J Korean Neurosurg Soc.  2017 Feb;60(2):130-137. 10.3340/jkns.2016.0101.006.

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

Affiliations
  • 1Department of Neurosurgery, Incheon St. Mary's Hospital, The Catholic University of Korea, Incheon, Korea.
  • 2Clinical Research Laboratory, Incheon St. Mary's Hospital, The Catholic University of Korea, Incheon, Korea.
  • 3Institute of Catholic Integrative Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, Incheon, Korea.
  • 4Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea. ssjeun@catholic.ac.kr

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.

Keyword

Memantine; Autophagy; N-methyl-D-aspartate; Glioma

MeSH Terms

Autophagy*
Cell Death
Cell Line
Gastrin-Secreting Cells
Glioma*
Memantine*
N-Methylaspartate
RNA, Small Interfering
Transfection
Vacuoles
Memantine
N-Methylaspartate
RNA, Small Interfering

Figure

  • Fig. 1 Memantine-induced cytotoxicity in glioma cell lines. After incubation with memantine (0–600 μM), malignant glioma cells were grown for 48 h starting at a density of 104/well. The number of viable cells was counted and expressed as a percentage of the untreated control. The results are representative of 3 independent experiments. *p<0.05, Student’s t-test.

  • Fig. 2 Expression levels of NMDAR1 and NMDAR2B in U-251 MG and T-98 G cells. The cell lines were analyzed using western blotting (A) and RT-PCR (B). NMDAR1 was present only in T-98 G cells, but NMDAR2B was detectable in both glioma cell lines. β-actin and GAPDH were used as internal controls. NMDAR: N-methyl-D-aspartate receptor, RT-PCR: reverse transcription polymerase chain reaction, GAPDH: glyceraldehyde-3-phosphate dehydrogenase.

  • Fig. 3 Memantine upregulated LC3-II and beclin-1 in T-98 G cells. A: Protein levels of LC3-I and LC3-II were analyzed in T-98 G cells treated with 0 and 400 μM memantine for 48 h. The graph shows the results of densitometric quantitation of western blots, presented as the LC3-II/LC3-I ratio relative to the control (0 μM of memantine). LC3-II was significantly increased by 400 μM memantine. B: Protein expression levels of AMPKα, beclin-1, APG5L, and ULK1 were evaluated using western blotting after incubation with memantine (0 and 400 μM) for 48 hr in T-98 G cells. The results of densitometric quantitation of immunoblots are presented as the ratio relative to the control (0 μM of memantine). Beclin-1 was significantly upregulated after the treatment with 400 μM of memantine. *p<0.05, Student’s t-test. AMPK: AMP-activated protein kinase.

  • Fig. 4 Memantine induced autophagic ultrastructural features in T-98 G cells according to TEM. A: The control cells showed the normal cytoplasm including mitochondria, ribosomes, and endoplasmic reticulum. B: After incubation with 400 μM memantine for 48 h, the cells developed autophagic vacuoles with lamellar structure in the cytoplasm. TEM: transmission electron microscopy.

  • Fig. 5 A knockdown of NMDAR1 using siRNA transfection in T-98 G cells. T-98 cells incubated in EMEM was used as the control value. A: After the transfection with anti-NMDAR1 siRNA (10 nM) for 48 hr, the expression of NMDAR1 was evaluated using western bloting. B: Cell viability of control and siRNA-transfected T-98 G cells was analyzed after incubation with various concentration of memantine (0–600 μM). Significant differences in cell viability was observed at 400, and 600 μM of memantine. C: Protein level of LC3-I and LC3-II was evaluated in control and NMDAR1 knockdown cells using western blotting after incubation with 400 μM memantine. The results of densitometric quantitation of immunoblots were represented as the LC3-II/LC3-I ratio relative to control cells. *p<0.05, Student’s t-test. NMDAR: N-methyl-D-aspartate receptor, siRNA: small interfering RNA, EMEM: Eagle’s minimum essential medium.


Cited by  1 articles

N-retinylidene-N-retinylethanolamine degradation in human retinal pigment epithelial cells via memantine- and ifenprodil-mediated autophagy
Jae Rim Lee, Kwang Won Jeong
Korean J Physiol Pharmacol. 2023;27(5):449-456.    doi: 10.4196/kjpp.2023.27.5.449.


Reference

References

1. Abdul M, Hoosein N. N-methyl-D-aspartate receptor in human prostate cancer. J Membr Biol. 205:125–128. 2005.
Article
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.
Article
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.
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.
Article
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.
Article
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.
Article
7. de Groot J, Sontheimer H. Glutamate and the biology of gliomas. Glia. 59:1181–1189. 2011.
Article
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.
Article
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.
Article
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.
Article
11. Johnson JW, Kotermanski SE. Mechanism of action of memantine. Curr Opin Pharmacol. 6:61–67. 2006.
Article
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.
Article
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.
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.
Article
15. McComb RD, Burger PC. Pathologic analysis of primary brain tumors. Neurol Clin. 3:711–728. 1985.
Article
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.
Article
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.
Article
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.
Article
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.
Article
22. Rzeski W, Turski L, Ikonomidou C. Glutamate antagonists limit tumor growth. Proc Natl Acad Sci USA. 98:6372–6377. 2001.
Article
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.
Article
24. Sontheimer H. A role for glutamate in growth and invasion of primary brain tumors. J Neurochem. 105:287–295. 2008.
Article
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.
Article
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.
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.
Article
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.
Article
29. Witt A, Macdonald N, Kirkpatrick P. Memantine hydrochloride. Nat Rev Drug Discov. 3:109–110. 2004.
Article
30. Ye ZC, Sontheimer H. Glioma cells release excitotoxic concentrations of glutamate. Cancer Res. 59:4383–4391. 1999.
Full Text Links
  • JKNS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr