MicroRNA‑93 promotes cell proliferation by directly targeting P21 in osteosarcoma cells

Retraction in: /10.3892/etm.2024.12389

  • Authors:
    • Yu He
    • Bo Yu
  • View Affiliations

  • Published online on: March 8, 2017     https://doi.org/10.3892/etm.2017.4204
  • Pages: 2003-2011
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs) are small, non-coding RNAs that are key regulators of gene expression by directly binding to the 3'‑untranslated region of their target mRNAs, resulting in translational repression or degradation of mRNA. It has been demonstrated that miRNAs have key roles in a variety of human malignancies, including osteosarcoma. The present study aimed to assess the molecular mechanism of miR‑93 in the regulation of osteosarcoma cell proliferation. Reverse‑transcription quantitative PCR and western blot assays were used to examine mRNA and protein expression. An MTT assay and flow cytometry were performed to determine the cell proliferation and cell cycle distribution. A luciferase reporter assay was performed to confirm the direct targeting of cyclin‑dependent kinase inhibitor 1A (CDKN1A), also known as P21, by miR‑93, which was suggested by a bioinformatics analysis. The results showed that the expression of miR‑93 was frequently and significantly increased in a total of 19 osteosarcoma tissues compared to their matched adjacent non‑tumor tissues, and the upregulation of miR‑93 was associated with the malignant progression of osteosarcoma. Furthermore, miR‑93 was also upregulated in the human osteosarcoma cell lines Saos‑2, U2OS, SW1353 and MG63 when compared with that in the human osteoblast cell line hFOB1.19. Transfection with miR‑93 inhibitor significantly reduced the miR‑93 levels and inhibited the proliferation of U2OS and MG63 osteosarcoma cells. The protein levels of P21 were negatively regulated by miR‑93 in U2OS and MG63 cells. Knockdown of miR‑93 caused cell cycle arrest at G1 stage in U2OS and MG63 cells, identical to the effect of P21 overexpression. Finally, P21 was found to be significantly downregulated in osteosarcoma tissues compared to their matched adjacent non‑tumor tissues, suggesting that the inhibition of P21 may be due to increased miR‑93 expression in osteosarcoma tissues. In conclusion, the present study demonstrated that miR‑93 enhances the proliferation of osteosarcoma cells, at least in part via inhibiting P21 expression and thus promoting cell cycle progression.

Introduction

Osteosarcoma is the most common type of malignant bone tumor and mainly affects adolescents and young adults (1,2). Deregulation of oncogenes or tumor suppressors have been found to have crucial roles in the tumorigenesis and malignant progression of osteosarcoma (1,3,4). Accordingly, revealing the exact roles of these oncogenes or tumor suppressors may help in identifying novel therapeutic targets or drug candidates for osteosarcoma.

MicroRNAs (miRNAs) are a class of non-coding small RNAs with 22–25 nucleotides (5,6). It has been well established that miRNAs have key roles in the regulation of gene expression through directly binding to the 3′ untranslated region (UTR) of their target mRNAs, thus leading to translational repression (7). Moreover, various miRNAs have been found to be deregulated in human cancers, and certain specific miRNAs participate in the development and progression of osteosarcoma through regulating the expression of oncogenes or tumor suppressors (8,9). However, the targets of numerous miRNAs in osteosarcoma have remained largely elusive. Investigations on these miRNAs and their target genes are important for identifying novel therapeutic targets for osteosarcoma.

miRNA-93 (miR-93) has been reported to be significantly upregulated and to have a promoting role in several cancer types (10,11). For instance, the expression of miR-93 was found to be significantly increased in esophageal squamous cell carcinoma (12). Moreover, miR-93 can promote the transforming growth factor (TGF)-β-induced epithelial-to-mesenchymal transition through directly targeting neural precursor cell expressed developmentally downregulated gene 4-like in lung cancer cells (13). Recently, miR-93 was found to promote the proliferation of osteosarcoma cells by suppressing the protein expression of the tumor suppressor phosphatase and tensin homolog (14). However, further targets of miR-93 involved in osteosarcoma growth remain to be identified.

