Open Access

MicroRNA-939-5p directly targets IGF-1R to inhibit the aggressive phenotypes of osteosarcoma through deactivating the PI3K/Akt pathway

Retraction in: /10.3892/ijmm.2022.5181

  • Authors:
    • Xiwu Zhao
    • Jian Li
    • Dapeng Yu
  • View Affiliations

  • Published online on: September 5, 2019     https://doi.org/10.3892/ijmm.2019.4333
  • Pages: 1833-1843
  • Copyright: © Zhao 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

The dysregulation of microRNA‑939‑5p (miR‑939) is involved in the development of multiple types of human cancer. However, the expression and roles of miR‑939 in osteosarcoma (OS) have yet to be clarified. The expression level of miR‑939 in OS was measured using reverse transcription quantitative polymerase chain reaction (RT‑qPCR). A Cell Counting Kit‑8 assay, flow cytometry analysis, Transwell migration and invasion assays, and a tumor xenograft assay were employed to explore the effects of miR‑939 in OS cells. Bioinformatics analysis, RT‑qPCR, western blot analysis and luciferase reporter assays were performed to explore its underlying mechanism. Expression of miR‑939 was decreased in both OS tissues and cell lines. The decreased miR‑939 expression was notably correlated with clinical stage and distant metastasis in patients with OS, where low miR‑939 levels were correlated with lower overall survival. miR‑939 overexpression decreased OS cell proliferation, migration and invasion in vitro; induced cell apoptosis, and impaired tumor growth in vivo. Mechanistically, insulin‑like growth factor 1 receptor (IGF‑1R) was characterized as direct target gene of miR‑939 in OS. The tumor‑suppressing effects of miR‑939 in OS cells were imitated by IGF‑1R knockdown. In addition, exogenous IGF‑1R expression abolished the tumor suppressive roles of miR‑939 in OS cells. miR‑939 was implicated in the deactivation of the PI3K/Akt pathway in OS in vitro and in vivo through regulating IGF‑1R expression. The present study demonstrated that miR‑939 exerted tumor‑suppressing roles in the malignancy of OS cells by directly targeting IGF‑1R and inactivating the PI3K/AKT pathway. Therefore, this miRNA may be a promising target for anticancer therapy in patients with OS.

Introduction

Osteosarcoma (OS) is a highly malignant bone tumor that normally occurs in the metaphysis of the long bones (1). It is characterized by osteoblastic differentiation and malignant osteoid production. Incipient symptoms for patients with OS are nonspecific (2). Globally, ~4.4/million individuals have OS, with a peak morbidity age of 15-19 years (3). Multiple factors, including genetic and epigenetic variations, and potential environmental factors, exert crucial roles in the etiology and progression of OS (4-6); however, studies on the detailed molecular events underlying OS pathogenesis are limited. Development of anticancer therapy for patients with OS, including surgical resection, and effective neoadjuvant and adjuvant chemotherapy, have resulted in notable improvements in the clinical outcomes over previous decades (7,8); however, the prognosis of patients with OS remains poor, and pulmonary metastasis is the most frequent cause of mortality (9). Therefore, an improved understanding of the mechanisms regulating the malignancy of OS is required for the identification of promising therapeutic approaches that may improve the prognosis of patients with OS.

MicroRNAs (miRNAs) are a series of single stranded and non-coding RNA molecules that range in length from 18-23 nucleotides (10). miRNAs negatively regulate gene expression via directly binding to the 3′-untranslated regions (3′-UTRs) of their target genes, which results in their degradation or destabilization (11). A total of >2,000 miRNAs have been identified in human cells, and it is estimated that they are implicated in the regulation of ~30% of the protein-coding genes in the human genome (12). It has been widely recognized that the aberration of miRNA expression is observed in almost all types of human malignancy, including OS (13), and pancreatic (14), gastric (15) and ovarian cancer (16). In OS, the dysregulation of miRNAs is involved in the modulation of OS occurrence and development as oncogenes or tumor suppressors (17,18). Therefore, investigating the detailed functions of OS-associated miRNAs may assist in developing novel targets for anticancer gene therapy.

Recently, the dysregulation of miR-939-5p (miR-939) has been demonstrated to participate in the aggressive behaviors of multiple types of human cancer (19-23). However, the expression profile and functional roles of miR-939 in OS have yet to be clarified. Therefore, the present study first detected miR-939 expression in OS tissues and cell lines. Then, the specific roles and associated molecular events of miR-939 in OS were elucidated in detail. The results of the present study verified that miR-939 overexpression suppressed the malignant phenotypes of OS by directly targeting insulin-like growth factor 1 receptor (IGF-1R) and deactivating the PI3K/Akt pathway. This suggests that the miR-939/IGF-1R/PI3K/Akt pathway is a potential target for anticancer therapy in patients with OS.

