838
Views
53
CrossRef citations to date
0
Altmetric
Review

The potential of targeting NMDA receptors outside the CNS

, PhD (Postdoctoral Fellow) & , PhD (Director of the Nephrology Research Department)

Bibliography

  • Haddad JJ. N-methyl-D-aspartate (NMDA) and the regulation of mitogen-activated protein kinase (MAPK) signaling pathways: a revolving neurochemical axis for therapeutic intervention? Prog Neurobiol 2005;77:252-82
  • Wang Y, Durkin JP. alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, but not N-methyl-D-aspartate, activates mitogen-activated protein kinase through G-protein beta gamma subunits in rat cortical neurons. J Biol Chem 1995;270:22783-7
  • Ahn YM, Oh SW, Kang UG, et al. An N-methyl-D-aspartate antagonist, MK-801, preferentially reduces electroconvulsive shock-induced phosphorylation of p38 mitogen-activated protein kinase in the rat hippocampus. Neurosci Lett 2000;296:101-4
  • Cammarota M, Bevilaqua LR, Ardenghi P, et al. Learning-associated activation of nuclear MAPK, CREB and Elk-1, along with Fos production, in the rat hippocampus after a one-trial avoidance learning: abolition by NMDA receptor blockade. Brain Res Mol Brain Res 2000;76:36-46
  • Chandler LJ, Sutton G, Dorairaj NR, et al. N-methyl D-aspartate receptor-mediated bidirectional control of extracellular signal-regulated kinase activity in cortical neuronal cultures. J Biol Chem 2001;276:2627-36
  • Jiang Q, Gu Z, Zhang G, et al. N-methyl-D-aspartate receptor activation results in regulation of extracellular signal-regulated kinases by protein kinases and phosphatases in glutamate-induced neuronal apototic-like death. Brain Res 2000;887:285-92
  • Zeng L, Lu L, Muller M, et al. Structure-based functional design of chemical ligands for AMPA-subtype glutamate receptors. J Mol Neurosci 2002;19:113-16
  • Barnstable CJ, Wei JY, Han MH. Modulation of synaptic function by cGMP and cGMP-gated cation channels. Neurochem Int 2004;45:875-84
  • Monaghan DT, Bridges RJ, Cotman CW. The excitatory amino acid receptors: their classes, pharmacology, and distinct properties in the function of the central nervous system. Annu Rev Pharmacol Toxicol 1989;29:365-402
  • Hollmann M, O’Shea-Greenfield A, Rogers SW, et al. Cloning by functional expression of a member of the glutamate receptor family. Nature 1989;342:643-8
  • Gladding CM, Fitzjohn SM, Molnar E. Metabotropic glutamate receptor-mediated long-term depression: molecular mechanisms. Pharmacol Rev 2009;61:395-412
  • Dingledine R, Borges K, Bowie D, et al. The glutamate receptor ion channels. Pharmacol Rev 1999;51:7-61
  • Cull-Candy SG, Brickley SG, Misra C, et al. NMDA receptor diversity in the cerebellum: identification of subunits contributing to functional receptors. Neuropharmacology 1998;37:1369-80
  • Cull-Candy S, Brickley S, Farrant M. NMDA receptor subunits: diversity, development and disease. Curr Opin Neurobiol 2001;11:327-35
  • Forrest D, Yuzaki M, Soares HD, et al. Targeted disruption of NMDA receptor 1 gene abolishes NMDA response and results in neonatal death. Neuron 1994;13:325-38
  • Scheetz AJ, Constantine-Paton M. Modulation of NMDA receptor function: implications for vertebrate neural development. FASEB J 1994;8:745-52
  • Hardingham GE, Bading H. The Yin and Yang of NMDA receptor signalling. Trends Neurosci 2003;26:81-9
  • Guttmann RP, Sokol S, Baker DL, et al. Proteolysis of the N-methyl-d-aspartate receptor by calpain in situ. J Pharmacol Exp Ther 2002;302:1023-30
  • Hardingham GE. Coupling of the NMDA receptor to neuroprotective and neurodestructive events. Biochem Soc Trans 2009;37:1147-60
