Open Access

Inhibition of long non‑coding RNA‑AWPPH decreases osteosarcoma cell proliferation, migration and invasion

  • Authors:
    • Wenbin Ding
    • Dajiang Wu
    • Fang Ji
    • Hao Zhang
  • View Affiliations

  • Published online on: September 20, 2019     https://doi.org/10.3892/ol.2019.10898
  • Pages: 5055-5062
  • Copyright: © Ding 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

Long non‑coding RNAs (lncRNAs) serve a crucial role in various types of cancer. The lncRNA AWPPH has been reported to promote hepatocellular carcinoma and bladder cancer progression. However, to the best of our knowledge, the biological roles of AWPPH in osteosarcoma (OS) remain unclear. In the present study, the levels of AWPPH in OS tissues and cell lines were determined by reverse transcription‑quantitative polymerase chain reaction. An MTT assay was used to detect OS cell proliferation. The levels of proteins associated with the PI3K/Akt signaling pathway and apoptosis were determined by western blotting. Wound‑healing and Transwell assays were conducted to determine cell migration and invasion, respectively. The results demonstrated that AWPPH was highly expressed in OS tissues and cells. Functional analyses revealed that AWPPH depletion significantly inhibited OS cell proliferation and migration, and promoted OS cell apoptosis. Furthermore, AWPPH downregulation significantly inhibited the PI3K/AKT pathway. The present study demonstrated that AWPPH was highly expressed in OS, and that AWPPH promoted OS cell proliferation and migration, and inhibited OS cell apoptosis, which may be mediated by PI3K/AKT pathway activation.

Introduction

Osteosarcoma (OS) is a bone malignancy that primarily affects adolescents and young adults. OS accounts for ~20% of all primary bone cancer cases in Europe and is the second highest cause of cancer-associated mortality in children (1). Despite advances in medical treatments, including surgery, transplantation and radiation, effective treatments for postsurgical recurrence and metastasis are not currently available, thus accounting for the poor patient outcome for OS (2). Although therapy for OS has improved, the overall survival rate of patients with OS has not substantially increased, and 35% of patients with OS die within 5 years of diagnosis (3). Furthermore, >20% of young patients with OS present with distant metastases at the time of diagnosis, and 40% of advanced stage cases progress to metastasis during therapy (4,5). To the best of our knowledge, the underlying molecular mechanism of metastasis in patients with OS has not yet been elucidated (6,7). It is therefore crucial to identify novel biomarkers and therapeutic strategies for the better management of OS.

Long non-coding RNAs (lncRNAs) are polyadenylated RNA polymerase II-transcribed RNAs that are ≥200 nucleotides in length and do not present obvious open reading frames to encode proteins (810). An increasing number of studies have demonstrated that lncRNAs serve crucial roles in tumor initiation, progression, metastasis, drug-resistance and recurrence (1113). A previous study reported that epigenetic activation of the lncRNA-maternally expressed gene 3 (MEG3) and/or inactivation of the mesenchymal-epithelial transition factor could represent a therapeutic strategy in pancreatic neuroendocrine tumor and insulinoma treatment (14). lncRNAs affect a variety of biological processes, including cellular proliferation, differentiation, migration, the immune response and apoptosis, which are all associated with tumorigenesis (15,16). However, the functional roles of lncRNAs remain commonly unknown. lncRNAs have been found to act as tumor suppressors or oncogenes (1719). It has been reported that Ewing sarcoma-associated transcript 1 (EWSAT1) facilitates the growth and metastasis of OS cells via MEG3 regulation, which suggests that the EWSAT1-MEG3 axis may represent a promising target for OS treatment (20,21). A meta-analysis indicated that MEG3 may be considered as a potential novel biomarker for the clinical outcome of certain types of human cancer (22), including bladder cancer (23), cervical carcinoma (24), hepatocellular cancers (25) and meningiomas (26). AWPPH is a novel IncRNA that has been reported to be involved in the development of several types of cancer and to have oncogenic functions (27,28). For examples, a recent study reported that AWPPH is overexpressed in hepatoma cells, where it can promote proliferation and migration of liver cancer cells, and stimulate the growth and metastasis of tumor cells by interacting with Y-box binding protein 1 (YBX1) (29). However, it remains unclear whether AWPPH could be considered as a critical regulator in OS that could predict patient prognosis.

