DOI QR코드

DOI QR Code

Memantine Induces NMDAR1-Mediated Autophagic Cell Death in Malignant Glioma Cells

  • Yoon, Wan-Soo (Department of Neurosurgery, Incheon St. Mary's Hospital, The Catholic University of Korea) ;
  • Yeom, Mi-Young (Clinical Research Laboratory, Incheon St. Mary's Hospital, The Catholic University of Korea) ;
  • Kang, Eun-Sun (Clinical Research Laboratory, Incheon St. Mary's Hospital, The Catholic University of Korea) ;
  • Chung, Yong-An (Institute of Catholic Integrative Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea) ;
  • Chung, Dong-Sup (Department of Neurosurgery, Incheon St. Mary's Hospital, The Catholic University of Korea) ;
  • Jeun, Sin-Soo (Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea)
  • Received : 2016.01.20
  • Accepted : 2016.08.30
  • Published : 2017.03.01

Abstract

Objective : Autophagy is one of the key responses of cells to programmed cell death. Memantine, an approved anti-dementia drug, has an antiproliferative effect on cancer cells but the mechanism is poorly understood. The aim of the present study was to test the possibility of induction of autophagic cell death by memantine in glioma cell lines. Methods : Glioma cell lines (T-98 G and U-251 MG) were used for this study. Results : The antiproliferative effect of memantine was shown on T-98 G cells, which expressed N-methyl-D-aspartate 1 receptor (NMDAR1). Memantine increased the autophagic-related proteins as the conversion ratio of light chain protein 3-II (LC3-II)-/LC3-I and the expression of beclin-1. Memantine also increased formation of autophagic vacuoles observed under a transmission electron microscope. Transfection of small interfering RNA (siRNA) to knock down NMDAR1 in the glioma cells induced resistance to memantine and decreased the LC3-II/LC3-I ratio in T-98 G cells. Conclusion : Our study demonstrates that in glioma cells, memantine inhibits proliferation and induces autophagy mediated by NMDAR1.

