Skip to main content
Erschienen in: Journal of Neural Transmission 8/2018

31.01.2018 | Neurology and Preclinical Neurological Studies - Original Article

Glutamatergic mechanisms in l-DOPA-induced dyskinesia and therapeutic implications

verfasst von: Manuela Mellone, Fabrizio Gardoni

Erschienen in: Journal of Neural Transmission | Ausgabe 8/2018

Einloggen, um Zugang zu erhalten

Abstract

Overactivation of the glutamatergic synapse leading to maladaptive synaptic plasticity in the basal ganglia is a well-demonstrated process involved in the onset of l-DOPA-induced dyskinesia (LID). Changes in glutamate release are paralleled by compensatory modifications of the expression and/or synaptic localization of both ionotropic and metabotropic glutamate receptors (mGluRs). Accordingly, compounds targeting N-methyl-d-aspartate glutamate receptors (NMDARs) and specific subtypes of metabotropic glutamate receptors (mGluR4 and mGluR5) have been tested both in preclinical and clinical studies. At present, amantadine, a low-affinity non-competitive NMDAR antagonist, represents the only recommended add-on agent with a moderate anti-dyskinetic activity. The present review describes recent advances in basic research, preclinical and early clinical studies in the attempt of identifying innovative strategies for an accurate modulation of both pre- and postsynaptic glutamate receptors to reduce the severity of LID. Even if a complete understanding of LID molecular bases is still lacking, several compounds demonstrated an anti-dyskinetic activity in preclinical and early clinical studies. These results indicate that modulation of the glutamatergic system remains one of the most promising pharmacological strategies in the field.
Literatur
Zurück zum Zitat Ba M, Kong M, Yang H, Ma G, Lu G, Chen S, Liu Z (2006) Changes in subcellular distribution and phosphorylation of GluR1 in lesioned striatum of 6-hydroxydopamine-lesioned and l-DOPA-treated rats. Neurochem Res 31(11):1337–1347PubMedCrossRef Ba M, Kong M, Yang H, Ma G, Lu G, Chen S, Liu Z (2006) Changes in subcellular distribution and phosphorylation of GluR1 in lesioned striatum of 6-hydroxydopamine-lesioned and l-DOPA-treated rats. Neurochem Res 31(11):1337–1347PubMedCrossRef
Zurück zum Zitat Bastide MF, Meissner WG, Picconi B, Fasano S, Fernagut PO, Feyder M, Francardo V, Alcacer C, Ding Y, Brambilla R, Fisone G, Jon Stoessl A, Bourdenx M, Engeln M, Navailles S, De Deurwaerdère P, Ko WK, Simola N, Morelli M, Groc L, Rodriguez MC, Gurevich EV, Quik M, Morari M, Mellone M, Gardoni F, Tronci E, Guehl D, Tison F, Crossman AR, Kang UJ, Steece-Collier K, Fox S, Carta M, Angela Cenci M, Bézard E (2015) Pathophysiology of l-DOPA-induced motor and non-motor complications in Parkinson’s disease. Prog Neurobiol 132:96–168. https://doi.org/10.1016/j.pneurobio.2015.07.002 PubMedCrossRef Bastide MF, Meissner WG, Picconi B, Fasano S, Fernagut PO, Feyder M, Francardo V, Alcacer C, Ding Y, Brambilla R, Fisone G, Jon Stoessl A, Bourdenx M, Engeln M, Navailles S, De Deurwaerdère P, Ko WK, Simola N, Morelli M, Groc L, Rodriguez MC, Gurevich EV, Quik M, Morari M, Mellone M, Gardoni F, Tronci E, Guehl D, Tison F, Crossman AR, Kang UJ, Steece-Collier K, Fox S, Carta M, Angela Cenci M, Bézard E (2015) Pathophysiology of l-DOPA-induced motor and non-motor complications in Parkinson’s disease. Prog Neurobiol 132:96–168. https://​doi.​org/​10.​1016/​j.​pneurobio.​2015.​07.​002 PubMedCrossRef
Zurück zum Zitat Berg D, Godau J, Trenkwalder C, Eggert K, Csoti I, Storch A, Huber H, Morelli-Canelo M, Stamelou M, Ries V, Wolz M, Schneider C, Di Paolo T, Gasparini F, Hariry S, Vandemeulebroecke M, Abi-Saab W, Cooke K, Johns D, Gomez-Mancilla B (2011) AFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trials. Mov Disord 26(7):1243–1250. https://doi.org/10.1002/mds.23616 PubMedCrossRef Berg D, Godau J, Trenkwalder C, Eggert K, Csoti I, Storch A, Huber H, Morelli-Canelo M, Stamelou M, Ries V, Wolz M, Schneider C, Di Paolo T, Gasparini F, Hariry S, Vandemeulebroecke M, Abi-Saab W, Cooke K, Johns D, Gomez-Mancilla B (2011) AFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trials. Mov Disord 26(7):1243–1250. https://​doi.​org/​10.​1002/​mds.​23616 PubMedCrossRef
Zurück zum Zitat Beurrier C, Lopez S, Révy D, Selvam C, Goudet C, Lhérondel M, Gubellini P, Kerkerian-LeGoff L, Acher F, Pin JP, Amalric M (2009) Electrophysiological and behavioral evidence that modulation of metabotropic glutamate receptor 4 with a new agonist reverses experimental parkinsonism. FASEB J 23(10):3619–3628. https://doi.org/10.1096/fj.09-131789 PubMedCrossRef Beurrier C, Lopez S, Révy D, Selvam C, Goudet C, Lhérondel M, Gubellini P, Kerkerian-LeGoff L, Acher F, Pin JP, Amalric M (2009) Electrophysiological and behavioral evidence that modulation of metabotropic glutamate receptor 4 with a new agonist reverses experimental parkinsonism. FASEB J 23(10):3619–3628. https://​doi.​org/​10.​1096/​fj.​09-131789 PubMedCrossRef