The present study aimed to evaluate the effects of AWPPH on OS cell proliferation, migration and apoptosis. Furthermore, this study will investigate the role of AWPPH on the regulation of phosphorylated (p)-PI3K, p-AKT, Bcl-2 and cleaved-caspase-3 expressions, in order to determine the underlying mechanisms of AWPPH in OS.

Materials and methods

Patients

A total of 30 pairs of OS tissues and paracancerous tissues were obtained from 30 patients (17 men and 13 women) undergoing tumor resection at the Changhai Hospital (Shanghai, China) between April 2010 and October 2017. The average age of the patients was 22.34 years (age range, 10–59 years). All tissues (4–8 cm thickness) were immediately snap-frozen in liquid nitrogen and stored at −80°C until further use. Written informed consent was obtained from all patients and the study protocol was approved by the Ethics Committee of the Changhai Hospital.

Cells and reagents

The OS MG-63 and U2OS, and the osteoblast hFOB1.19 cell lines were purchased from the American Type Culture Collection (ATCC). Fetal bovine serum (FBS), Dulbecco's modified Eagle's medium (DMEM) and the MTT assay were purchased from Sigma-Aldrich; Merck KGaA. Penicillin, streptomycin, glutamine and TRIzol® were obtained from Invitrogen; Thermo Fisher Scientific, Inc. Antibodies against p-PI3K (cat. no. 4228S), PI3K (cat. no. 4249S), p-Akt (cat. no. 4060S), Akt (cat. no. 9272S), Bcl-2 (cat. no. 15071S), Bax (cat. no. 2774) cleaved-caspase-3 (cat. no. 9661S), caspase-3 (cat. no. 9662), cleaved-caspase-9 (cat. no. 20750) and caspase-9 (cat. no. 9508) were obtained from Cell Signaling Technology, Inc. and used at 1:500 dilution according to the manufacturer's instructions. Antibody against GAPDH, (cat. no. sc-32233); mouse anti-rabbit IgG-HRP antibody, (cat. no. sc-2357) and m-IgGκ BP-HRP antibody (cat. no. sc-516102) were purchased from Santa Cruz Biotechnology, Inc. and used at 1:5,000 dilution.

Cell culture

The MG-63 and U2OS cell lines were cultured in DMEM supplemented with 10% FBS, 100 U/ml penicillin, 100 mg/ml streptomycin and 2 mM glutamine, and placed at 37°C in a humidified incubator containing 5% CO2.

The hFOB1.19 cell line was cultured as previously described (30) and according to ATCC procedures. For in vitro proliferation, the hFOB1.19 cells were maintained in a non-differentiation medium that consisted of 1:1 DMEM/F-12 (GE Healthcare Life Sciences) containing 10% FBS (Sigma-Aldrich; Merck KGaA) and 0.3 mg/l G418 (Sigma-Aldrich; Merck KGaA), and cultured in a humidified incubator with 5% CO2 at 34°C. The medium was replaced every 2–3 days.

Short hairpin (sh)RNA transfection

The AWPPH shRNA used in the MG-63 and U2OS cell lines was purchased from Guangzhou RiboBio Co., Ltd. Cells were transfected using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The sequence of the AWPPH shRNA was 5′-CTGGATGGTCGCTGCTTTTTA-3′. AWPPH shRNA (100 nM) was inserted into the shRNA expression vector pGPH1/Neo (40 nM; Clontech Laboratories, Inc.) and packaged using Platinum-A (Cell Biolabs, Inc.). Cells (1.2×106) were transfected with AWPPH shRNA or scrambled shControl (5′-UUUCCGAACGUGUCACGUdTdT-3′) for 48 h prior to further experimentation.

