Cite

Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev. 2010;62(3):405–96.2071666910.1124/pr.109.002451TraynelisSFWollmuthLPMcBainCJMennitiFSVanceKMOgdenKKHansenKBYuanHMyersSJDingledineRGlutamate receptor ion channels: structure, regulation, and functionPharmacol Rev201062340596Search in Google Scholar

Guttmann RP, Sokol S, Baker DL, Simpkins KL, Dong Y, Lynch DR. Proteolysis of the N-methyl-d-aspartate receptor by calpain in situ. J Pharmacol Exp Ther. 2002;302(3):1023–30.10.1124/jpet.102.03696212183659GuttmannRPSokolSBakerDLSimpkinsKLDongYLynchDRProteolysis of the N-methyl-d-aspartate receptor by calpain in situJ Pharmacol Exp Ther20023023102330Open DOISearch in Google Scholar

Lynch DR, Guttmann RP. NMDA receptor pharmacology: perspectives from molecular biology. Curr Drug Targets. 2001;2(3):215–31.1155454910.2174/1389450013348434LynchDRGuttmannRPNMDA receptor pharmacology: perspectives from molecular biologyCurr Drug Targets20012321531Search in Google Scholar

Dingledine R, Borges K, Bowie D, Traynelis SF. The glutamate receptor ion channels. Pharmacol Rev. 1999;51(1):7–61.10049997DingledineRBorgesKBowieDTraynelisSFThe glutamate receptor ion channelsPharmacol Rev1999511761Search in Google Scholar

Cull-Candy S, Brickley S, Farrant M. NMDA receptor subunits: diversity, development and disease. Curr Opin Neurobiol. 2001;11(3):327–35.10.1016/S0959-4388(00)00215-411399431Cull-CandySBrickleySFarrantMNMDA receptor subunits: diversity, development and diseaseCurr Opin Neurobiol200111332735Open DOISearch in Google Scholar

Hardingham GE. Coupling of the NMDA receptor to neuroprotective and neurodestructive events. Biochem Soc Trans. 2009;37(Pt 6):1147–60.10.1042/BST037114719909238HardinghamGECoupling of the NMDA receptor to neuroprotective and neurodestructive eventsBiochem Soc Trans200937Pt 6114760Open DOISearch in Google Scholar

Wang R, Reddy PH. Role of Glutamate and NMDA Receptors in Alzheimer's Disease. J Alzheimers Dis. 2017;57(4):1041–1048.10.3233/JAD-16076327662322WangRReddyPHRole of Glutamate and NMDA Receptors in Alzheimer's DiseaseJ Alzheimers Dis201757410411048Open DOISearch in Google Scholar

Olivares D, Deshpande VK, Shi Y, Lahiri DK, Greig NH, Rogers JT, Huang X. N-methyl D-aspartate (NMDA) receptor antagonists and memantine treatment for Alzheimer's disease, vascular dementia and Parkinson's disease. Curr Alzheimer Res. 2012;9(6):746–58.10.2174/15672051280132256421875407OlivaresDDeshpandeVKShiYLahiriDKGreigNHRogersJTHuangXN-methyl D-aspartate (NMDA) receptor antagonists and memantine treatment for Alzheimer's disease, vascular dementia and Parkinson's diseaseCurr Alzheimer Res20129674658Open DOISearch in Google Scholar

Bozic M, Valdivielso JM. The potential of targeting NMDA receptors outside the CNS. Expert Opin Ther Targets. 2015;19(3):399–413.10.1517/14728222.2014.98390025495517BozicMValdivielsoJMThe potential of targeting NMDA receptors outside the CNSExpert Opin Ther Targets2015193399413Open DOISearch in Google Scholar

D'Amico M, Di Filippo C, Rossi F, Rossi F. Arrhythmias induced by myocardial ischaemia-reperfusion are sensitive to ionotropic excitatory amino acid receptor antagonists. Eur J Pharmacol. 1999;366(2–3):167–74.1008219710.1016/S0014-2999(98)00914-5D'AmicoMDi FilippoCRossiFRossiFArrhythmias induced by myocardial ischaemia-reperfusion are sensitive to ionotropic excitatory amino acid receptor antagonistsEur J Pharmacol19993662–316774Search in Google Scholar

Srejovic I, Zivkovic V, Nikolic T, Jeremic N, Stojic I, Jeremic J, Djuric D, Jakovljevic V. Modulation of N-methyl-d-aspartate receptors in isolated rat heart. Can J Physiol Pharmacol. 2017;95(11):1327–1334.10.1139/cjpp-2017-005628758414SrejovicIZivkovicVNikolicTJeremicNStojicIJeremicJDjuricDJakovljevicVModulation of N-methyl-d-aspartate receptors in isolated rat heartCan J Physiol Pharmacol201795111327133428758414Open DOISearch in Google Scholar