Zurück zum Zitat Bloomfield C, O’Donnell P, French SJ, Totterdell S (2007) Cholinergic neurons of the adult rat striatum are immunoreactive for glutamatergic N-methyl-d-aspartate 2D but not N-methyl-d-aspartate 2C receptor subunits. Neuroscience 150(3):639–646PubMedPubMedCentralCrossRef Bloomfield C, O’Donnell P, French SJ, Totterdell S (2007) Cholinergic neurons of the adult rat striatum are immunoreactive for glutamatergic N-methyl-d-aspartate 2D but not N-methyl-d-aspartate 2C receptor subunits. Neuroscience 150(3):639–646PubMedPubMedCentralCrossRef
Zurück zum Zitat Calabresi P, Pisani A, Mercuri NB, Bernardi G (1992) Long-term Potentiation in the striatum is unmasked by removing the voltage-dependent magnesium block of NMDA receptor channels. Eur J Neurosci 4(10):929–935PubMedCrossRef Calabresi P, Pisani A, Mercuri NB, Bernardi G (1992) Long-term Potentiation in the striatum is unmasked by removing the voltage-dependent magnesium block of NMDA receptor channels. Eur J Neurosci 4(10):929–935PubMedCrossRef
Zurück zum Zitat Chen Y, Yang W, Li X, Li X, Yang H, Xu Z, Yu S (2015) α-Synuclein-induced internalization of NMDA receptors in hippocampal neurons is associated with reduced inward current and Ca(2 +) influx upon NMDA stimulation. Neuroscience 300:297–306PubMedCrossRef Chen Y, Yang W, Li X, Li X, Yang H, Xu Z, Yu S (2015) α-Synuclein-induced internalization of NMDA receptors in hippocampal neurons is associated with reduced inward current and Ca(2 +) influx upon NMDA stimulation. Neuroscience 300:297–306PubMedCrossRef
Zurück zum Zitat Dekundy A, Pietraszek M, Schaefer D, Cenci MA, Danysz W (2006) Effects of group I metabotropic glutamate receptors blockade in experimental models of Parkinson’s disease. Brain Res Bull 69(3):318–326PubMedCrossRef Dekundy A, Pietraszek M, Schaefer D, Cenci MA, Danysz W (2006) Effects of group I metabotropic glutamate receptors blockade in experimental models of Parkinson’s disease. Brain Res Bull 69(3):318–326PubMedCrossRef
Zurück zum Zitat Diógenes MJ, Dias RB, Rombo DM, Vicente Miranda H, Maiolino F, Guerreiro P, Näsström T, Franquelim HG, Oliveira LM, Castanho MA, Lannfelt L, Bergström J, Ingelsson M, Quintas A, Sebastião AM, Lopes LV, Outeiro TF (2012) Extracellular alpha-synuclein oligomers modulate synaptic transmission and impair LTP via NMDA-receptor activation. J Neurosci 32(34):11750–11762. https://doi.org/10.1523/JNEUROSCI.0234-12.2012 PubMedCrossRef Diógenes MJ, Dias RB, Rombo DM, Vicente Miranda H, Maiolino F, Guerreiro P, Näsström T, Franquelim HG, Oliveira LM, Castanho MA, Lannfelt L, Bergström J, Ingelsson M, Quintas A, Sebastião AM, Lopes LV, Outeiro TF (2012) Extracellular alpha-synuclein oligomers modulate synaptic transmission and impair LTP via NMDA-receptor activation. J Neurosci 32(34):11750–11762. https://​doi.​org/​10.​1523/​JNEUROSCI.​0234-12.​2012 PubMedCrossRef
Zurück zum Zitat Dunah AW, Standaert DG (2001) Dopamine D1 receptor-dependent trafficking of striatal NMDA glutamate receptors to the postsynaptic membrane. J Neurosci 21(15):5546–5558PubMedCrossRef Dunah AW, Standaert DG (2001) Dopamine D1 receptor-dependent trafficking of striatal NMDA glutamate receptors to the postsynaptic membrane. J Neurosci 21(15):5546–5558PubMedCrossRef
Zurück zum Zitat Dunah AW, Wang Y, Yasuda RP, Kameyama K, Huganir RL, Wolfe BB, Standaert DG (2000) Alterations in subunit expression, composition, and phosphorylation of striatal N-methyl-d-aspartate glutamate receptors in a rat 6-hydroxydopamine model of Parkinson’s disease. Mol Pharmacol 57(2):342–352PubMed Dunah AW, Wang Y, Yasuda RP, Kameyama K, Huganir RL, Wolfe BB, Standaert DG (2000) Alterations in subunit expression, composition, and phosphorylation of striatal N-methyl-d-aspartate glutamate receptors in a rat 6-hydroxydopamine model of Parkinson’s disease. Mol Pharmacol 57(2):342–352PubMed
Zurück zum Zitat Eggert K, Squillacote D, Barone P, Dodel R, Katzenschlager R, Emre M, Lees AJ, Rascol O, Poewe W, Tolosa E, Trenkwalder C, Onofrj M, Stocchi F, Nappi G, Kostic V, Potic J, Ruzicka E, Oertel W, German Competence Network on Parkinson’s Disease (2010) Safety and efficacy of perampanel in advanced Parkinson’s disease: a randomized, placebo-controlled study. Mov Disord 25(7):896–905. https://doi.org/10.1002/mds.22974 PubMedCrossRef Eggert K, Squillacote D, Barone P, Dodel R, Katzenschlager R, Emre M, Lees AJ, Rascol O, Poewe W, Tolosa E, Trenkwalder C, Onofrj M, Stocchi F, Nappi G, Kostic V, Potic J, Ruzicka E, Oertel W, German Competence Network on Parkinson’s Disease (2010) Safety and efficacy of perampanel in advanced Parkinson’s disease: a randomized, placebo-controlled study. Mov Disord 25(7):896–905. https://​doi.​org/​10.​1002/​mds.​22974 PubMedCrossRef
