메뉴 건너뛰기




Volumn 8, Issue DEC, 2014, Pages

Parkinson’s disease: Animal models and dopaminergic cell vulnerability

Author keywords

6 OHDA; ATP13A2; DJ1; LRRK2; MPTP; Parkin; Rotenone; Synuclein

Indexed keywords

1,2,3,6 TETRAHYDRO 1 METHYL 4 PHENYLPYRIDINE; ADENOSINE TRIPHOSPHATASE; ADENOSINE TRIPHOSPHATASE 13A2; ALPHA SYNUCLEIN; AUTOPHAGY PROTEIN 7; DJ 1 PROTEIN; ENGRAILED 1 PROTEIN; IMMUNOGLOBULIN ENHANCER BINDING PROTEIN; LEUCINE RICH REPEAT KINASE 2; MANEB; NUCLEAR RECEPTOR RELATED FACTOR 1; OXIDOPAMINE; PARAQUAT; PARKIN; PEPTIDES AND PROTEINS; PSYCHOSTIMULANT AGENT; ROTENONE; SONIC HEDGEHOG PROTEIN; TRANSCRIPTION FACTOR PITX3; UNCLASSIFIED DRUG; VESICULAR MONOAMINE TRANSPORTER 2;

EID: 84918784944     PISSN: None     EISSN: 16625129     Source Type: Journal    
DOI: 10.3389/fnana.2014.00155     Document Type: Article
Times cited : (381)

References (202)
  • 1
    • 79251543343 scopus 로고    scopus 로고
    • Increased mitochondrial calcium sensitivity and abnormal expression of innate immunity genes precede dopaminergic defects in Pink1-deficient mice
    • Akundi, R. S., Huang, Z., Eason, J., Pandya, J. D., Zhi, L., Cass, W. A., et al. (2011). Increased mitochondrial calcium sensitivity and abnormal expression of innate immunity genes precede dopaminergic defects in Pink1-deficient mice. PLoS ONE6:e16038. doi: 10.1371/journal.pone.0016038
    • (2011) Plos ONE , vol.6
    • Akundi, R.S.1    Huang, Z.2    Eason, J.3    Pandya, J.D.4    Zhi, L.5    Cass, W.A.6
  • 2
    • 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., and Schmidt, W. J. (2004). The neurobehavioral changes induced by bilateral rotenone lesion in medial forebrain bundle of rats are reversed by L-DOPA. Behav. Brain Res.151, 117–124. doi: 10.1016/j.bbr.2003.08.014
    • (2004) Behav. Brain Res. , vol.151 , pp. 117-124
    • Alam, M.1    Mayerhofer, A.2    Schmidt, W.J.3
  • 4
    • 84866377694 scopus 로고    scopus 로고
    • Late-onset Parkinsonism in NFκB/c-Rel-deficient mice
    • Baiguera, C., Alghisi, M., Pinna, A., Bellucci, A., De Luca, M. A., Frau, L., et al. (2012). Late-onset Parkinsonism in NFκB/c-Rel-deficient mice. Brain135, 2750–2765. doi: 10.1093/brain/aws193
    • (2012) Brain , vol.135 , pp. 2750-2765
    • Baiguera, C.1    Alghisi, M.2    Pinna, A.3    Bellucci, A.4    De Luca, M.A.5    Frau, L.6
  • 5
    • 84893527711 scopus 로고    scopus 로고
    • Loss of leucine-rich repeat kinase 2 (LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs
    • Baptista, M. A. S., Dave, K. D., Frasier, M. A., Sherer, T. B., Greeley, M., Beck, M. J., et al. (2013). Loss of leucine-rich repeat kinase 2 (LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs. PLoS ONE8:e80705. doi: 10.1371/journal.pone.0080705
    • (2013) Plos ONE , vol.8
    • Baptista, M.1    Dave, K.D.2    Frasier, M.A.3    Sherer, T.B.4    Greeley, M.5    Beck, M.J.6
  • 6
    • 84929516259 scopus 로고    scopus 로고
    • Combined exposure to agriculture pesticides, paraquat and maneb, induces alterations in the N/OFQ-NOPr and PDYN/KOPr systems in rats: Relevance to sporadic Parkinson’s disease
    • [Epub ahead of print]
    • Bastías-Candia, S., Di Benedetto, M., D’Addario, C., Candeletti, S., and Romualdi, P. (2013). Combined exposure to agriculture pesticides, paraquat and maneb, induces alterations in the N/OFQ-NOPr and PDYN/KOPr systems in rats: relevance to sporadic Parkinson’s disease. Environ. Toxicol. doi: 10.1002/tox.21943 [Epub ahead of print].
    • (2013) Environ. Toxicol
    • Bastías-Candia, S.1    Di Benedetto, M.2    D’Addario, C.3    Candeletti, S.4    Romualdi, P.5
  • 7
    • 84939889293 scopus 로고    scopus 로고
    • Human adipose-derived mesenchymal stem cells improve motor functions and are neuroprotective in the 6-hydroxydopamine-rat model for parkinson’s disease when cultured in monolayer cultures but suppress hippocampal neurogenesis and hippocampal memory functi
    • [Epub ahead of print]
    • Berg, J., Roch, M., Altschüler, J., Winter, C., Schwerk, A., Kurtz, A., et al. (2014). Human adipose-derived mesenchymal stem cells improve motor functions and are neuroprotective in the 6-hydroxydopamine-rat model for parkinson’s disease when cultured in monolayer cultures but suppress hippocampal neurogenesis and hippocampal memory functi. Stem Cell Rev. Rep. doi: 10.1007/s12015-014-9551-y [Epub ahead of print].
    • (2014) Stem Cell Rev. Rep
    • Berg, J.1    Roch, M.2    Altschüler, J.3    Winter, C.4    Schwerk, A.5    Kurtz, A.6
  • 8
    • 77953611763 scopus 로고    scopus 로고
    • Paraquat and Parkinson’s disease
    • Berry, C., La Vecchia, C., and Nicotera, P. (2010). Paraquat and Parkinson’s disease. Cell Death Differ. 17, 1115–1125. doi: 10.1038/cdd.2009.217
    • (2010) Cell Death Differ , vol.17 , pp. 1115-1125
    • Berry, C.1    La Vecchia, C.2    Nicotera, P.3
  • 10
    • 0030984413 scopus 로고    scopus 로고
    • A chronic MPTP model reproducing the slow evolution of Parkinson’s disease: Evolution of motor symptoms in the monkey
    • Bezard, E., Imbert, C., Deloire, X., Bioulac, B., and Gross, C. E. (1997). A chronic MPTP model reproducing the slow evolution of Parkinson’s disease: evolution of motor symptoms in the monkey. Brain Res. 766, 107–112. doi: 10.1016/S0006-8993(97)00531-3
    • (1997) Brain Res , vol.766 , pp. 107-112
    • Bezard, E.1    Imbert, C.2    Deloire, X.3    Bioulac, B.4    Gross, C.E.5
  • 11
    • 47049116273 scopus 로고    scopus 로고
    • The 6-hydroxydopamine model: News from the past
    • Blandini, F., Armentero, M.-T., and Martignoni, E. (2008). The 6-hydroxydopamine model: news from the past. Parkinsonism Relat. Disord.14(Suppl. 2), S124–S129. doi: 10.1016/j.parkreldis.2008.04.015
    • (2008) Parkinsonism Relat. Disord. , vol.14 , pp. S124-S129
    • Blandini, F.1    Armentero, M.-T.2    Martignoni, E.3
  • 12
    • 77952541246 scopus 로고    scopus 로고
    • Progression of dopaminergic depletion in a model of MPTP-induced Parkin-sonism in non-human primates. An (18)F-DOPA and (11)C-DTBZ PET study
    • Blesa, J., Juri, C., Collantes, M., Peñuelas, I., Prieto, E., Iglesias, E., et al. (2010). Progression of dopaminergic depletion in a model of MPTP-induced Parkin-sonism in non-human primates. An (18)F-DOPA and (11)C-DTBZ PET study. Neurobiol. Dis. 38, 456–463. doi: 10.1016/j.nbd.2010.03.006
    • (2010) Neurobiol. Dis , vol.38 , pp. 456-463
    • Blesa, J.1    Juri, C.2    Collantes, M.3    Peñuelas, I.4    Prieto, E.5    Iglesias, E.6
  • 13
    • 84856195961 scopus 로고    scopus 로고
    • Inter-hemispheric asymmetry of nigrostriatal dopamin-ergic lesion: A possible compensatory mechanism in Parkinson’s disease
    • Blesa, J., Juri, C., Garcia-Cabezas, M. A., Adanez, R., Sanchez-Gonzalez, M. A., Cavada, C., et al. (2011). Inter-hemispheric asymmetry of nigrostriatal dopamin-ergic lesion: a possible compensatory mechanism in Parkinson’s disease. Front. Syst. Neurosci.5:92. doi: 10.3389/fnsys.2011.00092
    • (2011) Front. Syst. Neurosci. , vol.5 , pp. 92
    • Blesa, J.1    Juri, C.2    Garcia-Cabezas, M.A.3    Adanez, R.4    Sanchez-Gonzalez, M.A.5    Cavada, C.6
  • 14
    • 84863760756 scopus 로고    scopus 로고
    • The nigrostriatal systeminthe presymptomatic and symptomatic stages in the MPTP monkey model: A PET, histological and biochemical study
    • Blesa, J., Pifl, C., Sánchez-González, M. A., Juri, C., García-Cabezas, M. A., Adánez, R., et al. (2012). The nigrostriatal systeminthe presymptomatic and symptomatic stages in the MPTP monkey model: a PET, histological and biochemical study. Neurobiol. Dis.48, 79–91. doi: 10.1016/j.nbd.2012.05.018
    • (2012) Neurobiol. Dis. , vol.48 , pp. 79-91
    • Blesa, J.1    Pifl, C.2    Sánchez-González, M.A.3    Juri, C.4    García-Cabezas, M.A.5    Adánez, R.6
  • 15
    • 84942828573 scopus 로고    scopus 로고
    • A genetic mouse model of Parkinson’s disease shows involuntary movements and increased postsynaptic sensitivity to apomorphine
    • [Epub ahead of print]
    • Brehm, N., Bez, F., Carlsson, T., Kern, B., Gispert, S., Auburger, G., et al. (2014). A genetic mouse model of Parkinson’s disease shows involuntary movements and increased postsynaptic sensitivity to apomorphine. Mol. Neurobiol. doi: 10.1007/s12035-014-8911–8916 [Epub ahead of print].