Srejovic I, Jakovljevic V, Zivkovic V, Djuric D. Possible Role of N-Methyl-D-Aspartate Receptors in Physiology and Pathophysiology of Cardiovascular System. Ser J Exp Clin Res 2019; 20(1):3–13.10.1515/sjecr-2017-0010SrejovicIJakovljevicVZivkovicVDjuricDPossible Role of N-Methyl-D-Aspartate Receptors in Physiology and Pathophysiology of Cardiovascular SystemSer J Exp Clin Res2019201313Open DOISearch in Google Scholar

Djuric D, Jakovljevic V, Zivkovic V, Srejovic I. Homocysteine and homocysteine-related compounds: an overview of the roles in the pathology of the cardiovascular and nervous systems. Can J Physiol Pharmacol. 2018;96(10):991–1003.10.1139/cjpp-2018-011230130426DjuricDJakovljevicVZivkovicVSrejovicIHomocysteine and homocysteine-related compounds: an overview of the roles in the pathology of the cardiovascular and nervous systemsCan J Physiol Pharmacol201896109911003Open DOISearch in Google Scholar

Kamat PK, Rai S, Swarnkar S, Shukla R, Nath C. Mechanism of synapse redox stress in Okadaic acid (ICV) induced memory impairment: Role of NMDA receptor. Neurochem Int. 2014;76:32–41.2498417010.1016/j.neuint.2014.06.012KamatPKRaiSSwarnkarSShuklaRNathCMechanism of synapse redox stress in Okadaic acid (ICV) induced memory impairment: Role of NMDA receptorNeurochem Int2014763241Search in Google Scholar

Hardingham GE, Bading H. Synaptic versus extrasynaptic NMDA receptor signalling: implications for neuro-degenerative disorders. Nat Rev Neurosci. 2010;11(10):682–96.10.1038/nrn2911HardinghamGEBadingHSynaptic versus extrasynaptic NMDA receptor signalling: implications for neuro-degenerative disordersNat Rev Neurosci2010111068296Open DOISearch in Google Scholar

Gao X, Xu X, Pang J, Zhang C, Ding JM, Peng X, Liu Y, Cao JM. NMDA receptor activation induces mitochondrial dysfunction, oxidative stress and apoptosis in cultured neonatal rat cardiomyocytes. Physiol Res. 2007;56(5):559–69.16925458GaoXXuXPangJZhangCDingJMPengXLiuYCaoJMNMDA receptor activation induces mitochondrial dysfunction, oxidative stress and apoptosis in cultured neonatal rat cardiomyocytesPhysiol Res20075655596910.33549/physiolres.931053Search in Google Scholar

Stojic I, Srejovic I, Zivkovic V, Jeremic N, Djuric M, Stevanovic A, Milanovic T, Djuric D, Jakovljevic V. The effects of verapamil and its combinations with glutamate and glycine on cardiodynamics, coronary flow and oxidative stress in isolated rat heart. J Physiol Biochem. 2017;73(1):141–153.10.1007/s13105-016-0534-027812957StojicISrejovicIZivkovicVJeremicNDjuricMStevanovicAMilanovicTDjuricDJakovljevicVThe effects of verapamil and its combinations with glutamate and glycine on cardiodynamics, coronary flow and oxidative stress in isolated rat heartJ Physiol Biochem2017731141153Open DOISearch in Google Scholar

Hausenloy DJ, Barrabes JA, Bøtker HE, Davidson SM, Di Lisa F, Downey J, Engstrom T, Ferdinandy P, Carbrera-Fuentes HA, Heusch G, Ibanez B, Iliodromitis EK, Inserte J, Jennings R, Kalia N, Kharbanda R, Lecour S, Marber M, Miura T, Ovize M, Perez-Pinzon MA, Piper HM, Przyklenk K, Schmidt MR, Redington A, Ruiz-Meana M, Vilahur G, Vinten-Johansen J, Yellon DM, Garcia-Dorado D. Ischaemic conditioning and targeting reperfusion injury: a 30 year voyage of discovery. Basic Res Cardiol. 2016;111(6):70.2776647410.1007/s00395-016-0588-8HausenloyDJBarrabesJABøtkerHEDavidsonSMDi LisaFDowneyJEngstromTFerdinandyPCarbrera-FuentesHAHeuschGIbanezBIliodromitisEKInserteJJenningsRKaliaNKharbandaRLecourSMarberMMiuraTOvizeMPerez-PinzonMAPiperHMPrzyklenkKSchmidtMRRedingtonARuiz-MeanaMVilahurGVinten-JohansenJYellonDMGarcia-DoradoDIschaemic conditioning and targeting reperfusion injury: a 30 year voyage of discoveryBasic Res Cardiol2016111670Search in Google Scholar

