Open Access

Long non‑coding RNA RNCR3 promotes glioma progression involving the Akt/GSK‑3β pathway

Retraction in: /10.3892/ol.2023.14055

  • Authors:
    • Bing Zhu
    • Shenyan Zhang
    • Ning Meng
    • He Zhang
    • Shaoji Yuan
    • Jin Zhang
  • View Affiliations

  • Published online on: October 18, 2019     https://doi.org/10.3892/ol.2019.11002
  • Pages: 6315-6322
  • Copyright: © Zhu 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

Increasing evidence has confirmed that long non‑coding RNAs (lncRNAs) serve critical roles in the development of a large number of human malignancies, including glioma. Several previously published studies have reported that the lncRNA retinal non‑coding RNA3 (RNCR3; also termed LINC00599) exerts important roles in certain human malignancies; however, the precise biological role and underlying molecular mechanisms of RNCR3 in the development of glioma are yet to be fully elucidated. In the present study, it was revealed that the expression of RNCR3 was increased in glioma tissues compared with in corresponding adjacent normal tissues. Furthermore, increased levels of RNCR3 expression were associated with tumor progression and poor survival rates of patients with glioma. In addition, the U87 and U251 cell lines were selected to investigate the biological function and potential mechanisms of RNCR3 in glioma, and it was observed that RNCR3 knockdown led to an impairment of the proliferative and invasive abilities of cells; furthermore, G1 phase arrest was induced in glioma cells in vitro. Finally, the results of western blot analyses revealed that knockdown of RNCR3 led to a decrease in the expression levels of phosphorylated Akt and glycogen synthase kinase‑3β (GSK‑3β), without any clear effect on the expression levels of total Akt and GSK‑3β. Collectively, these results suggested that RNCR3 is able to regulate cell proliferation, the cell cycle and cell invasion in glioma, potentially via the Akt/GSK‑3β signaling pathway.

Introduction

Glioma is the most common and most aggressive primary malignant tumor arising in the central nervous system of adults globally (1). It is also one of the main causes of cancerassociated mortality (2). Although certain progress has been made regarding effective early diagnosis of the disease, the majority of patients with glioma have progressed to the advanced stages by the time of diagnosis (3). Intensive research has been conducted to examine the biological mechanisms underlying the progression of glioma; however, the prognosis of patients in this category remains rather poor (4,5).

With the rapid development of genome and transcriptome sequencing technologies, numerous novel non-protein-coding RNAs have been identified (6). Whereas >80% of the genome is transcribed into mRNA transcripts, only ~2% of the genome codes for proteins (6,7). Long non-coding RNAs (lncRNAs) comprise a class of non-protein-coding RNAs that are >200 nucleotides in length (8). LncRNAs serve key regulatory roles in a variety of biological and pathological processes, including tumorigenesis, transcription regulation and epigenetic post-transcription regulation (911).

Retinal noncoding RNA3 (RNCR3, also termed LINC00599) is a lncRNA that is transcribed from the intergenic regions of the genome, and is highly conserved in mammals (12). A number of studies have reported that RNCR3 exerts important regulatory functions in cell proliferation and differentiation, and the process of atherosclerosis (13,14). Regarding human cancers, Tian et al (15) reported that RNCR promoted prostate cancer development by regulating microRNA (miR)-185-5p. However, the biological functions and molecular mechanisms of RNCR3 in glioma are yet to be fully elucidated.

In the present study, RNCR3 expression levels in glioma tissues and in paired normal tissues were evaluated, following which the biological function of RNCR3 in glioma cell lines, and the underlying mechanisms of RNCR3 in glioma, were further investigated. To the best of our knowledge, the present study is the first to demonstrate that RNCR3 functions as an oncogene in glioma development.

Materials and methods

Clinical samples

The present study was approved by the Ethics and Research Committees of the Sunshine Union Hospital of Shandong Province (Weifang, China), and performed in accordance with the principles of the Declaration of Helsinki. A total of 54 pairs of glioma tissue samples and paired adjacent normal tissues were obtained from patients undergoing resection at the Department of Neurosurgery of Sunshine Union Hospital between January 2005 and December 2010. The patients included 30 males and 24 females with a mean age of 53.9 years (range, 41–74 years). All the patients were pathologically confirmed, and none of the patients had received preoperative chemotherapy or radiation therapy. The tissues were collected during surgery and stored immediately in liquid nitrogen (−196°C). Written informed consent was collected from all subjects. The clinical characteristics of all the patients are summarized in Table I.