    • (2014) Mol. Neurobiol
    • Brehm, N.1    Bez, F.2    Carlsson, T.3    Kern, B.4    Gispert, S.5    Auburger, G.6
  • 16
    • 84883828888 scopus 로고    scopus 로고
    • Advances in the pharmacological treatment of Parkinson’s disease: Targeting neurotransmitter systems
    • Brichta, L., Greengard, P., and Flajolet, M. (2013). Advances in the pharmacological treatment of Parkinson’s disease: targeting neurotransmitter systems. Trends Neurosci. 36, 543–554. doi: 10.1016/j.tins.2013.06.003
    • (2013) Trends Neurosci , vol.36 , pp. 543-554
    • Brichta, L.1    Greengard, P.2    Flajolet, M.3
  • 17
    • 0033608811 scopus 로고    scopus 로고
    • Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss
    • Brooks, A. I., Chadwick, C. A., Gelbard, H. A., Cory-Slechta, D. A., and Federoff, H. J. (1999). Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Res. 823, 1–10. doi: 10.1016/S0006-8993(98)01192-5
    • (1999) Brain Res , vol.823 , pp. 1-10
    • Brooks, A.I.1    Chadwick, C.A.2    Gelbard, H.A.3    Cory-Slechta, D.A.4    Federoff, H.J.5
  • 18
    • 34548405973 scopus 로고    scopus 로고
    • Neurotoxicity of substituted amphetamines: Molecular and cellular mechanisms
    • Cadet, J. L., Krasnova, I. N., Jayanthi, S., and Lyles, J. (2007). Neurotoxicity of substituted amphetamines: molecular and cellular mechanisms. Neurotox. Res.11, 183–202. doi: 10.1007/BF03033567
    • (2007) Neurotox. Res. , vol.11 , pp. 183-202
    • Cadet, J.L.1    Krasnova, I.N.2    Jayanthi, S.3    Lyles, J.4
  • 21
    • 69449095628 scopus 로고    scopus 로고
    • Molecular and cellular mechanisms of ecstasy-induced neurotoxicity: An overview
    • Capela, J. P., Carmo, H., Remião, F., Bastos, M. L., Meisel, A., and Carvalho, F. (2009). Molecular and cellular mechanisms of ecstasy-induced neurotoxicity: an overview. Mol. Neurobiol. 39, 210–271. doi: 10.1007/s12035-009-8064–8061
    • (2009) Mol. Neurobiol , vol.39 , pp. 210-271
    • Capela, J.P.1    Carmo, H.2    Remião, F.3    Bastos, M.L.4    Meisel, A.5    Carvalho, F.6
  • 22
    • 84901314467 scopus 로고    scopus 로고
    • Caenorhabditis elegans
    • Chege, P. M., and McColl, G. (2014). Caenorhabditis elegans: a model to investigate oxidative stress and metal dyshomeostasis in Parkinson’s disease. Front. Aging Neurosci. 6:89. doi: 10.3389/fnagi.2014.00089
    • (2014) Front. Aging Neurosci. , vol.6 , pp. 89
    • Chege, P.M.1    McColl, G.2
  • 23
    • 84866065925 scopus 로고    scopus 로고
    • (G2019S) LRRK2 activates MKK4-JNK pathway and causes degeneration of SN dopaminergic neurons in a transgenic mouse model of PD
    • Chen, C.-Y., Weng, Y.-H., Chien, K.-Y., Lin, K.-J., Yeh, T.-H., Cheng, Y.-P., et al. (2012). (G2019S) LRRK2 activates MKK4-JNK pathway and causes degeneration of SN dopaminergic neurons in a transgenic mouse model of PD. Cell Death Differ. 19, 1623–1633. doi: 10.1038/cdd.2012.42
    • (2012) Cell Death Differ , vol.19 , pp. 1623-1633
    • Chen, C.-Y.1    Weng, Y.-H.2    Chien, K.-Y.3    Lin, K.-J.4    Yeh, T.-H.5    Cheng, Y.-P.6
  • 24
    • 33745140532 scopus 로고    scopus 로고
    • Proteasome dysfunction in aged human alpha-synuclein transgenic mice
    • Chen, L., Thiruchelvam, M. J., Madura, K., and Richfield, E. K. (2006). Proteasome dysfunction in aged human alpha-synuclein transgenic mice. Neurobiol. Dis.23, 120–126. doi: 10.1016/j.nbd.2006.02.004
    • (2006) Neurobiol. Dis. , vol.23 , pp. 120-126
    • Chen, L.1    Thiruchelvam, M.J.2    Madura, K.3    Richfield, E.K.4
  • 25
    • 0021259475 scopus 로고
    • Neurochemical and behavioral effects of 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) in rat, guinea pig, and monkey
    • Chiueh, C. C., Markey, S. P., Burns, R. S., Johannessen, J. N., Jacobowitz, D. M., and Kopin, I. J. (1984). Neurochemical and behavioral effects of 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP) in rat, guinea pig, and monkey. Psychopharmacol. Bull. 20, 548–553.
    • (1984) Psychopharmacol. Bull , vol.20 , pp. 548-553
    • Chiueh, C.C.1    Markey, S.P.2    Burns, R.S.3    Johannessen, J.N.4    Jacobowitz, D.M.5    Kopin, I.J.6
  • 26
    • 84889646696 scopus 로고    scopus 로고
    • MPTP-induced dopamine neuron degeneration and glia activation is potentiated in MDMA-pretreated mice
    • Costa, G., Frau, L., Wardas, J., Pinna, A., Plumitallo, A., and Morelli, M. (2013). MPTP-induced dopamine neuron degeneration and glia activation is potentiated in MDMA-pretreated mice. Mov. Disord.28, 1957–1965. doi: 10.1002/mds.25646
    • (2013) Mov. Disord. , vol.28 , pp. 1957-1965
    • Costa, G.1    Frau, L.2    Wardas, J.3    Pinna, A.4    Plumitallo, A.5    Morelli, M.6
  • 27
    • 84905257556 scopus 로고    scopus 로고
    • The ubiquitin-proteasome system in neu-rodegenerative diseases: Precipitating factor, yet part of the solution
    • Dantuma, N. P., and Bott, L. C. (2014). The ubiquitin-proteasome system in neu-rodegenerative diseases: precipitating factor, yet part of the solution. Front. Mol. Neurosci.7:70. doi: 10.3389/fnmol.2014.00070
    • (2014) Front. Mol. Neurosci. , vol.7 , pp. 70
    • Dantuma, N.P.1    Bott, L.C.2
  • 29
    • 0141741347 scopus 로고    scopus 로고
    • Parkinson’s disease: Mechanisms and models
    • Dauer, W., and Przedborski, S. (2003). Parkinson’s disease: mechanisms and models. Neuron39, 889–909. doi: 10.1016/S0896-6273(03)00568-3
    • (2003) Neuron , vol.39 , pp. 889-909
    • Dauer, W.1    Przedborski, S.2
  • 30
    • 77953666105 scopus 로고    scopus 로고
    • Genetic animal models of Parkinson’s disease
    • Dawson, T. M., Ko, H. S., and Dawson, V. L. (2010). Genetic animal models of Parkinson’s disease. Neuron66, 646–661. doi: 10.1016/j.neuron.2010.04.034
    • (2010) Neuron , vol.66 , pp. 646-661
    • Dawson, T.M.1    Ko, H.S.2    Dawson, V.L.3
  • 31
    • 0033607235 scopus 로고    scopus 로고
    • A mechanism of paraquat toxicity involving nitric oxide synthase
    • Day, B. J., Patel, M., Calavetta, L., Chang, L. Y., and Stamler, J. S. (1999). A mechanism of paraquat toxicity involving nitric oxide synthase. Proc. Natl. Acad. Sci. U.S.A.96, 12760–12765. doi: 10.1073/pnas.96.22.12760
    • (1999) Proc. Natl. Acad. Sci. U.S.A. , vol.96 , pp. 12760-12765
    • Day, B.J.1    Patel, M.2    Calavetta, L.3    Chang, L.Y.4    Stamler, J.S.5
  • 32
    • 84856570373 scopus 로고    scopus 로고
    • Progressive neurodegenerative and behavioural changes induced by AAV-mediated overexpression of α-synuclein in midbrain dopamine neurons
    • Decressac, M., Mattsson, B., Lundblad, M., Weikop, P., and Björklund, A. (2012). Progressive neurodegenerative and behavioural changes induced by AAV-mediated overexpression of α-synuclein in midbrain dopamine neurons. Neurobiol. Dis.45, 939–953. doi: 10.1016/j.nbd.2011.12.013
    • (2012) Neurobiol. Dis. , vol.45 , pp. 939-953
    • Decressac, M.1    Mattsson, B.2    Lundblad, M.3    Weikop, P.4    Björklund, A.5
  • 33
    • 0036787779 scopus 로고    scopus 로고
    • Overexpression of Parkinson’s disease-associated alpha-synucleinA53T by recombinant adeno-associated virus in mice does not increase the vulnerability of dopaminergic neurons to MPTP
    • Dong, Z., Ferger, B., Feldon, J., and Büeler, H. (2002). Overexpression of Parkinson’s disease-associated alpha-synucleinA53T by recombinant adeno-associated virus in mice does not increase the vulnerability of dopaminergic neurons to MPTP. J. Neurobiol.53, 1–10. doi: 10.1002/neu.10094
    • (2002) J. Neurobiol. , vol.53 , pp. 1-10
    • Dong, Z.1    Ferger, B.2    Feldon, J.3    Büeler, H.4
  • 34
    • 84894544413 scopus 로고    scopus 로고
    • Loss of dopaminergic nigrostriatal neurons accounts for the motivational and affective deficits in Parkinson’s disease
    • Drui, G., Carnicella, S., Carcenac, C., Favier, M., Bertrand, A., Boulet, S., et al. (2014). Loss of dopaminergic nigrostriatal neurons accounts for the motivational and affective deficits in Parkinson’s disease. Mol. Psychiatry19, 358–367. doi: 10.1038/mp.2013.3
    • (2014) Mol. Psychiatry , vol.19 , pp. 358-367
    • Drui, G.1    Carnicella, S.2    Carcenac, C.3    Favier, M.4    Bertrand, A.5    Boulet, S.6
  • 35
    • 77955383138 scopus 로고    scopus 로고
    • Behavioral analysis of motor and non-motor symptoms in rodent models of Parkinson’s disease
    • Dunnett, S. B., and Lelos, M. (2010). Behavioral analysis of motor and non-motor symptoms in rodent models of Parkinson’s disease. Prog. Brain Res.184, 35–51. doi: 10.1016/S0079-6123(10)840038
    • (2010) Prog. Brain Res. , vol.184 , pp. 35-51
    • Dunnett, S.B.1    Lelos, M.2
  • 36
    • 78751522558 scopus 로고    scopus 로고
    • A rat model of progressive nigral neurodegeneration induced by the Parkinson’s disease-associated G2019S mutation in LRRK2
    • Dusonchet, J., Kochubey, O., Stafa, K., Young, S. M., Zufferey, R., Moore, D. J., et al. (2011). A rat model of progressive nigral neurodegeneration induced by the Parkinson’s disease-associated G2019S mutation in LRRK2. J. Neurosci.31, 907–912. doi: 10.1523/JNEUROSCI.5092-10.2011
    • (2011) J. Neurosci. , vol.31 , pp. 907-912
    • Dusonchet, J.1    Kochubey, O.2    Stafa, K.3    Young, S.M.4    Zufferey, R.5    Moore, D.J.6
  • 38
    • 80053937625 scopus 로고    scopus 로고
    • Low concentrations of methamphetamine can protect dopaminergic cells against a larger oxidative stress injury: Mechanistic study
