메뉴 건너뛰기




Volumn 11, Issue 1, 2014, Pages 92-110

Animal Models of Parkinson's Disease: A Gateway to Therapeutics?

Author keywords

animal models; motor symptoms; neuroprotection; neurotoxins; non motor symptoms; Parkinson's disease; therapeutics; transgenic

Indexed keywords

1,2,3,6 TETRAHYDRO 1 METHYL 4 PHENYLPYRIDINE; ADENOSINE A2 RECEPTOR ANTAGONIST; ADRENERGIC RECEPTOR BLOCKING AGENT; ALPHA SYNUCLEIN; AMPA RECEPTOR ANTAGONIST; BAICALEIN; BENSERAZIDE; DJ 1 PROTEIN; GLUTATHIONE; IDEBENONE; IMMUNOGLOBULIN ENHANCER BINDING PROTEIN; LEUCINE RICH REPEAT KINASE 2; LEVODOPA; METABOTROPIC RECEPTOR; N METHYL DEXTRO ASPARTIC ACID RECEPTOR BLOCKING AGENT; NITRIC OXIDE SYNTHASE INHIBITOR; OXIDOPAMINE; PARAQUAT; PARKIN; PROTEASOME; PROTEIN; PROTEIN PINK1; RASAGILINE; RESVERATROL; ROTENONE; SEROTONIN AGONIST; TOXIN; TRANSCRIPTION FACTOR PITX3; UNCLASSIFIED DRUG; VESICULAR MONOAMINE TRANSPORTER 2;

EID: 84893043031     PISSN: 19337213     EISSN: 18787479     Source Type: Journal    
DOI: 10.1007/s13311-013-0234-1     Document Type: Review
Times cited : (82)

References (185)
  • 3
    • 41549114279 scopus 로고    scopus 로고
    • The role of autophagy-lysosome pathway in neurodegeneration associated with Parkinson's disease
    • Pan T, Kondo S, Le W, Jankovic J. The role of autophagy-lysosome pathway in neurodegeneration associated with Parkinson's disease. Brain 2008;131: 1969-1978.
    • (2008) Brain , vol.131 , pp. 1969-1978
    • Pan, T.1    Kondo, S.2    Le, W.3    Jankovic, J.4
  • 4
    • 84866610524 scopus 로고    scopus 로고
    • Lysosome-dependent pathways as a unifying theme in Parkinson's disease
    • Tofaris GK. Lysosome-dependent pathways as a unifying theme in Parkinson's disease. Mov Disord 2012;27: 1364-1369.
    • (2012) Mov Disord , vol.27 , pp. 1364-1369
    • Tofaris, G.K.1
  • 7
    • 84877923497 scopus 로고    scopus 로고
    • The VPS35 gene and Parkinson's disease
    • Deng H, Gao K, Jankovic J. The VPS35 gene and Parkinson's disease. Mov Disord 2013;28: 569-575.
    • (2013) Mov Disord , vol.28 , pp. 569-575
    • Deng, H.1    Gao, K.2    Jankovic, J.3
  • 8
    • 84872590599 scopus 로고    scopus 로고
    • F-box only protein 7 gene in parkinsonian-pyramidal disease
    • Deng H, Liang H, Jankovic J. F-box only protein 7 gene in parkinsonian-pyramidal disease. JAMA Neurol 2013;70: 20-24.
    • (2013) JAMA Neurol , vol.70 , pp. 20-24
    • Deng, H.1    Liang, H.2    Jankovic, J.3
  • 9
    • 59149101888 scopus 로고    scopus 로고
    • Etiopathogenesis of Parkinson's disease: A New Beginning?
    • Le W, Chen S, Jankovic J. Etiopathogenesis of Parkinson's disease: A New Beginning? Neuroscientist 2009;15: 28-35.
    • (2009) Neuroscientist , vol.15 , pp. 28-35
    • Le, W.1    Chen, S.2    Jankovic, J.3
  • 10
    • 84863728773 scopus 로고    scopus 로고
    • Therapies in Parkinson's disease
    • Jankovic J, Poewe W. Therapies in Parkinson's disease. Curr Opin Neurol 2012;25: 433-447.
    • (2012) Curr Opin Neurol , vol.25 , pp. 433-447
    • Jankovic, J.1    Poewe, W.2
  • 12
    • 84859108554 scopus 로고    scopus 로고
    • Animal models of Parkinson's disease
    • Blandini F and Armentero MT. Animal models of Parkinson's disease. FEBS J 2012; 279: 1156-1166.
    • (2012) FEBS J , vol.279 , pp. 1156-1166
    • Blandini, F.1    Armentero, M.T.2
  • 13
    • 84893105563 scopus 로고    scopus 로고
    • Animal models of Parkinson's disease
    • In: Jankovic J (ed.), Clinical Insights E-Book Series, Future Science Group, London, UK
    • Le W, Jankovic J. Animal models of Parkinson's disease. In: Jankovic J (ed.) Parkinson's disease: diagnosis, motor symptoms and non-motor features. Clinical Insights E-Book Series, Future Science Group, London, UK, www. futuremedicine. com, 2013, pp. 115-136.
    • (2013) Parkinson's disease: Diagnosis, motor symptoms and non-motor features , pp. 115-136
    • Le, W.1    Jankovic, J.2
  • 14
    • 84859699540 scopus 로고    scopus 로고
    • Animal models of the non-motor features of Parkinson's disease
    • McDowell K, Chesselet MF. Animal models of the non-motor features of Parkinson's disease. Neurobiol Dis 2012;46: 597-606.
    • (2012) Neurobiol Dis , vol.46 , pp. 597-606
    • McDowell, K.1    Chesselet, M.F.2
  • 15
    • 84867302438 scopus 로고    scopus 로고
    • A call for transparent reporting to optimize thepredictive value of preclinical research
    • Landis SC, Amara SG, Asadullah K, et al. A call for transparent reporting to optimize thepredictive value of preclinical research. Nature 2012;490: 187-191.
    • (2012) Nature , vol.490 , pp. 187-191
    • Landis, S.C.1    Amara, S.G.2    Asadullah, K.3
  • 16
    • 84876247951 scopus 로고    scopus 로고
    • Should clinicians care about preclinical animal research?
    • Silberberg SD. Should clinicians care about preclinical animal research? Neurology 2013;80: 1072-1073.
    • (2013) Neurology , vol.80 , pp. 1072-1073
    • Silberberg, S.D.1
  • 17
    • 0014376793 scopus 로고
    • 6-hydroxy-dopamine induced degeneration of central monoamine neurons
    • Ungerstedt U. 6-hydroxy-dopamine induced degeneration of central monoamine neurons. Eur J Pharmacol 1968;5: 107-110.
    • (1968) Eur J Pharmacol , vol.5 , pp. 107-110
    • Ungerstedt, U.1
  • 18
    • 0015060694 scopus 로고
    • Depletion of brain noradrenaline and dopamine by 6-hydroxydopamine
    • Breese GR, Traylor TD. Depletion of brain noradrenaline and dopamine by 6-hydroxydopamine. Br J Pharmacol 1971;42: 88-99.
    • (1971) Br J Pharmacol , vol.42 , pp. 88-99
    • Breese, G.R.1    Traylor, T.D.2
  • 19
    • 0017281887 scopus 로고
    • Specificity of dopaminergic neuronal degeneration induced by intracerebral injection of 6-hydroxydopamine in the nigrostriatal dopamine system
    • Javoy F, Sotelo C, Herbet A, Agid Y. Specificity of dopaminergic neuronal degeneration induced by intracerebral injection of 6-hydroxydopamine in the nigrostriatal dopamine system. Brain Res 1976;102: 201-215.
    • (1976) Brain Res , vol.102 , pp. 201-215
    • Javoy, F.1    Sotelo, C.2    Herbet, A.3    Agid, Y.4
  • 20
    • 0028220834 scopus 로고
    • Progressive degeneration of nigrostriatal dopamine neurons following intrastriatal terminal lesions with 6-hydroxydopamine: a combined retrograde tracing and immunocytochemical study in the rat
    • Sauer H, Oertel WH. Progressive degeneration of nigrostriatal dopamine neurons following intrastriatal terminal lesions with 6-hydroxydopamine: a combined retrograde tracing and immunocytochemical study in the rat. Neuroscience 1994;59: 401-415.
    • (1994) Neuroscience , vol.59 , pp. 401-415
    • Sauer, H.1    Oertel, W.H.2
  • 21
    • 84863188413 scopus 로고    scopus 로고
    • Alterations inaxonal transport motor proteins in sporadic and experimental Parkinson's disease
    • Chu Y, Morfini GA, Langhamer LB, He Y, Brady ST, Kordower JH. Alterations inaxonal transport motor proteins in sporadic and experimental Parkinson's disease. Brain 2012;135: 2058-2073.
    • (2012) Brain , vol.135 , pp. 2058-2073
    • Chu, Y.1    Morfini, G.A.2    Langhamer, L.B.3    He, Y.4    Brady, S.T.5    Kordower, J.H.6
  • 22
    • 0030175258 scopus 로고    scopus 로고
    • Unilateral 6-hydroxydopamine lesions of meso-striatal dopamine neurons and their physiological sequelae
    • Schwarting RKW, Huston P. Unilateral 6-hydroxydopamine lesions of meso-striatal dopamine neurons and their physiological sequelae. Prog Neurobiol 1996;49: 215-266.
    • (1996) Prog Neurobiol , vol.49 , pp. 215-266
    • Schwarting, R.K.W.1    Huston, P.2
  • 23
    • 0020680904 scopus 로고
    • Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis
    • Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science 1983;219: 979-980.
    • (1983) Science , vol.219 , pp. 979-980
    • Langston, J.W.1    Ballard, P.2    Tetrud, J.W.3    Irwin, I.4
  • 24
    • 0000340348 scopus 로고
    • Parkinsonism-inducing neurotoxin, N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine:Uptake of the metabolite N-methyl-4-phenylpyridine by dopamine neurons explains selective toxicity
    • Javitch JA, D'Amato RJ, Strittmatter SM, Snyder SH. Parkinsonism-inducing neurotoxin, N-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine: Uptake of the metabolite N-methyl-4-phenylpyridine by dopamine neurons explains selective toxicity. Proc Natl Acad Sci U S A 1985;82: 2173-2177.
    • (1985) Proc Natl Acad Sci U S A , vol.82 , pp. 2173-2177
    • Javitch, J.A.1    D'Amato, R.J.2    Strittmatter, S.M.3    Snyder, S.H.4
  • 25
    • 33751188956 scopus 로고    scopus 로고
    • Dopamine transporter (DAT) inhibitors alleviate specific Parkinsonian deficits in monkeys: association with DAT occupancy in vivo
    • Madras BK, Fahey MA, Goulet M, et al. Dopamine transporter (DAT) inhibitors alleviate specific Parkinsonian deficits in monkeys: association with DAT occupancy in vivo. J Pharmacol Exp Ther 2006;319: 2570-2585.
