Neuroglobin promotes the proliferation and suppresses the apoptosis of glioma cells by activating the PI3K/AKT pathway

  • Authors:
    • Bei Zhang
    • Yong Liu
    • Yajun Li
    • Xiao Zhe
    • Shijun Zhang
    • Lei Zhang
  • View Affiliations

  • Published online on: November 22, 2017     https://doi.org/10.3892/mmr.2017.8132
  • Pages: 2757-2763
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Our previous study demonstrated that neuroglobin (Ngb) functions as an independent predictive indicator of the prognosis of patients with glioma and promotes cancer cell growth by suppressing apoptosis. However, the understanding of the mechanisms underlying the survival‑enhancing function of Ngb in glioma is limited. In the present study, KEGG PathwayFinder by gene correlation analysis was performed on the R2: Genomics Analysis and Visualization Platform, which revealed a high association between Ngb and the phosphatidylinositol 3‑kinase (PI3K)/AKT pathway using glioma data (GSE4290) from the Gene Expression Omnibus database. Furthermore, western blotting experiments were performed in U251 and U87 glioma cells, and Ngb knockdown using short hairpin RNA reduced the protein levels of phosphorylated (p)‑AKT, p‑mammalian target of rapamycin (mTOR) and antiapoptotic factor Bcl‑2, and increased the expression of the proapoptotic protein Bcl‑2‑associated X, in U251 cells. In addition, Ngb overexpression promoted the activation of the PI3K/AKT pathway in U87 cells. MK2206, a PI3K/AKT signaling inhibitor, reduced the expression of p‑AKT and increased the levels of apoptosis‑associated proteins, including cleaved poly(ADP‑ribose) polymerase 1 and cleaved caspase‑3/7/8, in Ngb‑overexpressing U87 cells. Furthermore, MK2206 treatment reduced the proliferation and induced the apoptosis of Ngb‑overexpressing U87 cells, as indicated by the results of MTT, colony formation and flow cytometry assays. In addition, insulin‑like growth factor‑1, a PI3K/AKT signaling activator, reversed Ngb knockdown‑induced growth arrest and apoptosis in U251 cells. In conclusion, the results of the present study indicate that Ngb may facilitate a malignant phenotype of glioma cells by activating the PI3K/AKT pathway.

Introduction

Glioma, the most common form of brain malignancy, is one a major contributor to cancer-associated deaths worldwide (1). The strategies employed for the diagnosis and treatment of glioma have improved over the past several decades (2). However, the prognosis of patients with glioma remains poor, with a survival time of 12–18 months post-diagnosis (3). Therefore, the identification of the exact mechanisms underlying the malignant phenotype of glioma cells is required.

Neuroglobin (Ngb), a novel tumor-associated protein, functions as an oncogene or tumor suppressor in human cancer. Previous reports have demonstrated that overexpression of Ngb enhances reactive oxygen species scavenging and reverses oxidative stress-induced cell death in neuroblastoma cells (46). The expression of Ngb is upregulated under hypoxic conditions in glioblastoma cells and tumor xenografts, indicating a potential role of Ngb in cancer cell survival in hypoxic microenvironments (7,8). In addition to brain tumors, aberrant expression of Ngb has also been reported in other types of malignancies. For example, Ngb is reported to be overexpressed in certain non-small cell lung cancer cases, particularly in squamous cell carcinomas (9). Notably, 17β-estradiol induces Ngb upregulation, which renders cancer cells, including MCF-7, HepG2, SK-N-BE, HeLa and DLD-1, resistant to oxidative stress (1013). However, Ngb expression is downregulated in hepatocellular carcinoma tissues and its silencing promotes the proliferation and cell cycle progression of cancer cells (14). Our previous study demonstrated that Ngb functions as an independent prognostic biomarker for patients with glioma and promotes the growth of cancer cells by suppressing apoptosis (15). However, the mechanisms underlying the survival-enhancing effect of Ngb in glioma remains a challenge, therefore, the present study aimed to investigate the effect and mechanisms of Ngb in glioma.

The results of the present study demonstrated that Ngb promoted the proliferation and inhibited the apoptosis of glioma cells, which may occur through effects on the phosphatidylinositol 3-kinase (PI3K)/AKT pathway. To the best of our knowledge, the presents study is the first to indicate that Ngb may be a potential therapeutic target for glioma.