    • El Ayadi, A., and Zigmond, M. J. (2011). Low concentrations of methamphetamine can protect dopaminergic cells against a larger oxidative stress injury: mechanistic study. PLoS ONE6:e24722. doi: 10.1371/journal.pone.0024722
    • (2011) Plos ONE , vol.6
    • El Ayadi, A.1    Zigmond, M.J.2
  • 39
    • 84864150600 scopus 로고    scopus 로고
    • Mitochondrial dysfunction in Parkinson’s disease: Molecular mechanisms and pathophysiological consequences
    • Exner, N., Lutz, A. K., Haass, C., and Winklhofer, K. F. (2012). Mitochondrial dysfunction in Parkinson’s disease: molecular mechanisms and pathophysiological consequences. EMBO J. 31, 3038–3062. doi: 10.1038/emboj.2012.170
    • (2012) EMBO J , vol.31 , pp. 3038-3062
    • Exner, N.1    Lutz, A.K.2    Haass, C.3    Winklhofer, K.F.4
  • 40
    • 2342662120 scopus 로고    scopus 로고
    • Behavioral and immunohistochemical effects of chronic intravenous and subcutaneous infusions of varying doses of rotenone
    • Fleming, S. M., Zhu, C., Fernagut, P.-O., Mehta, A., DiCarlo, C. D., Seaman, R. L., et al. (2004). Behavioral and immunohistochemical effects of chronic intravenous and subcutaneous infusions of varying doses of rotenone. Exp. Neurol.187, 418–429. doi: 10.1016/j.expneurol.2004.01.023
    • (2004) Exp. Neurol. , vol.187 , pp. 418-429
    • Fleming, S.M.1    Zhu, C.2    Fernagut, P.-O.3    Mehta, A.4    Dicarlo, C.D.5    Seaman, R.L.6
  • 41
    • 0022466381 scopus 로고
    • Locus ceruleus lesions and eosinophilic inclusions in MPTP-treated monkeys
    • Forno, L. S., Langston, J. W., DeLanney, L. E., Irwin, I., and Ricaurte, G. A. (1986). Locus ceruleus lesions and eosinophilic inclusions in MPTP-treated monkeys. Ann. Neurol.20, 449–455. doi: 10.1002/ana.410200403
    • (1986) Ann. Neurol. , vol.20 , pp. 449-455
    • Forno, L.S.1    Langston, J.W.2    Delanney, L.E.3    Irwin, I.4    Ricaurte, G.A.5
  • 42
    • 84867230531 scopus 로고    scopus 로고
    • Pesticide exposure and Parkinson’s disease: Epidemiological evidence of association
    • Freire, C., and Koifman, S. (2012). Pesticide exposure and Parkinson’s disease: epidemiological evidence of association. Neurotoxicology33, 947–971. doi: 10.1016/j.neuro.2012.05.011
    • (2012) Neurotoxicology , vol.33 , pp. 947-971
    • Freire, C.1    Koifman, S.2
  • 43
    • 84861595545 scopus 로고    scopus 로고
    • Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation of α-synuclein and LRRK2 in the brain
    • Friedman, L. G., Lachenmayer, M. L., Wang, J., He, L., Poulose, S. M., Komatsu, M., et al. (2012). Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation of α-synuclein and LRRK2 in the brain. J. Neurosci.32, 7585–7593. doi: 10.1523/JNEUROSCI.5809-11.2012
    • (2012) J. Neurosci. , vol.32 , pp. 7585-7593
    • Friedman, L.G.1    Lachenmayer, M.L.2    Wang, J.3    He, L.4    Poulose, S.M.5    Komatsu, M.6
  • 44
    • 61649088435 scopus 로고    scopus 로고
    • PINK1-associated Parkinson’s disease is caused by neu-ronal vulnerability to calcium-induced cell death
    • Gandhi, S., Wood-Kaczmar, A., Yao, Z., Plun-Favreau, H., Deas, E., Klupsch, K., et al. (2009). PINK1-associated Parkinson’s disease is caused by neu-ronal vulnerability to calcium-induced cell death. Mol. Cell33, 627–638. doi: 10.1016/j.molcel.2009.02.013
    • (2009) Mol. Cell , vol.33 , pp. 627-638
    • Gandhi, S.1    Wood-Kaczmar, A.2    Yao, Z.3    Plun-Favreau, H.4    Deas, E.5    Klupsch, K.6
  • 45
    • 49649097747 scopus 로고    scopus 로고
    • Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress
    • Gautier, C. A., Kitada, T., and Shen, J. (2008). Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress. Proc. Natl. Acad. Sci. U.S.A.105, 11364–11369. doi: 10.1073/pnas.0802076105
    • (2008) Proc. Natl. Acad. Sci. U.S.A. , vol.105 , pp. 11364-11369
    • Gautier, C.A.1    Kitada, T.2    Shen, J.3
  • 46
    • 0037118259 scopus 로고    scopus 로고
    • Neuronal alpha-synucleinopathy with severe movement disorder in mice expressing A53T human alpha-synuclein
    • Giasson, B. I., Duda, J. E., Quinn, S. M., Zhang, B., Trojanowski, J. Q., and Lee, V. M. (2002). Neuronal alpha-synucleinopathy with severe movement disorder in mice expressing A53T human alpha-synuclein. Neuron34, 521–533. doi: 10.1016/S0896-6273(02)00682-7
    • (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.6
  • 47
    • 10744221260 scopus 로고    scopus 로고
    • Transgenic mice expressing mutant A53T human alpha-synuclein show neuronal dysfunction in the absence of aggregate formation
    • Gispert, S., Del Turco, D., Garrett, L., Chen, A., Bernard, D. J., Hamm-Clement, J., et al. (2003). Transgenic mice expressing mutant A53T human alpha-synuclein show neuronal dysfunction in the absence of aggregate formation. Mol. Cell. Neurosci.24, 419–429. doi: 10.1016/S1044-7431(03)00198-2
    • (2003) Mol. Cell. Neurosci. , vol.24 , pp. 419-429
    • Gispert, S.1    Del Turco, D.2    Garrett, L.3    Chen, A.4    Bernard, D.J.5    Hamm-Clement, J.6
  • 48
    • 66749163493 scopus 로고    scopus 로고
    • Parkinson phenotype in aged PINK1-deficient mice is accompanied by progressive mitochondrial dysfunction in absence of neurodegeneration
    • Gispert, S., Ricciardi, F., Kurz, A., Azizov, M., Hoepken, H.-H., Becker, D., et al. (2009). Parkinson phenotype in aged PINK1-deficient mice is accompanied by progressive mitochondrial dysfunction in absence of neurodegeneration. PLoS ONE4:e5777. doi: 10.1371/journal.pone.0005777
    • (2009) Plos ONE , vol.4
    • Gispert, S.1    Ricciardi, F.2    Kurz, A.3    Azizov, M.4    Hoepken, H.-H.5    Becker, D.6
  • 49
    • 84859264837 scopus 로고    scopus 로고
    • Sensorimotor assessment of the unilateral 6-hydroxydopamine mouse model of Parkinson’s disease
    • Glajch, K. E., Fleming, S. M., Surmeier, D. J., and Osten, P. (2012). Sensorimotor assessment of the unilateral 6-hydroxydopamine mouse model of Parkinson’s disease. Behav. Brain Res.230, 309–316. doi: 10.1016/j.bbr.2011.12.007
    • (2012) Behav. Brain Res. , vol.230 , pp. 309-316
    • Glajch, K.E.1    Fleming, S.M.2    Surmeier, D.J.3    Osten, P.4
  • 51
    • 0141891953 scopus 로고    scopus 로고
    • Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons
    • Goldberg, M. S., Fleming, S. M., Palacino, J. J., Cepeda, C., Lam, H. A., Bhat-nagar, A., et al. (2003). Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons. J. Biol. Chem.278, 43628–43635. doi: 10.1074/jbc.M308947200
    • (2003) J. Biol. Chem. , vol.278 , pp. 43628-43635
    • Goldberg, M.S.1    Fleming, S.M.2    Palacino, J.J.3    Cepeda, C.4    Lam, H.A.5    Bhat-Nagar, A.6
  • 52
    • 13844253723 scopus 로고    scopus 로고
    • Nigrostriatal dopaminergic deficits and hypokinesia caused by inactivation of the familial Parkinsonism-linked gene DJ-1
    • Goldberg, M. S., Pisani, A., Haburcak, M., Vortherms, T. A., Kitada, T., Costa, C., et al. (2005). Nigrostriatal dopaminergic deficits and hypokinesia caused by inactivation of the familial Parkinsonism-linked gene DJ-1. Neuron45, 489–496. doi: 10.1016/j.neuron.2005.01.041
    • (2005) Neuron , vol.45 , pp. 489-496
    • Goldberg, M.S.1    Pisani, A.2    Haburcak, M.3    Vortherms, T.A.4    Kitada, T.5    Costa, C.6
  • 53
    • 0037333642 scopus 로고    scopus 로고
    • Motor dysfunction and gliosis with preserved dopaminergic markers in human alpha-synuclein A30P transgenic mice
    • Gomez-Isla, T., Irizarry, M. C., Mariash, A., Cheung, B., Soto, O., Schrump, S., et al. (2003). Motor dysfunction and gliosis with preserved dopaminergic markers in human alpha-synuclein A30P transgenic mice. Neurobiol. Aging24, 245–258. doi: 10.1016/S0197-4580(02)00091-X
    • (2003) Neurobiol. Aging , vol.24 , pp. 245-258
    • Gomez-Isla, T.1    Irizarry, M.C.2    Mariash, A.3    Cheung, B.4    Soto, O.5    Schrump, S.6
  • 54
    • 84864307761 scopus 로고    scopus 로고
    • Sonic hedgehog maintains cellular and neurochem-ical homeostasis in the adult nigrostriatal circuit
    • Gonzalez-Reyes, L. E., Verbitsky, M., Blesa, J., Jackson-Lewis, V., Paredes, D., Tillack, K., et al. (2012). Sonic hedgehog maintains cellular and neurochem-ical homeostasis in the adult nigrostriatal circuit. Neuron75, 306–319. doi: 10.1016/j.neuron.2012.05.018
    • (2012) Neuron , vol.75 , pp. 306-319
    • Gonzalez-Reyes, L.E.1    Verbitsky, M.2    Blesa, J.3    Jackson-Lewis, V.4    Paredes, D.5    Tillack, K.6
  • 55
    • 79954599799 scopus 로고    scopus 로고
    • Impaired nigrostriatal function precedes behavioral deficits in a genetic mitochondrial model of Parkinson’s disease
    • Good, C. H., Hoffman, A. F., Hoffer, B. J., Chefer, V. I., Shippenberg, T. S., Bäckman, C. M., et al. (2011). Impaired nigrostriatal function precedes behavioral deficits in a genetic mitochondrial model of Parkinson’s disease. FASEB J. 25, 1333–1344. doi: 10.1096/fj.10–173625
    • (2011) FASEB J , vol.25 , pp. 1333-1344
    • Good, C.H.1    Hoffman, A.F.2    Hoffer, B.J.3    Chefer, V.I.4    Shippenberg, T.S.5    Bäckman, C.M.6
  • 56
    • 36348951506 scopus 로고    scopus 로고
    • Early loss of dopaminergic terminals in striosomes after MDMA administration to mice
    • Granado, N., Escobedo, I., O’Shea, E., Colado, I., and Moratalla, R. (2008a). Early loss of dopaminergic terminals in striosomes after MDMA administration to mice. Synapse62, 80–84. doi: 10.1002/syn.20466
    • (2008) Synapse , vol.62 , pp. 80-84
    • Granado, N.1    Escobedo, I.2    O’Shea, E.3    Colado, I.4    Moratalla, R.5
  • 57
    • 54349116574 scopus 로고    scopus 로고