    • (2006) J Pharmacol Exp Ther , vol.319 , pp. 2570-2585
    • Madras, B.K.1    Fahey, M.A.2    Goulet, M.3
  • 27
    • 0032829070 scopus 로고    scopus 로고
    • Evidence of active nerve cell degeneration in the substantia nigra of humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure
    • Langston JW, Forno LS, Tetrud J, Reeves AG, Kaplan JA, Karluk D. Evidence of active nerve cell degeneration in the substantia nigra of humans years after 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine exposure. Ann Neurol 1999;46: 598-605.
    • (1999) Ann Neurol , vol.46 , pp. 598-605
    • Langston, J.W.1    Forno, L.S.2    Tetrud, J.3    Reeves, A.G.4    Kaplan, J.A.5    Karluk, D.6
  • 28
    • 0027492176 scopus 로고
    • Age-related effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine treatment of common marmosets
    • Rose S, Nomoto M, Jackson EA, et al. Age-related effects of 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine treatment of common marmosets. Eur J Pharmacol 1993;230: 177-185.
    • (1993) Eur J Pharmacol , vol.230 , pp. 177-185
    • Rose, S.1    Nomoto, M.2    Jackson, E.A.3
  • 30
    • 79961228775 scopus 로고    scopus 로고
    • REM sleep behavior disorder in the marmoset MPTP model of early Parkinson disease
    • Verhave PS, Jongsma MJ, van den Berg RM, et al. REM sleep behavior disorder in the marmoset MPTP model of early Parkinson disease. Sleep 2011;34: 1119-1125.
    • (2011) Sleep , vol.34 , pp. 1119-1125
    • Verhave, P.S.1    Jongsma, M.J.2    van den Berg, R.M.3
  • 31
    • 67650215631 scopus 로고    scopus 로고
    • Third-generation antiepileptic drugs: mechanisms of action, pharmacokinetics and interactions
    • Luszczki JJ. Third-generation antiepileptic drugs: mechanisms of action, pharmacokinetics and interactions. Pharmacol Rep 2009;61: 197-216.
    • (2009) Pharmacol Rep , vol.61 , pp. 197-216
    • Luszczki, J.J.1
  • 32
    • 84875516790 scopus 로고    scopus 로고
    • Safinamide reduces dyskinesias and prolongs l-DOPA antiparkinsonian effect in parkinsonian monkeys
    • Grégoire L, Jourdain VA, Townsend M, Roach A, Di Paolo T. Safinamide reduces dyskinesias and prolongs l-DOPA antiparkinsonian effect in parkinsonian monkeys. Parkinsonism Relat Disord 2013;19: 508-514.
    • (2013) Parkinsonism Relat Disord , vol.19 , pp. 508-514
    • Grégoire, L.1    Jourdain, V.A.2    Townsend, M.3    Roach, A.4    Di Paolo, T.5
  • 33
    • 84855957418 scopus 로고    scopus 로고
    • A randomized, double-blind, placebo-controlled trial of safinamide as add-on therapy in early Parkinson's disease patients
    • Stocchi F, Borgohain R, Onofrj M, et al. A randomized, double-blind, placebo-controlled trial of safinamide as add-on therapy in early Parkinson's disease patients. Mov Disord 2012;27: 106-112.
    • (2012) Mov Disord , vol.27 , pp. 106-112
    • Stocchi, F.1    Borgohain, R.2    Onofrj, M.3
  • 35
    • 33845993229 scopus 로고    scopus 로고
    • Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys
    • Kordower JH, Herzog CD, Dass B, et al. Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys. Ann Neurol 2006;60: 706-715.
    • (2006) Ann Neurol , vol.60 , pp. 706-715
    • Kordower, J.H.1    Herzog, C.D.2    Dass, B.3
  • 36
    • 34447572589 scopus 로고    scopus 로고
    • Striatal delivery of CERE-120, an AAV2 vector encoding human neurturin, enhances activity of the dopaminergic nigrostriatal system in aged monkeys
    • Herzog CD, Dass B, Holden JE, et al. Striatal delivery of CERE-120, an AAV2 vector encoding human neurturin, enhances activity of the dopaminergic nigrostriatal system in aged monkeys. Mov Disord 2007;22: 1124-1132.
    • (2007) Mov Disord , vol.22 , pp. 1124-1132
    • Herzog, C.D.1    Dass, B.2    Holden, J.E.3
  • 37
    • 78349247631 scopus 로고    scopus 로고
    • Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomized, controlled trial
    • Marks WJ Jr, Bartus RT, Siffert J, et al. Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomized, controlled trial. Lancet Neurol 2010;9: 1164-1172.
    • (2010) Lancet Neurol , vol.9 , pp. 1164-1172
    • Marks Jr., W.J.1    Bartus, R.T.2    Siffert, J.3
  • 38
    • 79951476995 scopus 로고    scopus 로고
    • Bioactivity of AAV2-neurturin gene therapy (CERE-120): Differences between Parkinson's disease and nonhuman primate brains
    • Bartus RT, Herzog CD, Chu Y, et al. Bioactivity of AAV2-neurturin gene therapy (CERE-120): Differences between Parkinson's disease and nonhuman primate brains. Mov Disord 2011;26: 27-36.
    • (2011) Mov Disord , vol.26 , pp. 27-36
    • Bartus, R.T.1    Herzog, C.D.2    Chu, Y.3
  • 39
    • 84879086501 scopus 로고    scopus 로고
    • Safety/feasibility of targeting thesubstantia nigra with AAV2-neurturin in Parkinson patients
    • Bartus RT, Baumann TL, Siffert J, et al. Safety/feasibility of targeting thesubstantia nigra with AAV2-neurturin in Parkinson patients. Neurology 2013;80: 1698-1701.
    • (2013) Neurology , vol.80 , pp. 1698-1701
    • Bartus, R.T.1    Baumann, T.L.2    Siffert, J.3
  • 40
  • 42
    • 0035816365 scopus 로고    scopus 로고
    • Carrier-mediated processes in blood-brain barrier penetration and neural uptake of paraquat
    • Shimizu K, Ohtaki K, Matsubara K, et al. Carrier-mediated processes in blood-brain barrier penetration and neural uptake of paraquat. Brain Res 2001;906: 135-142.
    • (2001) Brain Res , vol.906 , pp. 135-142
    • Shimizu, K.1    Ohtaki, K.2    Matsubara, K.3
  • 46
    • 0036075892 scopus 로고    scopus 로고
    • Environmental risk factors and Parkinson's disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat
    • McCormack AL, Thiruchelvam M, Manning-Bog AB, et al. Environmental risk factors and Parkinson's disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol Dis 2002;10: 119-127.
    • (2002) Neurobiol Dis , vol.10 , pp. 119-127
    • McCormack, A.L.1    Thiruchelvam, M.2    Manning-Bog, A.B.3
  • 47
    • 84855515680 scopus 로고    scopus 로고
    • Paraquat neurotoxicity is mediated by the dopamine transporter and organic cation transporter-3
    • Rappold PM, Cui M, Chesser AS, et al. Paraquat neurotoxicity is mediated by the dopamine transporter and organic cation transporter-3. Proc Natl Acad Sci U S A. 2011;108: 20766-20771.
    • (2011) Proc Natl Acad Sci U S A. , vol.108 , pp. 20766-20771
    • Rappold, P.M.1    Cui, M.2    Chesser, A.S.3
  • 48
    • 0037127197 scopus 로고    scopus 로고
    • The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein
    • Manning-Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Di Monte DA. The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein. J Biol Chem 2002;277: 1641-1644.
    • (2002) J Biol Chem , vol.277 , pp. 1641-1644
    • Manning-Bog, A.B.1    McCormack, A.L.2    Li, J.3    Uversky, V.N.4    Fink, A.L.5    Di Monte, D.A.6
  • 49
    • 0030200933 scopus 로고    scopus 로고
    • Disulfiram and diethyldithiocarbamate intoxication affects the storage and release of striatal dopamine
    • Vaccari A, Saba PL, Ruiu S, Collu M, Devoto P. Disulfiram and diethyldithiocarbamate intoxication affects the storage and release of striatal dopamine. Toxicol Appl Pharmacol 1996; 139: 102-108.
    • (1996) Toxicol Appl Pharmacol , vol.139 , pp. 102-108
    • Vaccari, A.1    Saba, P.L.2    Ruiu, S.3    Collu, M.4    Devoto, P.5
  • 50
    • 0042421940 scopus 로고    scopus 로고
    • Age-related irreversible progressive nigrostriatal dopaminergic neurotoxicity in the paraquat and maneb model of the Parkinson's disease phenotype
    • Thiruchelvam M, McCormack A, Richfield EK, et al. Age-related irreversible progressive nigrostriatal dopaminergic neurotoxicity in the paraquat and maneb model of the Parkinson's disease phenotype. Eur J Neurosci 2003;18: 589-600.
    • (2003) Eur J Neurosci , vol.18 , pp. 589-600
    • Thiruchelvam, M.1    McCormack, A.2    Richfield, E.K.3
  • 52
    • 0037114971 scopus 로고    scopus 로고
    • Endoplasmic reticulum stress and the unfolded protein response in cellular models of Parkinson's disease
    • Ryu EJ, Harding HP, Angelastro JM, Vitolo OV, Ron D, Greene LA. Endoplasmic reticulum stress and the unfolded protein response in cellular models of Parkinson's disease. J Neurosci 2002;22: 10690-10698.
    • (2002) J Neurosci , vol.22 , pp. 10690-10698
    • Ryu, E.J.1    Harding, H.P.2    Angelastro, J.M.3    Vitolo, O.V.4    Ron, D.5    Greene, L.A.6
  • 53
    • 16544391570 scopus 로고    scopus 로고
    • The neurobehavioral changes induced by bilateral rotenone lesion in medial forebrain bundle of rats are reversed by L-DOPA
    • Alam M, Mayerhofer A, Schmidt WJ. The neurobehavioral changes induced by bilateral rotenone lesion in medial forebrain bundle of rats are reversed by L-DOPA. Behav Brain Res 2004;151: 117-124.
    • (2004) Behav Brain Res , vol.151 , pp. 117-124
    • Alam, M.1    Mayerhofer, A.2    Schmidt, W.J.3
  • 54
    • 77649284470 scopus 로고    scopus 로고
    • Progression of Parkinson's disease pathology is reproduced by intragastric administration of rotenone in mice
    • Pan-Montojo F, Anichtchik O, Dening Y, et al. Progression of Parkinson's disease pathology is reproduced by intragastric administration of rotenone in mice. PLos One 2010;5: e8762.
    • (2010) PLos One , vol.5
    • Pan-Montojo, F.1    Anichtchik, O.2    Dening, Y.3
  • 55
    • 84862609075 scopus 로고    scopus 로고
    • Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice
    • Luk KC, Kehm VM, Zhang B, O'Brien P, Trojanowski JQ, Lee VM. Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice. J Exp Med 2012;209: 975-986.
    • (2012) J Exp Med , vol.209 , pp. 975-986
    • Luk, K.C.1    Kehm, V.M.2    Zhang, B.3    O'Brien, P.4    Trojanowski, J.Q.5    Lee, V.M.6
  • 56
    • 84875415994 scopus 로고    scopus 로고
    • α-Synuclein oligomers and clinical implications for Parkinson disease
    • Kalia LV, Kalia SK, McLean PJ, Lozano AM, Lang AE. α-Synuclein oligomers and clinical implications for Parkinson disease. Ann Neurol 2013;73: 155-169.
