메뉴 건너뛰기




Volumn 19, Issue 9, 2005, Pages 723-743

Treatment of Parkinson's disease: What's on the Horizon?

Author keywords

[No Author keywords available]

Indexed keywords

5 AMINO 2 (2 FURYL) 7 (2 PHENYLETHYL)PYRAZOLO[4,3 E][1,2,4]TRIAZOLO[1,5 C]PYRIMIDINE; 6 NITRO 7 SULFAMOYLBENZO[F]QUINOXALINE 2,3 DIONE; 6 QUINOXALINECARBOXYLIC ACID PIPERIDIDE; ADENOSINE A2A RECEPTOR ANTAGONIST; AMANTADINE; AMPA RECEPTOR AGONIST; AMPA RECEPTOR ANTAGONIST; ANTIOXIDANT; ANTIPARKINSON AGENT; APOMORPHINE; CAFFEINE; CARBIDOPA PLUS LEVODOPA; CEP 1347; DECAHYDRO 6 [2 (1H TETRAZOL 5 YL)ETHYL] 3 ISOQUINOLINECARBOXYLIC ACID; DOPAMINE RECEPTOR BLOCKING AGENT; DOPAMINE RECEPTOR STIMULATING AGENT; GLIAL CELL LINE DERIVED NEUROTROPHIC FACTOR; GLUTAMATE ALPHA AMINO 3 HYDROXY 5 METHYL 4 ISOXAZOLEPROPIONIC ACID RECEPTOR ANTAGONIST; GLUTAMATE DECARBOXYLASE; ISTRADEFYLLINE; LEVODOPA; MEMANTINE; N METHYL DEXTRO ASPARTIC ACID RECEPTOR BLOCKING AGENT; NS 2330; PRAMIPEXOLE; SELEGILINE; TALAMPANEL; TOCOPHEROL; UBIDECARENONE; UNCLASSIFIED DRUG; UNINDEXED DRUG; VIRUS VECTOR;

EID: 24944582908     PISSN: 11727047     EISSN: None     Source Type: Journal    
DOI: 10.2165/00023210-200519090-00001     Document Type: Review
Times cited : (68)

References (130)
  • 1
    • 0037154184 scopus 로고    scopus 로고
    • Recent advances in the genetics and pathogenesis of Parkinson's disease
    • Mouradian M. Recent advances in the genetics and pathogenesis of Parkinson's disease. Neurology 2002; 58 (2): 179-85
    • (2002) Neurology , vol.58 , Issue.2 , pp. 179-185
    • Mouradian, M.1
  • 2
    • 0036316947 scopus 로고    scopus 로고
    • Impairment of the ubiquitin-proteasome system causes dopaminergic cell death and inclusion body formation in the ventral mesence-phalic cultures
    • McNaught K, Mytilineou C, Jnobaptiste R, et al. Impairment of the ubiquitin-proteasome system causes dopaminergic cell death and inclusion body formation in the ventral mesence-phalic cultures. J Neurochem 2002; 81: 301-6
    • (2002) J Neurochem , vol.81 , pp. 301-306
    • McNaught, K.1    Mytilineou, C.2    Jnobaptiste, R.3
  • 3
    • 0030778373 scopus 로고    scopus 로고
    • Entacapone improves motor fluctuations in levodopa-treated Parkinson's disease patients
    • Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson's disease patients. Ann Neurol 1997; 42 (5): 747-55
    • (1997) Ann Neurol , vol.42 , Issue.5 , pp. 747-755
  • 4
    • 0031773064 scopus 로고    scopus 로고
    • Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations
    • Rinne UK, Larsen JP, Siden A, et al. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. Neurology 1998; 51: 1309-14
    • (1998) Neurology , vol.51 , pp. 1309-1314
    • Rinne, U.K.1    Larsen, J.P.2    Siden, A.3
  • 5
    • 0029895288 scopus 로고    scopus 로고
    • A double-blind pharmacokinetic and clinical dose-response study of entacapone as an adjuvant to levodopa therapy in advanced Parkinson's disease
    • Ruottinen HM, Rinne UK. A double-blind pharmacokinetic and clinical dose-response study of entacapone as an adjuvant to levodopa therapy in advanced Parkinson's disease. Clin Neuropharmacol 1996; 19: 283-96
    • (1996) Clin Neuropharmacol , vol.19 , pp. 283-296
    • Ruottinen, H.M.1    Rinne, U.K.2
  • 6
    • 0345600914 scopus 로고    scopus 로고
    • 2A antagonists as novel nondopaminergic therapy for motor dysfunction in PD
    • 2A antagonists as novel nondopaminergic therapy for motor dysfunction in PD. Neurology 2003; 61 Suppl. 6: S33-8
    • (2003) Neurology , vol.61 , Issue.6 SUPPL.
    • Jenner, P.1
  • 8
    • 0022939250 scopus 로고
    • Striatal in homogeneities and basal ganglia origin
    • Penney Jr J, Young A. Striatal in homogeneities and basal ganglia origin. Mov Disord 1986; 1: 3-15
    • (1986) Mov Disord , vol.1 , pp. 3-15
    • Penney Jr., J.1    Young, A.2
  • 10
    • 0034105156 scopus 로고    scopus 로고
    • Striatal mechanisms and pathogenesis of parkinsonian signs and motor complications
    • Chase T, Oh J. Striatal mechanisms and pathogenesis of parkinsonian signs and motor complications. Ann Neurol 2000; 27 Suppl. 1: S122-9
    • (2000) Ann Neurol , vol.27 , Issue.1 SUPPL.
    • Chase, T.1    Oh, J.2
  • 11
    • 0027339887 scopus 로고
    • 2A receptors regulate the gene expression of striatopallidal and striatonigral neurons
    • 2A receptors regulate the gene expression of striatopallidal and striatonigral neurons. J Neurosci 1993; 13: 1080-7
    • (1993) J Neurosci , vol.13 , pp. 1080-1087
    • Schiffmann, S.1    Vanderhaeghen, J.2
  • 13
    • 0033806207 scopus 로고    scopus 로고
    • Evidence for adenosine/dopamine receptor interactions: Indications for heteromerization
    • Franco R, Ferre S, Agnati L, et al. Evidence for adenosine/dopamine receptor interactions: indications for heteromerization. Neuropsychopharmacology 2000; 23: S50-9
    • (2000) Neuropsychopharmacology , vol.23
    • Franco, R.1    Ferre, S.2    Agnati, L.3
  • 14
    • 0043126985 scopus 로고    scopus 로고
    • Nondopaminergic symptomatic therapies for Parkinson's disease
    • Feigin A. Nondopaminergic symptomatic therapies for Parkinson's disease. Neurology 2003; 61: 286-7
    • (2003) Neurology , vol.61 , pp. 286-287
    • Feigin, A.1
  • 15
    • 0344738704 scopus 로고    scopus 로고
    • 2A and dopamine receptor interactions in basal ganglia of dopamine denervated rats
    • 2A and dopamine receptor interactions in basal ganglia of dopamine denervated rats. Neurology 2003; 61 Suppl. 6: S39-42
    • (2003) Neurology , vol.61 , Issue.6 SUPPL.