MTT assay

The effect of AWPPH downregulation on MG63 and U2OS cell proliferation was measured by MTT assay. Cells (1×104/100 µl) were seeded in 96-well plates. MTT (50 µl) was added and the cells were incubated for 4 h. Subsequently, 150 µl DMSO was added to dissolve the purple formazan, and the absorbance was read at 570 nm with a microplate reader (Thermo Fisher Scientific, Inc.).

RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA from OS tissues, the two OS cell lines MG-63 and U2OS and the human osteoblast hFOB 1.19 cell line was isolated with TRIzol® according to the manufacturer's instructions. Total RNA concentration was determined with photometric method using a NanoQuant plate (Tecan Group, Ltd.). RNA (1 ng) was reverse transcribed using the Reverse EasyScript One-Step gDNA Removal and cDNA Synthesis SuperMix (Trangene) according to the manufacturer's instructions. RT-qPCR was performed using SYBR-Green Master mix (Roche Life Science) in an ABI 7500 Real-time PCR instrument according to the following reactions: Denaturation for 1 cycle at 95°C for 1 min, followed by 40 cycles of 20 sec at 95°C and 40 sec at 58°C. GAPDH was used as an endogenous control. The primers used were designed as follows: AWPPH forward, 5′-CTGGATGGTCGCTGCTTTTTA-3′ and reverse, 5′-AGGGGGATGAGTCGTGATTT-3′; and GAPDH forward, 5′-GGAGCGAGATCCCTCCAAAAT-3′ and reverse, 5′-GGCTGTTGTCATACTTCTCATGG-3′. The relative expression levels were normalized to endogenous controls and assessed using the 2−ΔΔCq method (31).

Cell migration and invasion assays

For the migration assay, 1×105 MG-63 and U2OS cells were seeded in six-well plates and cultured at 37°C overnight. A linear wound was carefully made with fine pipette-tips in the middle of the well, cell debris was removed and the cells were incubated with serum-free medium. The wounded monolayers were photographed at 0 and 24 h after wounding using an inverted microscope (magnification, 40×; Carl Zeiss AG).

For the invasion assay, 3×105 MG-63 and U2OS cells were seeded in FBS-free DMEM in the upper chamber of a 24-well Matrigel-coated Transwell invasion insert (diameter, 6.4 mm; pore size, 8 µm; BD Biosciences). The lower chamber was filled with DMEM supplemented with 20% FBS. Following 24 h of incubation, the cells in the upper chamber were removed, and the cells that had invaded the membrane were stained using crystal violet for 20 min at room temperature. Five randomly selected fields were captured with an upright optical microscope (magnification, ×10; Leica Microsystems GmbH) and the migrated cells were counted. All experiments were performed in triplicate at least three times.

Western blotting

MG-63 and U2OS cells were collected, and the total protein was extracted using RIPA lysis buffer (cat. no. P0013K; Beyotime Institute of Biotechnology) containing 1 mmol/l phenylmethylsulfonyl fluoride protease inhibitor (cat. no. ST506; Beyotime Institute of Biotechnology). Protein concentration of cell lysates was determined using a Bradford Protein Assay Kit (MultiSciences Biotech Co., Ltd.). Proteins (40 µg) were separated by 12% SDS-PAGE and transferred to polyvinylidene difluoride membranes. Membranes were incubated with antibodies against p-PI3K, PI3K, p-Akt, Akt, Bcl-2, Bax, cleaved-caspase-3, caspase-3, cleaved-caspase-9, caspase-9 and GAPDH at 4°C overnight. Membranes were washed with PBST and incubated with secondary antibodies for 1 h at room temperature. Enhanced chemiluminescence reagent (Thermo Fisher Scientific Inc.) was used to detect the signal on the membrane. Relative expression level of proteins was normalized to endogenous control GAPDH using Image-Pro Plus 6.0 (Media Cybernetics Inc.).