    • (2013) Ann Neurol , vol.73 , pp. 155-169
    • Kalia, L.V.1    Kalia, S.K.2    McLean, P.J.3    Lozano, A.M.4    Lang, A.E.5
  • 57
    • 77349083730 scopus 로고    scopus 로고
    • Non-invasive evaluation of nigrostriatal neuropathology in a proteasome inhibitor rodent model of Parkinson's disease
    • Vernon AC, Johansson SM, Modo MM. Non-invasive evaluation of nigrostriatal neuropathology in a proteasome inhibitor rodent model of Parkinson's disease. BMC Neurosci 2010;11: 1.
    • (2010) BMC Neurosci , vol.11 , pp. 1
    • Vernon, A.C.1    Johansson, S.M.2    Modo, M.M.3
  • 58
    • 77956596883 scopus 로고    scopus 로고
    • Proteasome inhibition modeling nigral neuron degeneration in Parkinson's disease
    • Xie W, Li X, Li C, Zhu W, Jankovic J, Le W. Proteasome inhibition modeling nigral neuron degeneration in Parkinson's disease. J Neurochem 2010;115: 188-199.
    • (2010) J Neurochem , vol.115 , pp. 188-199
    • Xie, W.1    Li, X.2    Li, C.3    Zhu, W.4    Jankovic, J.5    Le, W.6
  • 59
    • 77955346226 scopus 로고    scopus 로고
    • Novel D3 dopamine receptor preferring agonist D-264: Evidence of neuroprotective property in Parkinson's disease animal models induced by MPTP and lactacystin
    • Li C, Biswas S, Li X, Dutta AK, Le W. Novel D3 dopamine receptor preferring agonist D-264: Evidence of neuroprotective property in Parkinson's disease animal models induced by MPTP and lactacystin. J Neurosci Res 2010;15: 2513-2523.
    • (2010) J Neurosci Res , vol.15 , pp. 2513-2523
    • Li, C.1    Biswas, S.2    Li, X.3    Dutta, A.K.4    Le, W.5
  • 60
    • 84867742272 scopus 로고    scopus 로고
    • Anti-parkinsonian effects of Nurr1 activator in ubiquitin-proteasome system impairment induced Animal model of Parkinson's disease
    • Zhang Z, Li X, Xie W, et al. Anti-parkinsonian effects of Nurr1 activator in ubiquitin-proteasome system impairment induced Animal model of Parkinson's disease. CNS Neurol Disord Drug Targets 2012;11: 768-773.
    • (2012) CNS Neurol Disord Drug Targets , vol.11 , pp. 768-773
    • Zhang, Z.1    Li, X.2    Xie, W.3
  • 61
    • 84869041395 scopus 로고    scopus 로고
    • Development and characterization of a new Parkinson's disease model resulting from impaired autophagy
    • Ahmed I, Liang Y, Schools S, et al. Development and characterization of a new Parkinson's disease model resulting from impaired autophagy. J Neurosci 2012;32: 16503-16509.
    • (2012) J Neurosci , vol.32 , pp. 16503-16509
    • Ahmed, I.1    Liang, Y.2    Schools, S.3
  • 62
    • 77953666105 scopus 로고    scopus 로고
    • Genetic animal models of Parkinson's disease
    • Ted MD, Han SK, Valina LD. Genetic animal models of Parkinson's disease. Neuron 2010;66: 646-661.
    • (2010) Neuron , vol.66 , pp. 646-661
    • Ted, M.D.1    Han, S.K.2    Valina, L.D.3
  • 63
    • 33749240943 scopus 로고    scopus 로고
    • Mechanisms of Parkinson's disease linked to pathological alpha-synuclein: new targets for drug discovery
    • Lee VM, Trojanowski JQ. Mechanisms of Parkinson's disease linked to pathological alpha-synuclein: new targets for drug discovery. Neuron 2006;52: 33-38.
    • (2006) Neuron , vol.52 , pp. 33-38
    • Lee, V.M.1    Trojanowski, J.Q.2
  • 64
    • 0034531475 scopus 로고    scopus 로고
    • The α-synucleinopathies: Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy
    • Spillantini MG, Goedert M. The α-synucleinopathies: Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Ann NY Acad Sci 2000;920: 16-27.
    • (2000) Ann NY Acad Sci , vol.920 , pp. 16-27
    • Spillantini, M.G.1    Goedert, M.2
  • 65
    • 0035066885 scopus 로고    scopus 로고
    • Direct binding and functional coupling of α-synuclein to the dopamine transporters accelerate dopamine-induced apoptosis
    • Lee FJS, Liu F, Pristupa ZB, Niznik HB. Direct binding and functional coupling of α-synuclein to the dopamine transporters accelerate dopamine-induced apoptosis. FASEB J 2001;15: 916-926.
    • (2001) FASEB J , vol.15 , pp. 916-926
    • Lee, F.J.S.1    Liu, F.2    Pristupa, Z.B.3    Niznik, H.B.4
  • 66
    • 0034681471 scopus 로고    scopus 로고
    • Dopaminergic loss and inclusion body formation in alpha-synuclein mice: implications for neurodegenerative disorders
    • Masliah E, Rockenstein E, Veinbergs I, et al. Dopaminergic loss and inclusion body formation in alpha-synuclein mice: implications for neurodegenerative disorders. Science 2000;287: 1265-1269.
    • (2000) Science , vol.287 , pp. 1265-1269
    • Masliah, E.1    Rockenstein, E.2    Veinbergs, I.3
  • 67
    • 0037118259 scopus 로고    scopus 로고
    • Neuronal α-synucleinopathy with severe movement disorder in mice expressing A53T human α-synuclein
    • Giasson BI, Duda JE, Quinn SM, Zhang B, Trojanowski JQ, Lee VMY. Neuronal α-synucleinopathy with severe movement disorder in mice expressing A53T human α-synuclein. Neuron 2002;34: 521-533.
    • (2002) Neuron , vol.34 , pp. 521-533
    • Giasson, B.I.1    Duda, J.E.2    Quinn, S.M.3    Zhang, B.4    Trojanowski, J.Q.5    Lee, V.M.Y.6
  • 68
    • 37549030985 scopus 로고    scopus 로고
    • In vivo alpha-synuclein overexpression in rodents: a useful model of Parkinson's disease?
    • Chesselet MF. In vivo alpha-synuclein overexpression in rodents: a useful model of Parkinson's disease? Exp Neurol 2008;209: 22-27.
    • (2008) Exp Neurol , vol.209 , pp. 22-27
    • Chesselet, M.F.1
  • 69
    • 79955629341 scopus 로고    scopus 로고
    • Elevated tonic extracellular dopamine concentration and altered dopamine modulation of synaptic activity precede dopamine loss in the striatum of mice overexpressing human alpha-synuclein
    • Lam HA, Wu N, Cely I, et al. Elevated tonic extracellular dopamine concentration and altered dopamine modulation of synaptic activity precede dopamine loss in the striatum of mice overexpressing human alpha-synuclein. J Neurosci Res 2011;89: 1091-1102.
    • (2011) J Neurosci Res , vol.89 , pp. 1091-1102
    • Lam, H.A.1    Wu, N.2    Cely, I.3
  • 70
    • 39749196233 scopus 로고    scopus 로고
    • Selective loss of nigral dopamine neurons induced by overexpression of truncated human alpha-synuclein in mice
    • Wakamatsu M, Ishii A, Iwata S, et al. Selective loss of nigral dopamine neurons induced by overexpression of truncated human alpha-synuclein in mice. Neurobiol Aging 2008;29: 574-585.
    • (2008) Neurobiol Aging , vol.29 , pp. 574-585
    • Wakamatsu, M.1    Ishii, A.2    Iwata, S.3
  • 71
    • 84874594129 scopus 로고    scopus 로고
    • What's to like about the prion-like hypothesis for the spreading of aggregated α-synuclein in Parkinson disease?
    • Dunning CJ, George S, Brundin P. What's to like about the prion-like hypothesis for the spreading of aggregated α-synuclein in Parkinson disease? Prion 2013;7: 92-97.
    • (2013) Prion , vol.7 , pp. 92-97
    • Dunning, C.J.1    George, S.2    Brundin, P.3
  • 72
    • 8844233579 scopus 로고    scopus 로고
    • Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology
    • Zimprich A, Biskup S, Leitner P, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 2004;44: 601-607.
    • (2004) Neuron , vol.44 , pp. 601-607
    • Zimprich, A.1    Biskup, S.2    Leitner, P.3
  • 74
    • 33846562487 scopus 로고    scopus 로고
    • Parkinson's disease-associated mutations in LRRK2 link enhanced GTP-binding and kinase activities to neuronal toxicity
    • West AB, Moore DJ, Choi C, et al. Parkinson's disease-associated mutations in LRRK2 link enhanced GTP-binding and kinase activities to neuronal toxicity. Hum Mol Genet 2007;16: 223-232.
    • (2007) Hum Mol Genet , vol.16 , pp. 223-232
    • West, A.B.1    Moore, D.J.2    Choi, C.3
  • 76
    • 72149087091 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 regulates the progression ofneuropathology induced by Parkinson's-disease-related mutant αsynuclein
    • Lin X, Parisiadou L, Gu XL, et al. Leucine-rich repeat kinase 2 regulates the progression ofneuropathology induced by Parkinson's-disease-related mutant αsynuclein. Neuron 2009;64: 807-827.
    • (2009) Neuron , vol.64 , pp. 807-827
    • Lin, X.1    Parisiadou, L.2    Gu, X.L.3
  • 77
    • 67649813448 scopus 로고    scopus 로고
    • Mutant LRRK2 (R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson's disease
    • Li Y, Liu W, Oo TF, et al. Mutant LRRK2 (R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson's disease. Nat Neurosci 2009;12: 826-828.
    • (2009) Nat Neurosci , vol.12 , pp. 826-828
    • Li, Y.1    Liu, W.2    Oo, T.F.3
  • 78
    • 80053447194 scopus 로고    scopus 로고
    • Role of LRRK2 kinase dysfunction in Parkinson disease
    • Kumar A, Cookson MR. Role of LRRK2 kinase dysfunction in Parkinson disease. Expert Rev Mol Med 2011;13: e20.
    • (2011) Expert Rev Mol Med , vol.13
    • Kumar, A.1    Cookson, M.R.2
  • 79
    • 84861552733 scopus 로고    scopus 로고
    • LRRK2knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors
    • Hinkle KM, Yue M, Behrouz B, et al. LRRK2knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors. Mol Neurodegener 2012;7: 25.
    • (2012) Mol Neurodegener , vol.7 , pp. 25
    • Hinkle, K.M.1    Yue, M.2    Behrouz, B.3
  • 80
    • 84880877882 scopus 로고    scopus 로고
    • Spread of neuronal degeneration in a dopaminergic, Lrrk-G2019S model of Parkinson disease
    • Hindle SJ, Elliott CJ. Spread of neuronal degeneration in a dopaminergic, Lrrk-G2019S model of Parkinson disease. Autophagy 2013;9: 936-938.
