma cells by directly targeting ZEB1 Retraction in /10.3892/mmr.2023.12936". Molecular Medicine Reports 19.5 (2019): 3948-3954.
Chicago
He, J., Xiang, D., Lin, Y."MicroRNA‑708 inhibits the proliferation and invasion of osteosarcoma cells by directly targeting ZEB1 Retraction in /10.3892/mmr.2023.12936". Molecular Medicine Reports 19, no. 5 (2019): 3948-3954. https://doi.org/10.3892/mmr.2019.10013