207
Views
2
CrossRef citations to date
0
Altmetric
Original Article

Everolimus attenuates glutamate-induced PC12 cells death

, , , , &
Pages 457-466 | Received 02 Mar 2020, Accepted 01 May 2021, Published online: 02 Feb 2023

References

  • Hu Y, Li J, Liu P, et al. Protection of SH-SY5Y neuronal cells from glutamate-induced apoptosis by 3,6’-disinapoyl sucrose, a bioactive compound isolated from Radix Polygala. J Biomed Biotechnol. 2012;2012:1–5.
  • Lau A, Tymianski M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch. 2010;460(2):525–542.
  • Froissard P, Duval D. Cytotoxic effects of glutamic acid on PC12 cells. Neurochem Int. 1994;24(5):485–493.
  • Gerstner B, Lee J, DeSilva TM, et al. 17beta-estradiol protects against hypoxic/ischemic white matter damage in the neonatal rat brain. J Neurosci Res. 2009;87(9):2078–2086.
  • Fasolo A, Sessa C. mTOR inhibitors in the treatment of cancer. Expert Opin Investig Drugs. 2008;17(11):1717–1734.
  • Lebwohl D, Anak Ö, Sahmoud T, et al. Development of everolimus, a novel oral mTOR inhibitor, across a spectrum of diseases. Ann N Y Acad Sci. 2013;1291:14–32.
  • Bockaert J, Marin P. mTOR in brain physiology and pathologies. Physiol Rev. 2015;95(4):1157–1187.
  • Lipton JO, Sahin M. The neurology of mTOR. Neuron. 2014;84(2):275–291.
  • McCabe MP, Cullen ER, Barrows CM, et al. Genetic inactivation of mTORC1 or mTORC2 in neurons reveals distinct functions in glutamatergic synaptic transmission. Elife. 2020;9:e51440–e51469.
  • Takei N, Nawa H. mTOR signaling and its roles in normal and abnormal brain development. Front Mol Neurosci. 2014;28–40.
  • Aoki Y, Nakahara T, Asano D, et al. Preventive effects of rapamycin on inflammation and capillary degeneration in a rat model of NMDA-induced retinal injury. Biol Pharm Bull. 2015;38(2):321–324.
  • Erlich S, Alexandrovich A, Shohami E, et al. Rapamycin is a neuroprotective treatment for traumatic brain injury. Neurobiol Dis. 2007;26(1):86–93.
  • Su W, Li Z, Jia Y, et al. Rapamycin is neuroprotective in a rat chronic hypertensive glaucoma model. PLoS One. 2014;9(6):e99719.
  • Huiras P, Gabardi S. Everolimus: a review of its pharmacologic properties and use in solid organ transplantation. Rev Health Care. 2011;2(4):229–241.
  • Suvanish Kumar VS, Pretorius E, Rajanikant GK. The Synergistic Combination of Everolimus and Paroxetine Exerts Post-ischemic Neuroprotection In Vitro. Cell Mol Neurobiol. 2018;38(7):1383–1397.
  • Saliba SW, Vieira EL, Santos RP, et al. Neuroprotective effects of intrastriatal injection of rapamycin in a mouse model of excitotoxicity induced by quinolinic acid. J Neuroinflammation. 2017;14(1):25–37.
  • Chauhan A, Sharma U, Jagannathan NR, et al. Rapamycin protects against middle cerebral artery occlusion induced focal cerebral ischemia in rats. Behav Brain Res. 2011;225(2):603–609.
  • Chen HC, Fong TH, Hsu PW, et al. Multifaceted effects of rapamycin on functional recovery after spinal cord injury in rats through autophagy promotion, anti-inflammation, and neuroprotection. J Surg Res. 2013;179(1):e203-210–e210.
  • Bove J, Martinez-Vicente M, Vila M. Fighting neurodegeneration with rapamycin: mechanistic insights. Nat Rev Neurosci. 2011;12(8):437–452.