Zurück zum Zitat Elahi B, Phielipp N, Chen R (2012) N-methyl-d-aspartate antagonists in levodopa induced dyskinesia: a meta-analysis. Can J Neurol Sci 39(4):465–472PubMedCrossRef Elahi B, Phielipp N, Chen R (2012) N-methyl-d-aspartate antagonists in levodopa induced dyskinesia: a meta-analysis. Can J Neurol Sci 39(4):465–472PubMedCrossRef
Zurück zum Zitat Engers DW, Blobaum AL, Gogliotti RD, Cheung YY, Salovich JM, Garcia-Barrantes PM, Daniels JS, Morrison R, Jones CK, Soars MG, Zhuo X, Hurley J, Macor JE, Bronson JJ, Conn PJ, Lindsley CW, Niswender CM, Hopkins CR (2016) Discovery, synthesis, and preclinical characterization of N-(3-Chloro-4-fluorophenyl)-1H-pyrazolo[4,3-b]pyridin-3-amine (VU0418506), a novel positive allosteric modulator of the metabotropic glutamate receptor 4 (mGlu4). ACS Chem Neurosci 7(9):1192–1200. https://doi.org/10.1021/acschemneuro.6b00035 PubMedPubMedCentralCrossRef Engers DW, Blobaum AL, Gogliotti RD, Cheung YY, Salovich JM, Garcia-Barrantes PM, Daniels JS, Morrison R, Jones CK, Soars MG, Zhuo X, Hurley J, Macor JE, Bronson JJ, Conn PJ, Lindsley CW, Niswender CM, Hopkins CR (2016) Discovery, synthesis, and preclinical characterization of N-(3-Chloro-4-fluorophenyl)-1H-pyrazolo[4,3-b]pyridin-3-amine (VU0418506), a novel positive allosteric modulator of the metabotropic glutamate receptor 4 (mGlu4). ACS Chem Neurosci 7(9):1192–1200. https://​doi.​org/​10.​1021/​acschemneuro.​6b00035 PubMedPubMedCentralCrossRef
Zurück zum Zitat Errico F, Bonito-Oliva A, Bagetta V, Vitucci D, Romano R, Zianni E, Napolitano F, Marinucci S, Di Luca M, Calabresi P, Fisone G, Carta M, Picconi B, Gardoni F, Usiello A (2011) Higher free D-aspartate and N-methyl-d-aspartate levels prevent striatal depotentiation and anticipate l-DOPA-induced dyskinesia. Exp Neurol 232(2):240–250. https://doi.org/10.1016/j.expneurol.2011.09.013 PubMedCrossRef Errico F, Bonito-Oliva A, Bagetta V, Vitucci D, Romano R, Zianni E, Napolitano F, Marinucci S, Di Luca M, Calabresi P, Fisone G, Carta M, Picconi B, Gardoni F, Usiello A (2011) Higher free D-aspartate and N-methyl-d-aspartate levels prevent striatal depotentiation and anticipate l-DOPA-induced dyskinesia. Exp Neurol 232(2):240–250. https://​doi.​org/​10.​1016/​j.​expneurol.​2011.​09.​013 PubMedCrossRef
Zurück zum Zitat Ferreira DG, Temido-Ferreira M, Miranda HV, Batalha VL, Coelho JE, Szegö ÉM, Marques-Morgado I, Vaz SH, Rhee JS, Schmitz M, Zerr I, Lopes LV, Outeiro TF (2017) α-synuclein interacts with PrPC to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci 20(11):1569–1579. https://doi.org/10.1038/nn.4648 PubMedCrossRef Ferreira DG, Temido-Ferreira M, Miranda HV, Batalha VL, Coelho JE, Szegö ÉM, Marques-Morgado I, Vaz SH, Rhee JS, Schmitz M, Zerr I, Lopes LV, Outeiro TF (2017) α-synuclein interacts with PrPC to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci 20(11):1569–1579. https://​doi.​org/​10.​1038/​nn.​4648 PubMedCrossRef
Zurück zum Zitat Fiorentini C, Gardoni F, Spano P, Di Luca M, Missale C (2003) Regulation of dopamine D1 receptor trafficking and desensitization by oligomerization with glutamate N-methyl-d-aspartate receptors. J Biol Chem 278(22):20196–20202PubMedCrossRef Fiorentini C, Gardoni F, Spano P, Di Luca M, Missale C (2003) Regulation of dopamine D1 receptor trafficking and desensitization by oligomerization with glutamate N-methyl-d-aspartate receptors. J Biol Chem 278(22):20196–20202PubMedCrossRef
Zurück zum Zitat Gardoni F, Picconi B, Ghiglieri V, Polli F, Bagetta V, Bernardi G, Cattabeni F, Di Luca M, Calabresi P (2006) A critical interaction between NR2B and MAGUK in l-DOPA induced dyskinesia. J Neurosci 26(11):2914–2922PubMedCrossRef Gardoni F, Picconi B, Ghiglieri V, Polli F, Bagetta V, Bernardi G, Cattabeni F, Di Luca M, Calabresi P (2006) A critical interaction between NR2B and MAGUK in l-DOPA induced dyskinesia. J Neurosci 26(11):2914–2922PubMedCrossRef
Zurück zum Zitat Gubellini P, Pisani A, Centonze D, Bernardi G, Calabresi P (2004) Metabotropic glutamate receptors and striatal synaptic plasticity: implications for neurological diseases. Prog Neurobiol 74(5):271–300PubMedCrossRef Gubellini P, Pisani A, Centonze D, Bernardi G, Calabresi P (2004) Metabotropic glutamate receptors and striatal synaptic plasticity: implications for neurological diseases. Prog Neurobiol 74(5):271–300PubMedCrossRef