Table I.

Expression of RNCR3 in association with the clinicopathological variables.

Table I.

Expression of RNCR3 in association with the clinicopathological variables.

RNCR3 expression

Clinicopathological parametersNHighLowP-value
All543123
Age (years) 0.319
  <50291712
  ≥50251411
Sex 0.812
  Male281612
  Female261511
Tumor size (cm) 0.012a
  <5301416
  ≥524177
WHO grade 0.007a
  I + II251015
  III+ IV29218

a P<0.05. RNCR3, retinal noncoding RNA3; WHO, World Health Organization.

Cell culture

A human astroglia cell line, HA, was acquired from BeNa Culture Collection. Two human glioma cell lines, SHG-44 and U251, and a glioblastoma of unknown origin, U87 (cat. no. HTB14) were purchased from the American Type Culture Collection. Cell lines were authenticated via short tandem repeat cell authentication profiling. All the cell lines were maintained in Gibco™ DMEM (Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS, Invitrogen, USA), and 50 U/ml penicillin and 0.1 mg/ml streptomycin (Biowest). All the cell cultures were incubated at 37°C with 5% CO2.

RNA extraction and reverse transcription-quantitative PCR (RT-qPCR)

Following the manufacturer's protocol, TRIzol® reagent (Thermo Fisher Scientific, Inc.) was used to isolate the RNA of tissues or cells, and the isolated RNA was subsequently reverse-transcribed using an Invitrogen™ PrimeScript RT Reagent kit (Thermo Fisher Scientific, Inc.). SYBR Premix Ex Taq™ reagent (Takara Biotechnology Co., Ltd.) was used for the qPCR assay, and qPCR was performed using an Applied Biosystems™ ABI PRISM 7500 PCR system (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocols. PCRs were performed in a total volume of 20 µl with 10 µl 2× SYBR premix ex-taq, 5 µl cDNA, 0.8 µl primers (2.5 µM) and 4.2 µl ddH2O). The PCR thermocycling conditions were; denaturation at 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec, 55°C for 15 sec and 72°C for 10 sec with a final extension step at 60°C for 1 min. Melting curves of the amplified products were analyzed at the end of each PCR to confirm that only one product was amplified and detected. Expression was calculated via the relative quantification cycle (Cq) value and was normalized to the expression of the internal control gene GAPDH. Relative expression of RNCR3 was calculated using the 2−ΔΔCq method (16). The primer sequences for RNCR3 and GAPDH are presented in Table II. The mean value was selected as the cut-off between high and low RNCR3 expression in patients with glioma. The relative expression of control groups was normalized to 1.

Table II.

Sequences of primers used.

Table II.

Sequences of primers used.

GeneForward primerReverse primer
RNCR3 5′-CAACACCTTCCTCCGTGACTGTG-3′ 5′-GCTGGCTCCTTCTTGTCCACATA3′.
GAPDH 5′-CGCTCTCTGCTCCTCCTGTTC-3′ 5′-ATCCGTTGACTCCGACCTTCAC-3′

[i] RNCR3, retinal noncoding RNA3.

RNCR3 short hairpin RNA (shRNA)

RNCR3 shRNA (sh-RNCR3) was purchased from Shanghai GenePharma Co. Ltd. And transfected into cells at a final concentration of 50 nM. An Invitrogen™ Lipofectamine® 3000 kit (Thermo Fisher Scientific, Inc.) was used to perform the transfections, according to the manufacturer's protocol. RT-qPCR was subsequently used to evaluate the knockdown efficiency. For all experiments involving transfected cells, cells were transfected for 48 h before subsequent experiments were performed.

Cell Counting Kit-8 (CCK-8) assay

Cell proliferation was examined every 24 h, according to the manufacturer's protocol. Cells were placed into the 96-well plates at a density of ~3,000 cells/well. Subsequently, 10 µl CCK-8 (Dojindo Molecular technologies, Inc.) was added, and the cells were incubated at 37°C for a further 2 h. Finally, the absorbance at 450 nm was determined using a spectrometer.