Statistical analysis

P-values were calculated using Student's t-test or one-way analysis of variance with SPSS software version 19.0 (IBM Corp.). All data are expressed as the mean ± standard error of the mean. P<0.05 was considered to indicate a statistically significant difference.

Results

AWPPH is highly expressed in OS tissues and cells

The expression levels of AWPPH in OS tissues, in the two OS cell lines MG-63 and U2OS, and in the human osteoblast hFOB 1.19 cell line were detected by RT-qPCR. As presented in Fig. 1A, AWPPH expression levels were significantly elevated in OS tissues compared with those in non-cancerous matched bone tissues. In addition, the expression levels of AWPPH were significantly increased in the MG-63 and U2OS cell lines (Fig. 1B) compared with those in hFOB1.19 cells. In order to investigate the biological roles of AWPPH in OS cells, the MG-63 and U2OS cell lines were stably depleted of AWPPH using AWPPH-specific shRNA. RT-qPCR analysis reported that transfection with AWPPH shRNA effectively decreased AWPPH mRNA expression in the MG-63 and U2OS cells, compared with that in the control and shControl groups (Fig. 1C and D).

AWPPH downregulation inhibits OS cell proliferation, migration and invasion

As presented in Fig. 2A and B, MG63 and U2OS cell proliferation was significantly decreased following 48 and 72 h of transfection with AWPPH shRNA, compared with the negative control. The wound-healing assay demonstrated that AWPPH depletion decreased the migration capacity of the MG-63 (Fig. 2C) and U2OS (Fig. 2D) cells. In addition, the Transwell assay revealed that AWPPH depletion significantly inhibited MG-63 and U2OS cell invasion (Fig. 2E). These data suggested that AWPPH may serve a crucial role in OS cell proliferation, migration and invasion.

AWPPH downregulation promotes OS cell apoptosis-associated factors

In order to investigate whether cell apoptosis contributes to the decrease in cell viability, western blotting was used to detect cell apoptosis-associated proteins. The results demonstrated that cleaved-caspase-3, cleaved-caspase-9 and Bax expressions were significantly increased, whereas Bcl-2 expression was significantly decreased in the MG-63 and U2OS cells following transfection with AWPPH shRNA (Fig. 3A and B).

AWPPH downregulation inhibits the PI3K/AKT pathway

As presented in Fig. 4A and B, western blotting demonstrated that AWPPH depletion significantly decreased the protein levels of p-PI3K and p-AKT in the MG-63 and U2OS cells. These results suggested that AWPPH may partly function by activating the PI3K/AKT pathway.

Discussion

OS is the most frequent type of primary bone cancer, with an incidence of 0.2–3/100,000 per year (32,33). An increasing number of studies have suggested that the aberrant expression of lncRNAs is a potential driver event of tumorigenesis and cancer progression (18,34,35). In the present study, the AWPPH expression level was significantly increased in OS tissues and cells compared with that in control groups. Furthermore, AWPPH depletion decreased OS cell proliferation, migration and invasion, promoted OS cell apoptosis and inhibited the PI3K/AKT pathway. These results demonstrated that AWPPH may be considered as a potential prognostic biomarker for OS.