    • (2013) Autophagy , vol.9 , pp. 936-938
    • Hindle, S.J.1    Elliott, C.J.2
  • 81
    • 0342368772 scopus 로고    scopus 로고
    • Association between early-onset Parkinson's disease and mutations in the parkin gene
    • French Parkinson's Disease Genetics Study Group; European Consortium on Genetic Susceptibility in Parkinson's Disease
    • Lücking CB, Dürr A, Bonifati V, et al; French Parkinson's Disease Genetics Study Group; European Consortium on Genetic Susceptibility in Parkinson's Disease. Association between early-onset Parkinson's disease and mutations in the parkin gene. N Engl J Med 2000;342: 1560-1567.
    • (2000) N Engl J Med , vol.342 , pp. 1560-1567
    • Lücking, C.B.1    Dürr, A.2    Bonifati, V.3
  • 82
    • 0032499264 scopus 로고    scopus 로고
    • Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism
    • Kitada T, Asakawa S, Hattori N, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998;392: 605-608.
    • (1998) Nature , vol.392 , pp. 605-608
    • Kitada, T.1    Asakawa, S.2    Hattori, N.3
  • 83
    • 84855929223 scopus 로고    scopus 로고
    • SIRT1 protects against α-synuclein aggregation by activating molecular chaperones
    • Doherty KM, Silveira-Moriyama L, Parkkinen L, et al. SIRT1 protects against α-synuclein aggregation by activating molecular chaperones. J Neurosci 2012;32: 124-132.
    • (2012) J Neurosci , vol.32 , pp. 124-132
    • Doherty, K.M.1    Silveira-Moriyama, L.2    Parkkinen, L.3
  • 84
    • 0034700158 scopus 로고    scopus 로고
    • Parkin functions as an E2-dependent ubiquitin-protein ligase and promotes the degradation of the synaptic vesicle-associated protein, CDCrel-1
    • Zhang Y, Gao J, Chung KK, Huang H, Dawson VL, Dawson TM. Parkin functions as an E2-dependent ubiquitin-protein ligase and promotes the degradation of the synaptic vesicle-associated protein, CDCrel-1. Proc Natl Acad Sci U S A. 2000;97: 13354-13359.
    • (2000) Proc Natl Acad Sci U S A , vol.97 , pp. 13354-13359
    • Zhang, Y.1    Gao, J.2    Chung, K.K.3    Huang, H.4    Dawson, V.L.5    Dawson, T.M.6
  • 85
    • 0141891953 scopus 로고    scopus 로고
    • Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons
    • Goldberg MS, Fleming SM, Palacino JJ, et al. Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons. J Biol Chem 2003;278: 43628-43635.
    • (2003) J Biol Chem , vol.278 , pp. 43628-43635
    • Goldberg, M.S.1    Fleming, S.M.2    Palacino, J.J.3
  • 86
    • 3242677684 scopus 로고    scopus 로고
    • Loss of locus coeruleus neurons and reduced startle in parkin null mice
    • Von Coelln R, Thomas B, Savitt JM, et al. Loss of locus coeruleus neurons and reduced startle in parkin null mice. Proc Natl Acad Sci U S A 2004;101: 10744-10749.
    • (2004) Proc Natl Acad Sci U S A , vol.101 , pp. 10744-10749
    • Von Coelln, R.1    Thomas, B.2    Savitt, J.M.3
  • 87
    • 13844313915 scopus 로고    scopus 로고
    • Parkin-deficient mice are not a robust model of parkinsonism
    • Perez FA, Palmiter RD. Parkin-deficient mice are not a robust model of parkinsonism. Proc Natl Acad Sci U S A 2005;102: 2174-2179.
    • (2005) Proc Natl Acad Sci U S A , vol.102 , pp. 2174-2179
    • Perez, F.A.1    Palmiter, R.D.2
  • 88
    • 2442481789 scopus 로고    scopus 로고
    • Mitochondrial dysfunction and oxidative damage in parkin-deficient mice
    • Palacino JJ, Sagi D, Goldberg MS, et al. Mitochondrial dysfunction and oxidative damage in parkin-deficient mice. J Biol Chem 2004;279: 18614-18622.
    • (2004) J Biol Chem , vol.279 , pp. 18614-18622
    • Palacino, J.J.1    Sagi, D.2    Goldberg, M.S.3
  • 89
    • 10744226640 scopus 로고    scopus 로고
    • DJ-1 (PARK7) mutations are less frequent than Parkin (PARK2) mutations in early-onset Parkinson disease
    • Hedrich K, Djarmati A, Schafer N, et al. DJ-1 (PARK7) mutations are less frequent than Parkin (PARK2) mutations in early-onset Parkinson disease. Neurology 2004;62: 389-394.
    • (2004) Neurology , vol.62 , pp. 389-394
    • Hedrich, K.1    Djarmati, A.2    Schafer, N.3
  • 90
    • 24944534660 scopus 로고    scopus 로고
    • Mitochondrial localization of the Parkinson's disease related protein DJ-1: implications for pathogenesis
    • Zhang L, Shimoji M, Thomas B, et al. Mitochondrial localization of the Parkinson's disease related protein DJ-1: implications for pathogenesis. Hum Mol Genet 2005;14: 2063-2073.
    • (2005) Hum Mol Genet , vol.14 , pp. 2063-2073
    • Zhang, L.1    Shimoji, M.2    Thomas, B.3
  • 91
    • 0345357664 scopus 로고    scopus 로고
    • Down regulation of DJ-1 enhances cell death by oxidative stress, ER stress, and proteasome inhibition
    • Yokota T, Sugawara K, Ito K, Takahashi R, Ariga H, Mizusawa H. Down regulation of DJ-1 enhances cell death by oxidative stress, ER stress, and proteasome inhibition. Biochem Biophys Res Commun 2003;312: 1342-1348.
    • (2003) Biochem Biophys Res Commun , vol.312 , pp. 1342-1348
    • Yokota, T.1    Sugawara, K.2    Ito, K.3    Takahashi, R.4    Ariga, H.5    Mizusawa, H.6
  • 92
    • 13944279784 scopus 로고    scopus 로고
    • Sensitivity to oxidative stress in DJ-1-deficient dopamine neurons: an ES-derived cell model of primary parkinsonism
    • Martinat C, Shendelman S, Jonason A, et al. Sensitivity to oxidative stress in DJ-1-deficient dopamine neurons: an ES-derived cell model of primary parkinsonism. PLoS Biol 2004;2: e327.
    • (2004) PLoS Biol , vol.2
    • Martinat, C.1    Shendelman, S.2    Jonason, A.3
  • 93
    • 13944267769 scopus 로고    scopus 로고
    • DJ-1 is a redox-dependent molecular chaperone that inhibits α-synuclein aggregate formation
    • Shendelman S, Jonason A, Martinat C, Leete T, Abeliovich A. DJ-1 is a redox-dependent molecular chaperone that inhibits α-synuclein aggregate formation. PLoS Biol 2004;2: e362.
    • (2004) PLoS Biol , vol.2
    • Shendelman, S.1    Jonason, A.2    Martinat, C.3    Leete, T.4    Abeliovich, A.5
  • 94
    • 33947282153 scopus 로고    scopus 로고
    • DJ-1 and Parkin modulate dopamine-dependent behavior and inhibit MPTP-induced nigral dopamine neuron loss in mice
    • Paterna JC, Leng A, Weber E, Feldon J, Bueler H. DJ-1 and Parkin modulate dopamine-dependent behavior and inhibit MPTP-induced nigral dopamine neuron loss in mice. Mol Ther 2007;15: 698-704.
    • (2007) Mol Ther , vol.15 , pp. 698-704
    • Paterna, J.C.1    Leng, A.2    Weber, E.3    Feldon, J.4    Bueler, H.5
  • 95
    • 79955369578 scopus 로고    scopus 로고
    • Phenylbutyrate up-regulates the DJ-1 protein and protects neurons in cell culture and in animal models of Parkinson disease
    • Zhou W, Bercury K, Cummiskey J, Luong N, Lebin J, Freed CR. Phenylbutyrate up-regulates the DJ-1 protein and protects neurons in cell culture and in animal models of Parkinson disease. J Biol Chem 2011;286: 14941-14951.
    • (2011) J Biol Chem , vol.286 , pp. 14941-14951
    • Zhou, W.1    Bercury, K.2    Cummiskey, J.3    Luong, N.4    Lebin, J.5    Freed, C.R.6
  • 96
    • 13844253723 scopus 로고    scopus 로고
    • Nigrostriatal dopaminergic deficits and hypokinesia caused by inactivation of the familial Parkinsonism-linked gene DJ-1
    • Goldberg MS, Pisani A, Haburcak M, et al. Nigrostriatal dopaminergic deficits and hypokinesia caused by inactivation of the familial Parkinsonism-linked gene DJ-1. Neuron 2005;45: 489-496.
    • (2005) Neuron , vol.45 , pp. 489-496
    • Goldberg, M.S.1    Pisani, A.2    Haburcak, M.3
  • 97
    • 20144389422 scopus 로고    scopus 로고
    • Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress
    • Kim RH, Smith PD, Aleyasin H, et al. Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci U S A 2005;102: 5215-5220.
    • (2005) Proc Natl Acad Sci U S A , vol.102 , pp. 5215-5220
    • Kim, R.H.1    Smith, P.D.2    Aleyasin, H.3
  • 99
    • 27844547616 scopus 로고    scopus 로고
    • The role of Nurr1 in the development of dopaminergic neurons and Parkinson's disease
    • Jankovic J, Chen S, Le W. The role of Nurr1 in the development of dopaminergic neurons and Parkinson's disease. Prog Neurobiol 2005;77: 128-138.
    • (2005) Prog Neurobiol , vol.77 , pp. 128-138
    • Jankovic, J.1    Chen, S.2    Le, W.3
  • 100
    • 74249098205 scopus 로고    scopus 로고
    • Role of Pitx3 in the dopamine neuron development and Parkinson's disease
    • Li J, Dani J, Le W. Role of Pitx3 in the dopamine neuron development and Parkinson's disease. Curr Top Med Chem 2009;9: 855-859.
    • (2009) Curr Top Med Chem , vol.9 , pp. 855-859
    • Li, J.1    Dani, J.2    Le, W.3
  • 101
    • 72449191782 scopus 로고    scopus 로고
    • Nurr1 Is required for maintenance of maturing and adult midbrain dopamine neurons
    • Kadkhodaei B, Ito T, Joodmardi E, et al. Nurr1 Is required for maintenance of maturing and adult midbrain dopamine neurons. J Neurosci 2009;29: 15923-15932.
    • (2009) J Neurosci , vol.29 , pp. 15923-15932
    • Kadkhodaei, B.1    Ito, T.2    Joodmardi, E.3
  • 102
    • 33846636481 scopus 로고    scopus 로고
    • Progressive parkinsonism in mice with respiratory-chain-deficient dopamine neurons
    • Ekstrand MI, Terzioglu M, Galter D, et al. Progressive parkinsonism in mice with respiratory-chain-deficient dopamine neurons. Proc Natl Acad Sci U S A 2007;104: 1325-1330.