Materials and methods

Cell culture and transfection

U87MG ATCC and U251MG human glioma cell lines were obtained from the American Type Culture Collection (Manassas, VA, USA) and were cultivated in Dulbecco's modified Eagle's medium (DMEM; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal calf serum (Gibco; Thermo Fisher Scientific, Inc.) and antibiotics (100 units/ml penicillin and 100 µg/ml streptomycin; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) at 37°C in a humidified incubator containing 5% CO2.

Short hairpin RNA (shRNA) targeting Ngb (5′-GUGAGUCCCUGCUCUACAU-3′) and non-targeting (NT) shRNA (5′-GCCACACGAUUGCUGUCUU-3′) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Vectors (1 µg) were transfected into cells at 50–70% confluency using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Retroviral vector pMMP-Ngb was generated, packaged and transduced as previously described (16). pMMP is a MFG-based vector with modifications from the myeloproliferative sarcoma virus (17) and primer binding sequence (18) vector systems. pMMP vector alone was used as the control for Ngb overexpression experiments. AKT inhibitor, MK2206 (1 µM; 37°C for 48 h; Selleck Chemicals, Houston, TX, USA), and insulin-like growth factor-1 (IGF-1; 10 ng/ml; 37°C for 48 h; Sigma-Aldrich; Merck KGaA) were used to treat glioma cells 24 h post-transfection, according to the manufacturer's protocol. Control cells were treated with dimethylsulphoxide (DMSO; EMD Millipore, Billerica, MA, USA).

Bioinformatics analysis

KEGG PathwayFinder by gene correlation analysis in the R2: Genomics Analysis and Visualization Platform (http://r2.amc.nl) was performed to investigate the association between Ngb and various signaling pathways according to glioma data (GSE4290) (19).

MTT assay

Glioma cells (2×103 cells/well) were seeded in 96-well plates containing 100 µl DMEM per well. U87 cells were treated with either DMSO or MK2206 (1 µM; 37°C for 48 h) 24 h following Ngb retrovirus infection. U251 cells were treated with either DMSO or IGF-1 (10 ng/ml; 37°C for 48 h) 24 h post-transfection with Ngb shRNA. Following transfection for 24, 48, 72 and 96 h time intervals, 10 µl of MTT was added into each well and incubated at 37°C for 4 h. Subsequently, 150 µl DMSO was added per well and the absorbance was determined using microplate reader at 490 nm.

Colony formation assay

Glioma cells (1×103 cells/well) that were transfected with the corresponding vectors were cultured in 6-well plates and maintained at 37°C in humidified cell incubators containing 5% CO2 for 14–21 days. U87 cells were treated with either DMSO or MK2206 (1 µM; 37°C for 48 h) 24 h following infection with Ngb retroviruses. U251 cells were treated with either DMSO or IGF-1 (10 ng/ml; 37°C for 48 h) 24 h following transfection with Ngb shRNA. The formed cell colonies were stained with crystal violet (0.05%; 20 min at room temperature) and counted to represent the cell proliferation of glioma cells.

Apoptosis analysis

Apoptosis in glioma cells following transfection was detected by using an Annexin V/propidium iodide (PI) kit (BD Pharmingen; BD Biosciences, San Jose, CA, USA). U87 cell were treated with either DMSO or MK2206 (1 µM; 37°C for 48 h) 24 h post-infection with Ngb retroviruses. U251 cells were treated with DMSO or IGF-1 (10 ng/ml; 37°C for 48 h) 24 h post-transfection with Ngb shRNA. Briefly, glioma cells were resuspended in 1X binding buffer at a concentration of 1×106 cells/ml. A total of 100 µl of solution (1×105 cells) was transferred to a 5 ml culture tube and supplemented with 5 µl of Annexin V-fluorescein isothiocyanate and 5 µl of PI. Following incubation for 15 min at 25°C in the dark and with supplementation of 1X binding buffer (400 µl), the percentage ratio of apoptotic glioma cells was detected using FACSCalibur flow cytometer (BD Biosciences) and CellQuest Pro software (version 5.1.1; BD Biosciences).