A recent study reported that AWPPH is highly expressed in hepatocellular carcinoma (HCC) tissues, that it promotes HCC cell proliferation and migration in vitro, and that it promotes tumor growth and metastasis in vivo through YBX1 (29). In addition, AWPPH is highly expressed in bladder cancer (BC) and promotes cell proliferation, autophagy and migration, and inhibits cell apoptosis in BC by inhibiting SMAD family member 4 via enhancer of zeste 2 polycomb repressive complex 2 subunit (27). A previous study reported that increased expression of AWPPH is correlated with incomplete encapsulation, microvascular invasion, and advanced Tumor-Node-Metastasis stage and Barcelona clinic liver cancer stage in HCC (29). Additionally, survival analysis and Cox proportional hazards regression analysis revealed not only a correlation between AWPPH and poor recurrence-free survival and overall survival, but the fact that it is an independent prognostic factor of these measurements too (29). However, to the best of our knowledge, there are currently no reagents that target AWPPH and no studies or clinical tests on AWPHH expression in patients with OS, and the expression and role of AWPPH in OS therefore remain unknown. The present study demonstrated that AWPPH expression level was higher in OS cells compared with that in the osteoblast hFOB1.19 cell line. These results were consistent with those observed for AWPPH expression in certain subtypes of HCC and BC (19,22). In the present study, the role of AWPPH in OS carcinogenesis and the one of the potential mechanisms underlying AWPPH function were elucidated via AWPPH-knockdown. MTT assay confirmed that AWPPH-knockdown decreased OS cell proliferation. In addition, the results from the wound-healing and Transwell assays demonstrated that AWPPH depletion suppressed OS cell migration and invasion. Furthermore, western blotting analysis indicated that AWPPH-knockdown induced cell apoptosis by regulating Bcl-2, Bax, cleaved-caspase-3 and cleaved-caspase-9 protein expressions. These results indicated that AWPPH may have an oncogenic role in OS. Further investigation using a larger clinical sample size is required to validate the expression of AWPPH in OS and to determine its potential prognostic value.

Disease recurrence and metastasis occurrence frequently result in the poor outcome of patients with OS. Numerous studies have reported that the functional roles of lncRNAs in cancer involve the carcinogenic or metastatic signaling pathways (34,35). Investigating the association between signaling pathways and lncRNAs is therefore critical in order to develop novel strategies for the early diagnosis and treatment of OS. A thorough investigation has been performed of the role of the PI3K/AKT signaling pathway in cancer pathogenesis, and numerous drugs targeting this pathway are currently being developed (36). PI3K can change the protein structure of AKT and activate it, when binding to growth factor receptors, including EGFR, RAS and PTEN. PI3K can activate or inhibit a series of downstream effectors, regulating therefore cell proliferation, differentiation, apoptosis and migration. The activation of Akt by PI3k leads to the phosphorylation of apoptosis-related proteins, including Bax (37), Bad (38) and caspase-9 (39), inhibiting therefore their function of apoptosis promoters. Furthermore, the structures of growth factor receptors are all altered at some level in most solid tumors, including colorectal cancer (36), epithelial ovarian cancer (40) and cervical cancer (41). The PI3K/AKT signaling cascade and downstream effectors are therefore considered as attractive pharmacological targets (42,43). In addition, it has been reported that the downregulation of DEP domain-containing mTOR-interacting protein inhibits the proliferation, migration and survival of OS cells through the PI3K/Akt/mTOR pathway (44). A recent study reported that AWPPH activates the PI3K/AKT pathway in HCC cells (29). In the present study, AWPPH downregulation significantly decreased p-PI3K and p-AKT expression levels, which indicated that the PI3K/AKT pathway may participate in the oncogenic function of AWPPH.

In conclusion, the present study demonstrated that AWPPH was highly expressed in OS cells, and that AWPPH-knockdown suppressed OS cell proliferation, migration and invasion, and promoted OS cell apoptosis, which may be mediated by PI3K/AKT pathway activation. These data suggested that AWPPH may be considered as a prognostic biomarker and a potential therapeutic target in OS.

Acknowledgements

Not applicable.

Funding

This research was supported by the National Natural Science Foundation of China (grants nos. 81272942, 81702666 and 81502328).

Availability of data and materials

The analyzed data sets generated during the present study are available from the corresponding author on reasonable request.

Authors' contributions

WD and DW wrote the manuscript, and collected and interpreted the data. FJ and HZ designed the study and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study protocol was approved by the Ethics Committee of Changhai Hospital. Written informed consent was obtained from all patients.