    • (2007) Proc Natl Acad Sci U S A , vol.104 , pp. 1325-1330
    • Ekstrand, M.I.1    Terzioglu, M.2    Galter, D.3
  • 103
    • 67649379066 scopus 로고    scopus 로고
    • Non-motor symptoms of Parkinson's disease revealed in an animal model with reduced monoamine storage capacity
    • Taylor TN, Caudle WM, Shepherd KR, et al. Non-motor symptoms of Parkinson's disease revealed in an animal model with reduced monoamine storage capacity. J Neurosci 2009;29: 8103.
    • (2009) J Neurosci , vol.29 , pp. 8103
    • Taylor, T.N.1    Caudle, W.M.2    Shepherd, K.R.3
  • 104
    • 84866377694 scopus 로고    scopus 로고
    • Late-onset Parkinsonism in NFκB/c-Rel-deficient mice
    • Baiguera C, Alghisi M, Pinna A, et al. Late-onset Parkinsonism in NFκB/c-Rel-deficient mice. Brain 2012;135: 2750-2765.
    • (2012) Brain , vol.135 , pp. 2750-2765
    • Baiguera, C.1    Alghisi, M.2    Pinna, A.3
  • 105
    • 67649506191 scopus 로고    scopus 로고
    • Tests to assess motor phenotype in mice: a user's guide
    • Brooks SP, Dunnett SB. Tests to assess motor phenotype in mice: a user's guide. Nat Rev Neurosci 2009;10: 519-529.
    • (2009) Nat Rev Neurosci , vol.10 , pp. 519-529
    • Brooks, S.P.1    Dunnett, S.B.2
  • 106
    • 77952319464 scopus 로고    scopus 로고
    • Behavioral phenotyping of mouse models of Parkinson's disease
    • Taylor TN, Greene JG, Miller GW. Behavioral phenotyping of mouse models of Parkinson's disease. Behav Brain Res 2010;211: 1-10.
    • (2010) Behav Brain Res , vol.211 , pp. 1-10
    • Taylor, T.N.1    Greene, J.G.2    Miller, G.W.3
  • 107
    • 84876491370 scopus 로고    scopus 로고
    • Idiopathic REM sleep behaviour disorder in the development of Parkinson's disease
    • Boeve BF. Idiopathic REM sleep behaviour disorder in the development of Parkinson's disease. Lancet Neurol 2013;12: 469-482.
    • (2013) Lancet Neurol , vol.12 , pp. 469-482
    • Boeve, B.F.1
  • 108
    • 77957819556 scopus 로고    scopus 로고
    • Sleep alterations in an environmental neurotoxin-induced model of parkinsonism
    • McDowell KA, Hadjimarkou MM, Viechweg S, et al. Sleep alterations in an environmental neurotoxin-induced model of parkinsonism. Exp Neurol 2010;226: 84-89.
    • (2010) Exp Neurol , vol.226 , pp. 84-89
    • McDowell, K.A.1    Hadjimarkou, M.M.2    Viechweg, S.3
  • 109
    • 84861692236 scopus 로고    scopus 로고
    • Alpha-synuclein in colonic submucosa in early untreated Parkinson's disease
    • Shannon KM, Keshavarzian A, Mutlu E, et al. Alpha-synuclein in colonic submucosa in early untreated Parkinson's disease. Mov Disord 2012;27: 709-715.
    • (2012) Mov Disord , vol.27 , pp. 709-715
    • Shannon, K.M.1    Keshavarzian, A.2    Mutlu, E.3
  • 110
    • 67349122370 scopus 로고    scopus 로고
    • Delayed gastric emptying and enteric nervous system dysfunction in the rotenone model of Parkinson's disease
    • Greene JG, Noorian AR, Srinivasan S. Delayed gastric emptying and enteric nervous system dysfunction in the rotenone model of Parkinson's disease. Exp Neurol 2009;218: 154-161.
    • (2009) Exp Neurol , vol.218 , pp. 154-161
    • Greene, J.G.1    Noorian, A.R.2    Srinivasan, S.3
  • 111
    • 34548230441 scopus 로고    scopus 로고
    • Loss of enteric dopaminergic neurons and associated changes in colon motility in an MPTP mouse model of Parkinson's disease
    • Anderson G, Noorian AR, Taylor G, et al. Loss of enteric dopaminergic neurons and associated changes in colon motility in an MPTP mouse model of Parkinson's disease. Exp Neurol 2007;207: 4-12.
    • (2007) Exp Neurol , vol.207 , pp. 4-12
    • Anderson, G.1    Noorian, A.R.2    Taylor, G.3
  • 112
    • 70350773935 scopus 로고    scopus 로고
    • Functional and neurochemical changes of the gastrointestinal tract in a rodent model of Parkinson's disease
    • Blandini F, Balestra B, Levandis G, et al. Functional and neurochemical changes of the gastrointestinal tract in a rodent model of Parkinson's disease. Neurosci Lett 2009;467: 203-207.
    • (2009) Neurosci Lett , vol.467 , pp. 203-207
    • Blandini, F.1    Balestra, B.2    Levandis, G.3
  • 113
    • 67649392374 scopus 로고    scopus 로고
    • Effects of Plantago ovata husk on levodopa (with Carbidopa) bioavailability in rabbits withautonomicgastrointestinal disorders
    • García JJ, Fernández N, Calle AP, Diez MJ, Sahagún A, Sierra M. Effects of Plantago ovata husk on levodopa (with Carbidopa) bioavailability in rabbits withautonomicgastrointestinal disorders. Drug Metab Dispos 2009;37: 1434-1442.
    • (2009) Drug Metab Dispos , vol.37 , pp. 1434-1442
    • García, J.J.1    Fernández, N.2    Calle, A.P.3    Diez, M.J.4    Sahagún, A.5    Sierra, M.6
  • 114
    • 77953021942 scopus 로고    scopus 로고
    • Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders
    • Beach TG, Adler CH, Sue LI, et al. Multi-organ distribution of phosphorylated alpha-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol 2010;119: 689-702.
    • (2010) Acta Neuropathol , vol.119 , pp. 689-702
    • Beach, T.G.1    Adler, C.H.2    Sue, L.I.3
  • 115
    • 81555214521 scopus 로고    scopus 로고
    • Bladder, bowel, and sexual dysfunction in Parkinson's Disease
    • Sakakibara R, Kishi M, Ogawa E, et al. Bladder, bowel, and sexual dysfunction in Parkinson's Disease. Parkinsons Dis 2011;924605.
    • (2011) Parkinsons Dis , pp. 924605
    • Sakakibara, R.1    Kishi, M.2    Ogawa, E.3
  • 117
    • 2542444472 scopus 로고    scopus 로고
    • Effects of high frequency deep brain stimulation on urine storage and voiding function in conscious minipigs
    • Dalmose AL, Bjarkam CR, Sorensen JC, Djurhuus JC, T. M. Jorgensen. Effects of high frequency deep brain stimulation on urine storage and voiding function in conscious minipigs. Neurourol Urodyn 2004;23: 265-272.
    • (2004) Neurourol Urodyn , vol.23 , pp. 265-272
    • Dalmose, A.L.1    Bjarkam, C.R.2    Sorensen, J.C.3    Djurhuus, J.C.4    Jorgensen, T.M.5
  • 118
    • 72649096093 scopus 로고    scopus 로고
    • Dopamine receptor agonists and depression in Parkinson's disease
    • Picillo M, Rocco M, Barone P. Dopamine receptor agonists and depression in Parkinson's disease. Parkinsonism Relat Disord 2009;15(Suppl. 4): S81-84.
    • (2009) Parkinsonism Relat Disord , vol.15 , Issue.SUPPL. 4
    • Picillo, M.1    Rocco, M.2    Barone, P.3
  • 119
    • 84856904557 scopus 로고    scopus 로고
    • Evaluation of antidepressant activity of ropinirole coadministered with fluoxetine in acute and chronic behavioral models of depression in rats
    • Ghorpade S, Tripathi R, Sonawane D, Manjrekar N. Evaluation of antidepressant activity of ropinirole coadministered with fluoxetine in acute and chronic behavioral models of depression in rats. J Basic Clin Physiol Pharmacol 2011;22: 109-114.
    • (2011) J Basic Clin Physiol Pharmacol , vol.22 , pp. 109-114
    • Ghorpade, S.1    Tripathi, R.2    Sonawane, D.3    Manjrekar, N.4
  • 120
    • 84855278236 scopus 로고    scopus 로고
    • Fluoxetine rescues impaired hippocampal neurogenesis in a transgenic A53T synuclein mouse model
    • Kohl Z, Winner B, Ubhi K, et al. Fluoxetine rescues impaired hippocampal neurogenesis in a transgenic A53T synuclein mouse model. Eur J Neurosci 2012;35: 10-19.
    • (2012) Eur J Neurosci , vol.35 , pp. 10-19
    • Kohl, Z.1    Winner, B.2    Ubhi, K.3
  • 121
    • 84864866633 scopus 로고    scopus 로고
    • The effects of short-term nicotine administration on behavioral and oxidative stress deficiencies induced in a rat model of Parkinson's disease
    • Ciobica A, Padurariu M, Hritcu L. The effects of short-term nicotine administration on behavioral and oxidative stress deficiencies induced in a rat model of Parkinson's disease. Psychiatr Danub 2012;24: 194-205.
    • (2012) Psychiatr Danub , vol.24 , pp. 194-205
    • Ciobica, A.1    Padurariu, M.2    Hritcu, L.3
  • 122
    • 84858058939 scopus 로고    scopus 로고
    • The effects of pergolide on memory and oxidative stress in a rat model of Parkinson's disease
    • Ciobica A, Olteanu Z, Padurariu M, Hritcu L. The effects of pergolide on memory and oxidative stress in a rat model of Parkinson's disease. J Physiol Biochem 2012;68: 59-69.
    • (2012) J Physiol Biochem , vol.68 , pp. 59-69
    • Ciobica, A.1    Olteanu, Z.2    Padurariu, M.3    Hritcu, L.4
  • 123
    • 84856599800 scopus 로고    scopus 로고
    • Intranasally applied L-DOPA alleviates parkinsonian symptoms in rats with unilateral nigro-striatal6-OHDA lesions
    • Chao OY, Mattern C, Silva AM, et al. Intranasally applied L-DOPA alleviates parkinsonian symptoms in rats with unilateral nigro-striatal6-OHDA lesions. Brain Res Bull 2012;87: 340-345.
    • (2012) Brain Res Bull , vol.87 , pp. 340-345
    • Chao, O.Y.1    Mattern, C.2    Silva, A.M.3
  • 124
    • 80051822766 scopus 로고    scopus 로고
    • Sustained-release formulation of levodopa methyl ester/benserazide for prolonged suppressing dyskinesia expression in 6-OHDA-leisoned rats
    • Ren T, Yang X, Wu N, Cai Y, Liu Z, Yuan W. Sustained-release formulation of levodopa methyl ester/benserazide for prolonged suppressing dyskinesia expression in 6-OHDA-leisoned rats. Neurosci Lett 2011;502: 117-122.