Western blotting

U87 cells were treated with either DMSO or MK2206 (1 µM; 37°C for 48 h) 24 h following infection with Ngb retroviruses. U251 cells were treated with either DMSO or IGF-1 (10 ng/ml; 37°C for 48 h) 24 h post-transfection with Ngb shRNA. The transfected cells were lysed by radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China) and phenylmethylsulfonyl fluoride (Beyotime Institute of Biotechnology) for total protein extracts, followed by quantification with a Bradford Protein assay kit (Beyotime Institute of Biotechnology). Cell lysates (40 µg/lane) were separated by 10% SDS-PAGE. After being transferred to polyvinylidene fluoride membranes (Sigma-Aldrich; Merck KGaA) and blocked with 5% non-fat milk for 1 h at room temperature, the membranes were incubated with primary antibodies against GAPDH (1:5,000; cat. no. G8140-01; US Biological, Salem, MA, USA), Ngb (1:1,000; cat. no. ab37258; Abcam, Cambridge, MA, USA), AKT (1:1,000; cat. no. 9272; Cell Signaling Technology, Inc., Danvers, MA, USA), phosphorylated (p)-AKT (Ser473; 1:2,000; cat. no. 4060; Cell Signaling Technology, Inc.), mammalian target of rapamycin (mTOR; 1:1,000; cat. no. 2983; Cell Signaling Technology, Inc.), p-mTOR (Ser2448; 1:1,000; cat. no. 5536; Cell Signaling Technology, Inc.), Bcl-2 (1:1,000; cat. no. 15071; Cell Signaling Technology, Inc.), Bcl-2-associated X (Bax; 1:1,000; cat. no. 5023; Cell Signaling Technology, Inc.), cleaved poly(ADP-ribose) polymerase 1 (PARP; 1:1,000; cat. no. 5625; Cell Signaling Technology, Inc.), cleaved caspase-3 (1:1,000, cat. no. 9664; Cell Signaling Technology, Inc.), cleaved caspase-7 (1:1,000; cat. no. 8438; Cell Signaling Technology, Inc.) and cleaved caspase-8 (1:1,000; cat. no. 9496; Cell Signaling Technology, Inc.) at 4°C overnight. Subsequently, membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit and horse anti-mouse secondary antibodies for 1 h at room temperature (1:1,000; cat. nos. 7074 and 7076, respectively; Cell Signaling Technology, Inc.). GAPDH was employed as a loading control. Luminata Fe Western HRP Substrate (EMD Millipore, Billerica, MA, USA) was used to visualize proteins. A Bio-Rad Gel imaging system (Bio-Rad Laboratories, Inc., Hercules, CA, USA) was used to quantify western blotting data. Image J software (version 1.41; National Institutes of Health, Bethesda, MD, USA) was used to quantify protein levels.

Statistical analysis

Data are presented as the mean ± standard deviation and were analyzed using GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA). Student's t-test or one-way ANOVA followed by the post hoc Tukey's test were employed to analyze continuous variables. P<0.05 was considered to indicate a statistically significant difference.

Results

PI3K/AKT pathway is a candidate target of Ngb in glioma

To investigate the potential mechanisms underlying the survival-enhancing effect of Ngb in glioma, KEGG PathwayFinder by gene correlation analysis using the R2: Genomics Analysis and Visualization Platform (http://r2.amc.nl) was performed to investigate the association between Ngb and various signaling pathways in glioma. Base on the Gene Expression Omnibus (GEO) data (GSE4290) from the R2: Genomics Analysis and Visualization Platform, the results demonstrated that Ngb was strongly associated with the PI3K/AKT pathway in glioma (P<0.001; Table I). Therefore, the PI3K/AKT pathway is a candidate target of Ngb in glioma.

Table I.

KEGG PathwayFinder by Gene correlation of the GSE4290 dataset.

Table I.

KEGG PathwayFinder by Gene correlation of the GSE4290 dataset.