Cyclin-dependent kinase inhibitor 1A (CDKN1A) encodes a potent cyclin-dependent kinase inhibitor named P21, which can directly bind to cyclin-cyclin-dependent kinase 2 or cyclin-cyclin-dependent kinase 4 complexes, and further inhibit their activity (15). Therefore, P21 can function as a regulator of cell cycle progression at the G1 checkpoint (15). Recently, P21 was found to be frequently downregulated in osteosarcoma, and upregulation of P21 was shown to inhibit the proliferation of osteosarcoma cells (16,17). However, the mechanisms of the regulation of P21 expression in osteosarcoma cells have remained to be fully elucidated.

The present study aimed to investigate the molecular mechanisms of miR-93 in enhancing osteosarcoma cell proliferation with regard to regulation of P21.

Materials and methods

Tissue collection and ethics statement

The present study was approved by the Ethics Committee of the Second Xiangya Hospital (Central South University, Changsha, China). A total of 19 paired human osteosarcoma tissues and adjacent non-tumor tissues were collected at the Second Xiangya Hospital (Changsha, China) from April 2012 to June 2014, and were histologically confirmed by pathologists. Among these 19 samples, 10 were T1-T2 stage, and 9 were T3-T4 stage (18). Written informed consent was obtained from all patients. None of the patients received any pre-operative radiotherapy or chemotherapy. Tissues were immediately snap-frozen in liquid nitrogen after surgical removal and stored at −80°C prior to analysis.

Cell culture

Human OS cell lines (Saos-2, U2OS, SW1353 and MG63), the hFOB1.19 human osteoblast cell line and the HEK293 human embryonic kidney cell line were obtained from the Cell Bank of Central South University (Changsha, China). All cell lines were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a humidified atmosphere with 5% CO2.

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

Total RNA was extracted using TRIzol Reagent (Life Technologies; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The expression levels of mRNA were determined using the standard SYBR-Green RT-PCR kit (Takara Bio Inc., Dalian, China) on an ABI 7500 PCR system (Thermo Fisher Scientific, Inc.), in accordance with the manufacturer's instructions. GAPDH was used as an internal reference. The reaction conditions were 95°C for 3 min, and 45 cycles of denaturation at 95°C for 15 sec and annealing/elongation at 58°C for 30 sec. The specific primers (Sangon Biotech Co., Ltd., Shanghai, China) as follows: P21 forward, 5′-CGATGGAACTTCGACTTTGTCA-3′ and reverse, 5′-GCACAAGGGTACAAGACAGTG-3′; GAPDH forward, 5′-GGAGCGAGATCCCTCCAAAAT-3′ and reverse, 5′-GGCTGTTGTCATACTTCTCATGG-3′. To determine the expression levels of miRNA, a mirVana™ real-time RT-PCR microRNA detection kit (Life Technologies; Thermo Fisher Scientific, Inc.) was used according to the manufacturer's instructions. U6 was used as an internal reference. The reaction conditions were 95°C for 3 min, and 45 cycles of denaturation at 95°C for 15 sec and annealing/elongation at 58°C for 30 sec. miR-93 (cat. no. miRQP0074) and U6 (cat. no. miRQP9901) primers were purchased from Genecopoeia (Guangzhou, China; sequence not provided). Fold changes were calculated by relative quantification (2−ΔΔCt) (19).

Cell transfection

Negative control (NC) inhibitor (GenePharma, Shanghai, China), miR-93 inhibitor (GenePharma), pcDNA3.1-P21 open reading frame (ORF) plasmid (Amspring, Changsha, China) or pcDNA3.1 vector (NC) was individually diluted with OPTI-MEM (Life Technologies; Thermo Fisher Scientific, Inc.). The diluted Lipofectamine 2000 was then added to each diluted vector. Subsequent to incubation at room temperature for 20 min, each mixture was added to a cell suspension, which was then incubated at 37°C, followed by incubation for 6 h. The transfection mixture was replaced with RPMI-1640 medium with 10% FBS. After transfection for 48 h, the transfected cells were used for subsequent assays.