Materials and methods

Human tissue collection

The present study was approved by the Medical Ethics Committee of Shandong Provincial Western Hospital (Jinan, China). Written informed consent was obtained from all participants. A total of 58 pairs of OS tissue and adjacent non-tumorous tissue samples were obtained from patients at Shandong Provincial Western Hospital. None of the patients had been treated with radiotherapy, chemotherapy or other anticancer therapies. All tissues were immediately frozen in liquid nitrogen following surgical excision and stored at −80°C.

Cell culture

A total of 4 human OS cell lines, including SAOS-2, MG-63, HOS and U2OS, were purchased from the Shanghai Institute of Biochemistry and Cell Biology. The normal human osteoblast hFOB1.19 cell line was purchased from the American Type Culture Collection. All cell lines were grown in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (all from Gibco; Thermo Fisher Scientific, Inc.). All cells were maintained at 37°C in a humidified atmosphere containing 5% CO2.

Transfection

For miR-939 functional analysis, miR-939 mimics were chemically generated by Guangzhou RiboBio Co., Ltd., and transfected into cells to increase endogenous miR-939 expression. Similarly, small interfering RNA (siRNA) targeting the expression of IGF-1R (si-IGF-1R; Shanghai GenePharma Co., Ltd.) was utilized to silence IGF-1R expression. miRNA mimics negative control (miR-NC) and negative control siRNA (si-NC) served as the controls for miR-939 mimics and si-IGF-1R, respectively. The sequence of the si-IGF-1R was 5′-CACCGCGGCTGGAAACTCTTCTACACGAATGTAGAAGAGTTTCCAGCC GC-3′. The sequence of the si-NC was 5′-CACCGCTCACCGGCTCCAGATTTATCGAAATAAATCTGGAGCCGGTGAGC-3′. The empty vector (pcDNA3.1) and the plasmids containing IGF-1R pcDNA3.1-IGF-1R (pc-IGF-1R) were synthesized by (Youbao Bio; Hunan Keai Medical Devices Co. Ltd.). Cells were trans-fected with miR-939 mimics (100 pmol), miR-NC (100 pmol), si-IGF-1R (100 pmol), si-NC (100 pmol), pc-IGF-1R (4 μg) or pcDNA3.1 (4 μg) using Lipofectamine® 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) (24). Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), flow cytometry analysis and Transwell migration and invasion assays were carried out at 48 h post-transfection. After 24 h incubation, transfected cells were collected and used in Cell Counting Kit-8 (CCK-8) assay and tumor xenograft assay.

RNA preparation and RT-qPCR

Total RNA in tissues or cells was isolated using the RNeasy Plus mini kit (Qiagen GmbH). To detect miR-939 expression, total RNA was reverse transcribed into cDNA using the miScript Reverse Transcription kit (Qiagen GmbH). The temperature protocol for reverse transcription of miRNA was as follows: 37°C for 60 min, 95°C for 5 min, and the samples were subsequently kept at 4°C. miScript SYBR-Green PCR kit (Qiagen GmbH) was employed to conduct quantitative PCR using an ABI 7500 thermocycler (Thermo Fisher Scientific, Inc.). The thermocycling conditions were as follows: 95°C for 2 min, followed by 40 cycles of 95°C for 10 sec, 55°C for 30 sec and 72°C for 30 sec. For the analysis of IGF-1R mRNA expression, RT and qPCR were performed using PrimeScript RT Reagent kit and SYBR Premix Ex Taq™ kit (Takara Biotechnology Co., Ltd.), respectively. The temperature protocol for reverse transcription of RNA was as follows: 37°C for 15 min and 85°C for 5 sec. The thermo-cycling conditions 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. U6 small nuclear RNA and GAPDH served as the internal reference for miR-939 and IGF-1R, respectively. Relative gene expression was calculated using the 2−∆∆Cq formula (24). The primer sequences used for qPCR were: miR-939 forward, 5′-GGGTGGGGAGCTGAGGCTCTG-3′ and reverse, 5′-CAGTGCGTGTCGTGGAGT-3′; U6 forward, 5′-CGTTTTACTTCCTCATACAGCAC-3′ and reverse, 5′-GCACCAAGAGACCTGTGACA-3′; IGF-1R forward, 5′-AGGATATTGGGCTTTACAACCTG-3′; reverse, 5′-GAGGTAACAGAGGTCAGCATTTT-3′; and GAPDH forward, 5′-TGCACCACCAACTGCTTA-3′ and reverse, 5′-GGATGCAGGGATGATGTT C-3′.

CCK-8 assay

After 24 h culture, transfected cells were collected and used for the detection cell proliferation using a CCK-8 assay. Briefly, transected cells were inoculated into 96-well plates with an initial density of 2×103 cells/well. A total of 10 μl CCK-8 solution (Beyotime Institute of Biotechnology) was added into each well at 0, 24, 48 and 72 h after inoculation. After 2 h incubation, the optical density value of each well at a 450 nm wavelength was recorded using an ELx800 microplate Reader (Bio-Tek Instruments, Inc.).