  • Loftis JM, Janowsky A. The N-methyl-D-aspartate receptor subunit NR2B: localization, functional properties, regulation, and clinical implications. Pharmacol Ther 2003;97:55-85
  • Lipton SA, Rosenberg PA. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med 1994;330:613-22
  • Miglio G, Dianzani C, Fallarini S, et al. Stimulation of N-methyl-D-aspartate receptors modulates Jurkat T cell growth and adhesion to fibronectin. Biochem Biophys Res Commun 2007;361:404-9
  • Nahm WK, Philpot BD, Adams MM, et al. Significance of N-methyl-D-aspartate (NMDA) receptor-mediated signaling in human keratinocytes. J Cell Physiol 2004;200:309-17
  • Mentaverri R, Kamel S, Wattel A, et al. Regulation of bone resorption and osteoclast survival by nitric oxide: possible involvement of NMDA-receptor. J Cell Biochem 2003;88:1145-56
  • Rakic P, Komuro H. The role of receptor/channel activity in neuronal cell migration. J Neurobiol 1995;26:299-315
  • Shorte SL. N-methyl-D-aspartate evokes rapid net depolymerization of filamentous actin in cultured rat cerebellar granule cells. J Neurophysiol 1997;78:1135-43
  • Itzstein C, Espinosa L, Delmas PD, et al. Specific antagonists of NMDA receptors prevent osteoclast sealing zone formation required for bone resorption. Biochem Biophys Res Commun 2000;268:201-9
  • Parisi E, Almaden Y, Ibarz M, et al. N-methyl-D-aspartate receptors are expressed in rat parathyroid gland and regulate PTH secretion. Am J Physiol Renal Physiol 2009;296:F1291-6
  • Magnusson KR. The aging of the NMDA receptor complex. Front Biosci 1998;3:e70-80
  • Bellone C, Nicoll RA. Rapid bidirectional switching of synaptic NMDA receptors. Neuron 2007;55:779-85
  • Paoletti P, Neyton J. NMDA receptor subunits: function and pharmacology. Curr Opin Pharmacol 2007;7:39-47
  • Rebola N, Srikumar BN, Mulle C. Activity-dependent synaptic plasticity of NMDA receptors. J Physiol 2010;588:93-9
  • Luo J, Wang Y, Yasuda RP, et al. The majority of N-methyl-D-aspartate receptor complexes in adult rat cerebral cortex contain at least three different subunits (NR1/NR2A/NR2B). Mol Pharmacol 1997;51:79-86
  • Chazot PL. The NMDA receptor NR2B subunit: a valid therapeutic target for multiple CNS pathologies. Curr Med Chem 2004;11:389-96
  • Ishii T, Moriyoshi K, Sugihara H, et al. Molecular characterization of the family of the N-methyl-D-aspartate receptor subunits. J Biol Chem 1993;268:2836-43
  • Meguro H, Mori H, Araki K, et al. Functional characterization of a heteromeric NMDA receptor channel expressed from cloned cDNAs. Nature 1992;357:70-4
  • Nakanishi S. Molecular diversity of glutamate receptors and implications for brain function. Science 1992;258:597-603
  • Ciabarra AM, Sullivan JM, Gahn LG, et al. Cloning and characterization of chi-1: a developmentally regulated member of a novel class of the ionotropic glutamate receptor family. J Neurosci 1995;15:6498-508
  • Lynch DR, Guttmann RP. NMDA receptor pharmacology: perspectives from molecular biology. Curr Drug Targets 2001;2:215-31
  • Das S, Sasaki YF, Rothe T, et al. Increased NMDA current and spine density in mice lacking the NMDA receptor subunit NR3A. Nature 1998;393:377-81
  • Nishi M, Hinds H, Lu HP, et al. Motoneuron-specific expression of NR3B, a novel NMDA-type glutamate receptor subunit that works in a dominant-negative manner. J Neurosci 2001;21:RC185
  • Matsuda K, Fletcher M, Kamiya Y, et al. Specific assembly with the NMDA receptor 3B subunit controls surface expression and calcium permeability of NMDA receptors. J Neurosci 2003;23:10064-73
