Original Article

Neuroblastoma Cell Death Induced by eEF1A2 Knockdown Is Possibly Mediated by the Inhibition of Akt and mTOR Phosphorylation

Abstract

Background The protein kinase B/mammalian target of the rapamycin (Akt/mTOR) pathway is one of the most potent prosurvival signaling cascades that is constitutively active in neuroblastoma. The eukaryotic translation elongation factor-1, alpha-2 (eEF1A2) protein has been found to activate the Akt/mTOR pathway. However, there is a lack of data on the role of eEF1A2 in neuroblastoma. The present study investigated the effect of eEF1A2 silencing on the viability of neuroblastoma cells and its possible signaling.

Materials and Methods: Human SH-SY5Y neuroblastoma cells were transfected with small interfering RNA (siRNA) against eEF1A2. After 48 h of transfection, cell viability was assessed using an MTT assay. The mRNA expression of p53, Bax, Bcl-2, caspase-3 and members of the phosphoinositide 3-kinases (PI3K)/Akt/mTOR pathway was determined using quantitative real-time RT-PCR (qRT-PCR). The protein expression of Akt and mTOR was measured using Western blot analysis.

Results: eEF1A2 knockdown significantly decreased the viability of neuroblastoma cells. No significant changes were observed on the expression of p53, Bax/Bcl-2 ratio, and caspase-3 mRNAs; however, the upregulated trends were noted for the p53 and Bax/Bcl-2 ratio. eEF1A2 knockdown significantly inhibited the phosphorylation of both Akt and mTOR. Almost, all of the class I (PIK3CA, PIK3CB, and PIK3CD) and all of the class II PI3K genes were slightly increased in tumor cells with eEF1A2 knockdown. In addition, a slightly decreased expression of the Akt2, mTORC1, and mTORC2 was observed.

Conclusion: eEF1A2 knockdown induced neuroblastoma cell death, in part through the inhibition of Akt and mTOR, suggesting a potential role of eEF1A2 as a molecular target for neuroblastoma therapy.