    • Persistent MDMA-induced dopaminergic neurotoxicity in the striatum and substantia nigra of mice
    • Granado, N., O’Shea, E., Bove, J., Vila, M., Colado, M. I., and Moratalla, R. (2008b). Persistent MDMA-induced dopaminergic neurotoxicity in the striatum and substantia nigra of mice. J. Neurochem.107, 1102–1112. doi: 10.1111/j.1471-4159.2008.05705.x
    • (2008) J. Neurochem. , vol.107 , pp. 1102-1112
    • Granado, N.1    O’Shea, E.2    Bove, J.3    Vila, M.4    Colado, M.I.5    Moratalla, R.6
  • 58
    • 84886715175 scopus 로고    scopus 로고
    • Drosophila
    • Guo, M. (2012). Drosophila as a model to study mitochondrial dysfunction in Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2, pii:a009944. doi: 10.1101/cshperspect.a009944
    • (2012) Cold Spring Harb. Perspect. Med , vol.2
    • Guo, M.1
  • 59
    • 69849092676 scopus 로고    scopus 로고
    • No Lewy pathology in monkeys with over 10 years of severe MPTP Parkinsonism
    • Halliday, G., Herrero, M. T., Murphy, K., McCann, H., Ros-Bernal, F., Barcia, C., et al. (2009). No Lewy pathology in monkeys with over 10 years of severe MPTP Parkinsonism. Mov. Disord.24, 1519–1523. doi: 10.1002/mds.22481
    • (2009) Mov. Disord. , vol.24 , pp. 1519-1523
    • Halliday, G.1    Herrero, M.T.2    Murphy, K.3    McCann, H.4    Ros-Bernal, F.5    Barcia, C.6
  • 60
    • 84863305249 scopus 로고    scopus 로고
    • Inactivation of Pink1 gene in vivo sensitizes dopamine-producing neurons to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and can be rescued by autosomal recessive Parkinson disease genes, Parkin or DJ-1
    • Haque, M. E., Mount, M. P., Safarpour, F., Abdel-Messih, E., Callaghan, S., Mazerolle, C., et al. (2012). Inactivation of Pink1 gene in vivo sensitizes dopamine-producing neurons to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and can be rescued by autosomal recessive Parkinson disease genes, Parkin or DJ-1. J. Biol. Chem.287, 23162–23170. doi: 10.1074/jbc.M112.346437
    • (2012) J. Biol. Chem. , vol.287 , pp. 23162-23170
    • Haque, M.E.1    Mount, M.P.2    Safarpour, F.3    Abdel-Messih, E.4    Callaghan, S.5    Mazerolle, C.6
  • 61
    • 50049104725 scopus 로고    scopus 로고
    • Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: A case-control study
    • Healy, D. G., Falchi, M., O’Sullivan, S. S., Bonifati, V., Durr, A., Bressman, S., et al. (2008). Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: a case-control study. Lancet. Neurol. 7, 583–590. doi: 10.1016/S1474-4422(08)70117–70110
    • (2008) Lancet. Neurol , vol.7 , pp. 583-590
    • Healy, D.G.1    Falchi, M.2    O’Sullivan, S.S.3    Bonifati, V.4    Durr, A.5    Bressman, S.6
  • 63
    • 84885452439 scopus 로고    scopus 로고
    • Surprising behavioral and neurochemical enhancements in mice with combined mutations linked to Parkinson’s disease
    • Hennis, M. R., Marvin, M. A., Taylor, C. M., and Goldberg, M. S. (2014). Surprising behavioral and neurochemical enhancements in mice with combined mutations linked to Parkinson’s disease. Neurobiol. Dis.62, 113–123. doi: 10.1016/j.nbd.2013.09.009
    • (2014) Neurobiol. Dis. , vol.62 , pp. 113-123
    • Hennis, M.R.1    Marvin, M.A.2    Taylor, C.M.3    Goldberg, M.S.4
  • 64
    • 84893547450 scopus 로고    scopus 로고
    • Behavioral and neurotransmitter abnormalities in mice deficient for Parkin, DJ-1 and superoxide dismutase
    • Hennis, M. R., Seamans, K. W., Marvin, M. A., Casey, B. H., and Goldberg, M. S. (2013). Behavioral and neurotransmitter abnormalities in mice deficient for Parkin, DJ-1 and superoxide dismutase. PLoS ONE8:e84894. doi: 10.1371/journal.pone.0084894
    • (2013) Plos ONE , vol.8
    • Hennis, M.R.1    Seamans, K.W.2    Marvin, M.A.3    Casey, B.H.4    Goldberg, M.S.5
  • 65
    • 80053968304 scopus 로고    scopus 로고
    • LRRK2 protein levels are determined by kinase function and are crucial for kidney and lung homeostasis in mice
    • Herzig, M. C., Kolly, C., Persohn, E., Theil, D., Schweizer, T., Hafner, T., et al. (2011). LRRK2 protein levels are determined by kinase function and are crucial for kidney and lung homeostasis in mice. Hum. Mol. Genet.20, 4209–4223. doi: 10.1093/hmg/ddr348
    • (2011) Hum. Mol. Genet. , vol.20 , pp. 4209-4223
    • Herzig, M.C.1    Kolly, C.2    Persohn, E.3    Theil, D.4    Schweizer, T.5    Hafner, T.6
  • 66
    • 0025297803 scopus 로고
    • Acute neuropatho-logical changes in the caudate nucleus caused by MPTP and metham-phetamine: Immunohistochemical studies
    • Hess, A., Desiderio, C., and McAuliffe, W. G. (1990). Acute neuropatho-logical changes in the caudate nucleus caused by MPTP and metham-phetamine: immunohistochemical studies. J. Neurocytol. 19, 338–342. doi: 10.1007/BF01188403
    • (1990) J. Neurocytol , vol.19 , pp. 338-342
    • Hess, A.1    Desiderio, C.2    McAuliffe, W.G.3
  • 67
    • 84861552733 scopus 로고    scopus 로고
    • LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors
    • Hinkle, K. M., Yue, M., Behrouz, B., Dächsel, J. C., Lincoln, S. J., Bowles, E. E., et al. (2012). LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors. Mol. Neurodegener.7:25. doi: 10.1186/1750-1326-725
    • (2012) Mol. Neurodegener. , vol.7 , pp. 25
    • Hinkle, K.M.1    Yue, M.2    Behrouz, B.3    Dächsel, J.C.4    Lincoln, S.J.5    Bowles, E.E.6
  • 68
    • 79960564790 scopus 로고    scopus 로고
    • Methamphetamine neurotoxicity decreases phasic, but not tonic, dopaminergic signaling in the rat striatum
    • Howard, C. D., Keefe, K. A., Garris, P. A., and Daberkow, D. P. (2011). Methamphetamine neurotoxicity decreases phasic, but not tonic, dopaminergic signaling in the rat striatum. J. Neurochem.118, 668–676. doi: 10.1111/j.1471-4159.2011.07342.x
    • (2011) J. Neurochem. , vol.118 , pp. 668-676
    • Howard, C.D.1    Keefe, K.A.2    Garris, P.A.3    Daberkow, D.P.4
  • 69
    • 0037786542 scopus 로고    scopus 로고
    • Selective loss of dopaminergic neurons in the substantia nigra of Pitx3-deficient aphakia mice
    • Hwang, D.-Y., Ardayfio, P., Kang, U. J., Semina, E. V, and Kim, K.-S. (2003). Selective loss of dopaminergic neurons in the substantia nigra of Pitx3-deficient aphakia mice. Brain Res. Mol. Brain Res. 114, 123–131. doi: 10.1016/S0169-328X(03)00162-1
    • (2003) Brain Res. Mol. Brain Res , vol.114 , pp. 123-131
    • Hwang, D.-Y.1    Ardayfio, P.2    Kang, U.J.3    Semina, E.V.4    Kim, K.-S.5
  • 70
    • 58149468381 scopus 로고    scopus 로고
    • Motor impairment and aberrant production of neuro-chemicals in human alpha-synuclein A30P+A53T transgenic mice with alpha-synuclein pathology
    • Ikeda, M., Kawarabayashi, T., Harigaya, Y., Sasaki, A., Yamada, S., Matsubara, E., et al. (2009). Motor impairment and aberrant production of neuro-chemicals in human alpha-synuclein A30P+A53T transgenic mice with alpha-synuclein pathology. Brain Res. 1250, 232–241. doi: 10.1016/j.brainres.2008.10.011
    • (2009) Brain Res , vol.1250 , pp. 232-241
    • Ikeda, M.1    Kawarabayashi, T.2    Harigaya, Y.3    Sasaki, A.4    Yamada, S.5    Matsubara, E.6
  • 71
    • 10744221310 scopus 로고    scopus 로고
    • Parkin gene inactivation alters behaviour and dopamine neurotransmission in the mouse
    • Itier, J.-M., Ibanez, P., Mena, M. A., Abbas, N., Cohen-Salmon, C., Bohme, G. A., et al. (2003). Parkin gene inactivation alters behaviour and dopamine neurotransmission in the mouse. Hum. Mol. Genet.12, 2277–2291. doi: 10.1093/hmg/ddg239
    • (2003) Hum. Mol. Genet. , vol.12 , pp. 2277-2291
    • Itier, J.-M.1    Ibanez, P.2    Mena, M.A.3    Abbas, N.4    Cohen-Salmon, C.5    Bohme, G.A.6
  • 72
    • 34347259269 scopus 로고    scopus 로고
    • Protocol for the MPTP mouse model of Parkinson’s disease
    • Jackson-Lewis, V., and Przedborski, S. (2007). Protocol for the MPTP mouse model of Parkinson’s disease. Nat. Protoc.2, 141–151. doi: 10.1038/nprot.2006.342
    • (2007) Nat. Protoc. , vol.2 , pp. 141-151
    • Jackson-Lewis, V.1    Przedborski, S.2
  • 73
    • 84885743813 scopus 로고    scopus 로고
    • Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model
    • Janezic, S., Threlfell, S., Dodson, P. D., Dowie, M. J., Taylor, T. N., Potgieter, D., et al. (2013). Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model. Proc. Natl. Acad. Sci. U.S.A.110, E4016–E4025. doi: 10.1073/pnas.1309143110
    • (2013) Proc. Natl. Acad. Sci. U.S.A. , vol.110 , pp. E4016-E4025
    • Janezic, S.1    Threlfell, S.2    Dodson, P.D.3    Dowie, M.J.4    Taylor, T.N.5    Potgieter, D.6
  • 74
    • 27844547616 scopus 로고    scopus 로고
    • The role of Nurr1 in the development of dopaminergic neurons and Parkinson’s disease
    • Jankovic, J., Chen, S., and Le, W. D. (2005). The role of Nurr1 in the development of dopaminergic neurons and Parkinson’s disease. Prog. Neurobiol. 77, 128–138. doi: 10.1016/j.pneurobio.2005.09.001
    • (2005) Prog. Neurobiol , vol.77 , pp. 128-138
    • Jankovic, J.1    Chen, S.2    Le, W.D.3
  • 75
    • 0027712869 scopus 로고
    • Mapping toxicant-induced nervous system damage with a cupric silver stain: A quantitative analysis of neural degeneration induced by 3,4-methylenedioxymethamphetamine
    • discussion 150–154
    • Jensen, K. F., Olin, J., Haykal-Coates, N., O’Callaghan, J., Miller, D. B., and de Olmos, J. S. (1993). Mapping toxicant-induced nervous system damage with a cupric silver stain: a quantitative analysis of neural degeneration induced by 3,4-methylenedioxymethamphetamine. NIDA Res. Monogr. 136, 133–149; discussion 150–154.