Zurück zum Zitat Hallett PJ, Dunah AW, Ravenscroft P, Zhou S, Bezard E, Crossman AR, Brotchie JM, Standaert DG (2005) Alterations of striatal NMDA receptor subunits associated with the development of dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Neuropharmacology 48(4):503–516PubMedCrossRef Hallett PJ, Dunah AW, Ravenscroft P, Zhou S, Bezard E, Crossman AR, Brotchie JM, Standaert DG (2005) Alterations of striatal NMDA receptor subunits associated with the development of dyskinesia in the MPTP-lesioned primate model of Parkinson’s disease. Neuropharmacology 48(4):503–516PubMedCrossRef
Zurück zum Zitat Johnston TH, Fox SH, McIldowie MJ, Piggott MJ, Brotchie JM (2010) Reduction of l-DOPA-induced dyskinesia by the selective metabotropic glutamate receptor 5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson’s disease. J Pharmacol Exp Ther 333(3):865–873. https://doi.org/10.1124/jpet.110.166629 PubMedCrossRef Johnston TH, Fox SH, McIldowie MJ, Piggott MJ, Brotchie JM (2010) Reduction of l-DOPA-induced dyskinesia by the selective metabotropic glutamate receptor 5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson’s disease. J Pharmacol Exp Ther 333(3):865–873. https://​doi.​org/​10.​1124/​jpet.​110.​166629 PubMedCrossRef
Zurück zum Zitat Konitsiotis S, Blanchet PJ, Verhagen L, Lamers E, Chase TN (2000) AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology 54(8):1589–1595PubMedCrossRef Konitsiotis S, Blanchet PJ, Verhagen L, Lamers E, Chase TN (2000) AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology 54(8):1589–1595PubMedCrossRef
Zurück zum Zitat Konradi C, Westin JE, Carta M, Eaton ME, Kuter K, Dekundy A, Lundblad M, Cenci MA (2004) Transcriptome analysis in a rat model of l-DOPA-induced dyskinesia. Neurobiol Dis 17(2):219–236PubMedPubMedCentralCrossRef Konradi C, Westin JE, Carta M, Eaton ME, Kuter K, Dekundy A, Lundblad M, Cenci MA (2004) Transcriptome analysis in a rat model of l-DOPA-induced dyskinesia. Neurobiol Dis 17(2):219–236PubMedPubMedCentralCrossRef
Zurück zum Zitat Kosinski CM, Risso Bradley S, Conn PJ, Levey AI, Landwehrmeyer GB, Penney JB Jr, Young AB, Standaert DG (1999) Localization of metabotropic glutamate receptor 7 mRNA and mGluR7a protein in the rat basal ganglia. J Comp Neurol 415(2):266–284PubMedCrossRef Kosinski CM, Risso Bradley S, Conn PJ, Levey AI, Landwehrmeyer GB, Penney JB Jr, Young AB, Standaert DG (1999) Localization of metabotropic glutamate receptor 7 mRNA and mGluR7a protein in the rat basal ganglia. J Comp Neurol 415(2):266–284PubMedCrossRef
Zurück zum Zitat Lachowicz JE, Sibley DR (1997) Molecular characteristics of mammalian dopamine receptors. Pharmacol Toxicol 81(3):105–113PubMedCrossRef Lachowicz JE, Sibley DR (1997) Molecular characteristics of mammalian dopamine receptors. Pharmacol Toxicol 81(3):105–113PubMedCrossRef
Zurück zum Zitat Lapper SR, Bolam JP (1992) Input from the frontal cortex and the parafascicular nucleus to cholinergic interneurons in the dorsal striatum of the rat. Neuroscience 51(3):533–545PubMedCrossRef Lapper SR, Bolam JP (1992) Input from the frontal cortex and the parafascicular nucleus to cholinergic interneurons in the dorsal striatum of the rat. Neuroscience 51(3):533–545PubMedCrossRef
Zurück zum Zitat Le Poul E, Boléa C, Girard F, Poli S, Charvin D, Campo B, Bortoli J, Bessif A, Luo B, Koser AJ, Hodge LM, Smith KM, DiLella AG, Liverton N, Hess F, Browne SE, Reynolds IJ (2012) A potent and selective metabotropic glutamate receptor 4 positive allosteric modulator improves movement in rodent models of Parkinson’s disease. J Pharmacol Exp Ther 343(1):167–177. https://doi.org/10.1124/jpet.112.196063 PubMedCrossRef Le Poul E, Boléa C, Girard F, Poli S, Charvin D, Campo B, Bortoli J, Bessif A, Luo B, Koser AJ, Hodge LM, Smith KM, DiLella AG, Liverton N, Hess F, Browne SE, Reynolds IJ (2012) A potent and selective metabotropic glutamate receptor 4 positive allosteric modulator improves movement in rodent models of Parkinson’s disease. J Pharmacol Exp Ther 343(1):167–177. https://​doi.​org/​10.​1124/​jpet.​112.​196063 PubMedCrossRef
Zurück zum Zitat Lee FJ, Xue S, Pei L, Vukusic B, Chéry N, Wang Y, Wang YT, Niznik HB, Yu XM, Liu F (2002) Dual regulation of NMDA receptor functions by direct protein-protein interactions with the dopamine D1 receptor. Cell 111(2):219–230PubMedCrossRef Lee FJ, Xue S, Pei L, Vukusic B, Chéry N, Wang Y, Wang YT, Niznik HB, Yu XM, Liu F (2002) Dual regulation of NMDA receptor functions by direct protein-protein interactions with the dopamine D1 receptor. Cell 111(2):219–230PubMedCrossRef