  • Perez-Otano I, Ehlers MD. Learning from NMDA receptor trafficking: clues to the development and maturation of glutamatergic synapses. Neurosignals 2004;13:175-89
  • Al-Hallaq R, Jarabek B, Fu Z, et al. Association of NR3A with the N-methyl-D-aspartate receptor NR1 and NR2 subunits. Mol Pharmacol 2002;62:1119-27
  • Ulbrich M, Isacoff E. Rules of engagement for NMDA receptor subunits. Proc Natl Acad Sci USA 2008;105:14163-8
  • Chatterton J, Awobuluyi M, Premkumar L, et al. Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature 2002;415:793-8
  • Cavara N, Orth A, Hollmann M. Effects of NR1 splicing on NR1/NR3B-type excitatory glycine receptors. BMC Neurisci 2009;10:1-11
  • Nishikawa T, Morita K, Kinjo K, et al. Stimulation of catecholamine release from isolated adrenal glands by some amino acids. Jpn J Pharmacol 1982;32:291-7
  • Moroni F, Luzzi S, Franchi-Micheli S, et al. The presence of N-methyl-D-aspartate-type receptors for glutamic acid in the guinea pig myenteric plexus. Neurosci Lett 1986;68:57-62
  • Morhenn VB, Waleh NS, Mansbridge JN, et al. Evidence for an NMDA receptor subunit in human keratinocytes and rat cardiocytes. Eur J Pharmacol 1994;268:409-14
  • Chen HJ, Fitzgerald R, Brown AT, et al. Identification of a homocysteine receptor in the peripheral endothelium and its role in proliferation. J Vasc Surg 2005;41:853-60
  • Mcgee MA, Abdel-Rahman AA. Enhanced vascular neuronal nitric-oxide synthase-derived nitric-oxide production underlies the pressor response caused by peripheral N-methyl-D-aspartate receptor activation in conscious rats. J Pharmacol Exp Ther 2012;342:461-71
  • Leung JC, Travis BR, Verlander JW, et al. Expression and developmental regulation of the NMDA receptor subunits in the kidney and cardiovascular system. Am J Physiol Regul Integr Comp Physiol 2002;283:R964-71
  • Patton AJ, Genever PG, Birch MA, et al. Expression of an N-methyl-D-aspartate-type receptor by human and rat osteoblasts and osteoclasts suggests a novel glutamate signaling pathway in bone. Bone 1998;22:645-9
  • Seeber S, Becker K, Rau T, et al. Transient expression of NMDA receptor subunit NR2B in the developing rat heart. J Neurochem 2000;75:2472-7
  • Deng A, Valdivielso JM, Munger KA, et al. Vasodilatory N-methyl-D-aspartate receptors are constitutively expressed in rat kidney. J Am Soc Nephrol 2002;13:1381-4
  • Gill SS, Pulido OM, Mueller RW, et al. Molecular and immunochemical characterization of the ionotropic glutamate receptors in the rat heart. Brain Res Bull 1998;46:429-34
  • Gill S, Pulido OM. Glutamate receptors in peripheral tissues: current knowledge, future research, and implications for toxicology. Toxicol Pathol 2001;29:208-23
  • Lin YJ, Bovetto S, Carver JM, et al. Cloning of the cDNA for the human NMDA receptor NR2C subunit and its expression in the central nervous system and periphery. Brain Res Mol Brain Res 1996;43:57-64
  • Gonzalez-Cadavid NF, Ryndin I, Vernet D, et al. Presence of NMDA receptor subunits in the male lower urogenital tract. J Androl 2000;21:566-78
  • Ma MC, Huang HS, Chen YS, et al. Mechanosensitive N-methyl-D-aspartate receptors contribute to sensory activation in the rat renal pelvis. Hypertension 2008;52:938-U545
  • Parisi E, Bozic M, Ibarz M, et al. Sustained activation of renal N-methyl-D-aspartate receptors decreases vitamin D synthesis: a possible role for glutamate on the onset of secondary HPT. Am J Physiol Endocrinol Metab 2010;299:E825-31
  • Bozic M, de Rooij J, Parisi E, et al. Glutamatergic Signaling Maintains the Epithelial Phenotype of Proximal Tubular Cells. J Am Soc Nephrol 2011;22:1099-111