1. Matthay KK, Maris JM, Schleiermacher G, et al. Neuroblastoma. Nat Rev Dis Primers. 2016; 2: 16078.
2. Johnsen JI, Dyberg C, Wickstrom M. Neuroblastoma-A Neural Crest Derived Embryonal Malignancy. Front Mol Neurosci. 2019; 12: 9.
3. Kumar MD, Dravid A, Kumar A, et al. Gene therapy as a potential tool for treating neuroblastoma-a focused review. Cancer Gene Ther. 2016; 23(5) :115-24.
4. Zhu Q, Zhang Y, Liu Y, et al. MLIF Alleviates SH-SY5Y Neuroblastoma Injury Induced by Oxygen-Glucose Deprivation by Targeting Eukaryotic Translation Elongation Factor 1A2. PLoS One. 2016; 11(2): e0149965.
5. Khwanraj K, Madlah S, Grataitong K, et al. Comparative mRNA Expression of eEF1A Isoforms and a PI3K/Akt/mTOR Pathway in a Cellular Model of Parkinson's Disease. Parkinsons Dis. 2016; 2016: 8716016.
6. Lee MH, Surh YJ. eEF1A2 as a putative oncogene. Ann N Y Acad Sci. 2009; 1171: 87-93.
7. Amiri A, Noei F, Jeganathan S, et al. eEF1A2 activates Akt and stimulates Akt-dependent actin remodeling, invasion and migration. Oncogene. 2007; 26(21): 3027-40.
8. Tomlinson VA, Newbery HJ, Wray NR, et al. Translation elongation factor eEF1A2 is a potential oncoprotein that is overexpressed in two-thirds of breast tumours. BMC Cancer. 2005; 5: 113.
9. Anand N, Murthy S, Amann G, et al. Protein elongation factor EEF1A2 is a putative oncogene in ovarian cancer. Nat Genet. 2002; 31(3): 301-5.
10. Chambers DM, Peters J, Abbott CM. The lethal mutation of the mouse wasted (wst) is a deletion that abolishes expression of a tissue-specific isoform of translation elongation factor 1alpha, encoded by the Eef1a2 gene. Proc Natl Acad Sci U S A. 1998; 95(8): 4463-8.
11. Knudsen SM, Frydenberg J, Clark BF, et al. Tissue-dependent variation in the expression of elongation factor-1 alpha isoforms: isolation and characterisation of a cDNA encoding a novel variant of human elongation-factor 1 alpha. Eur J Biochem. 1993; 215(3): 549-54.
12. Lee S, Francoeur AM, Liu S, et al. Tissue-specific expression in mammalian brain, heart, and muscle of S1, a member of the elongation factor-1 alpha gene family. J Biol Chem. 1992; 267(33): 24064-8.
13. Browne GJ, Proud CG. Regulation of peptide-chain elongation in mammalian cells. Eur J Biochem. 2002; 269(22): 5360-8.
14. Chang R, Wang E. Mouse translation elongation factor eEF1A-2 interacts with Prdx-I to protect cells against apoptotic death induced by oxidative stress. J Cell Biochem. 2007; 100(2): 267-78.
15. Ruest LB, Marcotte R, Wang E. Peptide elongation factor eEF1A-2/S1 expression in cultured differentiated myotubes and its protective effect against caspase-3-mediated apoptosis. J Biol Chem. 2002; 277(7): 5418-25.
16. Gross SR, Kinzy TG. Translation elongation factor 1A is essential for regulation of the actin cytoskeleton and cell morphology. Nat Struct Mol Biol. 2005; 12(9): 772-8.
17. Li Z, Qi CF, Shin DM, et al. Eef1a2 promotes cell growth, inhibits apoptosis and activates JAK/STAT and AKT signaling in mouse plasmacytomas. PLoS One. 2010; 5(5): e10755.
18. Pellegrino R, Calvisi DF, Neumann O, et al. EEF1A2 inactivates p53 by way of PI3K/AKT/mTOR-dependent stabilization of MDM4 in hepatocellular carcinoma. Hepatology. 2014; 59(5): 1886-99.
19. Sun Y, Wong N, Guan Y, et al. The eukaryotic translation elongation factor eEF1A2 induces neoplastic properties and mediates tumorigenic effects of ZNF217 in precursor cells of human ovarian carcinomas. Int J Cancer. 2008; 123(8): 1761-9.
20. King D, Yeomanson D, Bryant HE. PI3King the lock: targeting the PI3K/Akt/mTOR pathway as a novel therapeutic strategy in neuroblastoma. J Pediatr Hematol Oncol. 2015; 37(4): 245-51.
21. Fulda S. The PI3K/Akt/mTOR pathway as therapeutic target in neuroblastoma. Curr Cancer Drug Targets. 2009; 9(6): 729-37.
22. Moreno-Smith M, Lakoma A, Chen Z, et al. p53 Nongenotoxic Activation and mTORC1 Inhibition Lead to Effective Combination for Neuroblastoma Therapy. Clin Cancer Res. 2017; 23(21): 6629-39.
23. Pourdehnad M, Truitt ML, Siddiqi IN, et al. Myc and mTOR converge on a common node in protein synthesis control that confers synthetic lethality in Myc-driven cancers. Proc Natl Acad Sci U S A. 2013; 110(29): 11988-93.
24. Opel D, Poremba C, Simon T, et al. Activation of Akt predicts poor outcome in neuroblastoma. Cancer Res. 2007; 67(2): 735-45.
25. Pei H, Li L, Fridley BL, et al. FKBP51 affects cancer cell response to chemotherapy by negatively regulating Akt. Cancer Cell. 2009; 16(3): 259-66.
26. Altomare DA, Tanno S, De Rienzo A, et al. Frequent activation of AKT2 kinase in human pancreatic carcinomas. J Cell Biochem. 2002; 87(4): 470-6.
27. Cheng GZ, Chan J, Wang Q, et al. Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel. Cancer Res. 2007; 67(5): 1979-87.
28. Kang J, Rychahou PG, Ishola TA, et al. N-myc is a novel regulator of PI3K-mediated VEGF expression in neuroblastoma. Oncogene. 2008; 27(28): 3999-4007.
29. Qiao J, Lee S, Paul P, et al. Akt2 regulates metastatic potential in neuroblastoma. PLoS One. 2013; 8(2): e56382.
30. Mohlin S, Hamidian A, von Stedingk K, et al. PI3K-mTORC2 but not PI3K-mTORC1 regulates transcription of HIF2A/EPAS1 and vascularization in neuroblastoma. Cancer Res. 2015; 75(21): 4617-28.
31. Zhang H, Dou J, Yu Y, et al. mTOR ATP-competitive inhibitor INK128 inhibits neuroblastoma growth via blocking mTORC signaling. Apoptosis. 2015; 20(1): 50-62.
32. Mavrommati I, Cisse O, Falasca M, et al. Novel roles for class II Phosphoinositide 3-Kinase C2beta in signalling pathways involved in prostate cancer cell invasion. Sci Rep. 2016; 6: 23277.
33. Budanov AV, Karin M. p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling. Cell. 2008; 134(3): 451-60.
34. Cui Q, Yu JH, Wu JN, et al. P53-mediated cell cycle arrest and apoptosis through a caspase-3- independent, but caspase-9-dependent pathway in oridonin-treated MCF-7 human breast cancer cells. Acta Pharmacol Sin. 2007; 28(7): 1057-66.
35. Lund A, Knudsen SM, Vissing H, et al. Assignment of human elongation factor 1alpha genes: EEF1A maps to chromosome 6q14 and EEF1A2 to 20q13.3. Genomics. 1996; 36(2): 359-61.
Files
IssueVol 15, No 4 (2021) QRcode
SectionOriginal Article(s)
DOI https://doi.org/10.18502/ijhoscr.v15i4.7477
Keywords
Eukaryotic translation elongation factor-1, alpha-2 (eEF1A2); Neuroblastoma; Small interfering RNA (siRNA); SH-SY5Y cells; Phosphoinositide 3-kinases (PI3K); Akt; mTOR; p53

Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
How to Cite
1.
Khwanraj K, Dharmasaroja P. Neuroblastoma Cell Death Induced by eEF1A2 Knockdown Is Possibly Mediated by the Inhibition of Akt and mTOR Phosphorylation. Int J Hematol Oncol Stem Cell Res. 2021;15(4):221-229.