Zurück zum Zitat Lees A, Fahn S, Eggert KM, Jankovic J, Lang A, Micheli F, Mouradian MM, Oertel WH, Olanow CW, Poewe W, Rascol O, Tolosa E, Squillacote D, Kumar D (2012) Perampanel, an AMPA antagonist, found to have no benefit in reducing “off” time in Parkinson’s disease. Mov Disord 27(2):284–288. https://doi.org/10.1002/mds.23983 PubMedCrossRef Lees A, Fahn S, Eggert KM, Jankovic J, Lang A, Micheli F, Mouradian MM, Oertel WH, Olanow CW, Poewe W, Rascol O, Tolosa E, Squillacote D, Kumar D (2012) Perampanel, an AMPA antagonist, found to have no benefit in reducing “off” time in Parkinson’s disease. Mov Disord 27(2):284–288. https://​doi.​org/​10.​1002/​mds.​23983 PubMedCrossRef
Zurück zum Zitat Levandis G, Bazzini E, Armentero MT, Nappi G, Blandini F (2008) Systemic administration of an mGluR5 antagonist, but not unilateral subthalamic lesion, counteracts l-DOPA-induced dyskinesias in a rodent model of Parkinson’s disease. Neurobiol Dis 29(1):161–168PubMedCrossRef Levandis G, Bazzini E, Armentero MT, Nappi G, Blandini F (2008) Systemic administration of an mGluR5 antagonist, but not unilateral subthalamic lesion, counteracts l-DOPA-induced dyskinesias in a rodent model of Parkinson’s disease. Neurobiol Dis 29(1):161–168PubMedCrossRef
Zurück zum Zitat Marin C, Jiménez A, Bonastre M, Vila M, Agid Y, Hirsch EC, Tolosa E (2001) LY293558, an AMPA glutamate receptor antagonist, prevents and reverses levodopa-induced motor alterations in Parkinsonian rats. Synapse 42(1):40–47PubMedCrossRef Marin C, Jiménez A, Bonastre M, Vila M, Agid Y, Hirsch EC, Tolosa E (2001) LY293558, an AMPA glutamate receptor antagonist, prevents and reverses levodopa-induced motor alterations in Parkinsonian rats. Synapse 42(1):40–47PubMedCrossRef
Zurück zum Zitat Matsui T, Kita H (2003) Activation of group III metabotropic glutamate receptors presynaptically reduces both GABAergic and glutamatergic transmission in the rat globus pallidus. Neuroscience 122(3):727–737PubMedCrossRef Matsui T, Kita H (2003) Activation of group III metabotropic glutamate receptors presynaptically reduces both GABAergic and glutamatergic transmission in the rat globus pallidus. Neuroscience 122(3):727–737PubMedCrossRef
Zurück zum Zitat Mela F, Marti M, Dekundy A, Danysz W, Morari M, Cenci MA (2007) Antagonism of metabotropic glutamate receptor type 5 attenuates l-DOPA-induced dyskinesia and its molecular and neurochemical correlates in a rat model of Parkinson’s disease. J Neurochem 101(2):483–497PubMedCrossRef Mela F, Marti M, Dekundy A, Danysz W, Morari M, Cenci MA (2007) Antagonism of metabotropic glutamate receptor type 5 attenuates l-DOPA-induced dyskinesia and its molecular and neurochemical correlates in a rat model of Parkinson’s disease. J Neurochem 101(2):483–497PubMedCrossRef
Zurück zum Zitat Missale C, Nash SR, Robinson SW, Jaber M, Caron MG (1998) Dopamine receptors: from structure to function. Physiol Rev 78(1):189–225PubMedCrossRef Missale C, Nash SR, Robinson SW, Jaber M, Caron MG (1998) Dopamine receptors: from structure to function. Physiol Rev 78(1):189–225PubMedCrossRef
Zurück zum Zitat Moreau C, Delval A, Tiffreau V, Defebvre L, Dujardin K, Duhamel A, Petyt G, Hossein-Foucher C, Blum D, Sablonnière B, Schraen S, Allorge D, Destée A, Bordet R, Devos D (2013) Memantine for axial signs in Parkinson’s disease: a randomised, double-blind, placebo-controlled pilot study. J Neurol Neurosurg Psychiatry 84(5):552–555. https://doi.org/10.1136/jnnp-2012-303182 PubMedCrossRef Moreau C, Delval A, Tiffreau V, Defebvre L, Dujardin K, Duhamel A, Petyt G, Hossein-Foucher C, Blum D, Sablonnière B, Schraen S, Allorge D, Destée A, Bordet R, Devos D (2013) Memantine for axial signs in Parkinson’s disease: a randomised, double-blind, placebo-controlled pilot study. J Neurol Neurosurg Psychiatry 84(5):552–555. https://​doi.​org/​10.​1136/​jnnp-2012-303182 PubMedCrossRef
Zurück zum Zitat Morissette M, Dridi M, Calon F, Hadj Tahar A, Meltzer LT, Bédard PJ, Di Paolo T (2006) Prevention of dyskinesia by an NMDA receptor antagonist in MPTP monkeys: effect on adenosine A2A receptors. Synapse 60(3):239–250PubMedCrossRef Morissette M, Dridi M, Calon F, Hadj Tahar A, Meltzer LT, Bédard PJ, Di Paolo T (2006) Prevention of dyskinesia by an NMDA receptor antagonist in MPTP monkeys: effect on adenosine A2A receptors. Synapse 60(3):239–250PubMedCrossRef
Zurück zum Zitat Nash JE, Fox SH, Henry B, Hill MP, Peggs D, McGuire S, Maneuf Y, Hille C, Brotchie JM, Crossman AR (2000) Antiparkinsonian actions of ifenprodil in the MPTP-lesioned marmoset model of Parkinson’s disease. Exp Neurol 165(1):136–142PubMedCrossRef Nash JE, Fox SH, Henry B, Hill MP, Peggs D, McGuire S, Maneuf Y, Hille C, Brotchie JM, Crossman AR (2000) Antiparkinsonian actions of ifenprodil in the MPTP-lesioned marmoset model of Parkinson’s disease. Exp Neurol 165(1):136–142PubMedCrossRef