Western blot analysis

Cells were lysed in cold radioimmunoprecipitation buffer (Sigma-Aldrich; Merck-Millipore, Darmstadt, Germany) and the protein concentration was determined using the BCA Protein Assay kit (Pierce; Thermo Fisher Scientific, Inc.). Protein was separated by 10% SDS-PAGE, transferred to a polyvinylidene difluoride (PVDF) membrane (Life Technologies; Thermo Fisher Scientific, Inc.), and then blocked in 5% powdered milk dissolved in PBS (Life Technologies; Thermo Fisher Scientific, Inc.) containing 0.1% Tween-20 (Sigma-Aldrich; Merck-Millipore). Then the PVDF membrane was incubated with monoclonal mouse anti-human P21 (1:200 dilution; ab7903) or monoclonal mouse anti-human GAPDH (1:200 dilution; ab8245) primary antibodies (both Abcam, Cambridge, MA, USA) for 3 h at room temperature and then washed with PBS for 10 min. The PVDF membrane was then incubated with the rabbit anti-mouse secondary antibody (1:5,000 dilution; ab190475; Abcam) for 1 h at room temperature, and then washed 3 times with Tris-buffered saline containing Tween 20. The immune complexes were then detected using the ECL Western Blotting kit (Pierce; Thermo Fisher Scientific, Inc.) and X-ray film (Eastman Kodak, Rochester, NY, USA). ImageJ software (version 1.25; National Institutes of Health, Bethesda, MD, USA) was used to analyze the relative protein expression, represented as the density ratio vs. GAPDH.

Luciferase reporter assay

Targetscan software 3.1 (www.targetscan.org) was used to predict the putative target genes of miR-93. The predicted miR-93 binding sites in the wild-type (WT) 3′UTR of P21 were cloned into the pGL3 vector (Promega Corp., Madison, WI, USA), named as pGL3-P21-WT-3′UTR. The mutant type (MT) miR-93 binding sites in the 3′UTR of P21 (Yearthbio, Changsha, China) were constructed by using the Quick-Change Site-Directed Mutagenesis kit (Stratagene, La Jolla, CA, USA), in accordance with the manufacturer's protocol, which was also inserted into the pGL3 vector (Promega Corp.), named as pGL3-MUT-P21-3′UTR. HEK293 cells were seeded in 96-well plates and co-transfected with 300 ng pGL3-WT-P21-3′UTR or pGL3-MUT-P21-3′UTR, and 100 nM of miR-NC or miR-93 mimics, using Lipofectamine 2000, in accordance with the manufacturer's protocol. The dual-luciferase reporter assay system (Promega Corp.) was used to determine the activities of Renilla luciferase and firefly luciferase after co-transfection for 48 h. The firefly luciferase activity was normalized to Renilla luciferase activity.

Cell proliferation assay

Cells (104) were plated into a 96-well plate and cultured at 37°C with 5% CO2 for 0, 12, 24, 48 or 72 h. Subsequently, 20 µl MTT (5 mg/ml; Life Technologies; Thermo Fisher Scientific, Inc.) was added, followed by incubation at 37°C for 4 h. A total of 150 µl DMSO was then added, followed by incubation at room temperature for 10 min. The optical density at 570 nm was determined using the XT-96DJ ELISA analyzer (Safeda Technology, Beijing, China).

Cell cycle analysis

Cells (1×106) were washed twice with DPBS (Thermo Fisher Scientific, Inc.), resuspended in 70% ethanol, and fixed overnight at −20°C. Cells were subsequently washed twice in DPBS with 3% bovine serum albumin (BSA; Thermo Fisher Scientific, Inc.) and incubated for 30 min at room temperature in propidium iodide (PI) staining buffer containing 3% BSA, 40 µg/ml PI (Yearthbio), and 0.2 mg/ml RNase (Thermo Fisher Scientific, Inc.) in DPBS. DNA content analyses were carried out using a flow cytometer (C6; BD Biosciences, Franklin Lakes, NJ, USA). Accuri C6 software was used for analysis (version 1; BD Biosciences).

Statistical analysis

Values are expressed as the mean ± standard error of the mean. Differences between groups were analyzed using Student's t-test for 2-group comparisons and one-way analysis of variance for multiple-group comparisons using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference between groups.

Results

miR-93 is upregulated in osteosarcoma

To reveal the exact role of miR-93 in osteosarcoma, RT-qPCR analysis was used to examine the miR-93 levels in a total of 19 paired of osteosarcoma tissues and adjacent non-tumor tissues. It was found that the expression of miR-93 was frequently and significantly increased in osteosarcoma tissues compared to that in their matched non-tumor tissues (Fig. 1A and B). Moreover, the osteosarcoma tissues of stage T3-T4 showed higher miR-93 levels when compared to those in the osteosarcoma of the T1-T2 stage (Fig. 1C), suggesting that high expression of miR-93 was associated with the malignant progression of osteosarcoma.