Hausenloy DJ, Yellon DM. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. J Clin Invest. 2013;123(1):92–100.2328141510.1172/JCI62874HausenloyDJYellonDMMyocardial ischemia-reperfusion injury: a neglected therapeutic targetJ Clin Invest2013123192100Search in Google Scholar

Hausenloy DJ, Yellon DM. Ischaemic conditioning and reperfusion injury. Nat Rev Cardiol. 2016;13(4):193–209.10.1038/nrcardio.2016.526843289HausenloyDJYellonDMIschaemic conditioning and reperfusion injuryNat Rev Cardiol2016134193209Open DOISearch in Google Scholar

Ohkawa H, Ohishi N, Yagi K (1979) Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95:351–358.10.1016/0003-2697(79)90738-336810OhkawaHOhishiNYagiK1979Assay for lipid peroxides in animal tissues by thiobarbituric acid reactionAnal Biochem95351358Open DOISearch in Google Scholar

Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR (1982) Analysis of nitrate, nitrite and [15 N] nitrate in biological fluids. Anal Biochem 126:131–138.10.1016/0003-2697(82)90118-XGreenLCWagnerDAGlogowskiJSkipperPLWishnokJSTannenbaumSR1982Analysis of nitrate, nitrite and [15 N] nitrate in biological fluidsAnal Biochem126131138Open DOISearch in Google Scholar

Auclair C, Voisin E (1985) Nitroblue tetrazolium reduction. In: Greenvvald RA (ed) Handbook of methods for oxygen radical research. CRC Press, Boca Raton, pp 123–132.AuclairCVoisinE1985Nitroblue tetrazolium reductionInGreenvvaldRA(ed)Handbook of methods for oxygen radical researchCRC PressBoca Raton123132Search in Google Scholar

Pick E, Keisari Y (1980) A simple colorimetric method for the measurement of hydrogen peroxide produced by cells in culture. J Immunol Methods 38:161–170.10.1016/0022-1759(80)90340-36778929PickEKeisariY1980A simple colorimetric method for the measurement of hydrogen peroxide produced by cells in cultureJ Immunol Methods38161170Open DOISearch in Google Scholar

Vizi ES, Kisfali M, Lőrincz T. Role of nonsynaptic GluN2B-containing NMDA receptors in excitotoxicity: evidence that fluoxetine selectively inhibits these receptors and may have neuroprotective effects. Brain Res Bull. 2013;93:32–8.2308936210.1016/j.brainresbull.2012.10.005ViziESKisfaliMLőrinczTRole of nonsynaptic GluN2B-containing NMDA receptors in excitotoxicity: evidence that fluoxetine selectively inhibits these receptors and may have neuroprotective effectsBrain Res Bull201393328Search in Google Scholar

Dickie BG, Holmes C, Greenfield SA. Neurotoxic and neurotrophic effects of chronic N-methyl-D-aspartate exposure upon mesencephalic dopaminergic neurons in organotypic culture. Neuroscience. 1996;72(3):731–41.10.1016/0306-4522(95)00611-79157319DickieBGHolmesCGreenfieldSANeurotoxic and neurotrophic effects of chronic N-methyl-D-aspartate exposure upon mesencephalic dopaminergic neurons in organotypic cultureNeuroscience199672373141Open DOISearch in Google Scholar

Tuttolomondo A, Di Sciacca R, Di Raimondo D, Arnao V, Renda C, Pinto A, Licata G. Neuron protection as a therapeutic target in acute ischemic stroke. Curr Top Med Chem. 2009;9(14):1317–34.1984965910.2174/156802609789869646TuttolomondoADi SciaccaRDi RaimondoDArnaoVRendaCPintoALicataGNeuron protection as a therapeutic target in acute ischemic strokeCurr Top Med Chem200991413173419849659Search in Google Scholar

Makarewicz D, Sulejczak D, Duszczyk M, Małek M, Słomka M, Lazarewicz JW. Delayed preconditioning with NMDA receptor antagonists in a rat model of perinatal asphyxia. Folia Neuropathol. 2014;52(3):270–84.MakarewiczDSulejczakDDuszczykMMałekMSłomkaMLazarewiczJWDelayed preconditioning with NMDA receptor antagonists in a rat model of perinatal asphyxiaFolia Neuropathol20145232708410.5114/fn.2014.4556825310738Search in Google Scholar