    • Carta, A.1    Pinna, A.2    Tronci, E.3
  • 16
    • 0031594271 scopus 로고    scopus 로고
    • 2A antagonist: A novel antiparkinsonian agent that does not provoke dyskinesia in parkinsonian monkeys
    • 2A antagonist: a novel antiparkinsonian agent that does not provoke dyskinesia in parkinsonian monkeys. Ann Neurol 1998; 43: 507-13
    • (1998) Ann Neurol , vol.43 , pp. 507-513
    • Kanda, T.1    Jackson, M.2    Smith, L.3
  • 17
    • 0043126954 scopus 로고    scopus 로고
    • 2A receptor antagonist istradefylline in advanced PD
    • 2A receptor antagonist istradefylline in advanced PD. Neurology 2003; 61: 297-303
    • (2003) Neurology , vol.61 , pp. 297-303
    • Hauser, R.1    Hubble, J.2    Truong, D.3
  • 18
    • 0031708101 scopus 로고    scopus 로고
    • COMT inhibition in the treatment of Parkinson's disease
    • Ruottinen HM, Rinne UK. COMT inhibition in the treatment of Parkinson's disease. J Neurol 1998; 245 Suppl. 3: 25-34
    • (1998) J Neurol , vol.245 , Issue.3 SUPPL. , pp. 25-34
    • Ruottinen, H.M.1    Rinne, U.K.2
  • 19
    • 0034049544 scopus 로고    scopus 로고
    • 2A antagonist KW-6002 with L-dopa or with selec-tive D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in MPTP-treated monkeys
    • 2A antagonist KW-6002 with L-dopa or with selec-tive D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in MPTP-treated monkeys. Exp Neurol 2000; 162: 321-7
    • (2000) Exp Neurol , vol.162 , pp. 321-327
    • Kanda, T.1    Jackson, M.2    Smith, L.3
  • 21
    • 0033910931 scopus 로고    scopus 로고
    • Sleep episodes in Parkinson's disease: A wake-up call
    • Frucht S, Greene P, Fahn S. Sleep episodes in Parkinson's disease: a wake-up call. Mov Disord 2000; 15: 601-3
    • (2000) Mov Disord , vol.15 , pp. 601-603
    • Frucht, S.1    Greene, P.2    Fahn, S.3
  • 22
    • 0032975093 scopus 로고    scopus 로고
    • Falling asleep at the wheel: Motor vehicle mishaps in persons taking pramipexole and ropinirole
    • Frucht S, Rogers J, Greene P, et al. Falling asleep at the wheel: motor vehicle mishaps in persons taking pramipexole and ropinirole. Neurology 1999; 52: 1908-10
    • (1999) Neurology , vol.52 , pp. 1908-1910
    • Frucht, S.1    Rogers, J.2    Greene, P.3
  • 23
    • 0038760845 scopus 로고    scopus 로고
    • Sleep attacks, daytime sleepi-ness, and dopamine agonists in Parkinson's disease
    • Paus S, Brecht H, Köster J, et al. Sleep attacks, daytime sleepi-ness, and dopamine agonists in Parkinson's disease. Mov Disord 2003; 18: 659-67
    • (2003) Mov Disord , vol.18 , pp. 659-667
    • Paus, S.1    Brecht, H.2    Köster, J.3
  • 24
    • 0033065106 scopus 로고    scopus 로고
    • Pericardial, retroperitoneal, and pleural fibrosis induced by pergolide
    • Shaunak S, Wilkins A, Pillig J, et al. Pericardial, retroperitoneal, and pleural fibrosis induced by pergolide. J Neurol Neurosurg Psychiatry 1999; 66: 79-81
    • (1999) J Neurol Neurosurg Psychiatry , vol.66 , pp. 79-81
    • Shaunak, S.1    Wilkins, A.2    Pillig, J.3
  • 25
    • 0344306440 scopus 로고    scopus 로고
    • 2A receptor antagonists in animal models of Parkinson's disease
    • 2A receptor antagonists in animal models of Parkinson's disease. Neurology 2003; 61 Suppl. 6: S55-61
    • (2003) Neurology , vol.61 , Issue.6 SUPPL.
    • Schwarzschild, M.1    Xu, K.2    Oztas, E.3
  • 26
    • 0344738721 scopus 로고    scopus 로고
    • 2A receptors in neuroprotection of dopaminergic neurons
    • 2A receptors in neuroprotection of dopaminergic neurons. Neurology 2003; 61 Suppl. 6: S49-50
    • (2003) Neurology , vol.61 , Issue.6 SUPPL.
    • Pedata, F.1    Pugliese, A.2    Melani, A.3
  • 27
    • 0036714747 scopus 로고    scopus 로고
    • A meta-analysis of coffee drinking, cigarette smoking, and the risk of Parkinson's disease
    • Hernàn M, Takkouche B, Caamaño-Isorna F, et al. A meta-analysis of coffee drinking, cigarette smoking, and the risk of Parkinson's disease. Ann Neurol 2002; 52: 276-84
    • (2002) Ann Neurol , vol.52 , pp. 276-284
    • Hernàn, M.1    Takkouche, B.2    Caamaño-Isorna, F.3
  • 28
    • 0032924712 scopus 로고    scopus 로고
    • Nutritional and occupational factors influencing the risk of Parkinson's disease: A case-controlled study in southeastern Sweden
    • Fall P, Fredrikson M, Axelson O, et al. Nutritional and occupational factors influencing the risk of Parkinson's disease: a case-controlled study in southeastern Sweden. Mov Disord 1999; 14: 28-37
    • (1999) Mov Disord , vol.14 , pp. 28-37
    • Fall, P.1    Fredrikson, M.2    Axelson, O.3
  • 29
    • 0345492465 scopus 로고    scopus 로고
    • Caffeinated clues from epidemiology of Parkinson's disease
    • Ascherio A, Chen H. Caffeinated clues from epidemiology of Parkinson's disease. Neurology 2001; 61 Suppl. 6: S51-4
    • (2001) Neurology , vol.61 , Issue.6 SUPPL.
    • Ascherio, A.1    Chen, H.2
  • 30
    • 0027530638 scopus 로고
    • Effects of tocopherol and deprenyl on the progression of disability in early Parkinson's disease
    • The Parkinson Study Group
    • Effects of tocopherol and deprenyl on the progression of disability in early Parkinson's disease. The Parkinson Study Group. N Engl J Med 1993; 328 (3): 176-83
    • (1993) N Engl J Med , vol.328 , Issue.3 , pp. 176-183
  • 31
    • 0027972318 scopus 로고
    • Does selegiline delay progression of Parkinson's disease? A critical re-evaluation of the DATATOP study
    • Ward C. Does selegiline delay progression of Parkinson's disease? A critical re-evaluation of the DATATOP study. J Neurol Neurosurg Psychiatry 1994; 57: 217-20
    • (1994) J Neurol Neurosurg Psychiatry , vol.57 , pp. 217-220
    • Ward, C.1
  • 32
    • 0029690504 scopus 로고    scopus 로고
    • Controversies in the therapy of Parkinson's disease
    • Fahn S. Controversies in the therapy of Parkinson's disease. Adv Neurol 1996; 69: 477-86
    • (1996) Adv Neurol , vol.69 , pp. 477-486
    • Fahn, S.1
  • 33
    • 0031708168 scopus 로고    scopus 로고
    • Neostriatal mechanisms in Parkinson's disease
    • Chase TN, Oh JD, Blanchet PJ. Neostriatal mechanisms in Parkinson's disease. Neurology 1998; 51 (2 Suppl. 2): S30-5
    • (1998) Neurology , vol.51 , Issue.2 SUPPL. 2
    • Chase, T.N.1    Oh, J.D.2    Blanchet, P.J.3
  • 34
    • 0029689447 scopus 로고    scopus 로고
    • Contribution of dopaminergic and glutamatergic mechanisms to the pathogenesis of motor response complications in Parkinson's disease
    • Chase TN, Engber TM, Mouradian MM. Contribution of dopaminergic and glutamatergic mechanisms to the pathogenesis of motor response complications in Parkinson's disease. Adv Neurol 1996; 69: 497-501
    • (1996) Adv Neurol , vol.69 , pp. 497-501
    • Chase, T.N.1    Engber, T.M.2    Mouradian, M.M.3
  • 35
    • 0346218258 scopus 로고    scopus 로고
    • Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson's disease
    • Calon F, Rajput AH, Hornykiewicz O, et al. Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson's disease. Neurobiol Dis 2003; 14 (3): 404-16
    • (2003) Neurobiol Dis , vol.14 , Issue.3 , pp. 404-416
    • Calon, F.1    Rajput, A.H.2    Hornykiewicz, O.3
  • 36
    • 1542360582 scopus 로고    scopus 로고
    • Effect of a selective glutamate antagonist on L-dopa-induced dyskinesias in drug-naive parkinsonian monkeys
    • Hadj Tahar A, Gregoire L, Darre A, et al. Effect of a selective glutamate antagonist on L-dopa-induced dyskinesias in drug-naive parkinsonian monkeys. Neurobiol Dis 2004; 15 (2): 171-6
    • (2004) Neurobiol Dis , vol.15 , Issue.2 , pp. 171-176
    • Hadj Tahar, A.1    Gregoire, L.2    Darre, A.3
  • 37
    • 18744437329 scopus 로고    scopus 로고
    • The role of glutamatergic transmission in the pathogenesis of levodopa-induced dyskinesias: Potential therapeutic approaches
    • Merims D, Ziv I, Sherki Y, et al. The role of glutamatergic transmission in the pathogenesis of levodopa-induced dyskinesias: potential therapeutic approaches. Neurol Neurochir Pol 2001; 35 Suppl. 3: 65-8
    • (2001) Neurol Neurochir Pol , vol.35 , Issue.3 SUPPL. , pp. 65-68
    • Merims, D.1    Ziv, I.2    Sherki, Y.3
  • 38
    • 0033811715 scopus 로고    scopus 로고
    • Dyskinesia in Parkinson's disease: Pathophysiology and clinical risk factors
    • Baas H. Dyskinesia in Parkinson's disease: pathophysiology and clinical risk factors. J Neurol 2000; 247 Suppl. 4: IV/12-6
    • (2000) J Neurol , vol.247 , Issue.4 SUPPL.