Cell cycle assay

Cells were collected, washed 3 times with cold PBS, and then fixed in 70% ethanol at 4°C overnight. Subsequently, the cells were washed, re-suspended, and incubated in a solution of 10 µg/ml RNase and 1 mg/ml propidium iodide (Sigma-Aldrich; Merck KGaA) at 37°C for 30 min in the dark. Finally, a FACSCalibur™ Cell Analyzer with ModFit version 5.0 (both BD Biosciences) was used to analyze the cells and the resultant flow cytometry data.

Cell invasion assay

The invasive ability of the cells was determined using BD 24-well Transwell® chambers (Costar; Corning Inc.) pre-coated with Matrigel™ coating, according to the manufacturer's protocol. Briefly, 1×105 cells suspended in 200 µl serum-free medium were seeded into the upper chamber, and 800 µl DMEM supplemented with 10% FBS was placed into the lower chamber. After incubating the cells for 18 h at 37°C, cells on the lower chamber membranes were fixed with 4% formaldehyde 15 min at room temperature and stained with 1% crystal violet at room temperature for 10 min. Cells in five randomly selected fields of the membrane were counted under a light microscope (magnification, ×40).

Western blot analysis

RIPA buffer (Beyotime Institute of Biotechnology) was used to extract proteins from the cells. Protein concentration was determined using a bicinchoninic acid (BCA) protein assay kit. The proteins (30 µg per lane) were resolved on a 10% SDS-PAGE and then electrophoretically transferred to polyvinylidene fluoride membranes (Bio-Rad Laboratories, Inc.). After blocking for 2 h at room temperature with 5% non-fat milk, the membranes were incubated with the desired primary antibodies at 4°C overnight. The primary antibodies were as follows: Akt (dilution, 1:1,000; cat. no. ab8805; Abcam), phosphorylated (p)-Akt (dilution, 1:1,000; cat. no. ab38449; Abcam), glycogen synthase kinase-3β (GSK-3β; dilution, 1:1,000; cat. no. ab32391; Abcam), p-GSK-3β (dilution, 1:1,000; cat. no. ab75745; Abcam), and β-actin (1:1,000; cat. no. ab227387; Abcam). Then, the membranes were incubated with anti-rabbit (dilution, 1:5,000; cat. no. 66467-1-Ig; ProteinTech Group, Inc.) horseradish peroxidase-conjugated secondary antibodies for 2 h at room temperature. An enhanced chemiluminescence kit (Santa Cruz Biotechnology, Inc.) was used to detect the intensities of the immunoblots, which were visualized following exposure to X-ray film. Protein expression levels were quantified relative to β-actin levels using ImageJ (version 1.8.0 National Institutes of Health).

Statistical analysis

Experimental data from at least three independent experiments are presented as the mean ± standard deviation. SPSS 18.0 software (SPSS, Inc.) was used to perform the statistical analyses. The associations between the levels of RNCR3 and clinicopathological factors was assessed by χ2 test. Differences between multiple groups were evaluated by one-way analysis of variance followed by Tukey's post hoc test. Differences between two groups were evaluated using an unpaired or paired Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of RNCR3 is increased in glioma tissues and cell lines

RT-qPCR analysis was performed to measure the relative expression of RNCR3 in 54 pairs of glioma tissue and adjacent normal tissue, we have used the normal tissues as control for normalization. The results obtained suggested that RNCR3 expression was markedly higher in glioma tissues compared with the corresponding adjacent normal tissues (Fig. 1A). RNCR3 expression was further examined in cell lines: A human astroglia cell line (HA), two human glioma cell lines (SHG-44 and U251), and a glioblastoma of unknown origin (U87). These experiments revealed that the expression of RNCR3 was higher in the glioma cell lines compared with that in the HA cell line (Fig. 1B).

Upregulated expression of RNCR3 is associated with progression of the disease, and poor prognosis of patients with glioma

Subsequently, the correlation between RNCR3 expression levels and clinicopathological factors in 66 glioma individuals was investigated (Table I). This analysis revealed that an elevated expression of RNCR3 was associated with tumor size and clinical stage, although no significant correlations were identified between RNCR3 expression and age and gender. In addition, KaplanMeier survival curves suggested that glioma patients who had higher expression levels of RNCR3 in their glioma tissues had significantly poorer survival rates compared with glioma patients who had correspondingly lower expression levels of RNCR3 in their glioma tissues (Fig. 1C).