  • Nasstrom J, Boo E, Stahlberg M, et al. Tissue distribution of two NMDA receptor antagonists, [3H]CGS 19755 and [3H]MK-801, after intrathecal injection in mice. Pharmacol Biochem Behav 1993;44:9-15
  • Deng A, Thomson SC. Renal NMDA receptors independently stimulate proximal reabsorption and glomerular filtration. Am J Physiol Renal Physiol 2009;296:F976-82
  • Zhang C, Yi F, Xia M, et al. NMDA receptor-mediated activation of NADPH oxidase and glomerulosclerosis in hyperhomocysteinemic rats. Antioxid Redox Signal 2010;13:975-86
  • Anderson M, Suh JM, Kim EY, et al. Functional NMDA receptors with atypical properties are expressed in podocytes. Am J Physiol Cell Physiol 2011;300:C22-32
  • Giardino L, Armelloni S, Corbelli A, et al. Podocyte glutamatergic signaling contributes to the function of the glomerular filtration barrier. J Am Soc Nephrol 2009;20:1929-40
  • Sproul AD, Steele SL, Thai TL, et al. N-methyl-D-aspartate receptor subunit NR3a expression and function in principal cells of the collecting duct. Am J Physiol Renal Physiol 2011;301:F44-54
  • Gu Y, Genever PG, Skerry TM, et al. The NMDA type glutamate receptors expressed by primary rat osteoblasts have the same electrophysiological characteristics as neuronal receptors. Calcif Tissue Int 2002;70:194-203
  • Itzstein C, Cheynel H, Burt-Pichat B, et al. Molecular identification of NMDA glutamate receptors expressed in bone cells. J Cell Biochem 2001;82:134-44
  • Ho ML, Tsai TN, Chang JK, et al. Down-regulation of N-methyl D-aspartate receptor in rat-modeled disuse osteopenia. Osteoporos Int 2005;16:1780-8
  • Chenu C, Serre CM, Raynal C, et al. Glutamate receptors are expressed by bone cells and are involved in bone resorption. Bone 1998;22:295-9
  • Kalariti N, Lembessis P, Koutsilieris M. Characterization of the glutamatergic system in MG-63 ostoblast-like osteosarcoma cells. Anticancer Res 2004;24:3923-30
  • Merle B, Itzstein C, Delmas PD, et al. NMDA glutamate receptors are expressed by osteoclast precursors and involved in the regulation of osteoclastogenesis. J Cell Biochem 2003;90:424-36
  • Hinoi E, Fujimori S, Takarada T, et al. Facilitation of glutamate release by ionotropic glutamate receptors in osteobasts. Biochem Biophys Res Commun 2002;297:452-8
  • Gu YC, Publicover SJ. Expression of functional metabotropic glutamate receptors primary cultured rat osteoblasts - Cross-talk with N-methyl-D-aspartate receptors. J Biol Chem 2000;275:34252-9
  • Fujita H, Hinoi E, Nakatani E, et al. Possible modulation of process extension by N-methyl-D-aspartate receptor expressed in osteocytic MLO-Y4 cells. J Pharmacol Sci 2012;119:112-16
  • Corsi M, Fina P, Trist DG. Co-agonism in drug-receptor interaction: illustrated by the NMDA receptors. Trends Pharmacol Sci 1996;17:220-2
  • Hood WF, Sun ET, Compton RP, Monahan JB. 1-Aminocyclobutane-1-carboxylate (ACBC): a specific antagonist of the N-methyl-D-aspartate receptor coupled glycine receptor. Eur J Pharmacol 1989;161:281-2
  • Lynch DR, Guttmann RP. Excitotoxicity: perspectives based on N-methyl-D-aspartate receptor subtypes. J Pharmacol Exp Ther 2002;300:717-23
  • Johnson JW, Ascher P. Glycine potentiates the NMDA response in cultured mouse brain neurons. Nature 1987;325:529-31
  • Kupper J, Ascher P, Neyton J. Internal Mg2+ block of recombinant NMDA channels mutated within the selectivity filter and expressed in Xenopus oocytes. J Physiol 1998;507(Pt 1):1-12
  • Kuner T, Schoepfer R. Multiple structural elements determine subunit specificity of Mg2+ block in NMDA receptor channels. J Neurosci 1996;16:3549-58