    • Baas, H.1
  • 39
    • 0004521596 scopus 로고    scopus 로고
    • Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: Effect of dopaminergic denervation and L-DOPA administration
    • Oh JD, Russell DS, Vaughan CL, et al. Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and L-DOPA administration. Brain Res 1998; 813 (1): 150-9
    • (1998) Brain Res , vol.813 , Issue.1 , pp. 150-159
    • Oh, J.D.1    Russell, D.S.2    Vaughan, C.L.3
  • 40
    • 0033550940 scopus 로고    scopus 로고
    • Effect of dopamine denervation and dopamine agonist administration on serine phosphorylation of striatal NMDA receptor subunits
    • Oh JD, Vaughan CL, Chase TN. Effect of dopamine denervation and dopamine agonist administration on serine phosphorylation of striatal NMDA receptor subunits. Brain Res 1999; 821 (2): 433-42
    • (1999) Brain Res , vol.821 , Issue.2 , pp. 433-442
    • Oh, J.D.1    Vaughan, C.L.2    Chase, T.N.3
  • 41
    • 0031954732 scopus 로고    scopus 로고
    • Levodopa therapy: Consequences of the nonphysiologic replacement of dopamine
    • Chase TN. Levodopa therapy: consequences of the nonphysiologic replacement of dopamine. Neurology 1998; 50 (5 Suppl. 5): S17-25
    • (1998) Neurology , vol.50 , Issue.5 SUPPL. 5
    • Chase, T.N.1
  • 42
    • 0031689728 scopus 로고    scopus 로고
    • Amantadine reduces levodopa-induced dyskinesias in parkinsonian monkeys
    • Blanchet PJ, Konitsiotis S, Chase TN. Amantadine reduces levodopa-induced dyskinesias in parkinsonian monkeys. Mov Disord 1998; 13 (5): 798-802
    • (1998) Mov Disord , vol.13 , Issue.5 , pp. 798-802
    • Blanchet, P.J.1    Konitsiotis, S.2    Chase, T.N.3
  • 43
    • 0029665122 scopus 로고    scopus 로고
    • Levodopa-induced dyskinesias improved by a glutamate antagonist in Parkinsonian monkeys
    • Papa SM, Chase TN. Levodopa-induced dyskinesias improved by a glutamate antagonist in Parkinsonian monkeys. Ann Neurol 1996; 39 (5): 574-8
    • (1996) Ann Neurol , vol.39 , Issue.5 , pp. 574-578
    • Papa, S.M.1    Chase, T.N.2
  • 44
    • 0006647256 scopus 로고    scopus 로고
    • Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson's disease
    • Verhagen Metman L, Del Dotto P, van den Munckhof P, et al. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson's disease. Neurology 1998; 50 (5): 1323-6
    • (1998) Neurology , vol.50 , Issue.5 , pp. 1323-1326
    • Verhagen Metman, L.1    Del Dotto, P.2    Van Den Munckhof, P.3
  • 45
    • 0030990693 scopus 로고    scopus 로고
    • Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian agents: Preclinical studies
    • Danysz W, Parsons CG, Kornhuber J, et al. Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian agents: preclinical studies. Neurosci Biobehav Rev 1997; 21 (4): 455-68
    • (1997) Neurosci Biobehav Rev , vol.21 , Issue.4 , pp. 455-468
    • Danysz, W.1    Parsons, C.G.2    Kornhuber, J.3
  • 46
    • 0034090891 scopus 로고    scopus 로고
    • AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys
    • Konitsiotis S, Blanchet PJ, Verhagen L, et al. AMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeys. Neurology 2000; 54 (8): 1589-95
    • (2000) Neurology , vol.54 , Issue.8 , pp. 1589-1595
    • Konitsiotis, S.1    Blanchet, P.J.2    Verhagen, L.3
  • 47
    • 1842333252 scopus 로고    scopus 로고
    • Differential interaction of competitive NMDA and AMPA antagonists with selective dopamine D-1 and D-2 agonists in a rat model of Parkinson's disease
    • Löschmann PA, Wullner U, Heneka MT, et al. Differential interaction of competitive NMDA and AMPA antagonists with selective dopamine D-1 and D-2 agonists in a rat model of Parkinson's disease. Synapse 1997; 26 (4): 381-91
    • (1997) Synapse , vol.26 , Issue.4 , pp. 381-391
    • Löschmann, P.A.1    Wullner, U.2    Heneka, M.T.3
  • 48
    • 0028971445 scopus 로고
    • Effect of coadministration of glutamate receptor antagonists and dopaminergic agonists on locomotion in monoamine-depleted rats
    • Gossel M, Schmidt WJ, Loscher W, et al. Effect of coadministration of glutamate receptor antagonists and dopaminergic agonists on locomotion in monoamine-depleted rats. J Neural Transm Park Dis Dement Sect 1995; 10 (1): 27-39
    • (1995) J Neural Transm Park Dis Dement Sect , vol.10 , Issue.1 , pp. 27-39
    • Gossel, M.1    Schmidt, W.J.2    Loscher, W.3
  • 49
    • 0025992357 scopus 로고
    • Synergism of the AMPA-antagonist NBQX and the NMDA-antagonist CPP with L-dopa in models of Parkinson's disease
    • Löschmann PA, Lange KW, Kunow M, et al. Synergism of the AMPA-antagonist NBQX and the NMDA-antagonist CPP with L-dopa in models of Parkinson's disease. J Neural Transm Park Dis Dement Sect 1991; 3 (3): 203-13
    • (1991) J Neural Transm Park Dis Dement Sect , vol.3 , Issue.3 , pp. 203-213
    • Löschmann, P.A.1    Lange, K.W.2    Kunow, M.3
  • 50
    • 0026668693 scopus 로고
    • NBQX (6-nitrosulfamoyl-benzo-quinoxaline-dione) and CPP (3-carboxy-piperazin-propyl phosphonic acid) potentiate dopamine agonist induced rotations in substantia nigra lesioned rats
    • Wachtel H, Kunow M, Loschmann PA. NBQX (6-nitrosulfamoyl-benzo- quinoxaline-dione) and CPP (3-carboxy-piperazin-propyl phosphonic acid) potentiate dopamine agonist induced rotations in substantia nigra lesioned rats. Neurosci Lett 1992; 142 (2): 179-82
    • (1992) Neurosci Lett , vol.142 , Issue.2 , pp. 179-182
    • Wachtel, H.1    Kunow, M.2    Loschmann, P.A.3
  • 51
    • 0034659603 scopus 로고    scopus 로고
    • Non-NMDA receptor-mediated mechanisms are involved in levodopa-induced motor response alterations in Parkinsonian rats