Flow cytometry analysis

Transfected cells were collected after 48 h incubation at 37°C and subjected to the detection of cell apoptosis using an Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit (BioLegend, Inc.). The transfected cells were then washed 3 times with PBS (Gibco; Thermo Fisher Scientific, Inc.), resuspended in 100 μl binding buffer, and double-stained with 5 μl Annexin V-FITC and 5 μl propidium iodide. Then, the cells were analyzed by a FACScan flow cytometer (BD Biosciences). Data were analyzed with CellQuest Pro 4.0.2 software (BD Biosciences).

Transwell migration and invasion assays

Transwell chambers (8 μm pore size; Costar; Corning Incorporated) pre-coated with Matrigel (BD Biosciences) at 37°C for 30 min were utilized to determine the invasive ability, while uncoated Transwell chambers were used in the Transwell migration assay. Transfected cells were collected at 48 h post-transfection and resuspended in FBS-free DMEM. A total of 200 μl cell suspension containing 5×104 transfected cells was added in the upper compartments. The lower compartments were covered with 500 μl DMEM containing 10% FBS. After 24 h incubation at 37°C, cells remaining in the upper membranes were gently removed using a cotton swab. The migrated and invaded cells on the lower surface of the membranes were fixed with 4% paraformaldehyde at room temperature for 30 min, stained with 0.1% crystal violet at room temperature for 30 min, and washed with PBS 3 times. Finally, the numbers of migrated and invaded cells were counted in 5 randomly chosen visuals/chamber under an inverted light microscope (×200 magnification; Olympus Corporation).

Tumor xenograft assay

A total of 8 female BALB/c nude mice (20 g; 4-5 week-old) were obtained from the Model Animal Research Institute of Nanjing University. HOS cells transfected with miR-939 mimics or miR-NC were suspended in PBS, and inoculated subcutaneously into the flank of nude mice (n=4 for each group). The tumor size was detected every 4 days, and the tumor volume was calculated using the formula: Volume (mm3)=1/2 × (length × width2). A total of 4 weeks after injection, all nude mice were sacrificed by means of cervical dislocation. The tumor xenografts were separated, weighed and processed for RT-qPCR and western blot analysis. The animal experiments were approved by the Institutional Animal Care and Use Committee of Shandong Provincial Western Hospital.

Target identification and luciferase activity assay

Bioinformatics analysis was performed to search for the potential targets of miR-939. TargetScan (release 7.2; March 2018; http://targetscan.org/) (25) and miRDB (http://mirdb.org/) (26) validated IGF-1R as a putative target of miR-939.

The wild-type (wt) 3′-UTR of IGF-1R containing the predicted miR-939 binding site and mutant (mut) IGF-1R 3′-UTR was chemically amplified by Shanghai GenePharma Co., Ltd., and inserted into the pMIR-REPORT luciferase reporter plasmid (Ambion; Thermo Fisher Scientific, Inc.) to generate the luciferase plasmids and referred to as pMIR-IGF-1R-3′-UTR wt and pMIR-IGF-1R-3′-UTR mut, respectively. Cells were inoculated into 24-well plates 1 day prior to transfection, followed by co-transfection with pMIR-IGF-1R-3′-UTR wt or pMIR-IGF-1R-3'-UTR mut, and miR-939 mimics or miR-NC, using the Lipofectamine® 2000. Following 48 h transfection, luciferase activity was determined using a Dual-Luciferase® Reporter Assay system (E1910; Promega Corporation) according to the manufacturer′s instructions. Renilla luciferase activity was used as the internal control.

Western blot analysis

The isolation of total protein was performed using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology). A bicinchoninic acid assay kit (Pierce; Thermo Fisher Scientific, Inc.) was used to quantify protein concentration. Equivalent amounts of protein (30 μg) were separated using 10% SDS-PAGE gel, transferred to polyvinylidene fluoride membranes, blocked at room temperature with 5% evaporated skimmed milk for 2 h, and then subjected to overnight incubation at 4°C with primary antibodies. Thereafter, protein bands were detected by goat anti-rabbit (cat. no. ab6721) or goat anti-mouse (cat. no. ab6790) immunoglobulin G horseradish peroxidase-conjugated secondary antibody (Abcam) and were visualized using an enhanced chemiluminescence Western Blotting Detection kit (Applygen Technologies, Inc.). The primary antibodies used in this study were as follows: Mouse anti-human monoclonal IGF-1R antibody (cat. no. sc-81464; Santa Cruz Biotechnology, Inc.), rabbit anti-human polyclonal phosphorylated (p)-PI3K antibody (cat. no. ab182651; Abcam), mouse anti-human monoclonal PI3K (cat. No ab189403; Abcam), mouse anti-human monoclonal p-Akt antibody (cat. no. sc-81433; Santa Cruz Biotechnology), mouse anti-human monoclonal Akt antibody (cat. no. sc-81434; Santa Cruz Biotechnology) and mouse anti-human monoclonal GAPDH antibody (cat. no. sc-32233). All primary antibodies were used with dilution 1:1,000, while the secondary antibodies were used with dilution 1:5,000. Quantity One software (version 4.62; Bio-Rad Laboratories, Inc.) was used for performing the densitometric analysis.