In addition, the miR-93 levels in several common human osteosarcoma cell lines, namely Saos-2, U2OS, SW1353 and MG63, were determined. The results showed that miR-93 was also upregulated in osteosarcoma cell lines compared to the normal human osteoblast hFOB1.19 cells (Fig. 1D). Accordingly, miR-93 was significantly upregulated in osteosarcoma.

Knockdown of miR-93 inhibits the proliferation of osteosarcoma cells

As miR-93 was upregulated in osteosarcoma cell lines, U2OS and MG63 cells were further transfected with miR-93 inhibitor. RT-qPCR indicated that transfection with miR-93 inhibitor significantly reduced the miR-93 levels in U2OS and MG63 cells compared to those in the control group, while transfection with the NC inhibitor showed no effect on its expression (Fig. 2A and B). An MTT assay was then performed to determine the cell proliferation. Knockdown of miR-93 was found to significantly suppress the proliferation of U2OS and MG63 cells when compared to that in the control group (Fig. 2C and D). Therefore, knockdown of miR-93 inhibited the proliferation of osteosarcoma cells.

As cell cycle progression is responsible for cell proliferation, flow cytometry was performed to examine the cell cycle distribution in U2OS and MG63 cells with or without transfection with miR-93 inhibitor. The results indicated that knockdown of miR-93 led to a significant cell cycle arrest at the G1 stage in U2OS and MG63 cells (Fig. 2E and F), which probably contributed to the inhibitory effect of miR-93 knockdown on osteosarcoma cell proliferation.

miR-93 directly targets P21 in osteosarcoma cells

A bioinformatics analysis was performed to investigate the potential target genes of miR-93 involved in the regulation of cell cycle progression. Analysis with Targetscan software indicated that P21, a CDKN, is a putative target gene of miR-93 (Fig. 3A). Therefore, a luciferase reporter assay was performed to confirm whether P21 was a direct target of miR-93. The WT or MT miR-93-binding sequence in the 3′UTR of P21 was respectively subcloned into the pGL3 vector downstream of the firefly luciferase reporter gene (Fig. 3B and C). Subsequently, HEK293 cells were co-transfected with pGL3-WT-P21-3′UTR or pGL3-MT-P21-3′UTR, and miR-93 mimics or miR-NC, respectively. After transfection for 48 h, the luciferase activity in HEK293 cells co-transfected with pGL3-WT-P21-3′UTR and miR-93 mimics was significantly decreased compared to that in the control group. However, the luciferase activity in cells co-transfected with pGL3-MT-P21-3′UTR and miR-93 mimics showed no difference from the control group (Fig. 3D), indicating that the P21 3′UTR indeed contained a binding sites for miR-93.

As miRNAs generally inhibit the expression of their target genes, the effect of miR-93 on the expression of P21 at the mRNA and protein level was then examined. RT-qPCR showed that knockdown of miR-93 had no effect on the mRNA levels of P21 in U2OS and MG63 cells (Fig. 4A and B). However, western blot analysis indicated that U2OS and MG63 cells showed a higher protein level of P21 after transfection with miR-93 inhibitor (Fig. 4C and D), indicating that miR-93 negatively regulates the expression of P21 at the post-transcriptional level in osteosarcoma cells.

P21 is involved in the miR-93-mediated proliferation of osteosarcoma cells

As knockdown of miR-93 caused an upregulation of P21, as well as a cell cycle arrest at G1 stage in osteosarcoma cells, it was speculated that P21 may act as a downstream effector in miR-93-mediated osteosarcoma cell proliferation. Thus, U2OS and MG63 cells were transfected with the pcDNA3.1-P21 ORF plasmid. After transfection, the mRNA and protein expression of P21 was markedly increased compared to that in the control group; however, transfection with the blank pcDNA3.1 vector did not affect the P21 expression in U2OS and MG63 cells (Fig. 5A and B). MTT assay data further indicated that overexpression of P21 also suppressed the proliferation of U2OS and MG63 cells, identical to the effect of miR-93 knockdown (Fig. 5C and D). Moreover, flow cytometry data showed that P21 upregulation indeed induced cell cycle arrest at the G1 stage in osteosarcoma cells, similar to the effect of miR-93 knockdown (Fig. 5E and F). Accordingly, the results suggested that P21 is indeed involved in the miR-93-mediated proliferation of osteosarcoma cells.