Knockdown of RNCR3 inhibits glioma cell proliferation

The results of the RT-qPCR assay confirmed that the RNCR3 expression levels of U87 and U251 cells transfected with sh-RNCR3 were lower compared with that of the control untransfected cells, or those U87 and U251 cells that were transfected with NC shRNA (Fig. 2A and B). RNCR3 knockdown led to a repression of cell proliferation in the U87 (Fig. 2C) and U251 (Fig. 2D) glioma cell lines.

Knockdown of RNCR3 induces G1 phase arrest of glioma cells

The results of the cell cycle assay experiments revealed that knocked-down RNCR3 induced the G1 phase arrest of U87 and U251 cells (Fig. 3).

Knockdown of RNCR3 inhibits cell invasion in the glioma cell lines

As shown in Fig. 4, the cell invasion assays confirmed that RNCR3 knockdown repressed cell invasion in the U87 and U251 cells.

Knockdown of RNCR3 inhibits the Akt/GSK-3β pathway in glioma cells

As has been well established, the Akt/GSK-3β signaling pathway is constitutively active in a large number of different types of human cancer. To further investigate whether RNCR3 regulates glioma development through Akt/GSK-3β pathway activation, western blot assays were performed to evaluate the levels of total and phosphorylated Akt and GSK-3β in the glioma cells. As shown in Fig. 5, silencing of RNCR3 did not exert a clear influence on the total level of Akt and GSK-3β, but the levels of phosphorylated Akt and GSK-3β were significantly reduced. These results indicated that overexpression of RNCR3 activated the Akt/GSK-3β pathway in human glioma cells.

Discussion

In view of the increasing number of studies that are being published focusing on the functional attributes of lncRNAs, emerging evidence has indicated that lncRNAs have important roles in various physiological and pathological processes, including cell proliferation, apoptosis and differentiation, and the development of different types of cancer (9,17,18). LncRNAs fulfill a range of different roles, including the regulation of gene transcription in basal transcription machinery, post-transcriptional regulation of RNA splicing and epigenetic regulation (19). For example, the lncRNA small ubiquitin-like modifier 1 pseudogene 3 promoted breast cancer progression by negatively regulating miR-320a (9). LncRNA highly upregulated in liver cancer triggers autophagy by stabilizing sirtuin-1, and impairs the chemosensitivity of hepatocellular carcinoma cells (18). LncRNA HOXA transcript at the distal tip enhances tumorigenesis and metastasis in esophageal squamous carcinoma cells through the transcriptional and post-transcriptional regulation of homeobox protein A13 (19).

In the present study it was revealed for the first time, to the best of our knowledge, that the expression of lncRNA RNCR3 was upregulated in glioma tissues and cell lines. Additionally, the increased expression levels of RNCR3 in glioma tissues were associated with tumor progression and poor survival rates in patients with glioma. Furthermore, RNCR3 knockdown inhibited the proliferative and invasive abilities, and induced the G1 phase arrest of glioma cells. Finally, the present study suggested that the effects of RNCR3 on cell proliferation, the cell cycle and cellular invasion may involve regulation of the Akt/GSK-3β signaling pathway.

A number of studies have suggested that aberrant signaling of the Akt pathway contributes to cell proliferation and invasion in various human malignancies (20). The results of the present study suggested that RNCR3 knockdown led to an inhibition of the phosphorylation of Akt, although there was no clear influence on the total levels of Akt. Akt phosphorylation has previously been reported to inhibit the activity of GSK-3β, which is an important downstream target protein of Akt (20). These results are consistent with previous studies, which reported on the function of Akt/GSK-3β in glioma (21,22). In addition, a large number of lncRNAs have been reported to regulate human cancers via actions on the Akt/GSK-3β pathway. For example, the lncRNA lung cancer-associated transcript 1 was revealed to enhance proliferation and invasion in clear cell renal cell carcinoma via the Akt/GSK-3β pathway (23). The lncRNA urothelial carcinoma-associated 1 (UCA1) was identified to promote tumorigenesis by regulating the AKT/GSK-3β pathway in cholangiocarcinoma (24). LncRNA UCA1, induced by SP1 transcription factor, was also shown to enhance cell proliferation by recruiting enhancer of zeste 2 polycomb repressive complex 2 subunit, the catalytic subunit of the polycomb repressive complex 2, and activating the Akt signaling pathway in gastric cancer (25).