    • (1993) NIDA Res. Monogr , vol.136 , pp. 133-149
    • Jensen, K.F.1    Olin, J.2    Haykal-Coates, N.3    O’Callaghan, J.4    Miller, D.B.5    De Olmos, J.S.6
  • 76
    • 10044247254 scopus 로고    scopus 로고
    • Age-dependent dopaminergic dysfunction in Nurr1 knockout mice
    • Jiang, C., Wan, X., He, Y., Pan, T., Jankovic, J., and Le, W. (2005). Age-dependent dopaminergic dysfunction in Nurr1 knockout mice. Exp. Neurol.191, 154–162. doi: 10.1016/j.expneurol.2004.08.035
    • (2005) Exp. Neurol. , vol.191 , pp. 154-162
    • Jiang, C.1    Wan, X.2    He, Y.3    Pan, T.4    Jankovic, J.5    Le, W.6
  • 77
    • 84868087279 scopus 로고    scopus 로고
    • ROS-dependent regulation of Parkin and DJ-1 localization during oxidative stress in neurons
    • Joselin, A. P., Hewitt, S. J., Callaghan, S. M., Kim, R. H., Chung, Y.-H., Mak, T. W., et al. (2012). ROS-dependent regulation of Parkin and DJ-1 localization during oxidative stress in neurons. Hum. Mol. Genet.21, 4888–4903. doi: 10.1093/hmg/dds325
    • (2012) Hum. Mol. Genet. , vol.21 , pp. 4888-4903
    • Joselin, A.P.1    Hewitt, S.J.2    Callaghan, S.M.3    Kim, R.H.4    Chung, Y.-H.5    Mak, T.W.6
  • 78
    • 72449191782 scopus 로고    scopus 로고
    • Nurr1 is required for maintenance of maturing and adult midbrain dopamine neurons
    • Kadkhodaei, B., Ito, T., Joodmardi, E., Mattsson, B., Rouillard, C., Carta, M., et al. (2009). Nurr1 is required for maintenance of maturing and adult midbrain dopamine neurons. J. Neurosci. 29, 15923–15932. doi: 10.1523/JNEUROSCI.3910-09.2009
    • (2009) J. Neurosci , vol.29 , pp. 15923-15932
    • Kadkhodaei, B.1    Ito, T.2    Joodmardi, E.3    Mattsson, B.4    Rouillard, C.5    Carta, M.6
  • 79
    • 84907487576 scopus 로고    scopus 로고
    • A 6.4 Mb duplication of the α-synuclein locus causing frontotemporal dementia and parkinsonism: Phenotype-genotype correlations
    • Kara, E., Kiely, A. P., Proukakis, C., Giffin, N., Love, S., Hehir, J., et al. (2014). A 6.4 Mb duplication of the α-synuclein locus causing frontotemporal dementia and parkinsonism: phenotype-genotype correlations.JAMA Neurol. 71, 1162–1171. doi: 10.1001/jamaneurol.2014.994
    • (2014) JAMA Neurol , vol.71 , pp. 1162-1171
    • Kara, E.1    Kiely, A.P.2    Proukakis, C.3    Giffin, N.4    Love, S.5    Hehir, J.6
  • 80
    • 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, R. H., Smith, P. D., Aleyasin, H., Hayley, S., Mount, M. P., Pownall, S., et al. (2005). 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.102, 5215–5220. doi: 10.1073/pnas.0501282102
    • (2005) Proc. Natl. Acad. Sci. U.S.A. , vol.102 , pp. 5215-5220
    • Kim, R.H.1    Smith, P.D.2    Aleyasin, H.3    Hayley, S.4    Mount, M.P.5    Pownall, S.6
  • 81
    • 0345269757 scopus 로고    scopus 로고
    • Nigrostriatal alpha-synucleinopathy induced by viral vector-mediated overexpression of human alpha-synuclein: A new primate model of Parkinson’s disease
    • Kirik, D., Annett, L. E., Burger, C., Muzyczka, N., Mandel, R. J., and Björk-lund, A. (2003). Nigrostriatal alpha-synucleinopathy induced by viral vector-mediated overexpression of human alpha-synuclein: a new primate model of Parkinson’s disease. Proc. Natl. Acad. Sci. U.S.A.100, 2884–2889. doi: 10.1073/pnas.0536383100
    • (2003) Proc. Natl. Acad. Sci. U.S.A. , vol.100 , pp. 2884-2889
    • Kirik, D.1    Annett, L.E.2    Burger, C.3    Muzyczka, N.4    Mandel, R.J.5    Björk-Lund, A.6
  • 82
    • 0036550101 scopus 로고    scopus 로고
    • Parkinson-like neurodegeneration induced by targeted overexpression of alpha-synuclein in the nigrostriatal system
    • Kirik, D., Rosenblad, C., Burger, C., Lundberg, C., Johansen, T. E., Muzy-czka, N., et al. (2002). Parkinson-like neurodegeneration induced by targeted overexpression of alpha-synuclein in the nigrostriatal system. J. Neurosci.22, 2780–2791.
    • (2002) J. Neurosci. , vol.22 , pp. 2780-2791
    • Kirik, D.1    Rosenblad, C.2    Burger, C.3    Lundberg, C.4    Johansen, T.E.5    Muzy-Czka, N.6
  • 83
    • 67649479622 scopus 로고    scopus 로고
    • Impaired dopamine release and synaptic plasticity in the striatum of parkin-/- mice
    • Kitada, T., Pisani, A., Karouani, M., Haburcak, M., Martella, G., Tscherter, A., et al. (2009a). Impaired dopamine release and synaptic plasticity in the striatum of parkin-/- mice. J. Neurochem.110, 613–621. doi: 10.1111/j.1471-4159.2009.06152.x
    • (2009) J. Neurochem. , vol.110 , pp. 613-621
    • Kitada, T.1    Pisani, A.2    Karouani, M.3    Haburcak, M.4    Martella, G.5    Tscherter, A.6
  • 84
    • 70349923087 scopus 로고    scopus 로고
    • Absence of nigral degeneration in aged parkin/DJ-1/PINK1 triple knockout mice
    • Kitada, T., Tong, Y., Gautier, C. A., and Shen, J. (2009b). Absence of nigral degeneration in aged parkin/DJ-1/PINK1 triple knockout mice. J. Neurochem.111, 696–702. doi: 10.1111/j.1471-4159.2009.06350.x
    • (2009) J. Neurochem. , vol.111 , pp. 696-702
    • Kitada, T.1    Tong, Y.2    Gautier, C.A.3    Shen, J.4
  • 85
    • 0036202813 scopus 로고    scopus 로고
    • Dopamin-ergic cell loss induced by human A30P alpha-synuclein gene transfer to the rat substantia nigra
    • Klein, R. L., King, M. A., Hamby, M. E., and Meyer, E. M. (2002). Dopamin-ergic cell loss induced by human A30P alpha-synuclein gene transfer to the rat substantia nigra. Hum. Gene Ther. 13, 605–612. doi: 10.1089/1043034025 2837206
    • (2002) Hum. Gene Ther , vol.13 , pp. 605-612
    • Klein, R.L.1    King, M.A.2    Hamby, M.E.3    Meyer, E.M.4
  • 86
    • 0017287310 scopus 로고
    • Influence of methamphetamine on nigral and striatal tyrosine hydroxylase activity and on striatal dopamine levels
    • Kogan, F. J., Nichols, W. K., and Gibb, J. W. (1976). Influence of methamphetamine on nigral and striatal tyrosine hydroxylase activity and on striatal dopamine levels. Eur. J. Pharmacol. 36, 363–371. doi: 10.1016/0014-2999(76)90090-X
    • (1976) Eur. J. Pharmacol , vol.36 , pp. 363-371
    • Kogan, F.J.1    Nichols, W.K.2    Gibb, J.W.3
  • 88
    • 84870903681 scopus 로고    scopus 로고
    • Genetic animal models for evaluating the role of autophagy in etiopathogenesis of Parkinson disease
    • Lachenmayer, M. L., and Yue, Z. (2012). Genetic animal models for evaluating the role of autophagy in etiopathogenesis of Parkinson disease. Autophagy8, 1837–1838. doi: 10.4161/auto.21859
    • (2012) Autophagy , vol.8 , pp. 1837-1838
    • Lachenmayer, M.L.1    Yue, Z.2
  • 89
    • 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 overex-pressing human α-synuclein
    • Lam, H. A., Wu, N., Cely, I., Kelly, R. L., Hean, S., Richter, F., et al. (2011). Elevated tonic extracellular dopamine concentration and altered dopamine modulation of synaptic activity precede dopamine loss in the striatum of mice overex-pressing human α-synuclein. J. Neurosci. Res.89, 1091–1102. doi: 10.1002/jnr.22611
    • (2011) J. Neurosci. Res. , vol.89 , pp. 1091-1102
    • Lam, H.A.1    Wu, N.2    Cely, I.3    Kelly, R.L.4    Hean, S.5    Richter, F.6
  • 90
    • 0020680904 scopus 로고
    • Chronic Parkinsonism inhumansduetoaproductofmeperidine-analogsynthesis
    • Langston, J. W., Ballard, P., Tetrud, J. W., and Irwin, I. (1983). Chronic Parkinsonism inhumansduetoaproductofmeperidine-analogsynthesis. Science219, 979–980. doi: 10.1126/science.6823561
    • (1983) Science , vol.219 , pp. 979-980
    • Langston, J.W.1    Ballard, P.2    Tetrud, J.W.3    Irwin, I.4
  • 91
    • 33845482072 scopus 로고    scopus 로고
    • Non-invasive imaging of neuropathology in a rat model of alpha-synuclein overexpression
    • Lauwers, E., Bequé, D., Van Laere, K., Nuyts, J., Bormans, G., Mortel-mans, L., et al. (2007). Non-invasive imaging of neuropathology in a rat model of alpha-synuclein overexpression. Neurobiol. Aging28, 248–257. doi: 10.1016/j.neurobiolaging.2005.12.005
    • (2007) Neurobiol. Aging , vol.28 , pp. 248-257
    • Lauwers, E.1    Bequé, D.2    Van Laere, K.3    Nuyts, J.4    Bormans, G.5    Mortel-Mans, L.6
  • 92
    • 0041625923 scopus 로고    scopus 로고
    • Neuropathology and neurodegeneration in rodent brain induced by lentiviral vector-mediated overexpression of alpha-synuclein
    • Lauwers, E., Debyser, Z., Van Dorpe, J., De Strooper, B., Nuttin, B., and Baekelandt, V. (2003). Neuropathology and neurodegeneration in rodent brain induced by lentiviral vector-mediated overexpression of alpha-synuclein. Brain Pathol. 13, 364–372. doi: 10.1111/j.1750-3639.2003.tb00035.x
    • (2003) Brain Pathol , vol.13 , pp. 364-372
    • Lauwers, E.1    Debyser, Z.2    Van Dorpe, J.3    De Strooper, B.4    Nuttin, B.5    Baekelandt, V.6
  • 93
    • 77956441086 scopus 로고    scopus 로고
    • Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson’s disease
    • Lee, B. D., Shin, J.-H., VanKampen, J., Petrucelli, L., West, A. B., Ko, H. S., et al. (2010). Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson’s disease. Nat. Med.16, 998–1000. doi: 10.1038/nm.2199
    • (2010) Nat. Med. , vol.16 , pp. 998-1000
    • Lee, B.D.1    Shin, J.-H.2    Vankampen, J.3    Petrucelli, L.4    West, A.B.5    Ko, H.S.6
  • 94
    • 74249098205 scopus 로고    scopus 로고
    • The role of transcription factor Pitx3 in dopamine neuron development and Parkinson’s disease
    • Li, J., Dani, J. A., and Le, W. (2009a). The role of transcription factor Pitx3 in dopamine neuron development and Parkinson’s disease. Curr. Top. Med. Chem. 9, 855–859.