P21 is downregulated in osteosarcoma

Finally, the expression of P21 in human osteosarcoma tissues and cell lines was examined. RT-qPCR data indicated that the mRNA expression of P21 was significantly decreased in osteosarcoma tissues compared to that in their matched adjacent non-tumor tissues (Fig. 6A). In contrast to miR-93, the osteosarcoma of T3-T4 stage showed a lower P21 expression level when compared to that in the osteosarcoma of the T1-T2 stage (Fig. 6B), suggesting that the decreased expression of P21 was associated with the malignant progression of osteosarcoma. In addition, the mRNA and protein expression of P21 was also significantly reduced in osteosarcoma cell lines when compared to that in the normal human osteoblast hFOB1.19 cells (Fig. 6C and D). Therefore, it was demonstrated that the expression of P21 is decreased in osteosarcoma, suggesting that the downregulation of P21 may be due to the upregulation of miR-93 in osteosarcoma tissues and cell lines.

Discussion

miRNAs have been found to have key roles in the tumorigenesis and malignant progression of osteosarcoma (4,20,21). However, the underlying mechanisms remain to be fully investigated. The present study aimed to reveal the role and molecular mechanisms of miR-93 in the regulation of osteosarcoma cell proliferation. It was found that the expression of miR-93 was significantly increased in 19 osteosarcoma tissues compared to that in their matched adjacent non-tumor tissues, and its levels were higher in osteosarcoma tissues with advanced stage. Besides, miR-93 was also upregulated in the human osteosarcoma cell lines Saos-2, U2OS, SW1353 and MG63 when compared with that in the hFOB1.19 human osteoblast cell line. In vitro experiments indicated that knockdown of miR-93 inhibited U2OS and MG63 cell proliferation, accompanied with a cell cycle arrest at G1 stage. P21 was further identified as a direct target of miR-93, and was involved in the miR-93-mediated osteosarcoma cell proliferation. In addition, it was found that P21 was significantly downregulated in osteosarcoma tissues compared to that in their matched adjacent non-tumor tissues, suggesting that the inhibition of P21 may be due to the increased miR-93 expression in osteosarcoma tissues.

miR-93 is a member of the miR-106b-25 cluster, which includes miR-106b, miR-93 and miR-25, and also a paralog of members of the miR-17-92 cluster (22). In recent years, miR-93 has been found to be deregulated and to generally have a promoting role in certain human cancer types (23,24). For instance, miR-93 can directly inhibit the expression of the tumor suppressor gene FUS1, and is thus involved in the development of lung cancer (25). Besides, miR-93 enhances angiogenesis and metastasis by targeting large tumor suppressor kinase 2 (26). The present study found that the expression of miR-93 was significantly increased in osteosarcoma tissues and cell lines, and suggested that the upregulation of miR-93 was associated with the malignant progression of osteosarcoma. Therefore, miR-93 may have an oncogenic role in osteosarcoma. To verify this speculation, U2OS and MG63 cells were transfected with miR-93 inhibitor, which caused a marked decrease in miR-93 levels and suppressed the proliferation of U2OS and MG63 cells. Recently, Kawano et al (14) also reported that miR-93 promoted the proliferation of osteosarcoma cells, consistent with the present findings.

The present study further investigated the underlying mechanism by which knockdown of miR-93 inhibited the proliferation of osteosarcoma cells. It was shown that miR-93 knockdown caused cell cycle arrest at G1 stage, which contributed to the reduced cell proliferation. As miRNAs mainly function through directly suppressing the translation of their target genes (27), the potential target genes of miR-93 in osteosarcoma were then examined. Among the putative target genes of miR-93, P21 is a key regulator of cell cycle progression at the G1 checkpoint (28). In addition, P21 can also interact with proliferating cell nuclear antigen, a DNA polymerase accessory factor, and has a regulatory role in S phase DNA replication and DNA damage repair (29). To clarify whether P21 is a direct target of miR-93, a luciferase reporter assay was performed, which demonstrated that miR-93 directly bound to the 3′UTR of the P21 gene. Further investigation indicated that miR-93 knockdown increased the protein expression of P21 in U2OS and MG63 cells, and overexpression of P21 also suppressed the proliferation of U2OS and MG63 cells, as did knockdown of miR-93. Therefore, it is suggested that P21 acts as a downstream effector of miR-93 in the regulation of osteosarcoma cell proliferation. In contrast to miR-93, the expression levels of P21 were found to be significantly reduced in osteosarcoma tissues and cell lines. Moreover, its expression levels were lower in osteosarcoma tissues of T3-T4 stage when compared with that in osteosarcoma tissues of T1-T2 stage. Therefore, the results suggested that the downregulation of P21 is at least partly due to the upregulation of miR-93, which is associated with the malignant progression of osteosarcoma.