Keywords

References

  1. Abdul M, Hoosein N : N-methyl-D-aspartate receptor in human prostate cancer. J Membr Biol 205 : 125-128, 2005 https://doi.org/10.1007/s00232-005-0777-0
  2. Adamson C, Kanu OO, Mehta AI, Di C, Lin N, Mattox AK, et al. : Glioblastoma multiforme: a review of where we have been and where we are going. Expert Opin Investig Drugs 18 : 1061-1083, 2009 https://doi.org/10.1517/13543780903052764
  3. Behrens PF, Langemann H, Strohschein R, Draeger J, Hennig J : Extracellular glutamate and other metabolites in and around RG2 rat glioma: an intracerebral microdialysis study. J Neurooncol 47 : 11-22, 2000 https://doi.org/10.1023/A:1006426917654
  4. Besshoh S, Bawa D, Teves L, Wallace MC, Gurd JW : Increased phosphorylation and redistribution of NMDA receptors between synaptic lipid rafts and postsynaptic densities following transient global ischemia in the rat brain. J Neurochem 93 : 186-194, 2005 https://doi.org/10.1111/j.1471-4159.2004.03009.x
  5. Bigford GE, Alonso OF, Dietrich D, Keane RW : A novel protein complex in membrane rafts linking the NR2B glutamate receptor and autophagy is disrupted following traumatic brain injury. J Neurotrauma 26 : 703-720, 2009 https://doi.org/10.1089/neu.2008.0783
  6. Chen M, Lu TJ, Chen XJ, Zhou Y, Chen Q, Feng XY, et al. : Differential roles of NMDA receptor subtypes in ischemic neuronal cell death and ischemic tolerance. Stroke 39 : 3042-3048, 2008 https://doi.org/10.1161/STROKEAHA.108.521898
  7. de Groot J, Sontheimer H : Glutamate and the biology of gliomas. Glia 59 : 1181-1189, 2011 https://doi.org/10.1002/glia.21113
  8. de Groot JF, Piao Y, Lu L, Fuller GN, Yung WK : Knockdown of GluR1 expression by RNA interference inhibits glioma proliferation. J Neurooncol 88 : 121-133, 2008 https://doi.org/10.1007/s11060-008-9552-2
  9. Fan G, Sun B, Wu Z, Guo Q, Guo Y : In vivo single-voxel proton MR spectroscopy in the differentiation of high-grade gliomas and solitary metastases. Clin Radiol 59 : 77-85, 2004 https://doi.org/10.1016/j.crad.2003.08.006
  10. Ishiuchi S, Yoshida Y, Sugawara K, Aihara M, Ohtani T, Watanabe T, et al. : Ca2+-permeable AMPA receptors regulate growth of human glioblastoma via Akt activation. J Neurosci 27 : 7987-8001, 2007 https://doi.org/10.1523/JNEUROSCI.2180-07.2007
  11. Johnson JW, Kotermanski SE : Mechanism of action of memantine. Curr Opin Pharmacol 6 : 61-67, 2006 https://doi.org/10.1016/j.coph.2005.09.007
  12. Kihara K, Kabeya Y, Ohsumi Y, Yoshimori T : Beclin-phosphatidylinositol 3-kinase complex functions at the trans-Golgi network. EMBO Rep 2 : 330-335, 2001 https://doi.org/10.1093/embo-reports/kve061
  13. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, et al. : Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 8 : 445-544, 2012 https://doi.org/10.4161/auto.19496
  14. Liu Y, Wong TP, Aarts M, Rooyakkers A, Liu L, Lai TW, et al. : NMDA receptor subunits have differential roles in mediating excitotoxic neuronal death both in vitro and in vivo. J Neurosci 27 : 2846-2857, 2007 https://doi.org/10.1523/JNEUROSCI.0116-07.2007
  15. McComb RD, Burger PC : Pathologic analysis of primary brain tumors. Neurol Clin 3 : 711-728, 1985 https://doi.org/10.1016/S0733-8619(18)31005-3
  16. Moots PL, Maciunas RJ, Eisert DR, Parker RA, Laporte K, Abou-Khalil B : The course of seizure disorders in patients with malignant gliomas. Arch Neurol 52 : 717-724, 1995 https://doi.org/10.1001/archneur.1995.00540310091021
  17. North WG, Gao G, Jensen A, Memoli VA, Du J : NMDA receptors are expressed by small-cell lung cancer and are potential targets for effective treatment. Clin Pharmacol 2 : 31-40, 2010
  18. Piao Y, Lu L, de Groot J : AMPA receptors promote perivascular glioma invasion via beta1 integrin dependent adhesion to the extracellular matrix. Neuro Oncol 11 : 260-273, 2009 https://doi.org/10.1215/15228517-2008-094
  19. Rijpkema M, Schuuring J, van der MY, van der GM, Bernsen H, Boerman R, et al. : Characterization of oligodendrogliomas using short echo time 1H MR spectroscopic imaging. NMR Biomed 16 : 12-18, 2003 https://doi.org/10.1002/nbm.807
  20. Rothman SM, Olney JW : Excitotoxicity and the NMDA receptor-still lethal after eight years. Trends Neurosci 18 : 57-58, 1995
  21. Ruban A, Berkutzki T, Cooper I, Mohar B, Teichberg VI : Blood glutamate scavengers prolong the survival of rats and mice with brain-implanted gliomas. Invest New Drugs 30 : 2226-2235, 2012 https://doi.org/10.1007/s10637-012-9794-x
  22. Rzeski W, Turski L, Ikonomidou C : Glutamate antagonists limit tumor growth. Proc Natl Acad Sci USA 98 : 6372-6377, 2001 https://doi.org/10.1073/pnas.091113598
  23. Schunemann DP, Grivicich I, Regner A, Leal LF, de Araujo DR, Jotz GP, et al. : Glutamate promotes cell growth by EGFR signaling on U-87MG human glioblastoma cell line. Pathol Oncol Res 16 : 285-293, 2010 https://doi.org/10.1007/s12253-009-9223-4
  24. Sontheimer H : A role for glutamate in growth and invasion of primary brain tumors. J Neurochem 105 : 287-295, 2008 https://doi.org/10.1111/j.1471-4159.2008.05301.x
  25. Stepulak A, Sifringer M, Rzeski W, Endesfelder S, Gratopp A, Pohl EE, et al. : NMDA antagonist inhibits the extracellular signal-regulated kinase pathway and suppresses cancer growth. Proc Natl Acad Sci USA 102 : 15605-15610, 2005 https://doi.org/10.1073/pnas.0507679102
  26. Surawicz TS, Davis F, Freels S, Laws ER Jr, Menck HR : Brain tumor survival: results from the National Cancer Data Base. J Neurooncol 40 : 151-160, 1998 https://doi.org/10.1023/A:1006091608586
  27. Takano T, Lin JH, Arcuino G, Gao Q, Yang J, Nedergaard M : Glutamate release promotes growth of malignant gliomas. Nat Med 7 : 1010-1015, 2001 https://doi.org/10.1038/nm0901-1010
  28. Watanabe K, Kanno T, Oshima T, Miwa H, Tashiro C, Nishizaki T : The NMDA receptor NR2A subunit regulates proliferation of MKN45 human gastric cancer cells. Biochem Biophys Res Commun 367 : 487-490, 2008 https://doi.org/10.1016/j.bbrc.2007.12.167
  29. Witt A, Macdonald N, Kirkpatrick P : Memantine hydrochloride. Nat Rev Drug Discov 3 : 109-110, 2004 https://doi.org/10.1038/nrd1311
  30. Ye ZC, Sontheimer H : Glioma cells release excitotoxic concentrations of glutamate. Cancer Res 59 : 4383-4391, 1999