    • (2009) Curr. Top. Med. Chem , vol.9 , pp. 855-859
    • Li, J.1    Dani, J.A.2    Le, W.3
  • 95
    • 67649813448 scopus 로고    scopus 로고
    • Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson’s disease
    • Li, Y., Liu, W., Oo, T. F., Wang, L., Tang, Y., Jackson-Lewis, V., et al. (2009b). Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson’s disease. Nat. Neurosci.12, 826–828. doi: 10.1038/nn.2349
    • (2009) Nat. Neurosci. , vol.12 , pp. 826-828
    • Li, Y.1    Liu, W.2    Oo, T.F.3    Wang, L.4    Tang, Y.5    Jackson-Lewis, V.6
  • 96
    • 76149134717 scopus 로고    scopus 로고
    • Enhanced striatal dopamine transmission and motor performance with LRRK2 overexpression in mice is eliminated by familial Parkinson’s disease mutation G2019S
    • Li, X., Patel, J. C., Wang, J., Avshalumov, M. V., Nicholson, C., Buxbaum, J. D., et al. (2010). Enhanced striatal dopamine transmission and motor performance with LRRK2 overexpression in mice is eliminated by familial Parkinson’s disease mutation G2019S. J. Neurosci.30, 1788–1797. doi: 10.1523/JNEUROSCI.5604-09.2010
    • (2010) J. Neurosci. , vol.30 , pp. 1788-1797
    • Li, X.1    Patel, J.C.2    Wang, J.3    Avshalumov, M.V.4    Nicholson, C.5    Buxbaum, J.D.6
  • 97
    • 72149087091 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson’s-disease-related mutant alpha-synuclein
    • Lin, X., Parisiadou, L., Gu, X.-L., Wang, L., Shim, H., Sun, L., et al. (2009). Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson’s-disease-related mutant alpha-synuclein. Neuron64, 807–827. doi: 10.1016/j.neuron.2009.11.006
    • (2009) Neuron , vol.64 , pp. 807-827
    • Lin, X.1    Parisiadou, L.2    Gu, X.-L.3    Wang, L.4    Shim, H.5    Sun, L.6
  • 98
    • 84863456841 scopus 로고    scopus 로고
    • Conditional expression of Parkinson’s disease-related mutant α-synuclein in the midbrain dopaminergic neurons causes progressive neurodegeneration and degradation of transcription factor nuclear receptor related 1
    • Lin, X., Parisiadou, L., Sgobio, C., Liu, G., Yu, J., Sun, L., et al. (2012). Conditional expression of Parkinson’s disease-related mutant α-synuclein in the midbrain dopaminergic neurons causes progressive neurodegeneration and degradation of transcription factor nuclear receptor related 1. J. Neurosci.32, 9248–9264. doi: 10.1523/JNEUROSCI.1731-12.2012
    • (2012) J. Neurosci. , vol.32 , pp. 9248-9264
    • Lin, X.1    Parisiadou, L.2    Sgobio, C.3    Liu, G.4    Yu, J.5    Sun, L.6
  • 99
    • 0036679197 scopus 로고    scopus 로고
    • Alpha -Synucleinopathy and selective dopaminergic neuron loss in a rat lentiviral-based model of Parkinson’s disease
    • Lo Bianco, C., Ridet, J.-L., Schneider, B. L., Deglon, N., and Aebischer, P. (2002). alpha -Synucleinopathy and selective dopaminergic neuron loss in a rat lentiviral-based model of Parkinson’s disease. Proc. Natl. Acad. Sci. U.S.A. 99, 10813–10818. doi: 10.1073/pnas.152339799
    • (2002) Proc. Natl. Acad. Sci. U.S.A , vol.99 , pp. 10813-10818
    • Lo Bianco, C.1    Ridet, J.-L.2    Schneider, B.L.3    Deglon, N.4    Aebischer, P.5
  • 100
    • 84904012715 scopus 로고    scopus 로고
    • Increased vesicular monoamine transporter enhances dopamine release and opposes Parkinson disease-related neurodegeneration in vivo
    • Lohr, K. M., Bernstein, A. I., Stout, K. A., Dunn, A. R., Lazo, C. R., Alter, S. P., et al. (2014). Increased vesicular monoamine transporter enhances dopamine release and opposes Parkinson disease-related neurodegeneration in vivo. Proc. Natl. Acad. Sci. U.S.A.111, 9977–9982. doi: 10.1073/pnas.1402134111
    • (2014) Proc. Natl. Acad. Sci. U.S.A. , vol.111 , pp. 9977-9982
    • Lohr, K.M.1    Bernstein, A.I.2    Stout, K.A.3    Dunn, A.R.4    Lazo, C.R.5    Alter, S.P.6
  • 101
    • 60849106352 scopus 로고    scopus 로고
    • Bacterial artificial chromosome transgenic mice expressing a truncated mutant parkin exhibit age-dependent hypokinetic motor deficits, dopaminergic neuron degeneration, and accumulation of proteinase K-resistant alpha-synuclein
    • Lu, X.-H., Fleming, S. M., Meurers, B., Ackerson, L. C., Mortazavi, F., Lo, V., et al. (2009). Bacterial artificial chromosome transgenic mice expressing a truncated mutant parkin exhibit age-dependent hypokinetic motor deficits, dopaminergic neuron degeneration, and accumulation of proteinase K-resistant alpha-synuclein. J. Neurosci.29, 1962–1976. doi: 10.1523/JNEUROSCI.5351-08.2009
    • (2009) J. Neurosci. , vol.29 , pp. 1962-1976
    • Lu, X.-H.1    Fleming, S.M.2    Meurers, B.3    Ackerson, L.C.4    Mortazavi, F.5    Lo, V.6
  • 102
    • 0342368772 scopus 로고    scopus 로고
    • Association between early-onset Parkinson’s disease and mutations in the parkin gene
    • Lücking, C. B., Dürr, A., Bonifati, V., Vaughan, J., De Michele, G., Gasser, T., et al. (2000). Association between early-onset Parkinson’s disease and mutations in the parkin gene. N. Engl. J. Med. 342, 1560–1567. doi: 10.1056/NEJM200005253422103
    • (2000) N. Engl. J. Med , vol.342 , pp. 1560-1567
    • Lücking, C.B.1    Dürr, A.2    Bonifati, V.3    Vaughan, J.4    De Michele, G.5    Gasser, T.6
  • 103
    • 84869109864 scopus 로고    scopus 로고
    • Pathological α-synuclein transmission initiates Parkinsonlike neurodegeneration in nontransgenic mice
    • Luk, K. C., Kehm, V., Carroll, J., Zhang, B., O’Brien, P., Trojanowski, J. Q., et al. (2012a). Pathological α-synuclein transmission initiates Parkinsonlike neurodegeneration in nontransgenic mice. Science338, 949–953. doi: 10.1126/science.1227157
    • (2012) Science , vol.338 , pp. 949-953
    • Luk, K.C.1    Kehm, V.2    Carroll, J.3    Zhang, B.4    O’Brien, P.5    Trojanowski, J.Q.6
  • 104
    • 84862609075 scopus 로고    scopus 로고
    • Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice
    • Luk, K. C., Kehm, V. M., Zhang, B., O’Brien, P., Trojanowski, J. Q., and Lee, V. M. Y. (2012b). Intracerebral inoculation of pathological α-synuclein initiates a rapidly progressive neurodegenerative α-synucleinopathy in mice. J. Exp. Med. 209, 975–986. doi: 10.1084/jem.20112457
    • (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.6
  • 105
    • 84887984409 scopus 로고    scopus 로고
    • Modeling Lewy pathology propagation in Parkinson’s disease
    • Luk, K. C., and Lee, V. M.-Y. (2014). Modeling Lewy pathology propagation in Parkinson’s disease. Parkinsonism Relat. Disord. 20(Suppl. 1), S85–S87. doi: 10.1016/S1353-8020(13)70022–70021
    • (2014) Parkinsonism Relat. Disord , vol.20 , pp. S85-S87
    • Luk, K.C.1    Lee, V.M.2
  • 106
    • 84862230156 scopus 로고    scopus 로고
    • The I2020T Leucine-rich repeat kinase 2 transgenic mouse exhibits impaired locomotive ability accompanied by dopaminergic neuron abnormalities
    • Maekawa, T., Mori, S., Sasaki, Y., Miyajima, T., Azuma, S., Ohta, E., et al. (2012). The I2020T Leucine-rich repeat kinase 2 transgenic mouse exhibits impaired locomotive ability accompanied by dopaminergic neuron abnormalities. Mol. Neurodegener.7, 15. doi: 10.1186/1750-1326-715
    • (2012) Mol. Neurodegener. , vol.7 , pp. 15
    • Maekawa, T.1    Mori, S.2    Sasaki, Y.3    Miyajima, T.4    Azuma, S.5    Ohta, E.6
  • 107
    • 0037127197 scopus 로고    scopus 로고
    • The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein
    • Manning-Bog, A. B., McCormack, A. L., Li, J., Uversky, V. N., Fink, A. L., and Di Monte, D. A. (2002). The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein. J. Biol. Chem.277, 1641–1644. doi: 10.1074/jbc.C100560200
    • (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
  • 108
    • 58149527808 scopus 로고    scopus 로고
    • Enhanced sensitivity to group II mGlu receptor activation at corticostriatal synapses in mice lacking the familial parkinsonism-linked genes PINK1 or Parkin
    • Martella, G., Platania, P., Vita, D., Sciamanna, G., Cuomo, D., Tassone, A., et al. (2009). Enhanced sensitivity to group II mGlu receptor activation at corticostriatal synapses in mice lacking the familial parkinsonism-linked genes PINK1 or Parkin. Exp. Neurol. 215, 388–396. doi: 10.1016/j.expneurol.2008.11.001
    • (2009) Exp. Neurol , vol.215 , pp. 388-396
    • Martella, G.1    Platania, P.2    Vita, D.3    Sciamanna, G.4    Cuomo, D.5    Tassone, A.6
  • 109
    • 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., Mallory, M., Hashimoto, M., Takeda, A., et al. (2000). Dopaminergic loss and inclusion body formation in alpha-synuclein mice: implications for neurodegenerative disorders. Science287, 1265–1269. doi: 10.1126/science.287.5456.1265
    • (2000) Science , vol.287 , pp. 1265-1269
    • Masliah, E.1    Rockenstein, E.2    Veinbergs, I.3    Mallory, M.4    Hashimoto, M.5    Takeda, A.6
  • 111
    • 84901835532 scopus 로고    scopus 로고
    • Exploring the pathogenetic mechanisms underlying Parkinson’s disease in medaka fish
    • Matsui, H., Uemura, N., Yamakado, H., Takeda, S., and Takahashi, R. (2014). Exploring the pathogenetic mechanisms underlying Parkinson’s disease in medaka fish. J. Parkinsons. Dis.4, 301–310. doi: 10.3233/JPD-130289
    • (2014) J. Parkinsons. Dis. , vol.4 , pp. 301-310
    • Matsui, H.1    Uemura, N.2    Yamakado, H.3    Takeda, S.4    Takahashi, R.5
  • 112
    • 0034979314 scopus 로고    scopus 로고
    • Lack of nigral pathology in transgenic mice expressing human alpha-synuclein driven by the tyrosine hydroxylase promoter