    • Marin C, Jimenez A, Bonastre M, et al. Non-NMDA receptor-mediated mechanisms are involved in levodopa-induced motor response alterations in Parkinsonian rats. Synapse 2000; 36 (4): 267-74
    • (2000) Synapse , vol.36 , Issue.4 , pp. 267-274
    • Marin, C.1    Jimenez, A.2    Bonastre, M.3
  • 52
    • 0034860914 scopus 로고    scopus 로고
    • LY293558, an AMPA glutamate receptor antagonist, prevents and reverses levodopa-induced motor alterations in Parkinsonian rats
    • Marin C, Jimenez A, Bonastre M, et al. LY293558, an AMPA glutamate receptor antagonist, prevents and reverses levodopa-induced motor alterations in Parkinsonian rats. Synapse 2001; 42 (1): 40-7
    • (2001) Synapse , vol.42 , Issue.1 , pp. 40-47
    • Marin, C.1    Jimenez, A.2    Bonastre, M.3
  • 53
    • 0025342419 scopus 로고
    • Excitatory amino acid receptors in the brain: Membrane binding and receptor autoradiographic approaches
    • Young AB, Fagg GE. Excitatory amino acid receptors in the brain: membrane binding and receptor autoradiographic approaches. Trends Pharmacol Sci 1990; 11 (3): 126-33
    • (1990) Trends Pharmacol Sci , vol.11 , Issue.3 , pp. 126-133
    • Young, A.B.1    Fagg, G.E.2
  • 54
    • 0026527118 scopus 로고
    • Excitatory amino acid binding sites in the basal ganglia of the rat: A quantitative autoradiographic study
    • Albin RL, Makowiec RL, Hollingsworth ZR, et al. Excitatory amino acid binding sites in the basal ganglia of the rat: a quantitative autoradiographic study. Neuroscience 1992; 46 (1): 35-48
    • (1992) Neuroscience , vol.46 , Issue.1 , pp. 35-48
    • Albin, R.L.1    Makowiec, R.L.2    Hollingsworth, Z.R.3
  • 55
    • 0031012008 scopus 로고    scopus 로고
    • Cellular, subcellular, and subsynaptic distribution of AMPA-type glutamate receptor subunits in the neostriatum of the rat
    • Bernard V, Somogyi P, Bolam JP. Cellular, subcellular, and subsynaptic distribution of AMPA-type glutamate receptor subunits in the neostriatum of the rat. J Neurosci 1997; 17 (2): 819-33
    • (1997) J Neurosci , vol.17 , Issue.2 , pp. 819-833
    • Bernard, V.1    Somogyi, P.2    Bolam, J.P.3
  • 56
    • 0031912995 scopus 로고    scopus 로고
    • Glutamate receptors in the mammalian central nervous system
    • Ozawa S, Kamiya H, Tsuzuki K. Glutamate receptors in the mammalian central nervous system. Prog Neurobiol 1998; 54 (5): 581-618
    • (1998) Prog Neurobiol , vol.54 , Issue.5 , pp. 581-618
    • Ozawa, S.1    Kamiya, H.2    Tsuzuki, K.3
  • 57
    • 0033564041 scopus 로고    scopus 로고
    • Characterization of phosphorylation sites on the glutamate receptor 4 subunit of the AMPA receptors
    • Carvalho AL, Kameyama K, Huganir RL. Characterization of phosphorylation sites on the glutamate receptor 4 subunit of the AMPA receptors. J Neurosci 1999; 19 (12): 4748-54
    • (1999) J Neurosci , vol.19 , Issue.12 , pp. 4748-4754
    • Carvalho, A.L.1    Kameyama, K.2    Huganir, R.L.3
  • 58
    • 0032588030 scopus 로고    scopus 로고
    • Ca2+/calmodulin-kinase II enhances channel conductance of alpha-amino-3-hydrox-y-5-methyl-4-isoxazolepropionate type glutamate receptors
    • U S A
    • Derkach V, Barria A, Soderling TR. Ca2+/calmodulin-kinase II enhances channel conductance of alpha-amino-3-hydrox-y-5-methyl-4-isoxazolepropionate type glutamate receptors. Proc Natl Acad Sci U S A 1999; 96 (6): 3269-74
    • (1999) Proc Natl Acad Sci , vol.96 , Issue.6 , pp. 3269-3274
    • Derkach, V.1    Barria, A.2    Soderling, T.R.3
  • 59
    • 0025117827 scopus 로고
    • 2,3-Dihydrox-y-6-nitro-7-sulfamoyl-benzo (F)quinoxaline: A neuroprotectant for cerebral ischemia
    • Sheardown MJ, Nielsen EO, Hansen AJ, et al. 2,3-Dihydrox-y-6-nitro-7- sulfamoyl-benzo (F)quinoxaline: a neuroprotectant for cerebral ischemia. Science 1990; 247 (4942): 571-4
    • (1990) Science , vol.247 , Issue.4942 , pp. 571-574
    • Sheardown, M.J.1    Nielsen, E.O.2    Hansen, A.J.3
  • 60
    • 0344178117 scopus 로고    scopus 로고
    • Synergistic interactions between ampakines and antipsychotic drugs
    • Johnson SA, Luu NT, Herbst TA, et al. Synergistic interactions between ampakines and antipsychotic drugs. J Pharmacol Exp Ther 1999; 289 (1): 392-7
    • (1999) J Pharmacol Exp Ther , vol.289 , Issue.1 , pp. 392-397
    • Johnson, S.A.1    Luu, N.T.2    Herbst, T.A.3
  • 61
    • 0030483662 scopus 로고    scopus 로고
    • Stereoselective effects-of 2,3-benzodiazepines in vivo: Electrophysiology and neuroprotection studies
    • Lodge D, Bond A, O'Neill MJ, et al. Stereoselective effects-of 2,3-benzodiazepines in vivo: electrophysiology and neuroprotection studies. Neuropharmacology 1996; 35 (12): 1681-8
    • (1996) Neuropharmacology , vol.35 , Issue.12 , pp. 1681-1688
    • Lodge, D.1    Bond, A.2    O'Neill, M.J.3
  • 62
    • 0030483692 scopus 로고    scopus 로고
    • Activity of 2,3-benzodiazepines at native rat and recombinant human glutamate receptors in vitro: Stereospecificity and selectivity profiles
    • Bleakman D, Ballyk BA, Schoepp DD, et al. Activity of 2,3-benzodiazepines at native rat and recombinant human glutamate receptors in vitro: stereospecificity and selectivity profiles. Neuropharmacology 1996; 35 (12): 1689-702
    • (1996) Neuropharmacology , vol.35 , Issue.12 , pp. 1689-1702
    • Bleakman, D.1    Ballyk, B.A.2    Schoepp, D.D.3
  • 63
    • 7244224929 scopus 로고    scopus 로고
    • Effect of monoamine reuptake inhibitor NS 2330 in advanced Parkinson's Disease
    • Bara-Jiminez W, Dimitrova T, Sherzai A, et al. Effect of monoamine reuptake inhibitor NS 2330 in advanced Parkinson's Disease. Mv Disord 2004; 19 (10): 1183-6
    • (2004) Mv Disord , vol.19 , Issue.10 , pp. 1183-1186