GroupIn_SetTotalPercentage, %P-value
Retrograde_endocannabinoid_signaling  79  9285.90 1.30×10−6
Nicotine_addiction  33  3594.30 6.10×10−5
PI3K_Akt_signaling_pathway16722574.20 6.70×10−5
GABAergic_synapse  65  7982.30 1.30×10−4
Synaptic_vesicle_cycle  51  6085.00 1.60×10−4
Glutamatergic_synapse  8310678.30 3.20×10−4
Dopaminergic_synapse  8811477.20 4.90×10−4
Axon_guidance  9412376.40 5.70×10−4
DNA_replication  32  3688.90 6.70×10−4
Circadian_entrainment  65  8279.30 8.30×10−4
Morphine_addiction  66  8478.60 1.10×10−3
Long_term_potentiation  49  6081.70 1.20×10−3
Spliceosome  8911875.40 1.60×10−3
Cholinergic_synapse  74  9776.30 2.40×10−3
Endocytosis16723670.80 2.80×10−3
Pancreatic_cancer  52  6678.80 3.50×10−3
Protein_processing_in_endoplasmic_reticulum11015172.80 3.50×10−3
Serotonergic_synapse  67  8876.10 4.20×10−3
Amphetamine_addiction  46  5879.30 4.80×10−3
Oxytocin_signaling_pathway10214072.90 4.90×10−3
Long_term_depression  45  5778.90 6.20×10−3
Amyotrophic_lateral_sclerosis__ALS_  40  5080.00 6.60×10−3
Aldosterone_synthesis_and_secretion  50  6576.90 9.60×10−3
Fc_gamma_R_mediated_phagocytosis  63  8475.00 9.80×10−3
Hepatitis_B  9813672.10 9.90×10−3

[i] In_Set, number of genes in pathway that significantly correlate with Ngb expression; Total, total number of genes in pathway; Percentage, %, ratio of Ngb-associated genes in total number of genes in pathway.

Ngb regulates the PI3K/AKT pathway in glioma cells

Subsequently, the present study investigated the activation of the PI3K/AKT pathway following modulation of Ngb levels by transfection. Ngb was silenced in U251 cells following transfection with shRNA targeting Ngb, compared with the NT shRNA group, as protein levels were reduced (P<0.05; Fig. 1). In addition, Ngb knockdown notably reduced the level of p-AKT (Ser473) compared with the NT shRNA group, without affecting total AKT levels, in U251 cells (P<0.05; Fig. 1). Furthermore, p-mTOR (Ser2448), Bcl-2 and Bax, which are downstream targets of the PI3K/ATK pathway, were also modulated by Ngb knockdown. Ngb knockdown led to reduced levels of p-mTOR (Ser2448) and Bcl-2, and increased BAX expression, in U251 cells, compared with the NT shRNA group (P<0.05; Fig. 1). By contrast, Ngb overexpression promoted the activation of the PI3K/AKT pathway, with increased levels of p-AKT (Ser473), p-mTOR (Ser2448) and Bcl-2, and decreased BAX expression, in U87 cells, compared with the empty vector control cells (P<0.05; Fig. 1). These data indicate that Ngb may promote the activation of the PI3K/AKT pathway in glioma cells.

Ngb promotes cellular malignant phenotypes of glioma by targeting the PI3K/AKT pathway

The present study further investigated whether Ngb regulated the proliferation and apoptosis of glioma cells through targeting the PI3K/AKT pathway. Overexpression of Ngb led to increased p-AKT expression, and reduced levels of cleaved PARP and cleaved caspase-3/7/8, in U87 cells, compared with cells transfected with empty vector (P<0.05; Fig. 2A). Functionally, Ngb overexpression promoted the proliferation and reduced the apoptosis of U87 cells, compared with cells transfected with empty vector (P<0.05; Fig. 2B-D). An AKT inhibitor, MK2206, was employed to block the activation of PI3K/AKT in Ngb-overexpressing U87 cells. MK2206 treatment led to reduced p-AKT expression and increased levels of cleaved PARP, cleaved caspase-3, cleaved caspase-7 and cleaved caspase-8 in Ngb-overexpressing U87 cells (P<0.05; Fig. 2A). Furthermore, MK2206 treatment reduced the proliferation and induced the apoptosis of Ngb-overexpressing U87 cells (P<0.05; Fig. 2B-D). Ngb knockdown using shRNA led to reduced expression of p-AKT, and increased levels of cleaved PARP and cleaved caspase-3/7/8, in U251 cells, compared with cells transfected with NT shRNA (P<0.05; Fig. 3A). In addition, Ngb knockdown reduced the proliferation and induced the apoptosis of U251 cells, compared with cells transfected with NT shRNA (P<0.05; Fig. 3B-D). IGF-1, an activator of the PI3K/AKT pathway, increased p-AKT levels, and decreased cleaved PARP and cleaved caspase-3/7/8 expression, in Ngb-knockdown U251 cells (P<0.05; Fig. 3A). Furthermore, IGF-1 treatment resulted in enhanced proliferation and reduced apoptosis in Ngb-knockdown U251 cells (P<0.05; Fig. 3A-D). Therefore, these results further confirm that Ngb may promote a cellular malignant phenotype in glioma, which may occur via the PI3K/AKT pathway.