Zurück zum Zitat Nash JE, Johnston TH, Collingridge GL, Garner CC, Brotchie JM (2005a) Subcellular redistribution of the synapse-associated proteins PSD-95 and SAP97 in animal models of Parkinson’s disease and l-DOPA-induced dyskinesia. FASEB J 19(6):583–585PubMedCrossRef Nash JE, Johnston TH, Collingridge GL, Garner CC, Brotchie JM (2005a) Subcellular redistribution of the synapse-associated proteins PSD-95 and SAP97 in animal models of Parkinson’s disease and l-DOPA-induced dyskinesia. FASEB J 19(6):583–585PubMedCrossRef
Zurück zum Zitat Nash JE, Ravenscroft P, McGuire S, Crossman AR, Menniti FS, Brotchie JM (2005b) The NR2B-selective NMDA receptor antagonist CP-101,606 exacerbates l-DOPA-induced dyskinesia and provides mild potentiation of anti-parkinsonian effects of l-DOPA in the MPTP-lesioned marmoset model of Parkinson’s disease. Exp Neurol 188(2):471–479CrossRef Nash JE, Ravenscroft P, McGuire S, Crossman AR, Menniti FS, Brotchie JM (2005b) The NR2B-selective NMDA receptor antagonist CP-101,606 exacerbates l-DOPA-induced dyskinesia and provides mild potentiation of anti-parkinsonian effects of l-DOPA in the MPTP-lesioned marmoset model of Parkinson’s disease. Exp Neurol 188(2):471–479CrossRef
Zurück zum Zitat Niswender CM, Johnson KA, Weaver CD, Jones CK, Xiang Z, Luo Q, Rodriguez AL, Marlo JE, de Paulis T, Thompson AD, Days EL, Nalywajko T, Austin CA, Williams MB, Ayala JE, Williams R, Lindsley CW, Conn PJ (2008) Discovery, characterization, and antiparkinsonian effect of novel positive allosteric modulators of metabotropic glutamate receptor 4. Mol Pharmacol 74(5):1345–1358. https://doi.org/10.1124/mol.108.049551 PubMedPubMedCentralCrossRef Niswender CM, Johnson KA, Weaver CD, Jones CK, Xiang Z, Luo Q, Rodriguez AL, Marlo JE, de Paulis T, Thompson AD, Days EL, Nalywajko T, Austin CA, Williams MB, Ayala JE, Williams R, Lindsley CW, Conn PJ (2008) Discovery, characterization, and antiparkinsonian effect of novel positive allosteric modulators of metabotropic glutamate receptor 4. Mol Pharmacol 74(5):1345–1358. https://​doi.​org/​10.​1124/​mol.​108.​049551 PubMedPubMedCentralCrossRef
Zurück zum Zitat Niswender CM, Jones CK, Lin X, Bubser M, Thompson Gray A, Blobaum AL, Engers DW, Rodriguez AL, Loch MT, Daniels JS, Lindsley CW, Hopkins CR, Javitch JA, Conn PJ (2016) Development and antiparkinsonian activity of VU0418506, a selective positive allosteric modulator of metabotropic glutamate receptor 4 homomers without activity at mGlu2/4 heteromers. ACS Chem Neurosci 7(9):1201–1211. https://doi.org/10.1021/acschemneuro.6b00036 PubMedPubMedCentralCrossRef Niswender CM, Jones CK, Lin X, Bubser M, Thompson Gray A, Blobaum AL, Engers DW, Rodriguez AL, Loch MT, Daniels JS, Lindsley CW, Hopkins CR, Javitch JA, Conn PJ (2016) Development and antiparkinsonian activity of VU0418506, a selective positive allosteric modulator of metabotropic glutamate receptor 4 homomers without activity at mGlu2/4 heteromers. ACS Chem Neurosci 7(9):1201–1211. https://​doi.​org/​10.​1021/​acschemneuro.​6b00036 PubMedPubMedCentralCrossRef
Zurück zum Zitat Oh JD, Russell DS, Vaughan CL, Chase TN (1998) Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and l-DOPA administration. Brain Res 813(1):150–159PubMedCrossRef Oh JD, Russell DS, Vaughan CL, Chase TN (1998) Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and l-DOPA administration. Brain Res 813(1):150–159PubMedCrossRef
Zurück zum Zitat Paillé V, Picconi B, Bagetta V, Ghiglieri V, Sgobio C, Di Filippo M, Viscomi MT, Giampà C, Fusco FR, Gardoni F, Bernardi G, Greengard P, Di Luca M, Calabresi P (2010) Distinct levels of dopamine denervation differentially alter striatal synaptic plasticity and NMDA receptor subunit composition. J Neurosci 30(42):14182–14193. https://doi.org/10.1523/JNEUROSCI.2149-10.2010 PubMedCrossRef Paillé V, Picconi B, Bagetta V, Ghiglieri V, Sgobio C, Di Filippo M, Viscomi MT, Giampà C, Fusco FR, Gardoni F, Bernardi G, Greengard P, Di Luca M, Calabresi P (2010) Distinct levels of dopamine denervation differentially alter striatal synaptic plasticity and NMDA receptor subunit composition. J Neurosci 30(42):14182–14193. https://​doi.​org/​10.​1523/​JNEUROSCI.​2149-10.​2010 PubMedCrossRef
Zurück zum Zitat Pei L, Lee FJ, Moszczynska A, Vukusic B, Liu F (2004) Regulation of dopamine D1 receptor function by physical interaction with the NMDA receptors. J Neurosci 24(5):1149–1158PubMedCrossRef Pei L, Lee FJ, Moszczynska A, Vukusic B, Liu F (2004) Regulation of dopamine D1 receptor function by physical interaction with the NMDA receptors. J Neurosci 24(5):1149–1158PubMedCrossRef