Statistical analysis

All experiments were repeated at least 3 times, and the data are presented as the mean ± standard deviation. The correlation analysis between miR-939 expression and clinicopathological features in patients with OS was analyzed using a χ2 test. A two-tailed student's t-test was used to analyze differences between two groups. The comparisons between multiple groups were examined using one-way analysis of variance followed by Tukey's post-hoc test. Spearman's correlation analysis was utilized to test the expression correlation between miR-939 and IGF-1R mRNA in OS tissues. Survival curves were assessed by Kaplan-Meier analysis and compared with log-rank test. All statistical analysis was performed using SPSS software (version 19.0; IBM Corp.). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-939 is downregulated in OS and predicts poor prognosis

To analyze the clinical significance of miR-939 in OS, its expression was first measured in 58 pairs of OS tissues and adjacent non-tumorous tissues using RT-qPCR. The results indicated that miR-939 expression was decreased in OS tissues compared with that in non-tumorous tissue (Fig. 1A; P<0.05). In addition, the expression level of miR-939 was determined in a panel of OS cell lines, including SAOS-2, MG-63, HOS and U2OS. The normal human osteoblast hFOB1.19 cell line served as the control. Data from the RT-qPCR analysis indicated that expression level of miR-939 was decreased in all OS cell lines examined compared with that in the hFOB1.19 cell line (Fig. 1B; P<0.05).

The high (≥0.35) and low (<0.35) miR-939 expression groups were defined based on the median value of miR-939 in OS tissues (0.35). The association between miR-939 expression and clinicopathological features in patients with OS was explored. As indicated in Table I, low miR-939 expression was notably correlated with clinical stage (P=0.033) and distant metastasis (P=0.028) in patients with OS. Furthermore, the prognostic value of miR-939, which was determined by Kaplan-Meier survival cures, indicated that the low miR-939 expression group exhibited a decrease in overall survival compared to the high miR-939 expression group (Fig. 1C; P=0.0135). These observations suggest that miR-939 may be closely associated with the progression and development of OS.

Table I

Association between clinicopathological characteristics and the expression of miR-939 in patients with osteosarcoma.

Table I

Association between clinicopathological characteristics and the expression of miR-939 in patients with osteosarcoma.

miR-939 expression
CharacteristicsLow (n=29)High (n=29)P-value
Age at diagnosis, years0.565
 <181922
 ≥18107
Sex0.292
 Male1813
 Female1116
Tumor size, cm0.592
 <51619
 ≥51310
Clinical stage0.033a
 I-IIA1221
 IIB/III178
Distant metastasis0.028a
 Negative1423
 Positive156

{ label (or @symbol) needed for fn[@id='tfn1-ijmm-44-05-1833'] } miR, microRNA.

a P<0.05.

miR-939 exerts a tumor-suppressing role in OS cells in vitro

To understand the roles of miR-939 in the malignancy of OS, miR-939 mimics or miR-NC were transfected into HOS and U2OS cells. Following transfection, RT-qPCR confirmed that miR-939 was evidently overexpressed in HOS and U2OS cells following miR-939 mimics transfection (Fig. 2A; P<0.05). Cell proliferation was subsequently determined via CCK-8 assay, and the results revealed that miR-939 mimics transfection resulted in a significant decrease in HOS and U2OS cell proliferation rates (Fig. 2B; P<0.05). To additionally examine the inhibitory effect of miR-939 in OS cell proliferation, the apoptosis rates of HOS and U2OS cells following miR-939 upregulation were detected. The data demonstrated that when miR-939 was upregulated in HOS and U2OS cells, the proportion of apoptotic cells was increased markedly (Fig. 2C; P<0.05). Furthermore, the migratory (Fig. 2D; P<0.05) and invasive (Fig. 2E; P<0.05) capacities were prominently decreased following ectopic miR-939 expression in HOS and U2OS cells, as demonstrated by the Transwell migration and invasion assays. These results implied that miR-939 exerted a tumor suppressive role in the aggressive behaviors of OS in vitro.

IGF-1R is a direct target gene of miR-939 in OS

The specific function of miRNAs relies on the negative regulation of their target genes; therefore, bioinformatics analysis was applied to search for the potential target of miR-939. Among these candidates, IGF-1R (Fig. 3A) was identified, as the gene has been implicated in the malignant phenotypes of OS (27-30). Next, a luciferase reporter assay was employed to explore whether miR-939 directly targeted IGF-1R in OS. Upregulation of miR-939 notably suppressed the luciferase activity of reporter plasmid containing the wild-type miR-939 binding sites (1 and 2) in both HOS and U2OS cells (P<0.05). Notably, miR-939 overexpression had no effect on the luciferase activity of the mutant reporter plasmid (1 and 2), suggesting that miR-939 was able to recognize and bind to the 3′-UTR of IGF-1R in OS (Fig. 3B). In addition, the results of the RT-qPCR and western blot analysis revealed that the mRNA (Fig. 3C; P<0.05) and protein (Fig. 3D; P<0.05) levels of IGF-1R were markedly decreased in HOS and U2OS cells overexpressing miR-939.