Patients consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Rothermundt C, Seddon BM, Dileo P, Strauss SJ, Coleman J, Briggs TW, Haile SR and Whelan JS: Follow-up practices for high-grade extremity Osteosarcoma. BMC Cancer. 16:3012016. View Article : Google Scholar : PubMed/NCBI

2 

Kager L, Zoubek A, Pötschger U, Kastner U, Flege S, Kempf-Bielack B, Branscheid D, Kotz R, Salzer-Kuntschik M, Winkelmann W, et al: Primary metastatic osteosarcoma: Presentation and outcome of patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol. 21:2011–2018. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Anderson ME: Update on survival in osteosarcoma. Orthop Clin North Am. 47:283–292. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Senerchia AA, Macedo CR, Ferman S, Scopinaro M, Cacciavillano W, Boldrini E, Lins de Moraes VL, Rey G, de Oliveira CT, Castillo L, et al: Results of a randomized, prospective clinical trial evaluating metronomic chemotherapy in nonmetastatic patients with high-grade, operable osteosarcomas of the extremities: A report from the Latin American Group of Osteosarcoma Treatment. Cancer. 123:1003–1010. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Chung SW, Han I, Oh JH, Shin SH, Cho HS and Kim HS: Prognostic effect of erroneous surgical procedures in patients with osteosarcoma: Evaluation using propensity score matching. J Bone Joint Surg Am. 96:e602014. View Article : Google Scholar : PubMed/NCBI

6 

Cheng C, Chen ZQ and Shi XT: MicroRNA-320 inhibits osteosarcoma cells proliferation by directly targeting fatty acid synthase. Tumour Biol. 35:4177–4183. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Li E, Zhang J, Yuan T and Ma B: MiR-145 inhibits osteosarcoma cells proliferation and invasion by targeting ROCK1. Tumour Biol. 35:7645–7650. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Rinn JL and Chang HY: Genome regulation by long noncoding RNAs. Annu Rev Biochem. 81:145–166. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Ponjavic J, Ponting CP and Lunter G: Functionality or transcriptional noise? Evidence for selection within long noncoding RNAs. Genome Res. 17:556–565. 2007. View Article : Google Scholar : PubMed/NCBI

10 

ENCODE Project Consortium, ; Birney E, Stamatoyannopoulos JA, Dutta A, Guigó R, Gingeras TR, Margulies EH, Weng Z, Snyder M, Dermitzakis ET, et al: Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 447:799–816. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Hirano T, Yoshikawa R, Harada H, Harada Y, Ishida A and Yamazaki T: Long noncoding RNA, CCDC26, controls myeloid leukemia cell growth through regulation of KIT expression. Mol Cancer. 14:902015. View Article : Google Scholar : PubMed/NCBI

12 

Chen F, Mo J and Zhang L: Long noncoding RNA BCAR4 promotes osteosarcoma progression through activating GLI2-dependent gene transcription. Tumour Biol. 37:13403–13412. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Raveh E, Matouk IJ, Gilon M and Hochberg A: The H19 Long non-coding RNA in cancer initiation, progression and metastasis-a proposed unifying theory. Mol Cancer. 14:1842015. View Article : Google Scholar : PubMed/NCBI

14 

Modali SD, Parekh VI, Kebebew E and Agarwal SK: Epigenetic regulation of the lncRNA MEG3 and its target c-MET in pancreatic neuroendocrine tumors. Mol Endocrinol. 29:224–237. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Wu XS, Wang F, Li HF, Hu YP, Jiang L, Zhang F, Li ML, Wang XA, Jin YP, Zhang YJ, et al: LncRNA-PAGBC acts as a microRNA sponge and promotes gallbladder tumorigenesis. EMBO Rep. 18:1837–1853. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Xiao J, Lv Y, Jin F, Liu Y, Ma Y, Xiong Y, Liu L, Zhang S, Sun Y, Tipoe GL, et al: LncRNA HANR promotes tumorigenesis and increase of chemoresistance in hepatocellular carcinoma. Cell Physiol Biochem. 43:1926–1938. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Hu X, Feng Y, Zhang D, Zhao SD, Hu Z, Greshock J, Zhang Y, Yang L, Zhong X, Wang LP, et al: A functional genomic approach identifies FAL1 as an oncogenic long noncoding RNA that associates with BMI1 and represses p21 expression in cancer. Cancer Cell. 26:344–357. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Peng W, Si S, Zhang Q, Li C, Zhao F, Wang F, Yu J and Ma R: Long non-coding RNA MEG3 functions as a competing endogenous RNA to regulate gastric cancer progression. J Exp Clin Cancer Res. 34:792015. View Article : Google Scholar : PubMed/NCBI