In conclusion, the present study demonstrated that the expression level of RNCR3 was increased in glioma tissues and in cell lines, and that RNCR3 may therefore serve as a novel prognostic indicator for patients with glioma. Silencing RNCR3 in glioma cells led to a suppression of the proliferative and invasive abilities of the cells, and induced cell cycle arrest involving the Akt/GSK3β pathway.

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

BZ and JZ designed the study. SYZ, NM and HZ analyzed the data. SJY collected and analyzed clinical samples and was also a major contributor in writing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics and Research Committees of the Sunshine Union Hospital of Shandong Province, and performed in accordance with the principles of the Declaration of Helsinki. Written informed consent was collected from all subjects.

Patient consent for publication

Not applicable.

Competing interests

The authors declare they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Zhi T, Jiang K, Xu X, Yu T, Wu W, Nie E, Zhou X, Jin X, Zhang J, Wang Y and Liu N: MicroRNA-520d-5p inhibits human glioma cell proliferation and induces cell cycle arrest by directly targeting PTTG1. Am J Transl Res. 9:4872–4887. 2017.PubMed/NCBI

3 

Wang Z, Yuan J, Li L, Yang Y, Xu X and Wang Y: Long non-coding RNA XIST exerts oncogenic functions in human glioma by targeting miR-137. Am J Transl Res. 9:1845–1855. 2017.PubMed/NCBI

4 

Boussiotis VA and Charest A: Immunotherapies for malignant glioma. Oncogene. 37:1121–1141. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Kesanakurti D, Maddirela D, Banasavadi-Siddegowda YK, Lai TH, Qamri Z, Jacob NK, Sampath D, Mohanam S, Kaur B and Puduvalli VK: A novel interaction of PAK4 with PPARgamma to regulate Nox1 and radiation-induced epithelial-to-mesenchymal transition in glioma. Oncogene. 36:5309–5320. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Chen Z, Chen X, Chen P, Yu S, Nie F, Lu B, Zhang T, Zhou Y, Chen Q, Wei C, et al: Long non-coding RNA SNHG20 promotes non-small cell lung cancer cell proliferation and migration by epigenetically silencing of P21 expression. Cell Death Dis. 8:e30922017. View Article : Google Scholar : PubMed/NCBI

7 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–108. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Kotzin JJ, Mowel WK and Henao-Mejia J: Viruses hijack a host lncRNA to replicate. Science. 358:993–994. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Liu J, Song Z, Feng C, Lu Y, Zhou Y, Lin Y and Dong C: The long non-coding RNA SUMO1P3 facilitates breast cancer progression by negatively regulating miR-320a. Am J Transl Res. 9:5594–5602. 2017.PubMed/NCBI

10 

Li LJ, Zhu JL, Bao WS, Chen DK, Huang WW and Weng ZL: Long noncoding RNA GHET1 promotes the development of bladder cancer. Int J Clin Exp Pathol. 7:7196–7205. 2014.PubMed/NCBI

11 

Battistelli C, Cicchini C, Santangelo L, Tramontano A, Grassi L, Gonzalez FJ, de Nonno V, Grassi G, Amicone L and Tripodi M: The Snail repressor recruits EZH2 to specific genomic sites through the enrollment of the lncRNA HOTAIR in epithelial-to-mesenchymal transition. Oncogene. 36:942–955. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Mercer TR, Qureshi IA, Gokhan S, Dinger ME, Li G, Mattick JS and Mehler MF: Long noncoding RNAs in neuronal-glial fate specification and oligodendrocyte lineage maturation. BMC Neurosci. 11:142010. View Article : Google Scholar : PubMed/NCBI

13 

Shan K, Jiang Q, Wang XQ, Wang YN, Yang H, Yao MD, Liu C, Li XM, Yao J, Liu B, et al: Role of long non-coding RNA-RNCR3 in atherosclerosis-related vascular dysfunction. Cell Death Dis. 7:e22482016. View Article : Google Scholar : PubMed/NCBI

14 

Sanuki R, Onishi A, Koike C, Muramatsu R, Watanabe S, Muranishi Y, Irie S, Uneo S, Koyasu T, Matsui R, et al: miR-124a is required for hippocampal axogenesis and retinal cone survival through Lhx2 suppression. Nat Neurosci. 14:1125–1134. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Tian C, Jin Y and Shi S: Long non-coding RNA SUMO1P3 may promote cell proliferation, migration, and invasion of pancreatic cancer via EMT signaling pathway. Oncol Lett. 16:6109–6115. 2018.PubMed/NCBI