  • Dai C, Ciccotosto GD, Cappai R, et al. Rapamycin Confers Neuroprotection against Colistin-Induced Oxidative Stress, Mitochondria Dysfunction, and Apoptosis through the Activation of Autophagy and mTOR/Akt/CREB Signaling Pathways. ACS Chem Neurosci. 2018;9(4):824–837.
  • Hayashi I, Aoki Y, Asano D, et al. Protective Effects of Everolimus against N-Methyl-D-aspartic Acid-Induced Retinal Damage in Rats. Biol Pharm Bull. 2015;38(11):1765–1771.
  • Ichikawa A, Nakahara T, Kurauchi Y, et al. Rapamycin prevents N-methyl-D-aspartate-induced retinal damage through an ERK-dependent mechanism in rats. J Neurosci Res. 2014;92(6):692–702.
  • Fanoudi S, Hosseini M, Alavi MS, et al. Everolimus, a mammalian target of rapamycin inhibitor, ameliorated streptozotocin-induced learning and memory deficits via neurochemical alterations in male rats. Excli J. 2018;17:999–1017.
  • Sadowski K, Kotulska-Jóźwiak K, Jóźwiak S. Role of mTOR inhibitors in epilepsy treatment. Pharmacol Rep. 2015;67(3):636–646.
  • Yang M-T, Lin Y-C, Ho W-H, et al. Everolimus is better than rapamycin in attenuating neuroinflammation in kainic acid-induced seizures. J Neuroinflammation. 2017;14(1):15–25.
  • Gorshtein A, Rubinfeld H, Kendler E, et al. Mammalian target of rapamycin inhibitors rapamycin and RAD001 (everolimus) induce anti-proliferative effects in GH-secreting pituitary tumor cells in vitro. Endocr Relat Cancer. 2009;16(3):1017–1027.
  • Witzig TE, Reeder C, Han JJ, et al. The mTORC1 inhibitor everolimus has anti-tumor activity in vitro and produces tumor responses in patients with relapsed T-cell lymphoma. Blood: blood. 2015;126(3):328–335. ) 2015-2002-629543
  • Hosseini A, Shafiee-Nick R, Mousavi SH. Combination of Nigella sativa with Glycyrrhiza glabra and Zingiber officinale augments their protective effects on doxorubicin-induced toxicity in h9c2 cells. Iran J Basic Med Sci. 2014;17(12):993–1000.
  • Yu W, Sheng M, Xu R, et al. Berberine protects human renal proximal tubular cells from hypoxia/reoxygenation injury via inhibiting endoplasmic reticulum and mitochondrial stress pathways. J Transl Med. 2013;11(1):25–35.
  • Ghorbani A, Sadeghnia HR, Asadpour E. Mechanism of protective effect of lettuce against glucose/serum deprivation-induced neurotoxicity. Nutr Neurosci. 2015;18(3):103–109.
  • Rajabian A, Boroushaki MT, Hayatdavoudi P, et al. Boswellia serrata Protects Against Glutamate-Induced Oxidative Stress and Apoptosis in PC12 and N2a Cells. DNA Cell Biol. 2016;35(11):666–679.
  • Wang C-J, Hu C-P, Xu K-P, et al. Protective effect of selaginellin on glutamate-induced cytotoxicity and apoptosis in differentiated PC12 cells. Naunyn Schmiedebergs Arch Pharmacol. 2010;381(1):73–81.
  • Li N, Liu B, Dluzen DE, et al. Protective effects of ginsenoside Rg2 against glutamate-induced neurotoxicity in PC12 cells. J Ethnopharmacol. 2007;111(3):458–463.
  • Penugonda S, Mare S, Goldstein G, et al. Effects of N-acetylcysteine amide (NACA), a novel thiol antioxidant against glutamate-induced cytotoxicity in neuronal cell line PC12. Brain Res. 2005;1056(2):132–138.
  • Sadeghnia HR, Rajabian A, Ghorbani A, et al. Effects of standardized extract of Ferula gummosa root on glutamate-induced neurotoxicity. Folia Neuropathol. 2017;55(4):340–346.