Discussion

The expression and role of Ngb in human cancer is a novel and controversial topic. Several studies have reported that Ngb may protect against oxidative stress-induced cell injury in brain cancer (4,5,7). In addition, our previous study demonstrated that Ngb was overexpressed in glioma tissues compared with normal brain tissues, and its overexpression was associated with poor prognostic features and shorter overall survival (15). Ngb has also been reported to promote the proliferation and inhibit the apoptosis of glioma cells in vitro and in vivo (15). However, the potential mechanisms underlying the effects of Ngb in glioma are yet to be established. The present study investigated the molecular mechanisms involved in the antiapoptotic effect of Ngb in glioma cells. KEGG PathwayFinder by gene expression analysis using the R2: Genomics Analysis and Visualization Platform revealed that Ngb was associated with the PI3K/AKT pathway in glioma tissues from the GSE4290 dataset of the GEO database. Further experiments in the current study demonstrated that Ngb enhanced the activation of the PI3K/AKT pathway in glioma cells.

Aberrant activation of the PI3K/AKT pathway has been widely reported in various cancers types during progression, including glioma (2022). The PI3K/AKT pathway has roles in the cell proliferation, cell cycle progression and apoptosis resistance of glioma cells via its downstream targets, which include mTOR, Bcl-2, Bax, cyclin D1 and Bcl-2-like 1 (20,2326). mTOR was reported to have an essential role in the cell survival, proliferation and apoptosis of glioma cells (27,28). Bcl-2 is an antiapoptotic protein, while Bax is a proapoptotic factor. Altered Bcl-2/Bax expression was associated with altered apoptosis levels glioma cells (29). In the present study, treatment with the AKT inhibitor MK2206 blocked the activation of the PI3K/AKT pathway, and subsequently resulted in decreased proliferation and increased apoptosis in Ngb-overexpressing U87 cells. Furthermore, IGF-1 treatment in U251 cells with Ngb knockdown enhanced the PI3K/AKT pathway activation, cell proliferation and apoptosis resistance. Therefore, Ngb may promote malignant phenotypes of glioma cells by targeting the PI3K/AKT pathway.

In conclusion, the results of the present study demonstrated that Ngb enhanced the activation of the PI3K/AKT pathway in glioma cells. Furthermore, Ngb regulated the proliferation and apoptosis of glioma cells, and these effects may occur via the PI3K/AKT pathway. Therefore, Ngb may serve as a potential target for the treatment of glioma.

Acknowledgements

The present study was supported by the Scientific Research Plan Projects of Shaanxi Education Department (grant nos. 14JK1629, 2010JK811 and 11JK0715) and the Edge Discipline Construction Project in Shaanxi Province.

References

1 

Awad AJ, Burns TC, Zhang Y and Abounader R: Targeting MET for glioma therapy. Neurosurg Focus. 37:E102014. View Article : Google Scholar : PubMed/NCBI