    • (2011) Neurosci Lett , vol.502 , pp. 117-122
    • Ren, T.1    Yang, X.2    Wu, N.3    Cai, Y.4    Liu, Z.5    Yuan, W.6
  • 125
    • 33847797214 scopus 로고    scopus 로고
    • Transdermal rotigotine: a new non-ergot dopamine agonist for the treatment of Parkinson's disease
    • Naidu Y, Chaudhuri KR. Transdermal rotigotine: a new non-ergot dopamine agonist for the treatment of Parkinson's disease. Expert Opin Drug Deliv 2007;4: 111-118.
    • (2007) Expert Opin Drug Deliv , vol.4 , pp. 111-118
    • Naidu, Y.1    Chaudhuri, K.R.2
  • 126
    • 37249035652 scopus 로고    scopus 로고
    • Constant dopaminergic stimulation by transdermal delivery of dopaminergic drugs: a new treatment paradigm in Parkinson's disease
    • Steiger M. Constant dopaminergic stimulation by transdermal delivery of dopaminergic drugs: a new treatment paradigm in Parkinson's disease. Eur J Neurol 2008;15: 6-15.
    • (2008) Eur J Neurol , vol.15 , pp. 6-15
    • Steiger, M.1
  • 127
    • 82455175532 scopus 로고    scopus 로고
    • Spotlight on rotigotine transdermal patch in Parkinson's disease
    • Sanford M, Scott LJ. Spotlight on rotigotine transdermal patch in Parkinson's disease. Drugs Aging 2011;28: 1015-1017.
    • (2011) Drugs Aging , vol.28 , pp. 1015-1017
    • Sanford, M.1    Scott, L.J.2
  • 128
    • 42749085138 scopus 로고    scopus 로고
    • Continuous delivery of ropinirole reverses motor deficits without dyskinesia induction in MPTP-treated common marmosets
    • Stockwell KA, Virley DJ, Perren M, et al. Continuous delivery of ropinirole reverses motor deficits without dyskinesia induction in MPTP-treated common marmosets. Exp Neurol 2008;211: 172-179.
    • (2008) Exp Neurol , vol.211 , pp. 172-179
    • Stockwell, K.A.1    Virley, D.J.2    Perren, M.3
  • 129
    • 28144463965 scopus 로고    scopus 로고
    • Validation of the l-dopa-induced dyskinesia in the 6-OHDA model and evaluation of the effects of selective dopamine receptor agonists and antagonists
    • Monville C, Torres EM, Dunnett SB. Validation of the l-dopa-induced dyskinesia in the 6-OHDA model and evaluation of the effects of selective dopamine receptor agonists and antagonists. Brain Res Bull 2005;68: 16-23.
    • (2005) Brain Res Bull , vol.68 , pp. 16-23
    • Monville, C.1    Torres, E.M.2    Dunnett, S.B.3
  • 130
    • 57049160908 scopus 로고    scopus 로고
    • Effects of a NR2B selective NMDA glutamate antagonist, CP-101,606, on dyskinesia and Parkinsonism
    • Nutt JG, Gunzler SA, Kirchhoff T, et al. Effects of a NR2B selective NMDA glutamate antagonist, CP-101, 606, on dyskinesia and Parkinsonism. Mov Disord 2008;23: 1860-1866.
    • (2008) Mov Disord , vol.23 , pp. 1860-1866
    • Nutt, J.G.1    Gunzler, S.A.2    Kirchhoff, T.3
  • 132
    • 27744571228 scopus 로고    scopus 로고
    • Combined blockade of AMPA and NMDA glutamate receptors reduces levodopa-induced motor complications in animal models of PD
    • Bibbiani F, Oh JD, Kielaite A, Collins MA, Smith C, Chase TN. Combined blockade of AMPA and NMDA glutamate receptors reduces levodopa-induced motor complications in animal models of PD. Exp Neurol 2005;196: 422-429.
    • (2005) Exp Neurol , vol.196 , pp. 422-429
    • Bibbiani, F.1    Oh, J.D.2    Kielaite, A.3    Collins, M.A.4    Smith, C.5    Chase, T.N.6
  • 133
    • 84855205036 scopus 로고    scopus 로고
    • Pharmacological characterization of MRZ-8676, a novel negative allosteric modulator of subtype 5 metabotropic glutamate receptors (mGluR5): focus on L-DOPA-induced dyskinesia
    • Dekundy A, Gravius A, Hechenberger M, et al. Pharmacological characterization of MRZ-8676, a novel negative allosteric modulator of subtype 5 metabotropic glutamate receptors (mGluR5): focus on L-DOPA-induced dyskinesia. J Neural Transm 2011;118: 1703-1716.
    • (2011) J Neural Transm , vol.118 , pp. 1703-1716
    • Dekundy, A.1    Gravius, A.2    Hechenberger, M.3
  • 134
    • 79959372108 scopus 로고    scopus 로고
    • AFQ056 treatment of levodopa-induced dyskinesias: Results of 2 randomized controlled trials
    • Berg D, Godau J, Trenkwalder C, et al. AFQ056 treatment of levodopa-induced dyskinesias: Results of 2 randomized controlled trials. Mov Disord 2011;26: 1243-1250.
    • (2011) Mov Disord , vol.26 , pp. 1243-1250
    • Berg, D.1    Godau, J.2    Trenkwalder, C.3
  • 135
    • 84856022448 scopus 로고    scopus 로고
    • The metabotropic glutamate receptor 4-positive allosteric modulator VU0364770 produces efficacy alone and in combination with L-DOPA or an adenosine 2A antagonist in preclinical rodent models of Parkinson's disease
    • Jones CK, Bubser M, Thompson AD, et al. The metabotropic glutamate receptor 4-positive allosteric modulator VU0364770 produces efficacy alone and in combination with L-DOPA or an adenosine 2A antagonist in preclinical rodent models of Parkinson's disease. J Pharmacol Exp Ther 2012;340: 404-421.
    • (2012) J Pharmacol Exp Ther , vol.340 , pp. 404-421
    • Jones, C.K.1    Bubser, M.2    Thompson, A.D.3
  • 136
    • 84857613594 scopus 로고    scopus 로고
    • Adenosine A(2A) receptor gene disruption protects in an α-synuclein model of Parkinson's disease
    • Kachroo A, Schwarzschild MA. Adenosine A(2A) receptor gene disruption protects in an α-synuclein model of Parkinson's disease. Ann Neurol 2012;71: 278-282.
    • (2012) Ann Neurol , vol.71 , pp. 278-282
    • Kachroo, A.1    Schwarzschild, M.A.2
  • 137
    • 78650416384 scopus 로고    scopus 로고
    • Chronic A2A antagonist treatment alleviates parkinsonian locomotor deficiency in MitoPark mice
    • Marcellino D, Lindqvist E, Schneider M, et al. Chronic A2A antagonist treatment alleviates parkinsonian locomotor deficiency in MitoPark mice. Neurobiol Dis 2010;40: 460-466.
    • (2010) Neurobiol Dis , vol.40 , pp. 460-466
    • Marcellino, D.1    Lindqvist, E.2    Schneider, M.3
  • 138
    • 72449124213 scopus 로고    scopus 로고
    • The selective alpha1 adrenoceptor antagonist HEAT reduces L-DOPA-induced dyskinesia in a rat model of Parkinson's disease
    • Buck K, Ferger B. The selective alpha1 adrenoceptor antagonist HEAT reduces L-DOPA-induced dyskinesia in a rat model of Parkinson's disease. Synapse 2010;64: 117-126.
    • (2010) Synapse , vol.64 , pp. 117-126
    • Buck, K.1    Ferger, B.2
  • 139
    • 34447532344 scopus 로고    scopus 로고
    • The differential effects of 5-HT(1A) receptor stimulation on dopamine receptor-mediated abnormal involuntary movements and rotations in the primed hemiparkinsonian rat
    • Dupre KB, Eskow KL, Negron G, Bishop C. The differential effects of 5-HT(1A) receptor stimulation on dopamine receptor-mediated abnormal involuntary movements and rotations in the primed hemiparkinsonian rat. Brain Res 2007;1158: 135-143.
    • (2007) Brain Res , vol.1158 , pp. 135-143
    • Dupre, K.B.1    Eskow, K.L.2    Negron, G.3    Bishop, C.4
  • 140
    • 84860452656 scopus 로고    scopus 로고
    • Neuronal nitric oxide synthase inhibition attenuates the development of L-DOPA-induceddyskinesia in hemi-Parkinsonian rats
    • Takuma K, Tanaka T, Takahashi T, et al. Neuronal nitric oxide synthase inhibition attenuates the development of L-DOPA-induceddyskinesia in hemi-Parkinsonian rats. Eur J Pharmacol 2012;683: 166-173.
    • (2012) Eur J Pharmacol , vol.683 , pp. 166-173
    • Takuma, K.1    Tanaka, T.2    Takahashi, T.3
  • 141
    • 77952847092 scopus 로고    scopus 로고
    • Effect of cdk5 antagonist on L-dopa-induced dyskinesias in a rat model of Parkinson's disease
    • Guan Q, Liu X, He Y, Jin L, Zhao L. Effect of cdk5 antagonist on L-dopa-induced dyskinesias in a rat model of Parkinson's disease. Int J Neurosci 2010;120: 421-427.
    • (2010) Int J Neurosci , vol.120 , pp. 421-427
    • Guan, Q.1    Liu, X.2    He, Y.3    Jin, L.4    Zhao, L.5
  • 142
    • 84863434599 scopus 로고    scopus 로고
    • Influence of corticostriatal δ-opioid receptors on abnormal involuntary movements induced by L-DOPA in hemiparkinsonian rats
    • Billet F, Costentin J, Dourmap N. Influence of corticostriatal δ-opioid receptors on abnormal involuntary movements induced by L-DOPA in hemiparkinsonian rats. Exp Neurol 2012; 236: 339-350.
    • (2012) Exp Neurol , vol.236 , pp. 339-350
    • Billet, F.1    Costentin, J.2    Dourmap, N.3
  • 143
    • 33845392575 scopus 로고    scopus 로고
    • Paraquat-induced oxidative stress in drosophila melanogaster: effects of melatonin, glutathione, serotonin, minocycline, lipoic acid and ascorbic acid
    • Bonilla E, Medina-Leendertz S, Villalobos V, Molero L, Bohórquez A. Paraquat-induced oxidative stress in drosophila melanogaster: effects of melatonin, glutathione, serotonin, minocycline, lipoic acid and ascorbic acid. Neurochem Res 2006;31: 1425-1432.
    • (2006) Neurochem Res , vol.31 , pp. 1425-1432
    • Bonilla, E.1    Medina-Leendertz, S.2    Villalobos, V.3    Molero, L.4    Bohórquez, A.5
  • 144
    • 79961208135 scopus 로고    scopus 로고
    • Protective effect of lycopene on oxidative stress and cognitive decline in rotenone induced model of Parkinson's disease
    • Kaur H, Chauhan S, Sandhir R. Protective effect of lycopene on oxidative stress and cognitive decline in rotenone induced model of Parkinson's disease. Neurochem Res 2011;36: 1435-1443.
    • (2011) Neurochem Res , vol.36 , pp. 1435-1443
    • Kaur, H.1    Chauhan, S.2    Sandhir, R.3
  • 145
    • 67650094606 scopus 로고    scopus 로고
    • Effect of centrophenoxine against rotenone-induced oxidative stress in an animal model of Parkinson's disease
    • Verma R, Nehru B. Effect of centrophenoxine against rotenone-induced oxidative stress in an animal model of Parkinson's disease. Neurochem Int 2009;55: 369-375.