    • Bara-Jiminez, W.1    Dimitrova, T.2    Sherzai, A.3
  • 64
    • 0009916844 scopus 로고    scopus 로고
    • NS2330 enhances cognitive function in normal volunteers and volunteers with possible Alzheimer's disease
    • Wesnes K, Preskorn S, Friesen S, et al. NS2330 enhances cognitive function in normal volunteers and volunteers with possible Alzheimer's disease. Clin Pharmacol Ther 2002; 71 (2): 7
    • (2002) Clin Pharmacol Ther , vol.71 , Issue.2 , pp. 7
    • Wesnes, K.1    Preskorn, S.2    Friesen, S.3
  • 65
    • 0035185984 scopus 로고    scopus 로고
    • NS-2330 (Neurosearch)
    • Thatte U. NS-2330 (Neurosearch). Curr Opin Investig Drugs 2001; 2 (11): 1592-4
    • (2001) Curr Opin Investig Drugs , vol.2 , Issue.11 , pp. 1592-1594
    • Thatte, U.1
  • 67
    • 0034743523 scopus 로고    scopus 로고
    • Role of coenzyme Q10 in chronic heart failure, angina, and hypertension
    • Tran M, Mitchell T, Kennedy D, et al. Role of coenzyme Q10 in chronic heart failure, angina, and hypertension. Pharmacotherapy 2001; 7: 797-806
    • (2001) Pharmacotherapy , vol.7 , pp. 797-806
    • Tran, M.1    Mitchell, T.2    Kennedy, D.3
  • 68
    • 85009226418 scopus 로고    scopus 로고
    • A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington's disease
    • Huntington Study Group. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington's disease. Neurology 2001; 57 (3): 397-404
    • (2001) Neurology , vol.57 , Issue.3 , pp. 397-404
  • 69
    • 0031895005 scopus 로고    scopus 로고
    • Mechanisms of MPTP toxicity
    • Przedborski S, Jackson-Lewis V. Mechanisms of MPTP toxicity. Mov Disord 1998; 13 Suppl. 1: 35-8
    • (1998) Mov Disord , vol.13 , Issue.1 SUPPL. , pp. 35-38
    • Przedborski, S.1    Jackson-Lewis, V.2
  • 70
    • 0026484964 scopus 로고
    • Platelet mitochondrial dysfunction in Parkinson's disease
    • Krige D, Carroll M, Cooper J, et al. Platelet mitochondrial dysfunction in Parkinson's disease. Ann Neurol 1992; 32: 782-8
    • (1992) Ann Neurol , vol.32 , pp. 782-788
    • Krige, D.1    Carroll, M.2    Cooper, J.3
  • 71
    • 0024848034 scopus 로고
    • Abnormalities of the electron transport chain in idiopathic Parkinson's disease
    • Parker Jr W, Boyson S, Parks J. Abnormalities of the electron transport chain in idiopathic Parkinson's disease. Ann Neurol 1989; 26: 719-23
    • (1989) Ann Neurol , vol.26 , pp. 719-723
    • Parker Jr., W.1    Boyson, S.2    Parks, J.3
  • 72
    • 0034993538 scopus 로고    scopus 로고
    • Mitochondrial dysfunction in neurodegenerative disorders and ageing
    • Turner C, Schapira A. Mitochondrial dysfunction in neurodegenerative disorders and ageing. Adv Exp Med Biol 2001; 487: 229-51
    • (2001) Adv Exp Med Biol , vol.487 , pp. 229-251
    • Turner, C.1    Schapira, A.2
  • 73
    • 0037426566 scopus 로고    scopus 로고
    • 10 supple-mentation provides mild symptomatic benefit in patients with Parkinson's disease
    • 10 supple-mentation provides mild symptomatic benefit in patients with Parkinson's disease. Neurosci Lett 2003; 341: 201-4
    • (2003) Neurosci Lett , vol.341 , pp. 201-204
    • Müller, T.1    Büttner, T.2    Gholipour, A.3
  • 74
    • 0031594295 scopus 로고    scopus 로고
    • 10 attenuates the 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) induced loss of striatal dopamine and dopaminergic axons in aged mice
    • 10 attenuates the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced loss of striatal dopamine and dopaminergic axons in aged mice. Brain Res 1998; 783: 109-14
    • (1998) Brain Res , vol.783 , pp. 109-114
    • Beal, M.F.1    Matthews, R.2    Tieleman, A.3
  • 75
    • 0032555066 scopus 로고    scopus 로고
    • Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects
    • U S A
    • Matthews R, Yang L, Browne S, et al. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci U S A 1998; 95: 8892-7
    • (1998) Proc Natl Acad Sci , vol.95 , pp. 8892-8897
    • Matthews, R.1    Yang, L.2    Browne, S.3
  • 78
    • 0033407094 scopus 로고    scopus 로고
    • Dopaminergic neurons degenerate by apoptosis in Parkinson's disease
    • Hirsch EC, Hunot S, Faucheux B, et al. Dopaminergic neurons degenerate by apoptosis in Parkinson's disease. Mov Disord 1999; 14 (2): 383-5
    • (1999) Mov Disord , vol.14 , Issue.2 , pp. 383-385
    • Hirsch, E.C.1    Hunot, S.2    Faucheux, B.3
  • 79
    • 12944250987 scopus 로고    scopus 로고
    • Caspase-3: A vulnerability factor and final effector in apoptotic death of dopaminergic neurons in Parkinson's disease
    • U S A
    • Hartmann A, Hunot S, Michel PP, et al. Caspase-3: a vulnerability factor and final effector in apoptotic death of dopaminergic neurons in Parkinson's disease. Proc Natl Acad Sci U S A 2000; 97 (6): 2875-80
    • (2000) Proc Natl Acad Sci , vol.97 , Issue.6 , pp. 2875-2880
    • Hartmann, A.1    Hunot, S.2    Michel, P.P.3
  • 80
    • 0032692497 scopus 로고    scopus 로고
    • CEP-1347 (KT7515), an inhibitor of JNK activation, rescues sympathetic neurons and neuronally differentiated PC12 cells from death evoked by three distinct insults
    • Maroney AC, Finn JP, Bozyczko-Coyne D, et al. CEP-1347 (KT7515), an inhibitor of JNK activation, rescues sympathetic neurons and neuronally differentiated PC12 cells from death evoked by three distinct insults. J Neurochem 1999; 73 (5): 1901-12
    • (1999) J Neurochem , vol.73 , Issue.5 , pp. 1901-1912
    • Maroney, A.C.1    Finn, J.P.2    Bozyczko-Coyne, D.3
  • 81
    • 0031044561 scopus 로고    scopus 로고
    • MEKKs, GCKs, MLKs, PAKs, TAKs, and tpls: Upstream regulators of the c-Jun amino-terminal kinases?