2 

Huse JT and Aldape KD: The evolving role of molecular markers in the diagnosis and management of diffuse glioma. Clin Cancer Res. 20:5601–5611. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Chistiakov DA and Chekhonin VP: Extracellular vesicles shed by glioma cells: Pathogenic role and clinical value. Tumour Biol. 35:8425–8438. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Fordel E, Thijs L, Martinet W, Lenjou M, Laufs T, Van Bockstaele D, Moens L and Dewilde S: Neuroglobin and cytoglobin overexpression protects human SH-SY5Y neuroblastoma cells against oxidative stress-induced cell death. Neurosci Lett. 410:146–151. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Fordel E, Thijs L, Martinet W, Schrijvers D, Moens L and Dewilde S: Anoxia or oxygen and glucose deprivation in SH-SY5Y cells: A step closer to the unraveling of neuroglobin and cytoglobin functions. Gene. 398:114–122. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Peroni D, Negro A, Bähr M and Dietz GP: Intracellular delivery of Neuroglobin using HIV-1 TAT protein transduction domain fails to protect against oxygen and glucose deprivation. Neurosci Lett. 421:110–114. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Emara M, Salloum N and Allalunis-Turner J: Expression and hypoxic up-regulation of neuroglobin in human glioblastoma cells. Mol Oncol. 3:45–53. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Emara M, Turner AR and Allalunis-Turner J: Hypoxic regulation of cytoglobin and neuroglobin expression in human normal and tumor tissues. Cancer Cell Int. 10:332010. View Article : Google Scholar : PubMed/NCBI

9 

Oleksiewicz U, Daskoulidou N, Liloglou T, Tasopoulou K, Bryan J, Gosney JR, Field JK and Xinarianos G: Neuroglobin and myoglobin in non-small cell lung cancer: Expression, regulation and prognosis. Lung Cancer. 74:411–418. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Fiocchetti M, Nuzzo MT, Totta P, Acconcia F, Ascenzi P and Marino M: Neuroglobin, a pro-survival player in estrogen receptor α-positive cancer cells. Cell Death Dis. 5:e14492014. View Article : Google Scholar : PubMed/NCBI

11 

Fiocchetti M, Cipolletti M, Leone S, Naldini A, Carraro F, Giordano D, Verde C, Ascenzi P and Marino M: Neuroglobin in breast cancer cells: Effect of hypoxia and oxidative stress on protein level, localization, and anti-apoptotic function. PLoS One. 11:e01549592016. View Article : Google Scholar : PubMed/NCBI

12 

Fiocchetti M, Cipolletti M, Leone S, Ascenzi P and Marino M: Neuroglobin overexpression induced by the 17β-Estradiol-Estrogen receptor-α pathway reduces the sensitivity of MCF-7 breast cancer cell to paclitaxel. IUBMB Life. 68:645–651. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Fiocchetti M, Camilli G, Acconcia F, Leone S, Ascenzi P and Marino M: ERβ-dependent neuroglobin up-regulation impairs 17β-estradiol-induced apoptosis in DLD-1 colon cancer cells upon oxidative stress injury. J Steroid Biochem Mol Biol. 149:128–137. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Zhang J, Lan SJ, Liu QR, Liu JM and Chen XQ: Neuroglobin, a novel intracellular hexa-coordinated globin, functions as a tumor suppressor in hepatocellular carcinoma via Raf/MAPK/Erk. Mol Pharmacol. 83:1109–1119. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Zhang B, Chang M, Wang J and Liu Y: Neuroglobin functions as a prognostic marker and promotes the tumor growth of glioma via suppressing apoptosis. Biomed Pharmacother. 88:173–180. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Tu K, Li J, Verma VK, Liu C, Billadeau DD, Lamprecht G, Xiang X, Guo L, Dhanasekaran R, Roberts LR, et al: Vasodilator-stimulated phosphoprotein promotes activation of hepatic stellate cells by regulating Rab11-dependent plasma membrane targeting of transforming growth factor beta receptors. Hepatology. 61:361–374. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Riviere I, Brose K and Mulligan RC: Effects of retroviral vector design on expression of human adenosine deaminase in murine bone marrow transplant recipients engrafted with genetically modified cells. Proc Natl Acad Sci USA. 92:pp. 6733–6737. 1995; View Article : Google Scholar : PubMed/NCBI

18 

Colicelli J and Goff SP: Isolation of a recombinant murine leukemia virus utilizing a new primer tRNA. J Virol. 57:37–45. 1986.PubMed/NCBI