16 

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

17 

Liang WC, Ren JL, Wong CW, Chan SO, Waye MM, Fu WM and Zhang JF: LncRNA-NEF antagonized epithelial to mesenchymal transition and cancer metastasis via cis-regulating FOXA2 and inactivating Wnt/β-catenin signaling. Oncogene. 37:1445–1456. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Xiong H, Ni Z, He J, Jiang S, Li X, He J, Gong W, Zheng L, Chen S, Li B, et al: LncRNA HULC triggers autophagy via stabilizing Sirt1 and attenuates the chemosensitivity of HCC cells. Oncogene. 36:3528–3540. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Lin C, Wang Y, Wang Y, Zhang S, Yu L, Guo C and Xu H: Transcriptional and posttranscriptional regulation of HOXA13 by lncRNA HOTTIP facilitates tumorigenesis and metastasis in esophageal squamous carcinoma cells. Oncogene. 36:5392–5406. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Song Z, Feng C, Lu Y, Gao Y, Lin Y and Dong C: Overexpression and biological function of MEF2D in human pancreatic cancer. Am J Transl Res. 9:4836–4847. 2017.PubMed/NCBI

21 

Majewska E and Szeliga M: AKT/GSK3beta Signaling in Glioblastoma. Neurochem Res. 42:918–924. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Park JH, Shin YJ, Riew TR and Lee MY: The indolinone MAZ51 induces cell rounding and G2/M cell cycle arrest in glioma cells without the inhibition of VEGFR-3 phosphorylation: Involvement of the RhoA and Akt/GSK3β signaling pathways. PLoS One. 9:e1090552014. View Article : Google Scholar : PubMed/NCBI

23 

Zheng Z, Zhao F, Zhu D, Han J, Chen H, Cai Y, Chen Z and Xie W: Long non-coding RNA LUCAT1 promotes proliferation and invasion in clear cell renal cell carcinoma through AKT/GSK-3beta signaling pathway. Cell Physiol Biochem. 48:891–904. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Xu Y, Yao Y, Leng K, Li Z, Qin W, Zhong X, Kang P, Wan M, Jiang X and Cui Y: Long non-coding RNA UCA1 indicates an unfavorable prognosis and promotes tumorigenesis via regulating AKT/GSK-3β signaling pathway in cholangiocarcinoma. Oncotarget. 8:96203–96214. 2017.PubMed/NCBI

25 

Wang ZQ, Cai Q, Hu L, He CY, Li JF, Quan ZW, Liu BY, Li C and Zhu ZG: Long noncoding RNA UCA1 induced by SP1 promotes cell proliferation via recruiting EZH2 and activating AKT pathway in gastric cancer. Cell Death Dis. 8:e28392017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

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
Zhu B, Zhang S, Meng N, Zhang H, Yuan S and Zhang J: Long non‑coding RNA RNCR3 promotes glioma progression involving the Akt/GSK‑3β pathway Retraction in /10.3892/ol.2023.14055. Oncol Lett 18: 6315-6322, 2019
APA
Zhu, B., Zhang, S., Meng, N., Zhang, H., Yuan, S., & Zhang, J. (2019). Long non‑coding RNA RNCR3 promotes glioma progression involving the Akt/GSK‑3β pathway Retraction in /10.3892/ol.2023.14055. Oncology Letters, 18, 6315-6322. https://doi.org/10.3892/ol.2019.11002
MLA
Zhu, B., Zhang, S., Meng, N., Zhang, H., Yuan, S., Zhang, J."Long non‑coding RNA RNCR3 promotes glioma progression involving the Akt/GSK‑3β pathway Retraction in /10.3892/ol.2023.14055". Oncology Letters 18.6 (2019): 6315-6322.
Chicago
Zhu, B., Zhang, S., Meng, N., Zhang, H., Yuan, S., Zhang, J."Long non‑coding RNA RNCR3 promotes glioma progression involving the Akt/GSK‑3β pathway Retraction in /10.3892/ol.2023.14055". Oncology Letters 18, no. 6 (2019): 6315-6322. https://doi.org/10.3892/ol.2019.11002