    • Fanger GR, Gerwins P, Widmann C, et al. MEKKs, GCKs, MLKs, PAKs, TAKs, and tpls: upstream regulators of the c-Jun amino-terminal kinases? Curr Opin Genet Dev 1997; 7 (1): 67-74
    • (1997) Curr Opin Genet Dev , vol.7 , Issue.1 , pp. 67-74
    • Fanger, G.R.1    Gerwins, P.2    Widmann, C.3
  • 82
    • 0036731701 scopus 로고    scopus 로고
    • Mixed-lineage kinase control of JNK and p38 MAPK pathways
    • Gallo KA, Johnson GL. Mixed-lineage kinase control of JNK and p38 MAPK pathways. Nat Rev Mol Cell Biol 2002; 3 (9): 663-72
    • (2002) Nat Rev Mol Cell Biol , vol.3 , Issue.9 , pp. 663-672
    • Gallo, K.A.1    Johnson, G.L.2
  • 83
    • 0035816685 scopus 로고    scopus 로고
    • Cep-1347 (KT7515), a semisynthetic inhibitor of the mixed lineage kinase family
    • Maroney AC, Finn JP, Connors TJ, et al. Cep-1347 (KT7515), a semisynthetic inhibitor of the mixed lineage kinase family. J Biol Chem 2001; 276 (27): 25302-8
    • (2001) J Biol Chem , vol.276 , Issue.27 , pp. 25302-25308
    • Maroney, A.C.1    Finn, J.P.2    Connors, T.J.3
  • 84
    • 18244379883 scopus 로고    scopus 로고
    • Mixed lineage kinase activity of indolocarbazole analogues
    • Murakata C, Kaneko M, Gessner G, et al. Mixed lineage kinase activity of indolocarbazole analogues. Bioorg Med Chem Lett 2002; 12 (2): 147-50
    • (2002) Bioorg Med Chem Lett , vol.12 , Issue.2 , pp. 147-150
    • Murakata, C.1    Kaneko, M.2    Gessner, G.3
  • 85
    • 0033029823 scopus 로고    scopus 로고
    • CEP-1347/KT-7515, an inhibitor of c-jun N-terminal kinase activation, attenuates the 1-methyl-4-phenyl tetrahydropyridine-mediated loss of nigrostriatal dopaminergic neurons in vivo
    • Saporito MS, Brown EM, Miller MS, et al. CEP-1347/KT-7515, an inhibitor of c-jun N-terminal kinase activation, attenuates the 1-methyl-4-phenyl tetrahydropyridine-mediated loss of nigrostriatal dopaminergic neurons in vivo. J Pharmacol Exp Ther 1999; 288 (2): 421-7
    • (1999) J Pharmacol Exp Ther , vol.288 , Issue.2 , pp. 421-427
    • Saporito, M.S.1    Brown, E.M.2    Miller, M.S.3
  • 86
    • 0033840581 scopus 로고    scopus 로고
    • MPTP activates c-Jun NH (2)-terminal kinase (JNK) and its upstream regulatory kinase MKK4 in nigrostriatal neurons in vivo
    • Saporito MS, Thomas BA, Scott RW. MPTP activates c-Jun NH (2)-terminal kinase (JNK) and its upstream regulatory kinase MKK4 in nigrostriatal neurons in vivo. J Neurochem 2000; 75 (3): 1200-8
    • (2000) J Neurochem , vol.75 , Issue.3 , pp. 1200-1208
    • Saporito, M.S.1    Thomas, B.A.2    Scott, R.W.3
  • 87
    • 0842324952 scopus 로고    scopus 로고
    • The safety and tolerability of a mixed lineage kinase inhibitor (CEP-1347) in PD
    • Parkinson Study Group. The safety and tolerability of a mixed lineage kinase inhibitor (CEP-1347) in PD. Neurology 2004; 62 (2): 330-2
    • (2004) Neurology , vol.62 , Issue.2 , pp. 330-332
  • 89
    • 24944575933 scopus 로고    scopus 로고
    • PRECEPT [online]. Available from URL: http://bidmc.harvard.edu/display. asp?.node_id = 1214. [Accessed 2004 Aug 14]
    • PRECEPT [Online]
  • 90
    • 0034776282 scopus 로고    scopus 로고
    • Long term safety and efficacy of unilateral deep brain stimulation of the thalamus for parkinsonian tremor
    • Lyons KE, Koller WC, Wilkinson SB, et al. Long term safety and efficacy of unilateral deep brain stimulation of the thalamus for parkinsonian tremor. J Neurol Neurosurg Psychiatry 2001; 71 (5): 682-4
    • (2001) J Neurol Neurosurg Psychiatry , vol.71 , Issue.5 , pp. 682-684
    • Lyons, K.E.1    Koller, W.C.2    Wilkinson, S.B.3
  • 91
    • 10744227411 scopus 로고    scopus 로고
    • Two-year follow-up of subthalamic deep brain stimulation in Parkinson's disease
    • Herzog J, Volkmann J, Krack P, et al. Two-year follow-up of subthalamic deep brain stimulation in Parkinson's disease. Mov Disord 2003; 18 (11): 1332-7
    • (2003) Mov Disord , vol.18 , Issue.11 , pp. 1332-1337
    • Herzog, J.1    Volkmann, J.2    Krack, P.3
  • 92
    • 0037064165 scopus 로고    scopus 로고
    • Subthalamic GAD gene therapy in a Parkinson's disease rat model
    • Luo J, Kaplitt M, Fitzsimons H, et al. Subthalamic GAD gene therapy in a Parkinson's disease rat model. Science 2002; 298 (5592): 425-9
    • (2002) Science , vol.298 , Issue.5592 , pp. 425-429
    • Luo, J.1    Kaplitt, M.2    Fitzsimons, H.3
  • 93
    • 0035839562 scopus 로고    scopus 로고
    • Subthalamic GAD gene transfer in Parkinson disease patients who are candidates for deep brain stimulation
    • During M, Kaplitt M, Stern M, et al. Subthalamic GAD gene transfer in Parkinson disease patients who are candidates for deep brain stimulation. Hum Gene Ther 2001; 12 (12): 1589-91
    • (2001) Hum Gene Ther , vol.12 , Issue.12 , pp. 1589-1591
    • During, M.1    Kaplitt, M.2    Stern, M.3
  • 94
    • 24944527294 scopus 로고    scopus 로고
    • Gene therapy used to treat patients with Parkinson's
    • Aug 19
    • Grady D, Kolata G. Gene therapy used to treat patients with Parkinson's. New York Times 2003 Aug 19
    • (2003) New York Times
    • Grady, D.1    Kolata, G.2
  • 95
    • 0037378036 scopus 로고    scopus 로고
    • In vivo gene delivery of glial cell line-derived neurotrophic factor for Parkinson's disease
    • Kordower J. In vivo gene delivery of glial cell line-derived neurotrophic factor for Parkinson's disease. Ann Neurol 2003; 53 Suppl. 3: S120-34
    • (2003) Ann Neurol , vol.53 , Issue.3 SUPPL.
    • Kordower, J.1
  • 96
    • 0034672304 scopus 로고    scopus 로고
    • Towards a neuroprotective gene therapy for Parkinson's disease: Use of adenovirus, AAV and lentivirus vectors for gene transfer of GDNF to the nigrostriatal system in the rat Parkinson model
    • Björklund A, Kirik D, Rosenblad C, et al. Towards a neuroprotective gene therapy for Parkinson's disease: use of adenovirus, AAV and lentivirus vectors for gene transfer of GDNF to the nigrostriatal system in the rat Parkinson model. Brain Res 2000; 886: 82-98
    • (2000) Brain Res , vol.886 , pp. 82-98
    • Björklund, A.1    Kirik, D.2    Rosenblad, C.3
  • 97
    • 0034721690 scopus 로고    scopus 로고
    • Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease
    • Kordower JH, Emborg ME, Bloch J, et al. Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease. Science 2000; 290: 767-73
    • (2000) Science , vol.290 , pp. 767-773
    • Kordower, J.H.1    Emborg, M.E.2    Bloch, J.3
  • 98
    • 24944568064 scopus 로고    scopus 로고
    • Business wire. Update of neurologix phase I clinical trial of gene therapy for Parkinson's disease
    • Apr 16-21; New Orleans (LA)
    • Business wire. Update of neurologix phase I clinical trial of gene therapy for Parkinson's disease. Presented at AANS annual meeting; 2005 Apr 16-21; New Orleans (LA)
    • (2005) AANS Annual Meeting
  • 99
    • 0037344441 scopus 로고    scopus 로고
    • GAD Zooks! Excitement to inhibition in one easy step?
    • Federoff H. GAD Zooks! Excitement to inhibition in one easy step? Gene Ther 2003; 10: 365-6
    • (2003) Gene Ther , vol.10 , pp. 365-366
    • Federoff, H.1
  • 100
    • 0034750770 scopus 로고    scopus 로고
    • Sustained delivery of GDNF: Towards a treatment for Parkinson's disease
    • Zurn A, Widmer H, Aebischer P. Sustained delivery of GDNF: towards a treatment for Parkinson's disease. Brain Res Rev 2001; 36: 222-9
    • (2001) Brain Res Rev , vol.36 , pp. 222-229
    • Zurn, A.1    Widmer, H.2    Aebischer, P.3
  • 101
    • 0031104816 scopus 로고    scopus 로고
    • Glial cell line-derived neurotrophic factor: A novel therapeutic approach to treat motor dysfunction in Parkinson's disease
    • Lapchak P, Gash D, Jiao S, et al. Glial cell line-derived neurotrophic factor: a novel therapeutic approach to treat motor dysfunction in Parkinson's disease. Exp Neurol 1997; 144: 29-34
    • (1997) Exp Neurol , vol.144 , pp. 29-34
    • Lapchak, P.1    Gash, D.2    Jiao, S.3
  • 102
    • 0031678831 scopus 로고    scopus 로고
    • Glial cell line-derived neurotrophic factor (GDNF): A drug candidate for the treatment of Parkinson's disease
    • Grondin R, Gash D. Glial cell line-derived neurotrophic factor (GDNF): a drug candidate for the treatment of Parkinson's disease. J Neurol 1998; 245 Suppl. 3: P35-42
    • (1998) J Neurol , vol.245 , Issue.3 SUPPL.