    • (2009) Neurochem Int , vol.55 , pp. 369-375
    • Verma, R.1    Nehru, B.2
  • 146
    • 83155187331 scopus 로고    scopus 로고
    • Motor disorders and impaired electrical power of pallidal EEG improved by gallic acid in animalmodel of Parkinson's disease
    • Sameri MJ, Sarkaki A, Farbood Y, Mansouri SM. Motor disorders and impaired electrical power of pallidal EEG improved by gallic acid in animalmodel of Parkinson's disease. Pak J Biol Sci 2011;14: 1109-1116.
    • (2011) Pak J Biol Sci , vol.14 , pp. 1109-1116
    • Sameri, M.J.1    Sarkaki, A.2    Farbood, Y.3    Mansouri, S.M.4
  • 147
    • 36549060678 scopus 로고    scopus 로고
    • Protective effects of green tea polyphenols in the 6-OHDA rat model of Parkinson's disease through inhibition of ROS-NO pathway
    • Guo S, Yan J, Yang T, Yang X, Bezard E, Zhao B. Protective effects of green tea polyphenols in the 6-OHDA rat model of Parkinson's disease through inhibition of ROS-NO pathway. Biol Psychiatry 2007;62: 1353-1362.
    • (2007) Biol Psychiatry , vol.62 , pp. 1353-1362
    • Guo, S.1    Yan, J.2    Yang, T.3    Yang, X.4    Bezard, E.5    Zhao, B.6
  • 149
    • 43549107707 scopus 로고    scopus 로고
    • Comparison of neuroprotective and neurorestorative capabilities of rasagiline and selegiline against lactacystin induced nigrostriatal dopaminergic degeneration
    • Zhu W, Xie W, Pan T, et al. Comparison of neuroprotective and neurorestorative capabilities of rasagiline and selegiline against lactacystin induced nigrostriatal dopaminergic degeneration. J Neurochem 2008;105: 1970-1978.
    • (2008) J Neurochem , vol.105 , pp. 1970-1978
    • Zhu, W.1    Xie, W.2    Pan, T.3
  • 150
    • 38149073317 scopus 로고    scopus 로고
    • Aggregation of alpha-synuclein by DOPAL, the monoamine oxidase metabolite of dopamine
    • Burke WJ, Kumar VB, Pandey N, et al. Aggregation of alpha-synuclein by DOPAL, the monoamine oxidase metabolite of dopamine. Acta Neuropathol 2008;115: 193-203.
    • (2008) Acta Neuropathol , vol.115 , pp. 193-203
    • Burke, W.J.1    Kumar, V.B.2    Pandey, N.3
  • 151
    • 81955167502 scopus 로고    scopus 로고
    • Explaining ADAGIO: A critical review of the biological basis for the clinical effects of rasagiline
    • Jenner P, Langston JW. Explaining ADAGIO: A critical review of the biological basis for the clinical effects of rasagiline. Mov Disord. 2011;26: 2316-2323.
    • (2011) Mov Disord , vol.26 , pp. 2316-2323
    • Jenner, P.1    Langston, J.W.2
  • 152
    • 77951941750 scopus 로고    scopus 로고
    • Reply: Based on the available randomized trial patients should say no to glutathione for Parkinson's disease
    • Okun MS, Lang A, Jankovic J. Reply: Based on the available randomized trial patients should say no to glutathione for Parkinson's disease. Mov Disord 2010;25: 961-962.
    • (2010) Mov Disord , vol.25 , pp. 961-962
    • Okun, M.S.1    Lang, A.2    Jankovic, J.3
  • 153
    • 84863983617 scopus 로고    scopus 로고
    • Assessment of the treatment effect of baicalein on a model of Parkinsonian tremor and elucidation of the mechanism
    • Yu X, He GR, Sun L, Lan X, Shi LL, Xuan ZH, Du GH. Assessment of the treatment effect of baicalein on a model of Parkinsonian tremor and elucidation of the mechanism. Life Sci 2012;91: 5-13.
    • (2012) Life Sci , vol.91 , pp. 5-13
    • Yu, X.1    He, G.R.2    Sun, L.3    Lan, X.4    Shi, L.L.5    Xuan, Z.H.6    Du, G.H.7
  • 154
    • 84855525812 scopus 로고    scopus 로고
    • Protective effects of baicalein against rotenone-induced neurotoxicity in PC12 cells and isolated rat brain mitochondria
    • Li XX, He GR, Mu X, et al. Protective effects of baicalein against rotenone-induced neurotoxicity in PC12 cells and isolated rat brain mitochondria. Eur J Pharmacol 2012;674: 227-233.
    • (2012) Eur J Pharmacol , vol.674 , pp. 227-233
    • Li, X.X.1    He, G.R.2    Mu, X.3
  • 155
    • 84880750830 scopus 로고    scopus 로고
    • Diapocynin prevents early Parkinson's disease symptoms in the leucine-rich repeat kinase 2 (LRRK2-R1441G) transgenic mouse
    • Dranka BP, Gifford A, Ghosh A, et al. Diapocynin prevents early Parkinson's disease symptoms in the leucine-rich repeat kinase 2 (LRRK2-R1441G) transgenic mouse. Neurosci Lett 2013;S0304-3940.
    • (2013) Neurosci Lett , pp. 0304-3940
    • Dranka, B.P.1    Gifford, A.2    Ghosh, A.3
  • 156
    • 84863258358 scopus 로고    scopus 로고
    • Effect of quercetin in the 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse model of Parkinson's disease
    • Lv C, Hong T, Yang Z, et al. Effect of quercetin in the 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced mouse model of Parkinson's disease. Evid Based Complement Alternat Med 2012;2012: 928643.
    • (2012) Evid Based Complement Alternat Med , vol.2012 , pp. 928643
    • Lv, C.1    Hong, T.2    Yang, Z.3
  • 157
    • 84860842847 scopus 로고    scopus 로고
    • Therapeutic effect of near infrared (NIR) light on Parkinson's disease models
    • Quirk BJ, Desmet KD, Henry M, et al. Therapeutic effect of near infrared (NIR) light on Parkinson's disease models. Front Biosci (Elite Ed) 2012;4: 818-823.
    • (2012) Front Biosci (Elite Ed) , vol.4 , pp. 818-823
    • Quirk, B.J.1    Desmet, K.D.2    Henry, M.3
  • 158
    • 79955757052 scopus 로고    scopus 로고
    • Passive immunization reduces behavioral and neuropathological deficits in an alpha-synucleintransgenic model of Lewy body disease
    • Masliah E, Rockenstein E, Mante M, et al. Passive immunization reduces behavioral and neuropathological deficits in an alpha-synucleintransgenic model of Lewy body disease. PLoS One 2011;6: e19338.
    • (2011) PLoS One , vol.6
    • Masliah, E.1    Rockenstein, E.2    Mante, M.3
  • 159
    • 79961210760 scopus 로고    scopus 로고
    • Lithium protects against oxidative stress-mediated cell death in α-synuclein-overexpressing in vitro and in vivo models of Parkinson's disease
    • Kim YH, Rane A, Lussier S, Andersen JK. Lithium protects against oxidative stress-mediated cell death in α-synuclein-overexpressing in vitro and in vivo models of Parkinson's disease. J Neurosci Res 2011;89: 1666-1675.
    • (2011) J Neurosci Res , vol.89 , pp. 1666-1675
    • Kim, Y.H.1    Rane, A.2    Lussier, S.3    Andersen, J.K.4
  • 160
    • 84860871900 scopus 로고    scopus 로고
    • A prolyl oligopeptidase inhibitor, KYP-2047, reduces α-synuclein protein levels and aggregates in cellular and animal models of Parkinson's disease
    • Myöhänen TT, Hannula MJ, van Elzen R, et al. A prolyl oligopeptidase inhibitor, KYP-2047, reduces α-synuclein protein levels and aggregates in cellular and animal models of Parkinson's disease. Br J Pharmacol 2012;166: 1097-1113.
    • (2012) Br J Pharmacol , vol.166 , pp. 1097-1113
    • Myöhänen, T.T.1    Hannula, M.J.2    van Elzen, R.3
  • 161
    • 84861728900 scopus 로고    scopus 로고
    • The hypoxia imaging agent CuII(atsm) is neuroprotective and improves motor and cognitive functions in multiple animal models of Parkinson's disease
    • Hung LW, Villemagne VL, Cheng L, et al. The hypoxia imaging agent CuII(atsm) is neuroprotective and improves motor and cognitive functions in multiple animal models of Parkinson's disease. J Exp Med 2012;209: 837-854.
    • (2012) J Exp Med , vol.209 , pp. 837-854
    • Hung, L.W.1    Villemagne, V.L.2    Cheng, L.3
  • 162
    • 79956317900 scopus 로고    scopus 로고
    • Enhanced phosphatase activity attenuates α-synucleinopathy in a mouse model
    • Lee KW, Chen W, Junn E, et al. Enhanced phosphatase activity attenuates α-synucleinopathy in a mouse model. J Neurosci 2011;31: 6963-6971.
    • (2011) J Neurosci , vol.31 , pp. 6963-6971
    • Lee, K.W.1    Chen, W.2    Junn, E.3
  • 163
    • 80052473816 scopus 로고    scopus 로고
    • Toll-like receptor 4 promotes α-synuclein clearance and survival of nigral dopaminergic neurons
    • Stefanova N, Fellner L, Reindl M, et al. Toll-like receptor 4 promotes α-synuclein clearance and survival of nigral dopaminergic neurons. Am J Pathol 2011; 179: 954-963.
    • (2011) Am J Pathol , vol.179 , pp. 954-963
    • Stefanova, N.1    Fellner, L.2    Reindl, M.3
  • 164
    • 80053152848 scopus 로고    scopus 로고
    • Inhibitors of LRRK2 kinase attenuate neurodegenerationand Parkinson-like phenotypes in Caenorhabditis elegans and Drosophila Parkinson's disease models
    • Liu Z, Hamamichi S, Lee BD et al. Inhibitors of LRRK2 kinase attenuate neurodegenerationand Parkinson-like phenotypes in Caenorhabditis elegans and Drosophila Parkinson's disease models. Hum Mol Genet 2011;20: 3933-3942.
    • (2011) Hum Mol Genet , vol.20 , pp. 3933-3942
    • Liu, Z.1    Hamamichi, S.2    Lee, B.D.3
  • 165
    • 0037226797 scopus 로고    scopus 로고
    • Mutations in NR4A2 associated with familial Parkinson's disease
    • Le W, Xu PY, Jiang H, et al. Mutations in NR4A2 associated with familial Parkinson's disease. Nat Genet2003;33: 85-89.
    • (2003) Nat Genet , vol.33 , pp. 85-89
    • Le, W.1    Xu, P.Y.2    Jiang, H.3
  • 166
    • 33845642069 scopus 로고    scopus 로고
    • Identification of a series of highly potent activators of the Nurr1 signaling pathway
    • Hintermann S, Chiesi M, von Krosigk U, et al. Identification of a series of highly potent activators of the Nurr1 signaling pathway Bioorg Med Chem Lett 2007;17: 193-196.