  • Tan JW, Tham CL, Israf DA, et al. Neuroprotective effects of biochanin A against glutamate-induced cytotoxicity in PC12 cells via apoptosis inhibition. Neurochem Res. 2013;38(3):512–518.
  • Zhang Y, Wang W, Hao C, et al. Astaxanthin protects PC12 cells from glutamate-induced neurotoxicity through multiple signaling pathways. J Funct Foods. 2015;16:137–151.
  • Zhao H, Yenari MA, Cheng D, et al. Bcl-2 overexpression protects against neuron loss within the ischemic margin following experimental stroke and inhibits cytochrome c translocation and caspase-3 activity. J Neurochem. 2003;85(4):1026–1036.
  • Song Y, Xue H, Liu T, et al. Rapamycin plays a neuroprotective effect after spinal cord injury via anti-inflammatory effects. J Biochem Mol Toxicol. 2015;29(1):29–34.
  • Malagelada C, Jin ZH, Jackson-Lewis V, et al. Rapamycin protects against neuron death in in vitro and in vivo models of Parkinson’s disease. J Neurosci. 2010;30(3):1166–1175.
  • Yin L, Ye S, Chen Z, et al. Rapamycin preconditioning attenuates transient focal cerebral ischemia/reperfusion injury in mice. Int J Neurosci. 2012;122(12):748–756.
  • Caccamo A, Majumder S, Deng JJ, et al. Rapamycin rescues TDP-43 mislocalization and the associated low molecular mass neurofilament instability. J Biol Chem. 2009;284(40):27416–27424.
  • Gonzalez-Mejia ME, Morales M, Hernandez-Kelly LC, et al. Glutamate-dependent translational regulation in cultured Bergmann glia cells: involvement of p70S6K. Neuroscience. 2006;141(3):1389–1398.
  • Magnuson B, Ekim B, Fingar DC. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J. 2012;441(1):1–21.
  • Zepeda RC, Barrera I, Castelan F, et al. Glutamate-dependent phosphorylation of the mammalian target of rapamycin (mTOR) in Bergmann glial cells. Neurochem Int. 2009;55(5):282–287.
  • Kimura R, Okouchi M, Fujioka H, et al. Glucagon-like peptide-1 (GLP-1) protects against methylglyoxal-induced PC12 cell apoptosis through the PI3K/Akt/mTOR/GCLc/redox signaling pathway. Neuroscience. 2009;162(4):1212–1219.
  • Gong R, Park CS, Abbassi NR, et al. Roles of glutamate receptors and the mammalian target of rapamycin (mTOR) signaling pathway in activity-dependent dendritic protein synthesis in hippocampal neurons. J Biol Chem. 2006;281(27):18802–18815.
  • Ryskalin L, Lazzeri G, Flaibani M, et al. 2017. mTOR-dependent cell proliferation in the brain. 2017
  • Jiang Q, Gu Z, Zhang G, et al. Diphosphorylation and involvement of extracellular signal-regulated kinases (ERK1/2) in glutamate-induced apoptotic-like death in cultured rat cortical neurons. Brain Res. 2000;857(1-2):71–77.
  • Molz S, Dal‐Cim T, Budni J, et al. Neuroprotective effect of guanosine against glutamate-induced cell death in rat hippocampal slices is mediated by the phosphatidylinositol-3 kinase/Akt/ glycogen synthase kinase 3β pathway activation and inducible nitric oxide synthase inhibition . J Neurosci Res. 2011;89(9):1400–1408.
  • Pi R, Li W, Lee NT, et al. Minocycline prevents glutamate‐induced apoptosis of cerebellar granule neurons by differential regulation of p38 and Akt pathways. J Neurochem. 2004;91(5):1219–1230.
  • Szydlowska K, Gozdz A, Dabrowski M, et al. Prolonged activation of ERK triggers glutamate-induced apoptosis of astrocytes: neuroprotective effect of FK506 . J Neurochem. 2010;113(4):904–918.