    • Grondin, R.1    Gash, D.2
  • 103
    • 0030031116 scopus 로고    scopus 로고
    • Glial cell line-derived neurotrophic factor exerts neurotrophic effects on dopaminergic neurons in vitro and promotes their survival and regrowth after damage by 1-methyl-4-phenylpyridinium
    • Hou J, Lin L, Mytilinenou C. Glial cell line-derived neurotrophic factor exerts neurotrophic effects on dopaminergic neurons in vitro and promotes their survival and regrowth after damage by 1-methyl-4-phenylpyridinium. J Neurochem 1996; 66: 74-82
    • (1996) J Neurochem , vol.66 , pp. 74-82
    • Hou, J.1    Lin, L.2    Mytilinenou, C.3
  • 104
    • 0028898505 scopus 로고
    • Glial cell line-derived neurotrophic factor augments midbrain dopaminergic circuits in vivo
    • Hudson J, Granholm A-C, Gerhardt G, et al. Glial cell line-derived neurotrophic factor augments midbrain dopaminergic circuits in vivo. Brain Res Bull 1995; 36: 425-32
    • (1995) Brain Res Bull , vol.36 , pp. 425-432
    • Hudson, J.1    Granholm, A.-C.2    Gerhardt, G.3
  • 105
    • 0028874203 scopus 로고
    • GDNF protects nigral dopamine neurons against 6-hydroxydopamine in vivo
    • Kearns C, Gash DM. GDNF protects nigral dopamine neurons against 6-hydroxydopamine in vivo. Brain Res 1995; 672: 104-11
    • (1995) Brain Res , vol.672 , pp. 104-111
    • Kearns, C.1    Gash, D.M.2
  • 106
    • 0030774588 scopus 로고    scopus 로고
    • GDNF protection against 6-OHDA: Time dependence and requirement for protein syn-thesis
    • Kearns C, Cass WA, Smoot K, et al. GDNF protection against 6-OHDA: time dependence and requirement for protein syn-thesis. J Neurosci 1997; 17: 7111-8
    • (1997) J Neurosci , vol.17 , pp. 7111-7118
    • Kearns, C.1    Cass, W.A.2    Smoot, K.3
  • 107
    • 0028834063 scopus 로고
    • Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo
    • Tomac A, Lindqvist E, Lin L-FH, et al. Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo. Nature 1995; 373: 335-9
    • (1995) Nature , vol.373 , pp. 335-339
    • Tomac, A.1    Lindqvist, E.2    Lin, L.-F.H.3
  • 108
    • 0028109526 scopus 로고
    • Glial cell line-derived neurotrophic factor reverses toxin-induced injury to midbrain dopaminergic neurons in vivo
    • Hoffer BJ, Hoffman A, Bowenkamp K, et al. Glial cell line-derived neurotrophic factor reverses toxin-induced injury to midbrain dopaminergic neurons in vivo. Neurosci Lett 1994; 182: 107-11
    • (1994) Neurosci Lett , vol.182 , pp. 107-111
    • Hoffer, B.J.1    Hoffman, A.2    Bowenkamp, K.3
  • 109
    • 0030950227 scopus 로고    scopus 로고
    • Glial cell line-derived neurotrophic factor attenuates behavioural deficits and regulates nigrostriatal dopaminergic and peptidergic markers in 6-hydroxydopamine-lesioned adult rats: Comparison of intraventricular and intranigral delivery
    • Lapchak P, Miller P, Collins F, et al. Glial cell line-derived neurotrophic factor attenuates behavioural deficits and regulates nigrostriatal dopaminergic and peptidergic markers in 6-hydroxydopamine-lesioned adult rats: comparison of intraventricular and intranigral delivery. Neuroscience 1997; 78 (1): 61-72
    • (1997) Neuroscience , vol.78 , Issue.1 , pp. 61-72
    • Lapchak, P.1    Miller, P.2    Collins, F.3
  • 110
    • 13344277993 scopus 로고    scopus 로고
    • Functional recovery in parkinsonian monkeys treated with GDNF
    • Gash D, Zhang Z, Ovadia A, et al. Functional recovery in parkinsonian monkeys treated with GDNF. Nature 1996; 380: 252-5
    • (1996) Nature , vol.380 , pp. 252-255
    • Gash, D.1    Zhang, Z.2    Ovadia, A.3
  • 111
    • 0030967428 scopus 로고    scopus 로고
    • Dose response to intraventricular glial cell line-derived neurotrophic factor administration in parkinsonian monkeys
    • Zhang Z, Miyoshi Y, Lapchak P, et al. Dose response to intraventricular glial cell line-derived neurotrophic factor administration in parkinsonian monkeys. J Pharmacol Exp Ther 1997; 282: 1396-401
    • (1997) J Pharmacol Exp Ther , vol.282 , pp. 1396-1401
    • Zhang, Z.1    Miyoshi, Y.2    Lapchak, P.3
  • 112
    • 0030838127 scopus 로고    scopus 로고
    • Glial cell line-derived neurotrophic factor-levodopa interactions and reduction of side effects in parkinsonian monkeys
    • Miyoshi Y, Zhang Z, Ovadia A, et al. Glial cell line-derived neurotrophic factor-levodopa interactions and reduction of side effects in parkinsonian monkeys. Ann Neurol 1997; 42: 208-14
    • (1997) Ann Neurol , vol.42 , pp. 208-214
    • Miyoshi, Y.1    Zhang, Z.2    Ovadia, A.3
  • 113
    • 0036787532 scopus 로고    scopus 로고
    • Chronic, controlled GDNF infusion promotes structural and functional recovery in advanced parkinsonian monkeys
    • Grondin R, Zhang Z, Yi A, et al. Chronic, controlled GDNF infusion promotes structural and functional recovery in advanced parkinsonian monkeys. Brain 2002; 125: 2191-201
    • (2002) Brain , vol.125 , pp. 2191-2201
    • Grondin, R.1    Zhang, Z.2    Yi, A.3
  • 114
    • 0037435511 scopus 로고    scopus 로고
    • Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in Parkinson's disease
    • Nutt J, Burchiel K, Comella C, et al. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in Parkinson's disease. Neurology 2003; 60: 69-73
    • (2003) Neurology , vol.60 , pp. 69-73
    • Nutt, J.1    Burchiel, K.2    Comella, C.3
  • 115
    • 0032850453 scopus 로고    scopus 로고
    • Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson's disease
    • Kordower J, Palfi S, Chen E, et al. Clinicopathological findings following intraventricular glial-derived neurotrophic factor treatment in a patient with Parkinson's disease. Ann Neurol 1999; 47 (3): 419-24
    • (1999) Ann Neurol , vol.47 , Issue.3 , pp. 419-424
    • Kordower, J.1    Palfi, S.2    Chen, E.3
  • 116
    • 0038249170 scopus 로고    scopus 로고
    • Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson's disease
    • Gill S, Patel N, Hotton G, et al. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson's disease. Nat Med 2003; 9 (5): 589-95
    • (2003) Nat Med , vol.9 , Issue.5 , pp. 589-595
    • Gill, S.1    Patel, N.2    Hotton, G.3
  • 117
    • 0030296554 scopus 로고    scopus 로고
    • Immune responses to adenovius vectors in the nervous system
    • Wood MJ, Charlton HM, Wood KJ, et al. Immune responses to adenovius vectors in the nervous system. Trends Neurosci 1996; 19: 497-501
    • (1996) Trends Neurosci , vol.19 , pp. 497-501
    • Wood, M.J.1    Charlton, H.M.2    Wood, K.J.3
  • 118
    • 0030816509 scopus 로고    scopus 로고
    • Immune responses to adenoviral vectors during gene transfer in the brain