    • (2007) Bioorg Med Chem Lett , vol.17 , pp. 193-196
    • Hintermann, S.1    Chiesi, M.2    von Krosigk, U.3
  • 167
    • 84873730294 scopus 로고    scopus 로고
    • Wnt5a cooperates with canonical Wnts to generate midbrain dopaminergic neurons in vivo and in stem cells
    • Anderson E, Saltó C, Villaescusa JC, et al. Wnt5a cooperates with canonical Wnts to generate midbrain dopaminergic neurons in vivo and in stem cells PNAS 2013;E602-E610.
    • (2013) PNAS
    • Anderson, E.1    Saltó, C.2    Villaescusa, J.C.3
  • 168
    • 84856981228 scopus 로고    scopus 로고
    • Plasticity of Subventricular Zone Neuroprogenitors in MPTP (methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mouse of Parkinson's disease involves cross talk between inflammatory and Wnt/-catenin signaling pathways: functional consequences for neuroprotection and repair
    • L'Episcopo F, Tirolo C, Testa N, et al. Plasticity of Subventricular Zone Neuroprogenitors in MPTP (methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine) mouse of Parkinson's disease involves cross talk between inflammatory and Wnt/-catenin signaling pathways: functional consequences for neuroprotection and repair. J Neurosci 2012;32: 2062-2085.
    • (2012) J Neurosci , vol.32 , pp. 2062-2085
    • L'Episcopo, F.1    Tirolo, C.2    Testa, N.3
  • 169
    • 78651282742 scopus 로고    scopus 로고
    • A neuroprotective role for angiogenin in models of Parkinson's disease
    • Steidinger TU, Standaert DG, Yacoubian TA. A neuroprotective role for angiogenin in models of Parkinson's disease. J Neurochem 2011;116: 334-341.
    • (2011) J Neurochem , vol.116 , pp. 334-341
    • Steidinger, T.U.1    Standaert, D.G.2    Yacoubian, T.A.3
  • 170
    • 83555174800 scopus 로고    scopus 로고
    • Melatonin protects against rotenone-induced cell injury via inhibition of Omi and Bax-mediated autophagy in Hela cells
    • Zhou H, Chen J, Lu X, et al. Melatonin protects against rotenone-induced cell injury via inhibition of Omi and Bax-mediated autophagy in Hela cells. J Pineal Res 2012;52: 120-127.
    • (2012) J Pineal Res , vol.52 , pp. 120-127
    • Zhou, H.1    Chen, J.2    Lu, X.3
  • 171
    • 84855929223 scopus 로고    scopus 로고
    • SIRT1 protects against α-synuclein aggregation by activating molecular chaperones
    • Donmez G, Arun A, Chung CY, et al. SIRT1 protects against α-synuclein aggregation by activating molecular chaperones. J Neurosci 2012;32: 124-132.
    • (2012) J Neurosci , vol.32 , pp. 124-132
    • Donmez, G.1    Arun, A.2    Chung, C.Y.3
  • 172
    • 80052359850 scopus 로고    scopus 로고
    • Resveratrol-activated AMPK/SIRT1/Autophagy in cellular models of Parkinson's disease
    • Wu Y, Li X, Zhu JX, et al. Resveratrol-activated AMPK/SIRT1/Autophagy in cellular models of Parkinson's disease. Neurosignals 2011;19: 163-174.
    • (2011) Neurosignals , vol.19 , pp. 163-174
    • Wu, Y.1    Li, X.2    Zhu, J.X.3
  • 173
    • 79955945545 scopus 로고    scopus 로고
    • Granulocyte macrophage-colony stimulating factor protects against substantia nigra dopaminergic cell loss in an environmental toxin model of Parkinson's disease
    • Mangano EN, Peters S, Litteljohn D, et al. Granulocyte macrophage-colony stimulating factor protects against substantia nigra dopaminergic cell loss in an environmental toxin model of Parkinson's disease. Neurbiol Dis 2011;43: 99-112.
    • (2011) Neurbiol Dis , vol.43 , pp. 99-112
    • Mangano, E.N.1    Peters, S.2    Litteljohn, D.3
  • 174
    • 70350150325 scopus 로고    scopus 로고
    • A chemical chaperone, sodium 4-phenylbutyric acid, attenuates the pathogenic potency in human alpha-synuclein A30P + A53T transgenic mice
    • Ono K, Ikemoto M, Kawarabayashi T, et al. A chemical chaperone, sodium 4-phenylbutyric acid, attenuates the pathogenic potency in human alpha-synuclein A30P + A53T transgenic mice. Parkinsonism Relat Disord 2009;15: 649-654.
    • (2009) Parkinsonism Relat Disord , vol.15 , pp. 649-654
    • Ono, K.1    Ikemoto, M.2    Kawarabayashi, T.3
  • 175
    • 84862487151 scopus 로고    scopus 로고
    • Sodium phenylbutyrate controls neuroinflammatory and antioxidant activities and protects dopaminergic neurons in mouse models of Parkinson's disease
    • Roy A, Ghosh A, Jana A, et al. Sodium phenylbutyrate controls neuroinflammatory and antioxidant activities and protects dopaminergic neurons in mouse models of Parkinson's disease. PLoS One. 2012;7: e38113.
    • (2012) PLoS One , vol.7
    • Roy, A.1    Ghosh, A.2    Jana, A.3
  • 176
    • 84859322860 scopus 로고    scopus 로고
    • Riluzole neuroprotection in a Parkinson's disease model involves suppression of reactive astrocytosis but not GLT-1 regulation
    • Carbone M, Duty S, Rattray M. Riluzole neuroprotection in a Parkinson's disease model involves suppression of reactive astrocytosis but not GLT-1 regulation. BMC Neurosci 2012;5: 13: 38.
    • (2012) BMC Neurosci , vol.13 , pp. 38
    • Carbone, M.1    Duty, S.2    Rattray, M.3
  • 177
    • 84862619730 scopus 로고    scopus 로고
    • Meloxicam ameliorates motor dysfunction and dopaminergic neurodegeneration by maintaining Akt-signaling in a mouse Parkinson's disease model
    • Tasaki Y, Yamamoto J, Omura T, et al. Meloxicam ameliorates motor dysfunction and dopaminergic neurodegeneration by maintaining Akt-signaling in a mouse Parkinson's disease model. Neurosci Lett 2012;521: 15-19.
    • (2012) Neurosci Lett , vol.521 , pp. 15-19
    • Tasaki, Y.1    Yamamoto, J.2    Omura, T.3
  • 178
    • 79958024002 scopus 로고    scopus 로고
    • Edaravone guards dopamine neurons in a rotenone model for Parkinson's disease
    • Xiong N, Xiong J, Khare G, et al. Edaravone guards dopamine neurons in a rotenone model for Parkinson's disease. PLoS One. 2011;6: e20677.
    • (2011) PLoS One , vol.6
    • Xiong, N.1    Xiong, J.2    Khare, G.3
  • 179
    • 83655183720 scopus 로고    scopus 로고
    • Idebenone and resveratrol extend lifespan and improve motor function of HtrA2 knockout mice
    • Gerhardt E, Gräber S, Szego EM, et al. Idebenone and resveratrol extend lifespan and improve motor function of HtrA2 knockout mice. PLoS One 2011;6: e28855.
    • (2011) PLoS One , vol.6
    • Gerhardt, E.1    Gräber, S.2    Szego, E.M.3
  • 180
    • 62849096879 scopus 로고    scopus 로고
    • Nicotinic receptor stimulation protects nigral dopaminergic neurons in rotenone-induced Parkinson's disease models
    • Takeuchi H, Yanagida T, Inden M, et al. Nicotinic receptor stimulation protects nigral dopaminergic neurons in rotenone-induced Parkinson's disease models. J Neurosci Res. 2009;87: 576-585.
    • (2009) J Neurosci Res , vol.87 , pp. 576-585
    • Takeuchi, H.1    Yanagida, T.2    Inden, M.3
  • 181
    • 79960171961 scopus 로고    scopus 로고
    • Effects of pioglitazone and retinoic acid in a rotenone model of Parkinson's disease
    • Ulusoy GK, Celik T, Kayir H, Gürsoy M, Isik AT, Uzbay TI. Effects of pioglitazone and retinoic acid in a rotenone model of Parkinson's disease. Brain Res Bull 2011;85: 380-384.
    • (2011) Brain Res Bull , vol.85 , pp. 380-384
    • Ulusoy, G.K.1    Celik, T.2    Kayir, H.3    Gürsoy, M.4    Isik, A.T.5    Uzbay, T.I.6
  • 182
    • 84879052027 scopus 로고    scopus 로고
    • A blood-brain barrier (BBB) disrupter is Also a potent α-synuclein (α-syn) aggregation inhibitor: a novel dual mechanism of mannitol for the treatment of Parkinson disease (PD)
    • Shaltiel-Karyo R, Frenkel-Pinter M, Rockenstein E, et al. A blood-brain barrier (BBB) disrupter is Also a potent α-synuclein (α-syn) aggregation inhibitor: a novel dual mechanism of mannitol for the treatment of Parkinson disease (PD). J Biol Chem 2013;288: 17579-17588.
    • (2013) J Biol Chem , vol.288 , pp. 17579-17588
    • Shaltiel-Karyo, R.1    Frenkel-Pinter, M.2    Rockenstein, E.3
  • 183
    • 77249085779 scopus 로고    scopus 로고
    • Metabotropic glutamate 7 receptor subtype modulates motor symptoms in rodent models of Parkinson's disease
    • Greco B, Lopez S, van der Putten H, et al. Metabotropic glutamate 7 receptor subtype modulates motor symptoms in rodent models of Parkinson's disease. J Pharmacol Exp Ther 2010;332: 1064-1071.
    • (2010) J Pharmacol Exp Ther , vol.332 , pp. 1064-1071
    • Greco, B.1    Lopez, S.2    van der Putten, H.3
  • 184
    • 54349110319 scopus 로고    scopus 로고
    • Discovery, characterization, and antiparkinsonian effect of novel positive allosteric molulators of metabotropic glutamate receptor 4
    • Niswender CM, Johnson KA, Weaver CD, et al. Discovery, characterization, and antiparkinsonian effect of novel positive allosteric molulators of metabotropic glutamate receptor 4. Mol Pharmacol 2008;74: 1345-1358.
    • (2008) Mol Pharmacol , vol.74 , pp. 1345-1358
    • Niswender, C.M.1    Johnson, K.A.2    Weaver, C.D.3
  • 185
    • 77954340961 scopus 로고    scopus 로고
    • Symptomatic and neuroprotective effects following activation of nigral group III metabotropic glutamate receptors in rodent models of Parkinson's disease
    • Austin PJ, Betts MJ, Broadstock M, et al. Symptomatic and neuroprotective effects following activation of nigral group III metabotropic glutamate receptors in rodent models of Parkinson's disease. Br J Pharmacol 2010;160: 1741-1753.
    • (2010) Br J Pharmacol , vol.160 , pp. 1741-1753
    • Austin, P.J.1    Betts, M.J.2    Broadstock, M.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.