To additionally elucidate the association between miR-939 and IGF-1R in OS tissues, IGF-1R expression in OS tissues was measured, and it was indicated that the IGF-1R mRNA was overexpressed compared with non-tumorous tissues (Fig. 3E; P<0.05). Furthermore, a negative expression correlation between miR-939 and IGF-1R mRNA in the same OS tissue was validated via Spearman's correlation analysis (Fig. 3F; R2=0.3261; P<0.0001). Taken together, these results suggested that IGF-1R is a direct target of miR-939 in OS.

IGF-1R silencing imitates the tumor-suppressing roles of miR-939 in OS cells

Having identified IGF-1R as a direct target of miR-939 in OS, the roles of IGF-1R in the development of OS were further examined. si-IGF-1R was introduced into HOS and U2OS cells, and RT-qPCR analysis confirmed that si-IGF-1R transfection suppressed the expression of IGF-1R in HOS and U2OS cells at mRNA (Fig. 4A; P<0.05) and protein (Fig. 4B; P<0.05) levels. Thereafter, CCK-8 assay and flow cytometry analysis were performed, and the results revealed that silencing of IGF-1R expression significantly inhibited the proliferation rate (Fig. 4C; P<0.05) and promoted the apoptosis rate (Fig. 4D, P<0.05) of HOS and U2OS cells. The migration (Fig. 4E; P<0.05) and invasion (Fig. 4F; P<0.05) rates were also attenuated in HOS and U2OS cells following si-IGF-1R transfection. These results demonstrated that IGF-1R knockdown was able to imitate the tumor-suppressing roles of miR-939 in OS cells, additionally suggesting that IGF-1R served a downstream mediator of miR-939 in OS.

Restoring IGF-1R expression counteracts the tumor suppressive effects of miR-939 in OS cells

Rescue experiments were performed to validate that decreasing IGF-1R expression is essential for the functions of miR-939 in OS cells. Firstly, pc-IGF-1R or pcDNA3.1 plasmids were transfected into HOS and U2OS cells. Western blot analysis confirmed that the transfection of pc-IGF-1R notably increased IGF-1R protein expression levels in HOS and U2OS cells (Fig. 5A; P<0.05). miR-939 overexpressing-HOS and U2OS cells were transfected with pc-IGF-1R to recover expression, and the restoration of IGF-1R mRNA (Fig. 5B; P<0.05) and protein (Fig. 5C, P<0.05) was then confirmed by RT-qPCR and western blot analysis. Upregulation of miR-939 inhibited HOS and U2OS cell proliferation (Fig. 5D; P<0.05), induced cell apoptosis (Fig. 5E; P<0.05), and hindered cell migration (Fig. 5F; P<0.05) and invasion (Fig. 5G; P<0.05), whereas reintroduction of IGF-1R abolished these effects. In summary, miR-939 may serve tumor-suppressing roles in OS progression by decreasing IGF-1R expression.

miR-939 deactivates the PI3K/Akt pathway in OS via IGF-1R regulation

IGF-1R has recently been implicated in the activation of PI3K/Akt pathway (31-33). Therefore, the present study attempted to identify whether miR-939 was able to deactivate the PI3K/Akt pathway in OS cells. Western blot analysis revealed that ectopic miR-939 expression downregulated the expression levels of p-PI3K and p-Akt in HOS and U2OS cells (Fig. 6), suggesting that miR-939 may have inactivated the PI3K/Akt pathway in OS cells. Notably, restoration of IGF-1R expression reversed the downregulation of p-PI3K and p-Akt in miR-939 overexpressing-HOS and U2OS cells. These results suggest that miR-939 overexpression deactivates the PI3K/AKT pathway in OS cells by decreasing IGF-1R expression.

miR-939 suppresses the tumor growth of OS in vivo

As the data from the present study indicated that restoration of miR-939 expression decreased OS cell proliferation in vitro, the involvement of miR-939 in in vivo tumor growth was subsequently explored though a tumor xenograft assay. A total of 4 weeks after inoculation, the tumor growth curves demonstrated that miR-939 upregulation markedly decreased the tumor growth of OS cells (Fig. 7A and B; P<0.05). Tumor weight in nude mice injected with miR-939 mimics transfected-HOS cells was decreased compared with that in the miR-NC group (Fig. 7C; P<0.05). In addition, the levels of IGF-1R and key molecules associated with the PI3K/Akt pathway were measured in the tumor xenografts. The data indicated that the expression levels of IG-1R, p-PI3K and p-Akt in tumor xenografts derived from the miR-939 mimics group were downregulated compared with those in the miR-NC group (Fig. 7D; P<0.05). Furthermore, miR-939 expression was detected in the tumor xenografts, and the results confirmed that the suppression of OS tumor growth was attributed to miR-939 overexpression. As expected, the upregulation of miR-939 was observed in the tumor xenografts obtained from nude mice injected with miR-939 mimics transfected-HOS cells (Fig. 7E; P<0.05). Therefore, these data demonstrated that miR-939 impaired tumor growth in vivo through suppression of IGF-1R expression and deactivation of the PI3K/Akt pathway.