20 

Sun L, Yang C, Xu J, Feng Y, Wang L and Cui T: Long noncoding RNA EWSAT1 promotes osteosarcoma cell growth and metastasis through suppression of MEG3 expression. DNA Cell Biol. 35:812–818. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Li Z, Yu X and Shen J: Long non-coding RNAs: Emerging players in osteosarcoma. Tumour Biol. 37:2811–2816. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Cui X, Jing X, Long C, Tian J and Zhu J: Long noncoding RNA MEG3, a potential novel biomarker to predict the clinical outcome of cancer patients: A meta-analysis. Oncotarget. 8:19049–19056. 2017.PubMed/NCBI

23 

Ying L, Huang Y, Chen H, Wang Y, Xia L, Chen Y, Liu Y and Qiu F: Downregulated MEG3 activates autophagy and increases cell proliferation in bladder cancer. Mol Biosyst. 9:407–411. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Qin R, Chen Z, Ding Y, Hao J, Hu J and Guo F: Long noncoding RNA MEG3 inhibits the proliferation of cervical carcinoma cells through the induction of cell cycle arrest and apoptosis. Neoplasma. 60:486–492. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Chang L, Wang G, Jia T, Zhang L, Li Y, Han Y, Zhang K, Lin G, Zhang R, Li J and Wang L: Armored long non-coding RNA MEG3 targeting EGFR based on recombinant MS2 bacteriophage virus-like particles against hepatocellular carcinoma. Oncotarget. 7:23988–24004. 2016.PubMed/NCBI

26 

Cao X, Zhuang S, Hu Y, Xi L, Deng L, Sheng H and Shen W: Associations between polymorphisms of long noncoding RNA MEG3 and risk of colorectal cancer in Chinese. Oncotarget. 7:19054–19059. 2016.PubMed/NCBI

27 

Zhu F, Zhang X, Yu Q, Han G, Diao F, Wu C and Zhang Y: LncRNA AWPPH inhibits SMAD4 via EZH2 to regulate bladder cancer progression. J Cell Biochem. 119:4496–4505. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Yu G, Wang W, Deng J and Dong S: LncRNA AWPPH promotes the proliferation, migration and invasion of ovarian carcinoma cells via activation of the Wnt/β-catenin signaling pathway. Mol Med Rep. 19:3615–3621. 2019.PubMed/NCBI

29 

Zhao X, Liu Y and Yu S: Long noncoding RNA AWPPH promotes hepatocellular carcinoma progression through YBX1 and serves as a prognostic biomarker. Biochim Biophys Acta Mol Basis Dis. 1863:1805–1816. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Guo D, Li Q, Lv Q, Wei Q, Cao S and Gu J: MiR-27a targets sFRP1 in hFOB cells to regulate proliferation, apoptosis and differentiation. PLoS One. 9:e913542014. View Article : Google Scholar : PubMed/NCBI

31 

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

32 

Bielack S, Carrle D and Casali PG; ESMO Guidelines Working Group, : Osteosarcoma: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 20 (Suppl 4):S137–S139. 2009. View Article : Google Scholar

33 

Zhang J, Lan Q and Lin J: Identification of key gene modules for human osteosarcoma by co-expression analysis. World J Surg Oncol. 16:892018. View Article : Google Scholar : PubMed/NCBI