Zurück zum Zitat Picconi B, Gardoni F, Centonze D, Mauceri D, Cenci MA, Bernardi G, Calabresi P, Di Luca M (2004) Abnormal Ca2+-calmodulin-dependent protein kinase II function mediates synaptic and motor deficits in experimental parkinsonism. J Neurosci 24(23):5283–5291PubMedCrossRef Picconi B, Gardoni F, Centonze D, Mauceri D, Cenci MA, Bernardi G, Calabresi P, Di Luca M (2004) Abnormal Ca2+-calmodulin-dependent protein kinase II function mediates synaptic and motor deficits in experimental parkinsonism. J Neurosci 24(23):5283–5291PubMedCrossRef
Zurück zum Zitat Porras G, Berthet A, Dehay B, Li Q, Ladepeche L, Normand E, Dovero S, Martinez A, Doudnikoff E, Martin-Négrier ML, Chuan Q, Bloch B, Choquet D, Boué-Grabot E, Groc L, Bezard E (2012) PSD-95 expression controls l-DOPA dyskinesia through dopamine D1 receptor trafficking. J Clin Invest 122(11):3977–3989. https://doi.org/10.1172/JCI59426 PubMedPubMedCentralCrossRef Porras G, Berthet A, Dehay B, Li Q, Ladepeche L, Normand E, Dovero S, Martinez A, Doudnikoff E, Martin-Négrier ML, Chuan Q, Bloch B, Choquet D, Boué-Grabot E, Groc L, Bezard E (2012) PSD-95 expression controls l-DOPA dyskinesia through dopamine D1 receptor trafficking. J Clin Invest 122(11):3977–3989. https://​doi.​org/​10.​1172/​JCI59426 PubMedPubMedCentralCrossRef
Zurück zum Zitat Rouse ST, Marino MJ, Bradley SR, Awad H, Wittmann M, Conn PJ (2000) Distribution and roles of metabotropic glutamate receptors in the basal ganglia motor circuit: implications for treatment of Parkinson’s disease and related disorders. Pharmacol Ther 88(3):427–435PubMedCrossRef Rouse ST, Marino MJ, Bradley SR, Awad H, Wittmann M, Conn PJ (2000) Distribution and roles of metabotropic glutamate receptors in the basal ganglia motor circuit: implications for treatment of Parkinson’s disease and related disorders. Pharmacol Ther 88(3):427–435PubMedCrossRef
Zurück zum Zitat Samadi P, Grégoire L, Morissette M, Calon F, Hadj Tahar A, Dridi M, Belanger N, Meltzer LT, Bédard PJ, Di Paolo T (2008) mGluR5 metabotropic glutamate receptors and dyskinesias in MPTP monkeys. Neurobiol Aging 29(7):1040–1051PubMedCrossRef Samadi P, Grégoire L, Morissette M, Calon F, Hadj Tahar A, Dridi M, Belanger N, Meltzer LT, Bédard PJ, Di Paolo T (2008) mGluR5 metabotropic glutamate receptors and dyskinesias in MPTP monkeys. Neurobiol Aging 29(7):1040–1051PubMedCrossRef
Zurück zum Zitat Santini E, Valjent E, Usiello A, Carta M, Borgkvist A, Girault JA, Hervé D, Greengard P, Fisone G (2007) Critical involvement of cAMP/DARPP-32 and extracellular signal-regulated protein kinase signaling in l-DOPA-induced dyskinesia. J Neurosci 27(26):6995–7005PubMedCrossRef Santini E, Valjent E, Usiello A, Carta M, Borgkvist A, Girault JA, Hervé D, Greengard P, Fisone G (2007) Critical involvement of cAMP/DARPP-32 and extracellular signal-regulated protein kinase signaling in l-DOPA-induced dyskinesia. J Neurosci 27(26):6995–7005PubMedCrossRef
Zurück zum Zitat Scott L, Zelenin S, Malmersjö S, Kowalewski JM, Markus EZ, Nairn AC, Greengard P, Brismar H, Aperia A (2006) Allosteric changes of the NMDA receptor trap diffusible dopamine 1 receptors in spines. Proc Natl Acad Sci USA 103(3):762–767PubMedCrossRef Scott L, Zelenin S, Malmersjö S, Kowalewski JM, Markus EZ, Nairn AC, Greengard P, Brismar H, Aperia A (2006) Allosteric changes of the NMDA receptor trap diffusible dopamine 1 receptors in spines. Proc Natl Acad Sci USA 103(3):762–767PubMedCrossRef
Zurück zum Zitat Silverdale MA, Nicholson SL, Crossman AR, Brotchie JM (2005) Topiramate reduces levodopa-induced dyskinesia in the MPTP-lesioned marmoset model of Parkinson’s disease. Mov Disord 20(4):403–409PubMedCrossRef Silverdale MA, Nicholson SL, Crossman AR, Brotchie JM (2005) Topiramate reduces levodopa-induced dyskinesia in the MPTP-lesioned marmoset model of Parkinson’s disease. Mov Disord 20(4):403–409PubMedCrossRef
Zurück zum Zitat Tahar AH, Grégoire L, Darré A, Bélanger N, Meltzer L, Bédard PJ (2004) Effect of a selective glutamate antagonist on l-DOPA-induced dyskinesias in drug-naive parkinsonian monkeys. Neurobiol Dis 15(2):171–176CrossRef Tahar AH, Grégoire L, Darré A, Bélanger N, Meltzer L, Bédard PJ (2004) Effect of a selective glutamate antagonist on l-DOPA-induced dyskinesias in drug-naive parkinsonian monkeys. Neurobiol Dis 15(2):171–176CrossRef
Zurück zum Zitat Testa CM, Standaert DG, Young AB, Penney JB Jr (1994) Metabotropic glutamate receptor mRNA expression in the basal ganglia of the rat. J Neurosci 14(5 Pt 2):3005–3018PubMedCrossRef Testa CM, Standaert DG, Young AB, Penney JB Jr (1994) Metabotropic glutamate receptor mRNA expression in the basal ganglia of the rat. J Neurosci 14(5 Pt 2):3005–3018PubMedCrossRef