Discussion

The dysregulation of miRNAs with oncogenic or tumor-suppressing effects in OS has been demonstrated in a number of studies (33-35). Furthermore, restoring or silencing miRNAs with chemically synthesized oligonucleotides has the potential to alter the roles of miRNA in carcinogenesis and cancer progression, providing a theoretical foundation for miRNA-based targeted anticancer therapy (36,37). Accordingly, exploring the detailed roles of miRNAs in the malignancy of OS may promote the identification of attractive targets for therapy in patients with OS. Therefore, in the present study, miR-939 expression was detected in both OS tissues and cell lines. Then, the effects of miR-939 on the malignancy of OS cells in vitro and in vivo were investigated. Finally, the molecular mechanisms and potential signaling pathway underlying the tumor suppressive roles of miR-939 in OS were explored in detail. The results will be beneficial for the identification of potential molecular targets for the OS diagnosis, therapy and prevention.

miR-939 expression is increased in ovarian cancer (19) and hepatocellular carcinoma (20). In addition, miR-939 was validated as an independent factor for the prognosis of patients with hepatocellular carcinoma (20). By contrast, decreased expression of miR-939 is observed in colorectal cancer (21) and tongue squamous cell carcinoma (22). Low miR-939 expression was inversely correlated with tumor stage in patients with tongue squamous cell carcinoma (22). There is also low expression of miR-939 in gastric cancer, and miR-939 was demonstrated to be an independent indicator for predicting poor prognosis and recurrence (23). However, the expression status of miR-939 in OS has yet to be elucidated. The present study identified that miR-939 was downregulated in OS, and that its downregulation was correlated with clinical stage and distant metastasis. Notably, patients with OS in the low miR-939 expression group exhibited a decrease in overall survival compared with those in the high miR-939 expression group. These observations suggest that miR-939 is an effective biomarker for the diagnosis and prognosis of patients with OS.

miR-939 has been identified as an oncogenic miRNA in ovarian cancer that promotes cell proliferation and anchorage-independent growth (19). Conversely, miR-939 exerts tumor-suppressing roles in multiple types of human cancer. For example, miR-939 overexpression decreases the migration and invasion of colorectal cancer cells (21), and suppresses the growth and metastasis of gastric cancer in vitro and in vivo (23). In addition, upregulation of miR-939 increases the chemosensitivity of gastric cancer cells to 5-fluorouracil (23). Nevertheless, the specific roles of miR-939 in the malignant phenotypes of OS in vitro and in vivo remain unclear clear. In the present study, a series of functional experiments demonstrated that restoration of miR-939 expression inhibited cell proliferation, migration and invasion in vitro, induced cell apoptosis in vitro, and decreased tumor growth in vivo. These results suggested that restoring miR-939, resulting in the inhibition of OS progression, may be a potential therapeutic technique for patients with OS.

Multiple genes, including adenomatous polyposis coli 2 in ovarian cancer (19), LIM domain kinase 2 in colorectal cancer (21) and solute carrier family 34 member 2 in gastric cancer (23), have been previously characterized as direct targets of miR-939. IGF-1R is a transmembrane tyrosine kinase receptor in the insulin receptor family, and was identified as a direct target of miR-939 in OS. IGF-1R is upregulated in OS, and the upregulation of IGF-1R exhibits a close association with surgical stage and distant metastasis (27). Patients with OS and high IGF-1R expression exhibit poorer clinical outcomes (27). In addition, increased IGF1-1R expression was confirmed as an independent prognostic maker for patients with OS (27). Functionally, IGF-1R serves an important role in the onset and development of OS, and is involved in the regulation of various aggressive behaviors, including cell adhesion, proliferation, apoptosis, cell cycle, motility, metastasis, chemoresistance and radioresistance (27-30). A second notable result of the present study was that miR-939 overexpression deactivated the PI3K/Akt pathway in OS cells. However, whether miR-939 was able to affect other pathways in OS was not explored in detail. This was a limitation of the present study, and it will be resolved in subsequent studies. Combined with the observations of the present study, targeting molecules that silence IGF-1R expression or restore miR-939 expression, resulting in the deactivation of PI3K/Akt pathway, may be useful for the therapy of patients with OS.

To conclude, miR-939 expression was decreased in OS and predicted poor prognosis. Exogenous miR-939 expression suppressed the malignancy of OS in vitro and in vivo by directly targeting IGF-1R and deactivating the PI3K/Akt pathway. Therefore, miR-939 may be a potential therapeutic target and promising biomarker for the diagnosis and prognosis in patients with OS.

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 on reasonable request.

Authors' contributions

XZ and JL performed RT-qPCR analysis, CCK-8 assay, lucif-erase reporter assay and western blotting. DY conducted other functional experiments and analyzed the data of this study. All authors have made a significant contribution to the results and methods. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Medical Ethics Committee of Shandong Provincial Western Hospital. Written informed consent was obtained from all participants.