Doeppner TR, Pehlke JR, Kaltwasser B, Schlechter J, Kilic E, Bähr M, Hermann DM. The indirect NMDAR antagonist acamprosate induces postischemic neurologic recovery associated with sustained neuroprotection and neuroregeneration. J Cereb Blood Flow Metab. 2015;35(12):2089–97.10.1038/jcbfm.2015.17926219600DoeppnerTRPehlkeJRKaltwasserBSchlechterJKilicEBährMHermannDMThe indirect NMDAR antagonist acamprosate induces postischemic neurologic recovery associated with sustained neuroprotection and neuroregenerationJ Cereb Blood Flow Metab20153512208997467113226219600Open DOISearch in Google Scholar

Li H, Luo XB, Xu Y, Hou XY. A Brief Ischemic Postconditioning Protects Against Amyloid-β Peptide Neurotoxicity by Downregulating MLK3-MKK3/6-P38MAPK Signal in Rat Hippocampus. J Alzheimers Dis. 2019;71(2):671–684.3142439310.3233/JAD-190207LiHLuoXBXuYHouXYA Brief Ischemic Postconditioning Protects Against Amyloid-β Peptide Neurotoxicity by Downregulating MLK3-MKK3/6-P38MAPK Signal in Rat HippocampusJ Alzheimers Dis201971267168431424393Search in Google Scholar

McGee MA, Abdel-Rahman AA. Enhanced vascular PI3K/Akt-NOX signaling underlies the peripheral NMDAR-mediated pressor response in conscious rats. J Cardiovasc Pharmacol. 2014;63(5):395–405.10.1097/FJC.000000000000005924336015McGeeMAAbdel-RahmanAAEnhanced vascular PI3K/Akt-NOX signaling underlies the peripheral NMDAR-mediated pressor response in conscious ratsJ Cardiovasc Pharmacol2014635395405401321924336015Open DOISearch in Google Scholar

Srejovic I, Jakovljevic V, Zivkovic V, Barudzic N, Radovanovic A, Stanojlovic O, Djuric DM. The effects of the modulation of NMDA receptors by homocysteine thiolactone and dizocilpine on cardiodynamics and oxidative stress in isolated rat heart. Mol Cell Biochem. 2015;401(1–2):97–105.10.1007/s11010-014-2296-825467376SrejovicIJakovljevicVZivkovicVBarudzicNRadovanovicAStanojlovicODjuricDMThe effects of the modulation of NMDA receptors by homocysteine thiolactone and dizocilpine on cardiodynamics and oxidative stress in isolated rat heartMol Cell Biochem20154011–29710525467376Open DOISearch in Google Scholar

Betzen C, White R, Zehendner CM, Pietrowski E, Bender B, Luhmann HJ, Kuhlmann CR. Oxidative stress upregulates the NMDA receptor on cerebrovascular endothelium. Free Radic Biol Med. 2009;47(8):1212–20.1966054110.1016/j.freeradbiomed.2009.07.034BetzenCWhiteRZehendnerCMPietrowskiEBenderBLuhmannHJKuhlmannCROxidative stress upregulates the NMDA receptor on cerebrovascular endotheliumFree Radic Biol Med200947812122019660541Search in Google Scholar

Moshal KS, Kumar M, Tyagi N, Mishra PK, Metreveli N, Rodriguez WE, Tyagi SC. Restoration of contractility in hyperhomocysteinemia by cardiac-specific deletion of NMDA-R1. Am J Physiol Heart Circ Physiol. 2009;296(3):H887–92.1918196610.1152/ajpheart.00750.2008MoshalKSKumarMTyagiNMishraPKMetreveliNRodriguezWETyagiSCRestoration of contractility in hyperhomocysteinemia by cardiac-specific deletion of NMDA-R1Am J Physiol Heart Circ Physiol20092963H88792266023019181966Search in Google Scholar

Tyagi N, Vacek JC, Givvimani S, Sen U, Tyagi SC. 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(2):78–87.2017042610.3109/10799891003614808TyagiNVacekJCGivvimaniSSenUTyagiSCCardiac specific deletion of N-methyl-d-aspartate receptor 1 ameliorates mtMMP-9 mediated autophagy/mitophagy in hyperhomocysteinemiaJ Recept Signal Transduct Res20103027887292188920170426Search in Google Scholar

eISSN:
2335-075X
ISSN:
1820-8665
Language:
English
Publication timeframe:
4 times per year
Journal Subjects:
Medicine, Clinical Medicine, other