  • Vissel B, Krupp JJ, Heinemann SF, et al. Intracellular domains of NR2 alter calcium-dependent inactivation of N-methyl-D-aspartate receptors. Mol Pharmacol 2002;61:595-605
  • Traynelis SF, Burgess MF, Zheng F, et al. Control of voltage-independent zinc inhibition of NMDA receptors by the NR1 subunit. J Neurosci 1998;18:6163-75
  • Duguid IC, Smart TG. Retrograde activation of presynaptic NMDA receptors enhances GABA release at cerebellar interneuron-Purkinje cell synapses. Nat Neurosci 2004;7:525-33
  • Kashiwagi K, Tanaka I, Tamura M, et al. Anthraquinone polyamines: novel channel blockers to study N-methyl-D-aspartate receptors. J Pharmacol Exp Ther 2004;309:884-93
  • Paoletti P, Vergnano AM, Barbour B, et al. Zinc at glutamatergic synapses. Neuroscience 2009;158:126-36
  • Omelchenko IA, Nelson CS, Allen CN. Lead inhibition of N-methyl-D-aspartate receptors containing NR2A, NR2C and NR2D subunits. J Pharmacol Exp Ther 1997;282:1458-64
  • Guilarte TR, McGlothan JL. Selective decrease in NR1 subunit splice variant mRNA in the hippocampus of Pb2+-exposed rats: implications for synaptic targeting and cell surface expression of NMDAR complexes. Brain Res Mol Brain Res 2003;113:37-43
  • Nagy J. The NR2B subtype of NMDA receptor: a potential target for the treatment of alcohol dependence. Curr Drug Targets CNS Neurol Disord 2004;3:169-79
  • Mony L, Kew JN, Gunthorpe MJ, et al. Allosteric modulators of NR2B-containing NMDA receptors: molecular mechanisms and therapeutic potential. Br J Pharmacol 2009;157:1301-17
  • Kornhuber J, Weller M, Schoppmeyer K, et al. Amantadine and memantine are nmda receptor antagonists with neuroprotective properties. J Neural Transm Suppl 1994;43:91-104
  • Huettner JE, Bean BP. Block of N-methyl-D-aspartate-activated current by the anticonvulsant MK-801: selective binding to open channels. Proc Natl Acad Sci USA 1988;85:1307-11
  • Fagg GE. Phencyclidine and related drugs bind to the activated N-methyl-D-aspartate receptor-channel complex in rat brain membranes. Neurosci Lett 1987;76:221-7
  • Williams K. Ifenprodil discriminates subtypes of the N-methyl-D-aspartate receptor: selectivity and mechanisms at recombinant heteromeric receptors. Mol Pharmacol 1993;44:851-9
  • Perin-Dureau F, Rachline J, Neyton J, et al. Mapping the binding site of the neuroprotectant ifenprodil on NMDA receptors. J Neurosci 2002;22:5955-65
  • Malherbe P, Mutel V, Broger C, et al. Identification of critical residues in the amino terminal domain of the human NR2B subunit involved in the RO 25-6981 binding pocket. J Pharmacol Exp Ther 2003;307:897-905
  • Ganor Y, Levite M. The neurotransmitter glutamate and human T cells: glutamate receptors and glutamate-induced direct and potent effects on normal human T cells, cancerous human leukemia and lymphoma T cells, and autoimmune human T cells. J Neural Transm 2014;121:983-1006
  • Divino Filho J, Hazel S, Fürst P, et al. Glutamate concentration in plasma, erythrocyte and muscle in relation to plasma levels of insulin-like growth factor (IGF)-I, IGF binding protein-1 and insulin in patients on haemodialysis. J Endocrinol 1998;156:519-27
  • Liu Y, Zhou L, Xu HF, et al. A preliminary experimental study on the cardiac toxicity of glutamate and the role of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor in rats. Chin Med J (Engl) 2013;126:1323-32
  • Lin TH, Yang RS, Tang CH, et al. Regulation of the maturation of osteoblasts and osteoclastogenesis by glutamate. Eur J Pharmacol 2008;589:37-44
  • Mason D, Suva LJ, Genever P, et al. Mechanically regulated expression of a neural glutamate transporter in bone: a role for excitatory amino acids as osteotropic agents? Bone 1997;20:199-205