  • Abd-Elrahman KS, Ferguson S. Modulation of mTOR and CREB pathways following mGluR5 blockade contribute to improved Huntington’s pathology in zQ 175 mice. 2019; 12: 1–9
  • Zhang C, Yuan X-r, Li H-y, et al. Anti-cancer effect of metabotropic glutamate receptor 1 inhibition in human glioma U87 cells: involvement of PI3K/Akt/mTOR pathway. Cell Physiol Biochem. 2015;35(2):419–432.
  • Zhu M, Qin YC, Gao CQ, et al. Extracellular Glutamate-Induced mTORC1 Activation via the IR/IRS/PI3K/Akt Pathway Enhances the Expansion of Porcine Intestinal Stem Cells. J Agric Food Chem. 2019;67(34):9510–9521.
  • Li X-G, Sui W-G, Gao C-Q, et al. L-Glutamate deficiency can trigger proliferation inhibition via down regulation of the mTOR/S6K1 pathway in pig intestinal epithelial cells. 2016;94:1541–1549.
  • Ye J-l, Gao C-q, Li X-g, et al. EAAT3 promotes amino acid transport and proliferation of porcine intestinal epithelial cells. Oncotarget. 2016;7(25):38681–38692.
  • Swiatkowski P, Nikolaeva I, Kumar G, et al. Role of Akt-independent mTORC1 and GSK3β signaling in sublethal NMDA-induced injury and the recovery of neuronal electrophysiology and survival. Sci Rep. 2017;7(1):1539–1554.
  • Li X, Wu C, Chen N, et al. PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma. Oncotarget. 2016;7(22):33440–33450.
  • Morrison R, Kinoshita Y. The role of p53 in neuronal cell death. Cell Death Differ. 2000;7(10):868–879.
  • Qin Z-H, Chen R-W, Wang Y, et al. Nuclear factor κB nuclear translocation upregulates c-Myc and p53 expression during NMDA receptor-mediated apoptosis in rat striatum. J Neurosci. 1999;19(10):4023–4033.
  • Xiang H, Hochman DW, Saya H, et al. Evidence for p53-mediated modulation of neuronal viability. J Neurosci. 1996;16(21):6753–6765.
  • Castedo M, Ferri KF, Blanco J, et al. Human Immunodeficiency Virus 1 Envelope Glycoprotein Complex-induced Apoptosis Involves Mammalian Target of rapamycin/FKBP12-rapamycin-associated protein-mediated p53 phosphorylation . J Exp Med. 2001;194(8):1097–1110.
  • Ding K, Wang H, Wu Y, et al. Rapamycin protects against apoptotic neuronal death and improves neurologic function after traumatic brain injury in mice via modulation of the mTOR-p53-Bax axis. J Surg Res. 2015;194(1):239–247.
  • Jiang J, Jiang J, Zuo Y, et al. Rapamycin protects the mitochondria against oxidative stress and apoptosis in a rat model of Parkinson’s disease. Int J Mol Med. 2013;31(4):825–832.
  • Demerlé-Pallardy C, Lonchampt M-O, Chabrier P-E, et al. Nitric oxide synthase induction in glial cells: effect on neuronal survival. Life Sci. 1993;52(23):1883–1890.
  • Dello Russo C, Lisi L, Tringali G, et al. Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem Pharmacol. 2009;78(9):1242–1251.
  • Lisi L, Navarra P, Feinstein DL, et al. The mTOR kinase inhibitor rapamycin decreases iNOS mRNA stability in astrocytes. J Neuroinflammation. 2011;8(1):1–11.
  • Fox JH, Connor T, Chopra V, et al. The mTOR kinase inhibitor Everolimus decreases S6 kinase phosphorylation but fails to reduce mutant huntingtin levels in brain and is not neuroprotective in the R6/2 mouse model of Huntington’s disease. Mol Neurodegener. 2010;5:26–37.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.