Patient consent for publication

All patients proved consent for the publication of their data.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Kager L, Zoubek A, Dominkus M, Lang S, Bodmer N, Jundt G, Klingebiel T, Jürgens H and Gadner H: Osteosarcoma in very young children: Experience of the cooperative osteosarcoma study group. Cancer. 116:5316–5324. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Geller DS and Gorlick R: Osteosarcoma: A review of diagnosis, management, and treatment strategies. Clin Adv Hematol Oncol. 8:705–718. 2010.

4 

Sun L, Li Y, Zhang J, Li H, Li B and Ye Z: Prognostic value of pathologic fracture in patients with high grade localized osteo-sarcoma: A systemic review and meta-analysis of cohort studies. J Orthop Res. 33:131–139. 2015. View Article : Google Scholar

5 

Li S, Zhang T, Xu W, Ding J, Yin F, Xu J, Sun W, Wang H, Sun M, Cai Z and Hua Y: Sarcoma-targeting peptide-decorated polypeptide nanogel intracellularly delivers shikonin for upregulated osteosarcoma necroptosis and diminished pulmonary metastasis. Theranostics. 8:1361–1375. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Zhang Y, Wang F, Li M, Yu Z, Qi R, Ding J, Zhang Z and Chen X: Self-stabilized hyaluronate nanogel for intracellular codelivery of doxorubicin and cisplatin to osteosarcoma. Adv Sci (Weinh). 5:pp. 17008212018, View Article : Google Scholar

7 

Moore DD and Luu HH: Osteosarcoma. Cancer Treat Res. 162:65–92. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Blattmann C, Oertel S, Schulz-Ertner D, Rieken S, Haufe S, Ewerbeck V, Unterberg A, Karapanagiotou-Schenkel I, Combs SE, Nikoghosyan A, et al: Non-randomized therapy trial to determine the safety and efficacy of heavy ion radiotherapy in patients with non-resectable osteosarcoma. BMC Cancer. 10:962010. View Article : Google Scholar : PubMed/NCBI

9 

Errani C, Longhi A, Rossi G, Rimondi E, Biazzo A, Toscano A, Alì N, Ruggieri P, Alberghini M, Picci P, et al: Palliative therapy for osteosarcoma. Expert Rev Anticancer Ther. 11:217–227. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Shukla GC, Singh J and Barik S: MicroRNAs: Processing, maturation, target recognition and regulatory functions. Mol Cell Pharmacol. 3:83–92. 2011.PubMed/NCBI

11 

Pasquinelli AE, Hunter S and Bracht J: MicroRNAs: A developing story. Curr Opin Genet Dev. 15:200–205. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Jia F, Zhang Z and Zhang X: MicroRNA-338-3p inhibits tumor growth and metastasis in osteosarcoma cells by targeting RUNX2/CDK4 and inhibition of MAPK pathway. J Cell Biochem. 120:6420–6430. 2019. View Article : Google Scholar

14 

Ma L, Shao Z and Zhao Y: MicroRNA-374a promotes pancreatic cancer cell proliferation and epithelial to mesenchymal transition by targeting SRCIN1. Pathol Res Pract. 215:1523822019. View Article : Google Scholar : PubMed/NCBI

15 

Zhang Z and Dai DQ: MicroRNA-596 acts as a tumor suppressor in gastric cancer and is upregulated by promotor demethylation. World J Gastroenterol. 25:1224–1237. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Tang Z, Fang Y and Du R: MicroRNA-107 induces cell cycle arrests by directly targeting cyclin E1 in ovarian cancer. Biochem Biophys Res Commun. 512:331–337. 2019. 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

19 

Ying X, Li-ya Q, Feng Z, Yin W and Ji-hong L: MiR-939 promotes the proliferation of human ovarian cancer cells by repressing APC2 expression. Biomed Pharmacother. 71:64–69. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Fornari F, Ferracin M, Trerè D, Milazzo M, Marinelli S, Galassi M, Venerandi L, Pollutri D, Patrizi C, Borghi A, et al: Circulating microRNAs, miR-939, miR-595, miR-519d and miR-494, identify cirrhotic patients with HCC. PLoS One. 10:pp. e01414482015, View Article : Google Scholar : PubMed/NCBI

21 

Zhang Y, Liu X, Li Q and Zhang Y: lncRNA LINC00460 promoted colorectal cancer cells metastasis via miR-939-5p sponging. Cancer Manag Res. 11:1779–1789. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Chen Y, Guo Y and Yan W: lncRNA RP5-916L7.2 correlates with advanced tumor stage, and promotes cells proliferation while inhibits cells apoptosis through targeting miR-328 and miR-939 in tongue squamous cell carcinoma. Clin Biochem. 67:24–32. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Zhang JX, Xu Y, Gao Y, Chen C, Zheng ZS, Yun M, Weng HW, Xie D and Ye S: Decreased expression of miR-939 contributes to chemoresistance and metastasis of gastric cancer via dysregulation of SLC34A2 and Raf/MEK/ERK pathway. Mol Cancer. 16:182017. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Agarwal V, Bell GW, Nam JW and Bartel DP: Predicting effective microRNA target sites in mammalian mRNAs. Elife. 4:2015. View Article : Google Scholar