  • Hinoi E, Yoneda Y. Possible involvement of glutamatergic signaling machineries in pathophysiology of rheumatoid arthritis. J Pharmacol Sci 2011;116:248-56
  • Ralphe J, Segar J, Schutte B, et al. Localization and function of the brain excitatory amino acid transporter type 1 in cardiac mitochondria. J Mol Cell Cardiol 2004;37:33-41
  • Hediger M. Glutamate transporters in kidney and brain. Am J Physiol 1999;277:F487-92
  • Welbourne T, Matthews J. Glutamate transporters and renal function. Am J Physiol 1999;277:F501-5
  • Laketic-Ljubojevic I, Suva LJ, Maathuis F, et al. Functional characterization of N-methyl-D-aspartic acid-gated channels in bone cells. Bone 1999;25:631-7
  • Xue H, Field C. New role of glutamate as an immunoregulator via glutamate receptors and transporters. Front Biosci (Schol Ed) 2011;1:1007-20
  • Slomowitz LA, Gabbai FB, Khang SJ, et al. Protein intake regulates the vasodilatory function of the kidney and NMDA receptor expression. Am J Physiol Regul Integr Comp Physiol 2004;287:R1184-9
  • Kim EY, Anderson M, Dryer SE. Sustained activation of N-methyl-D-aspartate receptors in podoctyes leads to oxidative stress, mobilization of transient receptor potential canonical 6 channels, nuclear factor of activated T cells activation, and apoptotic cell death. Mol Pharmacol 2012;82:728-37
  • Franke WW, Schmid E, Osborn M, et al. Different intermediate-sized filaments distinguished by immunofluorescence microscopy. Proc Natl Acad Sci USA 1978;75:5034-8
  • Frokiaer J. Collecting duct expression of N-methyl-D-aspartate receptor subtype NR3a regulates urinary concentrating capacity. Am J Physiol Renal Physiol 2011;301:F42-3
  • Leung JC, Marphis T, Craver RD, et al. Altered NMDA receptor expression in renal toxicity: protection with a receptor antagonist. Kidney Int 2004;66:167-76
  • Leung JC, Ragland N, Marphis T, et al. NMDA Agonists and Antagonists Induce Renal Culture Cell Toxicity. Med Chem 2008;4:565-71
  • Basile AS, Huang JM, Xie C, et al. N-methyl-D-aspartate antagonists limit aminoglycoside antibiotic-induced hearing loss. Nat Med 1996;2:1338-43
  • Puel JL, Ladrech S, Chabert R, et al. Electrophysiological Evidence for the Presence of Nmda Receptors in the Guinea-Pig Cochlea. Hear Res 1991;51:255-64
  • Segal JA, Harris BD, Kustova Y, et al. Aminoglycoside neurotoxicity involves NMDA receptor activation. Brain Res 1999;815:270-7
  • Arora S, Kaur T, Kaur A, et al. Glycine aggravates ischemia reperfusion-induced acute kidney injury through N-Methyl-D-Aspartate receptor activation in rats. Mol Cell Biochem 2014;393:123-31
  • Yang CC, Chien CT, Wu MH, et al. NMDA receptor blocker ameliorates ischemia-reperfusion-induced renal dysfunction in rat kidneys. Am J Physiol Renal Physiol 2008;294:F1433-40
  • Pundir M, Arora S, Kaur T, et al. Effect of modulating the allosteric sites of N-methyl-D-aspartate receptors in ischemia-reperfusion induced acute kidney injury. J Surg Res 2013;183:668-77
  • Peet NM, Grabowski PS, Laketic-Ljubojevic I, et al. The glutamate receptor antagonist MK801 modulates bone resorption in vitro by a mechanism predominantly involving osteoclast differentiation. FASEB J 1999;13:2179-85
  • Gray C, Marie H, Arora M, et al. Glutamate does not play a major role in controlling bone growth. J Bone Miner Res 2001;16:742-9
  • Hinoi E, Fujimori S, Yoneda Y. Modulation of cellular differentiation by N-methyl-D-aspartate receptors in osteoblasts. FASEB J 2003;17:1532-4
  • Li JL, Zhao L, Cui B, et al. Multiple signaling pathways involved in stimulation of osteoblast differentiation by N-methyl-D-aspartate receptors activation in vitro. Acta Pharmacol Sin 2011;32:895-903