34 

Bassett AR, Akhtar A, Barlow DP, Bird AP, Brockdorff N, Duboule D, Ephrussi A, Ferguson-Smith AC, Gingeras TR, Haerty W, et al: Considerations when investigating lncRNA function in vivo. Elife. 3:e030582014. View Article : Google Scholar : PubMed/NCBI

35 

Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 464:1071–1076. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Danielsen SA, Eide PW, Nesbakken A, Guren T, Leithe E and Lothe RA: Portrait of the PI3K/AKT pathway in colorectal cancer. Biochim Biophys Acta. 1855:104–121. 2015.PubMed/NCBI

37 

Gardai SJ, Hildeman DA, Frankel SK, Whitlock BB, Frasch SC, Borregaard N, Marrack P, Bratton DL and Henson PM: Phosphorylation of Bax Ser184 by Akt regulates its activity and apoptosis in neutrophils. J Biol Chem. 279:21085–21095. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Quan JH, Cha GH, Zhou W, Chu JQ, Nishikawa Y and Lee YH: Involvement of PI 3 kinase/Akt-dependent Bad phosphorylation in Toxoplasma gondii-mediated inhibition of host cell apoptosis. Exp Parasitol. 133:462–471. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Cardone MH, Roy N, Stennicke HR, Salvesen GS, Franke TF, Stanbridge E, Frisch S and Reed JC: Regulation of cell death protease caspase-9 by phosphorylation. Science. 282:1318–1321. 1998. View Article : Google Scholar : PubMed/NCBI

40 

Li C, Yu S, Wu S, Ni Y and Pan Z: MicroRNA-936 targets FGF2 to inhibit epithelial ovarian cancer aggressiveness by deactivating the PI3K/Akt pathway. Onco Targets Ther. 12:5311–5322. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Lee MS, Jeong MH, Lee HW, Han HJ, Ko A, Hewitt SM, Kim JH, Chun KH, Chung JY, Lee C, et al: PI3K/AKT activation induces PTEN ubiquitination and destabilization accelerating tumourigenesis. Nat Commun. 6:77692015. View Article : Google Scholar : PubMed/NCBI

42 

Porta C, Paglino C and Mosca A: Targeting PI3K/Akt/mTOR signaling in cancer. Front Oncol. 4:642014. View Article : Google Scholar : PubMed/NCBI

43 

Hennessy BT, Smith DL, Ram PT, Lu Y and Mills GB: Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 4:988–1004. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Hu B, Lv X, Gao F, Chen S, Wang S, Qing X, Liu J, Wang B and Shao Z: Downregulation of DEPTOR inhibits the proliferation, migration, and survival of osteosarcoma through PI3K/Akt/mTOR pathway. Onco Targets Ther. 10:4379–4391. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 18 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ding W, Wu D, Ji F and Zhang H: Inhibition of long non‑coding RNA‑AWPPH decreases osteosarcoma cell proliferation, migration and invasion. Oncol Lett 18: 5055-5062, 2019
APA
Ding, W., Wu, D., Ji, F., & Zhang, H. (2019). Inhibition of long non‑coding RNA‑AWPPH decreases osteosarcoma cell proliferation, migration and invasion. Oncology Letters, 18, 5055-5062. https://doi.org/10.3892/ol.2019.10898
MLA
Ding, W., Wu, D., Ji, F., Zhang, H."Inhibition of long non‑coding RNA‑AWPPH decreases osteosarcoma cell proliferation, migration and invasion". Oncology Letters 18.5 (2019): 5055-5062.
Chicago
Ding, W., Wu, D., Ji, F., Zhang, H."Inhibition of long non‑coding RNA‑AWPPH decreases osteosarcoma cell proliferation, migration and invasion". Oncology Letters 18, no. 5 (2019): 5055-5062. https://doi.org/10.3892/ol.2019.10898