Cited by

  1. Glutamate Receptors and Glioblastoma Multiforme: An Old “Route” for New Perspectives vol.20, pp.7, 2017, https://doi.org/10.3390/ijms20071796
  2. Inhibitory effects of Dulcitol on rat C6 glioma by regulating autophagy pathway vol.34, pp.10, 2017, https://doi.org/10.1080/14786419.2018.1512994
  3. Transcriptional block of AMPK-induced autophagy promotes glutamate excitotoxicity in nutrient-deprived SH-SY5Y neuroblastoma cells vol.77, pp.17, 2017, https://doi.org/10.1007/s00018-019-03356-2
  4. Memantine for Prevention of Brain Irradiation–Induced Cognitive Toxicity: A Tale of an Underappreciated and Underused Intervention vol.6, pp.None, 2017, https://doi.org/10.1200/go.20.00342
  5. Interactions between Tumor Cells, Neurons, and Microglia in the Glioma Microenvironment vol.21, pp.22, 2017, https://doi.org/10.3390/ijms21228476
  6. Memantine activates LKB1-AMPK pathway in breast carcinoma cells vol.41, pp.4, 2017, https://doi.org/10.17776/csj.768907
  7. Memantine activates LKB1-AMPK pathway in breast carcinoma cells vol.41, pp.4, 2017, https://doi.org/10.17776/csj.768907
  8. Calcium Ions Aggravate Alzheimer’s Disease Through the Aberrant Activation of Neuronal Networks, Leading to Synaptic and Cognitive Deficits vol.14, pp.None, 2017, https://doi.org/10.3389/fnmol.2021.757515
  9. Transcriptomic signatures of treatment response to the combination of escitalopram and memantine or placebo in late-life depression vol.26, pp.9, 2017, https://doi.org/10.1038/s41380-020-0752-2