    • Kajiwara K, Byrnes AP, Charlton HM, et al. Immune responses to adenoviral vectors during gene transfer in the brain. Hum Gene Ther 1997; 8: 253-65
    • (1997) Hum Gene Ther , vol.8 , pp. 253-265
    • Kajiwara, K.1    Byrnes, A.P.2    Charlton, H.M.3
  • 119
    • 0033978389 scopus 로고    scopus 로고
    • In vivo protection of nigral dopmaine neurons by lentiviral gene transfer of the novel GDNF-family member Neublastin/Artemin
    • Rosenbald C, Gronborg M, Hansen C, et al. In vivo protection of nigral dopmaine neurons by lentiviral gene transfer of the novel GDNF-family member Neublastin/Artemin. Mol Cell Neurosci 2000; 15: 199-214
    • (2000) Mol Cell Neurosci , vol.15 , pp. 199-214
    • Rosenbald, C.1    Gronborg, M.2    Hansen, C.3
  • 120
    • 0032708890 scopus 로고    scopus 로고
    • Recombinant adeno-associated viral vector-mediated glial cell line-derived neurotrophic factor gene transfer protects nigral dopamine neurons after onset of progressive degeneration in a rat model of Parkinson's disease
    • Mandel RJ, Snyder RO, Leff SE. Recombinant adeno-associated viral vector-mediated glial cell line-derived neurotrophic factor gene transfer protects nigral dopamine neurons after onset of progressive degeneration in a rat model of Parkinson's disease. Exp Neurol 1999; 160: 205-14
    • (1999) Exp Neurol , vol.160 , pp. 205-214
    • Mandel, R.J.1    Snyder, R.O.2    Leff, S.E.3
  • 121
    • 0031456051 scopus 로고    scopus 로고
    • Midbrain injection of recombinant adeno-associated virus encoding rat glial cell line-derived neurotrophic factor protects nigral neurons in a progressive 6-hydroxydopamine-induced degeneration model of Parkinson's disease in rats
    • U S A
    • Mandel RJ, Spratt SK, Snyder RO, et al. Midbrain injection of recombinant adeno-associated virus encoding rat glial cell line-derived neurotrophic factor protects nigral neurons in a progressive 6-hydroxydopamine-induced degeneration model of Parkinson's disease in rats. Proc Natl Acad Sci U S A 1997; 94: 14083-8
    • (1997) Proc Natl Acad Sci , vol.94 , pp. 14083-14088
    • Mandel, R.J.1    Spratt, S.K.2    Snyder, R.O.3
  • 122
    • 0034659835 scopus 로고    scopus 로고
    • Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson's model: Intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system
    • Kirik D, Rosenbald C, Björklund A, et al. Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson's model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system. J Neurosci 2000; 20: 4686-700
    • (2000) J Neurosci , vol.20 , pp. 4686-4700
    • Kirik, D.1    Rosenbald, C.2    Björklund, A.3
  • 123
    • 0032538911 scopus 로고    scopus 로고
    • Age-related declines in nigral neuronal function correlate with motor impairments in Rhesus monkeys
    • Emborg M, Ma S, Mufson E, et al. Age-related declines in nigral neuronal function correlate with motor impairments in Rhesus monkeys. J Comp Neurol 1998; 401: 253-63
    • (1998) J Comp Neurol , vol.401 , pp. 253-263
    • Emborg, M.1    Ma, S.2    Mufson, E.3
  • 124
    • 0029764836 scopus 로고    scopus 로고
    • 6-hydroxydopamine induces the loss of the dopaminergic phenotype in substantia nigra neurons of the rat: A possible mechanism for restoration of the nigrostriatal circuit mediated by glial cell line-derived neurotrophic factor
    • Bowenkamp K, David D, Lapchak P, et al. 6-hydroxydopamine induces the loss of the dopaminergic phenotype in substantia nigra neurons of the rat: a possible mechanism for restoration of the nigrostriatal circuit mediated by glial cell line-derived neurotrophic factor. Exp Brain Res 1996; 111: 1-7
    • (1996) Exp Brain Res , vol.111 , pp. 1-7
    • Bowenkamp, K.1    David, D.2    Lapchak, P.3
  • 125
    • 0036434879 scopus 로고    scopus 로고
    • Aberrant sprouting and downregulation of tyrosine hydroxylase in lesioned nigrostriatal dopamine neurons induced by long-lasting overexpression of glial cell line derived neurotrophic factor in the striatum by lentiviral gene transfer
    • Georgievska B, Kirik D, Björklund A. Aberrant sprouting and downregulation of tyrosine hydroxylase in lesioned nigrostriatal dopamine neurons induced by long-lasting overexpression of glial cell line derived neurotrophic factor in the striatum by lentiviral gene transfer. Exp Neurol 2002; 177: 461-74
    • (2002) Exp Neurol , vol.177 , pp. 461-474
    • Georgievska, B.1    Kirik, D.2    Björklund, A.3
  • 126
    • 0033106489 scopus 로고    scopus 로고
    • Encapsulated cells as therapy
    • Lysaght MJ, Aebischer P. Encapsulated cells as therapy. Sci Am 1999; 280: 76-82
    • (1999) Sci Am , vol.280 , pp. 76-82
    • Lysaght, M.J.1    Aebischer, P.2
  • 127
    • 0028917168 scopus 로고
    • Implantation of encapsulated catecholamine and GDNF-producing cells in rats with unilateral dopamine depletions and parkinsonian symptoms
    • Lindner MD, Winn SR, Baetge EE, et al. Implantation of encapsulated catecholamine and GDNF-producing cells in rats with unilateral dopamine depletions and parkinsonian symptoms. Exp Neurol 1995; 132: 62-76
    • (1995) Exp Neurol , vol.132 , pp. 62-76
    • Lindner, M.D.1    Winn, S.R.2    Baetge, E.E.3
  • 128
    • 0031031008 scopus 로고    scopus 로고
    • GDNF reduces drug-induced rotational behavior after medial forebrain bundle transection by a mechanism not involving striatal dopamine
    • Tseng JL, Baetge EE, Zurn AD, et al. GDNF reduces drug-induced rotational behavior after medial forebrain bundle transection by a mechanism not involving striatal dopamine. J Neurosci 1997; 1: 325-33
    • (1997) J Neurosci , vol.1 , pp. 325-333
    • Tseng, J.L.1    Baetge, E.E.2    Zurn, A.D.3
  • 129
    • 0035887595 scopus 로고    scopus 로고
    • Neuroprotection through delivery of glial cell-lined derived neurotrophic factor by neural stem cells in a mouse model of Parkinson's disease
    • Akerud P, Canals JM, Snyder EY, et al. Neuroprotection through delivery of glial cell-lined derived neurotrophic factor by neural stem cells in a mouse model of Parkinson's disease. J Neurosci 2001; 21: 8108-18
    • (2001) J Neurosci , vol.21 , pp. 8108-8118
    • Akerud, P.1    Canals, J.M.2    Snyder, E.Y.3
  • 130
    • 0035826089 scopus 로고    scopus 로고
    • Transplantation of embryon-ic dopamine neurons for severe Parkinson's disease
    • Freed C, Greene P, Breeze R, et al. Transplantation of embryon-ic dopamine neurons for severe Parkinson's disease. N Eng J Med 2001; 344 (10): 710-9
    • (2001) N Eng J Med , vol.344 , Issue.10 , pp. 710-719
    • Freed, C.1    Greene, P.2    Breeze, R.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.