Zurück zum Zitat Testa CM, Standaert DG, Landwehrmeyer GB, Penney JB Jr, Young AB (1995) Differential expression of mGluR5 metabotropic glutamate receptor mRNA by rat striatal neurons. J Comp Neurol 354(2):241–252PubMedCrossRef Testa CM, Standaert DG, Landwehrmeyer GB, Penney JB Jr, Young AB (1995) Differential expression of mGluR5 metabotropic glutamate receptor mRNA by rat striatal neurons. J Comp Neurol 354(2):241–252PubMedCrossRef
Zurück zum Zitat Tison F, Keywood C, Wakefield M, Durif F, Corvol JC, Eggert K, Lew M, Isaacson S, Bezard E, Poli SM, Goetz CG, Trenkwalder C, Rascol O (2016) A Phase 2A trial of the novel mGluR5-negative allosteric modulator dipraglurant for levodopa-induced dyskinesia in Parkinson’s Disease. Mov Disord 31(9):1373–1380. https://doi.org/10.1002/mds.26659 PubMedCrossRef Tison F, Keywood C, Wakefield M, Durif F, Corvol JC, Eggert K, Lew M, Isaacson S, Bezard E, Poli SM, Goetz CG, Trenkwalder C, Rascol O (2016) A Phase 2A trial of the novel mGluR5-negative allosteric modulator dipraglurant for levodopa-induced dyskinesia in Parkinson’s Disease. Mov Disord 31(9):1373–1380. https://​doi.​org/​10.​1002/​mds.​26659 PubMedCrossRef
Zurück zum Zitat Tozzi A, de Iure A, Bagetta V, Tantucci M, Durante V, Quiroga-Varela A, Costa C, Di Filippo M, Ghiglieri V, Latagliata EC, Wegrzynowicz M, Decressac M, Giampà C, Dalley JW, Xia J, Gardoni F, Mellone M, El-Agnaf OM, Ardah MT, Puglisi-Allegra S, Björklund A, Spillantini MG, Picconi B, Calabresi P (2016) Alpha-Synuclein produces early behavioral alterations via striatal cholinergic synaptic dysfunction by interacting with GluN2D N-methyl-d-Aspartate receptor subunit. Biol Psychiatry 79(5):402–414. https://doi.org/10.1016/j.biopsych.2015.08.013 PubMedCrossRef Tozzi A, de Iure A, Bagetta V, Tantucci M, Durante V, Quiroga-Varela A, Costa C, Di Filippo M, Ghiglieri V, Latagliata EC, Wegrzynowicz M, Decressac M, Giampà C, Dalley JW, Xia J, Gardoni F, Mellone M, El-Agnaf OM, Ardah MT, Puglisi-Allegra S, Björklund A, Spillantini MG, Picconi B, Calabresi P (2016) Alpha-Synuclein produces early behavioral alterations via striatal cholinergic synaptic dysfunction by interacting with GluN2D N-methyl-d-Aspartate receptor subunit. Biol Psychiatry 79(5):402–414. https://​doi.​org/​10.​1016/​j.​biopsych.​2015.​08.​013 PubMedCrossRef
Zurück zum Zitat Valenti O, Marino MJ, Wittmann M, Lis E, DiLella AG, Kinney GG, Conn PJ (2003) Group III metabotropic glutamate receptor-mediated modulation of the striatopallidal synapse. J Neurosci 23(18):7218–7226PubMedCrossRef Valenti O, Marino MJ, Wittmann M, Lis E, DiLella AG, Kinney GG, Conn PJ (2003) Group III metabotropic glutamate receptor-mediated modulation of the striatopallidal synapse. J Neurosci 23(18):7218–7226PubMedCrossRef
Zurück zum Zitat Valenti O, Mannaioni G, Seabrook GR, Conn PJ, Marino MJ (2005) Group III metabotropic glutamate-receptor-mediated modulation of excitatory transmission in rodent substantia nigra pars compacta dopamine neurons. J Pharmacol Exp Ther 313(3):1296–1304PubMedCrossRef Valenti O, Mannaioni G, Seabrook GR, Conn PJ, Marino MJ (2005) Group III metabotropic glutamate-receptor-mediated modulation of excitatory transmission in rodent substantia nigra pars compacta dopamine neurons. J Pharmacol Exp Ther 313(3):1296–1304PubMedCrossRef
Zurück zum Zitat Wessell RH, Ahmed SM, Menniti FS, Dunbar GL, Chase TN, Oh JD (2004) NR2B selective NMDA receptor antagonist CP-101,606 prevents levodopa-induced motor response alterations in Hemi-Parkinsonian rats. Neuropharmacology 47(2):184–194PubMedCrossRef Wessell RH, Ahmed SM, Menniti FS, Dunbar GL, Chase TN, Oh JD (2004) NR2B selective NMDA receptor antagonist CP-101,606 prevents levodopa-induced motor response alterations in Hemi-Parkinsonian rats. Neuropharmacology 47(2):184–194PubMedCrossRef
Metadaten
Titel
Glutamatergic mechanisms in l-DOPA-induced dyskinesia and therapeutic implications
verfasst von
Manuela Mellone
Fabrizio Gardoni
Publikationsdatum
31.01.2018
Verlag
Springer Vienna
Erschienen in
Journal of Neural Transmission / Ausgabe 8/2018
Print ISSN: 0300-9564
Elektronische ISSN: 1435-1463
DOI
https://doi.org/10.1007/s00702-018-1846-8

Weitere Artikel der Ausgabe 8/2018

Journal of Neural Transmission 8/2018 Zur Ausgabe

Neurology and Preclinical Neurological Studies - Review Article

On the neuronal circuitry mediating l-DOPA-induced dyskinesia

Neurology and Preclinical Neurological Studies - Review Article

Dopamine receptors: homomeric and heteromeric complexes in l-DOPA-induced dyskinesia

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.