The targeting association between miR-93 and P21 has also been found in other cell types. For instance, Jiang et al (30) found that miR-93 promoted the proliferation of ovarian granulosa cells through directly targeting P21 in polycystic ovarian syndrome. Petrocca et al (31) demonstrated that miR-93 impaired TGF-β-mediated tumor inhibition by directly targeting P21 and Bim. Moreover, Kim et al (32) reported that miR-93 inhibited the expression of P21 and ectopic expression of miR-93 led to activation of CDK2 and facilitation of G1/S phase transition in gastric cancer cells. Therefore, the present study expanded the current understanding of the role of the miR-93/P21 axis in human cancers.

In conclusion, the present study demonstrated that miR-93 has a promoting role in the proliferation of osteosarcoma cells, at least partly via inhibiting P21 expression and facilitating cell cycle progression. Therefore, miR-93 may become a potential therapeutic target for osteosarcoma treatment.

References

1 

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

2 

Valery PC, Laversanne M and Bray F: Bone cancer incidence by morphological subtype: A global assessment. Cancer Causes Control. 26:1127–1139. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Debebe Z and Rathmell WK: Ror2 as a therapeutic target in cancer. Pharmacol Ther. 150:143–148. 2015. View Article : Google Scholar : PubMed/NCBI

4 

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

5 

Ambros V: microRNAs: Tiny regulators with great potential. Cell. 107:823–826. 2001. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

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

8 

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

9 

Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Fang L, Deng Z, Shatseva T, Yang J, Peng C, Du WW, Yee AJ, Ang LC, He C, Shan SW and Yang BB: MicroRNA miR-93 promotes tumor growth and angiogenesis by targeting integrin-β8. Oncogene. 30:806–821. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Ohta K, Hoshino H, Wang J, Ono S, Iida Y, Hata K, Huang SK, Colquhoun S and Hoon DS: MicroRNA-93 activates c-Met/PI3K/Akt pathway activity in hepatocellular carcinoma by directly inhibiting PTEN and CDKN1A. Oncotarget. 6:3211–3224. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Ansari MH, Irani S, Edalat H, Amin R and Roushandeh Mohammadi A: Deregulation of miR-93 and miR-143 in human esophageal cancer. Tumour Biol. 37:3097–3103. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Qu MH, Han C, Srivastava AK, Cul T, Zou N, Gao ZQ and Wang QE: miR-93 promotes TGF-β-induced epithelial-to-mesenchymal transition through downregulation of NEDD4L in lung cancer cells. Tumour Biol. 37:5645–5651. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Kawano M, Tanaka K, Itonaga I, Ikeda S, Iwasaki T and Tsumura H: microRNA-93 promotes cell proliferation via targeting of PTEN in osteosarcoma cells. J Exp Clin Cancer Res. 34:762015. View Article : Google Scholar : PubMed/NCBI

15 

Dutto I, Tillhon M, Cazzalini O, Stivala LA and Prosperi E: Biology of the cell cycle inhibitor p21 (CDKN1A): Molecular mechanisms and relevance in chemical toxicology. Arch Toxicol. 89:155–178. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Lu M, Huang W, Bao N, Zhou G and Zhao J: The flavonoid ampelopsin inhibited cell growth and induced apoptosis and G0/G1 arrest in human osteosarcoma MG-63 cells in vitro. Pharmazie. 70:388–393. 2015.PubMed/NCBI

17 

Chen X, Deng M, Ma L, Zhou J, Xiao Y, Zhou X, Zhang C and Wu M: Inhibitory effects of forkhead box L1 gene on osteosarcoma growth through the induction of cell cycle arrest and apoptosis. Oncol Rep. 34:265–271. 2015.PubMed/NCBI