    • Matsuoka, Y., Vila, M., Lincoln, S., McCormack, A., Picciano, M., LaFrancois, J., et al. (2001). Lack of nigral pathology in transgenic mice expressing human alpha-synuclein driven by the tyrosine hydroxylase promoter. Neurobiol. Dis.8, 535–539. doi: 10.1006/nbdi.2001.0392
    • (2001) Neurobiol. Dis. , vol.8 , pp. 535-539
    • Matsuoka, Y.1    Vila, M.2    Lincoln, S.3    McCormack, A.4    Picciano, M.5    Lafrancois, J.6
  • 113
    • 0036075892 scopus 로고    scopus 로고
    • Environmental risk factors and Parkinson’s disease: Selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat
    • McCormack, A. L., Thiruchelvam, M., Manning-Bog, A. B., Thiffault, C., Langston, J. W., Cory-Slechta, D. A., et al. (2002). Environmental risk factors and Parkinson’s disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol. Dis. 10, 119–127. doi: 10.1006/nbdi.2002.0507
    • (2002) Neurobiol. Dis , vol.10 , pp. 119-127
    • McCormack, A.L.1    Thiruchelvam, M.2    Manning-Bog, A.B.3    Thiffault, C.4    Langston, J.W.5    Cory-Slechta, D.A.6
  • 114
    • 84859699540 scopus 로고    scopus 로고
    • Animal models of the non-motor features of Parkinson’s disease
    • McDowell, K., and Chesselet, M.-F. (2012). Animal models of the non-motor features of Parkinson’s disease. Neurobiol. Dis.46, 597–606. doi: 10.1016/j.nbd.2011.12.040
    • (2012) Neurobiol. Dis. , vol.46 , pp. 597-606
    • McDowell, K.1    Chesselet, M.-F.2
  • 115
    • 77957794336 scopus 로고    scopus 로고
    • Impaired dopaminergic neurotransmission and microtubule-associated protein tau alterations in human LRRK2 transgenic mice
    • Melrose, H. L., Dächsel, J. C., Behrouz, B., Lincoln, S. J., Yue, M., Hinkle, K. M., et al. (2010). Impaired dopaminergic neurotransmission and microtubule-associated protein tau alterations in human LRRK2 transgenic mice. Neurobiol. Dis.40, 503–517. doi: 10.1016/j.nbd.2010.07.010
    • (2010) Neurobiol. Dis. , vol.40 , pp. 503-517
    • Melrose, H.L.1    Dächsel, J.C.2    Behrouz, B.3    Lincoln, S.J.4    Yue, M.5    Hinkle, K.M.6
  • 116
    • 47049122191 scopus 로고    scopus 로고
    • Modeling PD pathogenesis in mice: advantages of a chronic MPTP protocol
    • Meredith, G. E., Totterdell, S., Potashkin, J. A., and Surmeier, D. J. (2008). Modeling PD pathogenesis in mice: advantages of a chronic MPTP protocol. Park. Relat Disord14(Suppl. 2), S112–S115. doi: 10.1016/j.parkreldis.2008.04.012
    • (2008) Park. Relat Disord , vol.14 , pp. S112-S115
    • Meredith, G.E.1    Totterdell, S.2    Potashkin, J.A.3    Surmeier, D.J.4
  • 117
    • 35448981156 scopus 로고    scopus 로고
    • Paraquat: The red herring of Parkinson’s disease research
    • Miller, G. W. (2007). Paraquat: the red herring of Parkinson’s disease research. Toxicol. Sci.100, 1–2. doi: 10.1093/toxsci/kfm223
    • (2007) Toxicol. Sci. , vol.100 , pp. 1-2
    • Miller, G.W.1
  • 118
    • 33847363531 scopus 로고    scopus 로고
    • Wild-type and mutant alpha-synuclein induce a multi-component gene expression profile consistent with shared pathophysiology in different transgenic mouse models of PD
    • Miller, R. M., Kiser, G. L., Kaysser-Kranich, T., Casaceli, C., Colla, E., Lee, M. K., et al. (2007). Wild-type and mutant alpha-synuclein induce a multi-component gene expression profile consistent with shared pathophysiology in different transgenic mouse models of PD. Exp. Neurol.204, 421–432. doi: 10.1016/j.expneurol.2006.12.005
    • (2007) Exp. Neurol. , vol.204 , pp. 421-432
    • Miller, R.M.1    Kiser, G.L.2    Kaysser-Kranich, T.3    Casaceli, C.4    Colla, E.5    Lee, M.K.6
  • 119
    • 84898023373 scopus 로고    scopus 로고
    • PINK1 loss-of-function mutations affect mitochondrial complex I activity via NdufA10 ubiquinone uncoupling
    • Morais, V. A., Haddad, D., Craessaerts, K., De Bock, P.-J., Swerts, J., Vilain, S., et al. (2014). PINK1 loss-of-function mutations affect mitochondrial complex I activity via NdufA10 ubiquinone uncoupling. Science344, 203–207. doi: 10.1126/science.1249161
    • (2014) Science , vol.344 , pp. 203-207
    • Morais, V.A.1    Haddad, D.2    Craessaerts, K.3    De Bock, P.-J.4    Swerts, J.5    Vilain, S.6
  • 120
    • 0026587806 scopus 로고
    • Differential vulnerability of primate caudate-putamen and striosome-matrix dopamine systems to the neurotoxic effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
    • Moratalla, R., Quinn, B., DeLanney, L. E., Irwin, I., Langston, J. W., and Gray-biel, A. M. (1992). Differential vulnerability of primate caudate-putamen and striosome-matrix dopamine systems to the neurotoxic effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc. Natl. Acad. Sci. U.S.A.89, 3859–3863. doi: 10.1073/pnas.89.9.3859
    • (1992) Proc. Natl. Acad. Sci. U.S.A. , vol.89 , pp. 3859-3863
    • Moratalla, R.1    Quinn, B.2    Delanney, L.E.3    Irwin, I.4    Langston, J.W.5    Gray-Biel, A.M.6
  • 121
    • 84900820438 scopus 로고    scopus 로고
    • Modeling dyskinesia in animal models of Parkinson disease
    • Morin, N., Jourdain, V. A., and Di Paolo, T. (2014). Modeling dyskinesia in animal models of Parkinson disease. Exp. Neurol.256, 105–116. doi: 10.1016/j.expneurol.2013.01.024
    • (2014) Exp. Neurol. , vol.256 , pp. 105-116
    • Morin, N.1    Jourdain, V.A.2    Di Paolo, T.3
  • 122
    • 79960908654 scopus 로고    scopus 로고
    • Impact of methamphetamine on dopamine neurons in primates is dependent on age: Implications for development of Parkinson’s disease
    • Morrow, B. A., Roth, R. H., Redmond, D. E., and Elsworth, J. D. (2011). Impact of methamphetamine on dopamine neurons in primates is dependent on age: implications for development of Parkinson’s disease. Neuroscience189, 277–285. doi: 10.1016/j.neuroscience.2011.05.046
    • (2011) Neuroscience , vol.189 , pp. 277-285
    • Morrow, B.A.1    Roth, R.H.2    Redmond, D.E.3    Elsworth, J.D.4
  • 123
    • 0028302235 scopus 로고
    • Differences in nigral neuron number and sensitivity to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in C57/bl and CD-1 mice
    • Muthane, U., Ramsay, K. A., Jiang, H., Jackson-Lewis, V., Donaldson, D., Fernando, S., et al. (1994). Differences in nigral neuron number and sensitivity to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in C57/bl and CD-1 mice. Exp. Neurol.126, 195–204. doi: 10.1006/exnr.1994.1058
    • (1994) Exp. Neurol. , vol.126 , pp. 195-204
    • Muthane, U.1    Ramsay, K.A.2    Jiang, H.3    Jackson-Lewis, V.4    Donaldson, D.5    Fernando, S.6
  • 124
    • 84879129926 scopus 로고    scopus 로고
    • Leucine-rich repeat kinase 2 (LRRK2)-deficient rats exhibit renal tubule injury and perturbations in metabolic and immunological homeostasis
    • Ness, D., Ren, Z., Gardai, S., Sharpnack, D., Johnson, V. J., Brennan, R. J., et al. (2013). Leucine-rich repeat kinase 2 (LRRK2)-deficient rats exhibit renal tubule injury and perturbations in metabolic and immunological homeostasis. PLoS ONE8:e66164. doi: 10.1371/journal.pone.0066164
    • (2013) Plos ONE , vol.8
    • Ness, D.1    Ren, Z.2    Gardai, S.3    Sharpnack, D.4    Johnson, V.J.5    Brennan, R.J.6
  • 125
    • 33646476833 scopus 로고    scopus 로고
    • Increased sensitivity to MPTP in human alpha-synuclein A30P trans-genic mice
    • Nieto, M., Gil-Bea, F. J., Dalfó, E., Cuadrado, M., Cabodevilla, F., Sánchez, B., et al. (2006). Increased sensitivity to MPTP in human alpha-synuclein A30P trans-genic mice. Neurobiol. Aging 27, 848–856. doi: 10.1016/j.neurobiolaging.2005.04.010
    • (2006) Neurobiol. Aging , vol.27 , pp. 848-856
    • Nieto, M.1    Gil-Bea, F.J.2    Dalfó, E.3    Cuadrado, M.4    Cabodevilla, F.5    Sánchez, B.6
  • 126
    • 84908409010 scopus 로고    scopus 로고
    • Progressive nigrostriatal terminal dysfunction and degeneration in the engrailed1 heterozygous mouse model of Parkinson’s disease
    • Nordström, U., Beauvais, G., Ghosh, A., Sasidharan, B. C. P., Lundblad, M., Fuchs, J., et al. (2014). Progressive nigrostriatal terminal dysfunction and degeneration in the engrailed1 heterozygous mouse model of Parkinson’s disease. Neurobiol. Dis. doi: 10.1016/j.nbd.2014.09.012
    • (2014) Neurobiol. Dis
    • Nordström, U.1    Beauvais, G.2    Ghosh, A.3    Sasidharan, B.4    Lundblad, M.5    Fuchs, J.6
  • 127
    • 84875638056 scopus 로고    scopus 로고
    • Age-dependent effects of A53T alpha-synuclein on behavior and dopaminergic function
    • Oaks, A. W., Frankfurt, M., Finkelstein, D. I., and Sidhu, A. (2013). Age-dependent effects of A53T alpha-synuclein on behavior and dopaminergic function. PLoS ONE8:e60378. doi: 10.1371/journal.pone.0060378
    • (2013) Plos ONE , vol.8
    • Oaks, A.W.1    Frankfurt, M.2    Finkelstein, D.I.3    Sidhu, A.4
  • 128
    • 84906323786 scopus 로고    scopus 로고
    • The expanding universe of disorders of the basal ganglia
    • Obeso, J. A., Rodriguez-Oroz, M. C., Stamelou, M., Bhatia, K. P., and Burn, D. J. (2014). The expanding universe of disorders of the basal ganglia. Lancet 384, 523–531. doi: 10.1016/S0140-6736(13)62418–62416
    • (2014) Lancet , vol.384 , pp. 523-531
    • Obeso, J.A.1    Rodriguez-Oroz, M.C.2    Stamelou, M.3    Bhatia, K.P.4    Burn, D.J.5
  • 129
    • 0027931499 scopus 로고
    • Neurotoxicity profiles of substituted amphetamines in the C57BL/6J mouse
    • O’Callaghan, J. P., and Miller, D. B. (1994). Neurotoxicity profiles of substituted amphetamines in the C57BL/6J mouse. J. Pharmacol. Exp. Ther. 270, 741–751.