19 

Sun L, Hui AM, Su Q, Vortmeyer A, Kotliarov Y, Pastorino S, Passaniti A, Menon J, Walling J, Bailey R, et al: Neuronal and glioma-derived stem cell factor induces angiogenesis within the brain. Cancer Cell. 9:287–300. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Liu M and Wang J, Qi Q, Huang B, Chen A, Li X and Wang J: Nitidine chloride inhibits the malignant behavior of human glioblastoma cells by targeting the PI3K/AKT/mTOR signaling pathway. Oncol Rep. 36:2160–2168. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Yang J, Yang Q, Yu J, Li X, Yu S and Zhang X: SPOCK1 promotes the proliferation, migration and invasion of glioma cells through PI3K/AKT and Wnt/β-catenin signaling pathways. Oncol Rep. 35:3566–3576. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Yu JS and Cui W: Proliferation, survival and metabolism: The role of PI3K/AKT/mTOR signalling in pluripotency and cell fate determination. Development. 143:3050–3060. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Luo M, Liu Q, He M, Yu Z, Pi R, Li M, Yang X, Wang S and Liu A: Gartanin induces cell cycle arrest and autophagy and suppresses migration involving PI3K/Akt/mTOR and MAPK signalling pathway in human glioma cells. J Cell Mol Med. 21:46–57. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Zanotto-Filho A, Braganhol E, Battastini AM and Moreira JC: Proteasome inhibitor MG132 induces selective apoptosis in glioblastoma cells through inhibition of PI3K/Akt and NFkappaB pathways, mitochondrial dysfunction, and activation of p38-JNK1/2 signaling. Invest New Drugs. 30:2252–2262. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Yu Z, Xie G, Zhou G, Cheng Y, Zhang G, Yao G, Chen Y, Li Y and Zhao G: NVP-BEZ235, a novel dual PI3K-mTOR inhibitor displays anti-glioma activity and reduces chemoresistance to temozolomide in human glioma cells. Cancer Lett. 367:58–68. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Ding L, Ding L, Wang S, Wang S, Wang W, Wang W, Lv P, Lv P, Zhao D, Zhao D, et al: Tanshinone IIA affects autophagy and apoptosis of glioma cells by inhibiting phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling pathway. Pharmacology. 99:188–195. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Zhao N, Guo Y, Zhang M, Lin L and Zheng Z: Akt-mTOR signaling is involved in Notch-1-mediated glioma cell survival and proliferation. Oncol Rep. 23:1443–1447. 2010.PubMed/NCBI

28 

Koul N, Sharma V, Dixit D, Ghosh S and Sen E: Bicyclic triterpenoid Iripallidal induces apoptosis and inhibits Akt/mTOR pathway in glioma cells. BMC Cancer. 10:3282010. View Article : Google Scholar : PubMed/NCBI

29 

Wang P, Zhen H, Jiang X, Zhang W, Cheng X, Guo G, Mao X and Zhang X: Boron neutron capture therapy induces apoptosis of glioma cells through Bcl-2/Bax. BMC Cancer. 10:6612010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang B, Liu Y, Li Y, Zhe X, Zhang S and Zhang L: Neuroglobin promotes the proliferation and suppresses the apoptosis of glioma cells by activating the PI3K/AKT pathway. Mol Med Rep 17: 2757-2763, 2018
APA
Zhang, B., Liu, Y., Li, Y., Zhe, X., Zhang, S., & Zhang, L. (2018). Neuroglobin promotes the proliferation and suppresses the apoptosis of glioma cells by activating the PI3K/AKT pathway. Molecular Medicine Reports, 17, 2757-2763. https://doi.org/10.3892/mmr.2017.8132
MLA
Zhang, B., Liu, Y., Li, Y., Zhe, X., Zhang, S., Zhang, L."Neuroglobin promotes the proliferation and suppresses the apoptosis of glioma cells by activating the PI3K/AKT pathway". Molecular Medicine Reports 17.2 (2018): 2757-2763.
Chicago
Zhang, B., Liu, Y., Li, Y., Zhe, X., Zhang, S., Zhang, L."Neuroglobin promotes the proliferation and suppresses the apoptosis of glioma cells by activating the PI3K/AKT pathway". Molecular Medicine Reports 17, no. 2 (2018): 2757-2763. https://doi.org/10.3892/mmr.2017.8132