18 

Lindsey BA, Markel JE and Kleinerman ES: Osteosarcoma overview. Rheumatol Ther. Dec 8–2016.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

19 

Rao X, Huang X, Zhou Z and Lin X: An improvement of the 2^(−delta delta CT) method for quantitative real-time polymerase chain reaction data analysis. Biostat Bioinforma Biomath. 3:71–85. 2013.PubMed/NCBI

20 

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

21 

Liu X, Liu Y, Wu S, Shi X, Li L, Zhao J and Xu H: Tumor-suppressing effects of miR-429 on human osteosarcoma. Cell Biochem Biophys. 70:215–224. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Li Y, Tan W, Neo TW, Aung MO, Wasser S, Lim SG and Tan TM: Role of the miR-106b-25 microRNA cluster in hepatocellular carcinoma. Cancer Sci. 100:1234–1242. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Jiang L, Wang C, Lei F, Zhang L, Zhang X, Liu A, Wu G, Zhu J and Song L: miR-93 promotes cell proliferation in gliomas through activation of PI3K/Akt signaling pathway. Oncotarget. 6:8286–8299. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Li G, Ren S, Su Z, Liu C, Deng T, Huang D, Tian Y, Qiu Y and Liu Y: Increased expression of miR-93 is associated with poor prognosis in head and neck squamous cell carcinoma. Tumour Biol. 36:3949–3956. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Du L, Schageman JJ, Subauste MC, Saber B, Hammond SM, Prudkin L, Wistuba II, Ji L, Roth JA, Minna JD and Pertsemlidis A: miR-93, miR-98, and miR-197 regulate expression of tumor suppressor gene FUS1. Mol Cancer Res. 7:1234–1243. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Fang L, Du WW, Yang W, Rutnam ZJ, Peng C, Li H, O'Malley YQ, Askeland RW, Sugg S, Liu M, et al: MiR-93 enhances angiogenesis and metastasis by targeting LATS2. Cell Cycle. 11:4352–4365. 2012. View Article : Google Scholar : PubMed/NCBI

27 

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

28 

Ohkoshi S, Yano M and Matsuda Y: Oncogenic role of p21 in hepatocarcinogenesis suggests a new treatment strategy. World J Gastroenterol. 21:12150–12156. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Strzalka W and Ziemienowicz A: Proliferating cell nuclear antigen (PCNA): A key factor in DNA replication and cell cycle regulation. Ann Bot. 107:1127–1140. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Jiang L, Huang J, Li L, Chen Y, Chen X, Zhao X and Yang D: MicroRNA-93 promotes ovarian granulosa cells proliferation through targeting CDKN1A in polycystic ovarian syndrome. J Clin Endocrinol Metab. 100:E729–E738. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Petrocca F, Visone R, Onelli MR, Shah MH, Nicoloso MS, de Martino I, Iliopoulos D, Pilozzi E, Liu CG, Negrini M, et al: E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell. 13:272–286. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Kim YK, Yu J, Han TS, Park SY, Namkoong B, Kim DH, Hur K, Yoo MW, Lee HJ, Yang HK and Kim VN: Functional links between clustered microRNAs: Suppression of cell-cycle inhibitors by microRNA clusters in gastric cancer. Nucleic Acids Res. 37:1672–1681. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

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
He Y and He Y: MicroRNA‑93 promotes cell proliferation by directly targeting P21 in osteosarcoma cells Retraction in /10.3892/etm.2024.12389. Exp Ther Med 13: 2003-2011, 2017
APA
He, Y., & He, Y. (2017). MicroRNA‑93 promotes cell proliferation by directly targeting P21 in osteosarcoma cells Retraction in /10.3892/etm.2024.12389. Experimental and Therapeutic Medicine, 13, 2003-2011. https://doi.org/10.3892/etm.2017.4204
MLA
He, Y., Yu, B."MicroRNA‑93 promotes cell proliferation by directly targeting P21 in osteosarcoma cells Retraction in /10.3892/etm.2024.12389". Experimental and Therapeutic Medicine 13.5 (2017): 2003-2011.
Chicago
He, Y., Yu, B."MicroRNA‑93 promotes cell proliferation by directly targeting P21 in osteosarcoma cells Retraction in /10.3892/etm.2024.12389". Experimental and Therapeutic Medicine 13, no. 5 (2017): 2003-2011. https://doi.org/10.3892/etm.2017.4204