  • Li JL, Cui B, Qi L, et al. NMDA enhances stretching-induced differentiation of osteobalsts through the ERK1/2 signaling pathway. Bone 2008;43:469-75
  • Szczesniak AM, Gilbert RW, Mukhida M, et al. Mechanical loading modulates glutamate receptor subunit expression in bone. Bone 2005;37:63-73
  • Koizumi M, Komaba H, Fukagawa M. Parathyroid function in chronic kidney disease: role of FGF23-Klotho axis. Contrib Nephrol 2013;180:110-23
  • D’Amico M, Di Filippo C, Rossi F, et al. Arrhythmias induced by myocardial ischaemia-reperfusion are sensitive to ionotropic excitatory amino acid receptor antagonists. Eur J Pharmacol 1999;366:167-74
  • Shi S, Liu T, Li Y, et al. Chronic N-Methyl-D-aspartate receptor activation induces cardiac electrical remodeling and increases susceptibility to ventricular arrhythmias. Pacing Clin Electrophysiol 2014;37(10):1367-77
  • Maldonado C, Soni CV, Todnem ND, et al. Hyperhomocysteinemia and sudden cardiac death: potential arrhythmogenic mechanisms. Curr Vasc Pharmacol 2010;8:64-74
  • Sun XL, Zhong JQ, Wang DG, et al. Increasing glutamate promotes ischemia-reperfusion-induced ventricular arrhythmias in rats in vivo. Pharmacology 2014;93:4-9
  • Huang CF, Su MJ. Positive inotropic action of NMDA receptor antagonist (+)-MK801 in rat heart. J Biomed Sci 1999;6:387-98
  • Kleiger RE, Miller JP, Bigger JT, et al. Decreased heart-rate-variability and its association with increased mortality after acute myocardial-infarction. Am J Cardiol 1987;59:256-62
  • Bennett AJ, DePetrillo PB. Differential effects of MK801 and lorazepam on heart rate variability in adolescent rhesus monkeys (Macoca mulatta). J Cardiovasc Pharmacol 2005;45:383-8
  • Aya AGM, Robert E, Bruelle P, et al. Effects of ketamine on ventricular conduction, refractoriness, and wavelength - Potential antiarrhythmic effects: a high-resolution epicardial mapping in rabbit hearts. Anesthesiology 1997;87:1417-27
  • Baczko I, Lepran I, Papp JG. Influence of anesthetics on the incidence of reperfusion-induced arrhythmias and sudden death in rats. J Cardiovasc Pharmacol 1997;29:196-201
  • Hageman GR, Simor T. Attenuation of the cardiac effects of cocaine by dizocilpine. Am J Physiol 1993;264:H1890-5
  • Gao X, Xu X, Pang J, et al. NMDA receptor activation induces mitochondrial dysfunction, oxidative stress and apoptosis in cultured neonatal rat cardiomyocytes. Physiol Res 2007;56:559-69
  • Tyagi N, Vacek JC, Givvimani S, et al. Cardiac specific deletion of N-methyl-d-aspartate receptor 1 ameliorates mtMMP-9 mediated autophagy/mitophagy in hyperhomocysteinemia. J Recept Signal Transduct Res 2010;30:78-87
  • Moshal KS, Tipparaju SM, Vacek TP, et al. Mitochondrial matrix metalloproteinase activation decreases myocyte contractility in hyperhomocysteinemia. Am J Physiol Heart Circ Physiol 2008;295:H890-7
  • Tyagi N, Mishra PK, Tyagi SC. Homocysteine, hydrogen sulfide (H2S) and NMDA-receptor in heart failure. Indian J Biochem Biophys 2009;46:441-6
  • Meng L, Zhang ZX, Xu K, et al. HIV-1 gp120 induces autophagy in cardiomyocytes via the NMDA receptor. Int J Cardiol 2013;167:2517-23
  • Meneghini A, Ferreira C, de Abreu LC, et al. Memantine prevents cardiomyocytes nuclear size reduction in the left ventricle of rats exposed to cold stress. Clinics 2009;64:921-6
  • Watanabe M, Mishina M, Inoue Y. Distinct gene expression of the N-methyl-D-aspartate receptor channel subunit in peripheral neurons of the mouse sensory ganglia and adrenal gland. Neurosci Lett 1994;165:183-6

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.