26 

Liu W and Wang X: Prediction of functional microRNA targets by integrative modeling of microRNA binding and target expression data. Genome Biol. 20:182019. View Article : Google Scholar : PubMed/NCBI

27 

Wang YH, Han XD, Qiu Y, Xiong J, Yu Y, Wang B, Zhu ZZ, Qian BP, Chen YX, Wang SF, et al: Increased expression of insulin-like growth factor-1 receptor is correlated with tumor metastasis and prognosis in patients with osteosarcoma. J Surg Oncol. 105:235–243. 2012. View Article : Google Scholar

28 

Li YS, Liu Q, He HB and Luo W: The possible role of insulin-like growth factor-1 in osteosarcoma. Curr Prob Cancer. 43:228–235. 2019. View Article : Google Scholar

29 

Wang YH, Wang ZX, Qiu Y, Xiong J, Chen YX, Miao DS and De W: Lentivirus-mediated RNAi knockdown of insulin-like growth factor-1 receptor inhibits growth, reduces invasion, and enhances radiosensitivity in human osteosarcoma cells. Mol Cell Biochem. 327:257–266. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Wang YH, Xiong J, Wang SF, Yu Y, Wang B, Chen YX, Shi HF and Qiu Y: Lentivirus-mediated shRNA targeting insulin-like growth factor-1 receptor (IGF-1R) enhances chemosensitivity of osteosarcoma cells in vitro and in vivo. Mol Cell Biochem. 341:225–233. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Chen L, Jiang X, Chen H, Han Q, Liu C and Sun M: MicroRNA-628 inhibits the proliferation of acute myeloid leukemia cells by directly targeting IGF-1R. OncoTargets Ther. 12:907–919. 2019. View Article : Google Scholar

32 

Jiang W, Meng L, Xu G, Lv C, Wang H, Tian H, Chen R, Jiao B, Wang B and Huang C: Wentilactone A induces cell apoptosis by targeting AKR1C1 gene via the IGF-1R/IRS1/PI3K/AKT/ Nrf2/FLIP/Caspase-3 signaling pathway in small cell lung cancer. Oncol Lett. 16:6445–6457. 2018.PubMed/NCBI

33 

Huang YK, Kang WM, Ma ZQ, Liu YQ, Zhou L and Yu JC: NUCKS1 promotes gastric cancer cell aggressiveness by upregulating IGF-1R and subsequently activating the PI3K/Akt/mTOR signaling pathway. Carcinogenesis. 40:370–379. 2019. View Article : Google Scholar

34 

Zhang J, Yan YG, Wang C, Zhang SJ, Yu XH and Wang WJ: MicroRNAs in osteosarcoma. Clin Chim Acta. 444:9–17. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Heng L, Jia Z, Bai J, Zhang K, Zhu Y, Ma J, Zhang J and Duan H: Molecular characterization of metastatic osteosar-coma: Differentially expressed genes, transcription factors and microRNAs. Mol Med Rep. 15:2829–2836. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Gurbuz N and Ozpolat B: MicroRNA-based targeted therapeutics in pancreatic cancer. Anticancer Res. 39:529–532. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Henry JC, Azevedo-Pouly AC and Schmittgen TD: MicroRNA replacement therapy for cancer. Pharm Res. 28:3030–3042. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 44 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao X, Li J and Yu D: MicroRNA-939-5p directly targets IGF-1R to inhibit the aggressive phenotypes of osteosarcoma through deactivating the PI3K/Akt pathway Retraction in /10.3892/ijmm.2022.5181. Int J Mol Med 44: 1833-1843, 2019
APA
Zhao, X., Li, J., & Yu, D. (2019). MicroRNA-939-5p directly targets IGF-1R to inhibit the aggressive phenotypes of osteosarcoma through deactivating the PI3K/Akt pathway Retraction in /10.3892/ijmm.2022.5181. International Journal of Molecular Medicine, 44, 1833-1843. https://doi.org/10.3892/ijmm.2019.4333
MLA
Zhao, X., Li, J., Yu, D."MicroRNA-939-5p directly targets IGF-1R to inhibit the aggressive phenotypes of osteosarcoma through deactivating the PI3K/Akt pathway Retraction in /10.3892/ijmm.2022.5181". International Journal of Molecular Medicine 44.5 (2019): 1833-1843.
Chicago
Zhao, X., Li, J., Yu, D."MicroRNA-939-5p directly targets IGF-1R to inhibit the aggressive phenotypes of osteosarcoma through deactivating the PI3K/Akt pathway Retraction in /10.3892/ijmm.2022.5181". International Journal of Molecular Medicine 44, no. 5 (2019): 1833-1843. https://doi.org/10.3892/ijmm.2019.4333