    • (1994) J. Pharmacol. Exp. Ther , vol.270 , pp. 741-751
    • O’Callaghan, J.P.1    Miller, D.B.2
  • 131
    • 84888119924 scopus 로고    scopus 로고
    • RAAV2/7 vector-mediated overexpression of alpha-synuclein in mouse substantia nigra induces protein aggregation and progressive dose-dependent neurodegeneration
    • Oliveras-Salvá, M., Van der Perren, A., Casadei, N., Stroobants, S., Nuber, S., D’Hooge, R., et al. (2013). rAAV2/7 vector-mediated overexpression of alpha-synuclein in mouse substantia nigra induces protein aggregation and progressive dose-dependent neurodegeneration. Mol. Neurodegener. 8, 44. doi: 10.1186/1750-1326-8-44
    • (2013) Mol. Neurodegener , vol.8 , pp. 44
    • Oliveras-Salvá, M.1    Van Der Perren, A.2    Casadei, N.3    Stroobants, S.4    Nuber, S.5    D’Hooge, R.6
  • 132
    • 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., Ikeda, M., Nishinakagawa, T., Hosokawa, M., et al. (2009). A chemical chaperone, sodium 4-phenylbutyric acid, attenuates the pathogenic potency in human alpha-synuclein A30P + A53T transgenic mice. Parkinsonism Relat. Disord.15, 649–654. doi: 10.1016/j.parkreldis.2009.03.002
    • (2009) Parkinsonism Relat. Disord. , vol.15 , pp. 649-654
    • Ono, K.1    Ikemoto, M.2    Kawarabayashi, T.3    Ikeda, M.4    Nishinakagawa, T.5    Hosokawa, M.6
  • 133
    • 84876098997 scopus 로고    scopus 로고
    • The Parkinson disease-related protein DJ-1 counteracts mitochondrial impairment induced by the tumour suppressor protein p53 by enhancing endoplasmic reticulum-mitochondria tethering
    • Ottolini, D., Calì, T., Negro, A., and Brini, M. (2013). The Parkinson disease-related protein DJ-1 counteracts mitochondrial impairment induced by the tumour suppressor protein p53 by enhancing endoplasmic reticulum-mitochondria tethering. Hum. Mol. Genet.22, 2152–2168. doi: 10.1093/hmg/ddt068
    • (2013) Hum. Mol. Genet. , vol.22 , pp. 2152-2168
    • Ottolini, D.1    Calì, T.2    Negro, A.3    Brini, M.4
  • 134
    • 2442481789 scopus 로고    scopus 로고
    • Mitochondrial dysfunction and oxidative damage in parkin-deficient mice
    • Palacino, J. J., Sagi, D., Goldberg, M. S., Krauss, S., Motz, C., Wacker, M., et al. (2004). Mitochondrial dysfunction and oxidative damage in parkin-deficient mice. J. Biol. Chem.279, 18614–18622. doi: 10.1074/jbc.M401135200
    • (2004) J. Biol. Chem. , vol.279 , pp. 18614-18622
    • Palacino, J.J.1    Sagi, D.2    Goldberg, M.S.3    Krauss, S.4    Motz, C.5    Wacker, M.6
  • 135
    • 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., Knels, L., Pursche, S., Jung, R., et al. (2010). Progression of Parkinson’s disease pathology is reproduced by intragastric administration of rotenone in mice. PLoS ONE5:e8762. doi: 10.1371/journal.pone.0008762
    • (2010) Plos ONE , vol.5
    • Pan-Montojo, F.1    Anichtchik, O.2    Dening, Y.3    Knels, L.4    Pursche, S.5    Jung, R.6
  • 136
    • 84881018218 scopus 로고    scopus 로고
    • Behavioral characterization of A53T mice reveals early and late stage deficits related to Parkinson’s disease
    • Paumier, K. L., Sukoff Rizzo, S. J., Berger, Z., Chen, Y., Gonzales, C., Kaftan, E., et al. (2013). Behavioral characterization of A53T mice reveals early and late stage deficits related to Parkinson’s disease. PLoS ONE8:e70274. doi: 10.1371/journal.pone.0070274
    • (2013) Plos ONE , vol.8
    • Paumier, K.L.1    Sukoff Rizzo, S.J.2    Berger, Z.3    Chen, Y.4    Gonzales, C.5    Kaftan, E.6
  • 137
    • 13844313915 scopus 로고    scopus 로고
    • Parkin-deficient mice are not a robust model of parkinsonism
    • Perez, F. A., and Palmiter, R. D. (2005). Parkin-deficient mice are not a robust model of parkinsonism. Proc. Natl. Acad. Sci. U.S.A.102, 2174–2179. doi: 10.1073/pnas.0409598102
    • (2005) Proc. Natl. Acad. Sci. U.S.A. , vol.102 , pp. 2174-2179
    • Perez, F.A.1    Palmiter, R.D.2
  • 138
    • 0037648357 scopus 로고    scopus 로고
    • Parkin mutations are frequent in patients with isolated early-onset parkinsonism
    • Periquet, M., Latouche, M., Lohmann, E., Rawal, N., De Michele, G., Ricard, S., et al. (2003). Parkin mutations are frequent in patients with isolated early-onset parkinsonism. Brain126, 1271–1278. doi: 10.1093/brain/awg136
    • (2003) Brain , vol.126 , pp. 1271-1278
    • Periquet, M.1    Latouche, M.2    Lohmann, E.3    Rawal, N.4    De Michele, G.5    Ricard, S.6
  • 139
    • 77950852274 scopus 로고    scopus 로고
    • DJ-1-deficient mice show less TH-positive neurons in the ventral tegmental area and exhibit non-motoric behavioural impairments
    • Pham, T. T., Giesert, F., Röthig, A., Floss, T., Kallnik, M., Weindl, K., et al. (2010). DJ-1-deficient mice show less TH-positive neurons in the ventral tegmental area and exhibit non-motoric behavioural impairments. Genes Brain Behav. 9, 305–317. doi: 10.1111/j.1601-183X.2009.00559.x
    • (2010) Genes Brain Behav , vol.9 , pp. 305-317
    • Pham, T.T.1    Giesert, F.2    Röthig, A.3    Floss, T.4    Kallnik, M.5    Weindl, K.6
  • 140
    • 84902170828 scopus 로고    scopus 로고
    • Is Parkinson’s disease a vesicular dopamine storage disorder? Evidence from a study in isolated synaptic vesicles of human and nonhuman primate striatum
    • Pifl, C., Rajput, A., Reither, H., Blesa, J., Cavada, C., Obeso, J. A, et al. (2014). Is Parkinson’s disease a vesicular dopamine storage disorder? Evidence from a study in isolated synaptic vesicles of human and nonhuman primate striatum. J. Neurosci. 34, 8210–8218. doi: 10.1523/JNEUROSCI.5456-13.2014
    • (2014) J. Neurosci , vol.34 , pp. 8210-8218
    • Pifl, C.1    Rajput, A.2    Reither, H.3    Blesa, J.4    Cavada, C.5    Obeso, J.A.6
  • 141
    • 84886692989 scopus 로고    scopus 로고
    • Modeling Parkinson’s disease in primates: The MPTP model
    • Porras, G., Li, Q., and Bezard, E. (2012). Modeling Parkinson’s disease in primates: the MPTP model. Cold Spring Harb. Perspect. Med. 2, a009308. doi: 10.1101/cshperspect.a009308
    • (2012) Cold Spring Harb. Perspect. Med , vol.2
    • Porras, G.1    Li, Q.2    Bezard, E.3
  • 142
    • 84911423174 scopus 로고    scopus 로고
    • The prevalence of Parkinson’s disease: A systematic review and meta-analysis
    • Pringsheim, T., Jette, N., Frolkis, A., and Steeves, T. D. L. (2014). The prevalence of Parkinson’s disease: a systematic review and meta-analysis. Mov. Disord.29, 1583–1590. doi: 10.1002/mds.25945
    • (2014) Mov. Disord. , vol.29 , pp. 1583-1590
    • Pringsheim, T.1    Jette, N.2    Frolkis, A.3    Steeves, T.4
  • 143
    • 0029015394 scopus 로고
    • Dose-dependent lesions of the dopaminergic nigrostriatal pathway induced by intrastriatal injection of 6-hydroxydopamine
    • Przedborski, S., Levivier, M., Jiang, H., Ferreira, M., Jackson-Lewis, V., Donaldson, D., et al. (1995). Dose-dependent lesions of the dopaminergic nigrostriatal pathway induced by intrastriatal injection of 6-hydroxydopamine. Neuroscience67, 631–647. doi: 10.1016/0306-4522(95)00066-R
    • (1995) Neuroscience , vol.67 , pp. 631-647
    • Przedborski, S.1    Levivier, M.2    Jiang, H.3    Ferreira, M.4    Jackson-Lewis, V.5    Donaldson, D.6
  • 144
    • 27744450270 scopus 로고    scopus 로고
    • Alpha-synuclein expression in the substantia nigra of MPTP-lesioned non-human primates
    • Purisai, M. G., McCormack, A. L., Langston, W. J., Johnston, L. C., and Di Monte, D. A. (2005). Alpha-synuclein expression in the substantia nigra of MPTP-lesioned non-human primates. Neurobiol. Dis.20, 898–906. doi: 10.1016/j.nbd.2005.05.028
    • (2005) Neurobiol. Dis. , vol.20 , pp. 898-906
    • Purisai, M.G.1    McCormack, A.L.2    Langston, W.J.3    Johnston, L.C.4    Di Monte, D.A.5
  • 145
    • 84875277277 scopus 로고    scopus 로고
    • Monogenic Parkinson’s disease and parkinsonism: Clinical phenotypes and frequencies of known mutations
    • Puschmann, A. (2013). Monogenic Parkinson’s disease and parkinsonism: clinical phenotypes and frequencies of known mutations. Parkinsonism Relat. Disord.19, 407–415. doi: 10.1016/j.parkreldis.2013.01.020
    • (2013) Parkinsonism Relat. Disord. , vol.19 , pp. 407-415
    • Puschmann, A.1
  • 146
    • 79953758383 scopus 로고    scopus 로고
    • Dopaminergic neuronal loss, reduced neurite complexity and autophagic abnormalities in transgenic mice expressing G2019S mutant LRRK2
    • Ramonet, D., Daher, J. P. L., Lin, B. M., Stafa, K., Kim, J., Banerjee, R., et al. (2011). Dopaminergic neuronal loss, reduced neurite complexity and autophagic abnormalities in transgenic mice expressing G2019S mutant LRRK2. PLoS ONE6:e18568. doi: 10.1371/journal.pone.0018568
    • (2011) Plos ONE , vol.6
    • Ramonet, D.1    Daher, J.2    Lin, B.M.3    Stafa, K.4    Kim, J.5    Banerjee, R.6
  • 147
    • 84855515680 scopus 로고    scopus 로고
    • Paraquat neurotoxicity is mediated by the dopamine transporter and organic cation transporter-3
    • Rappold, P. M., Cui, M., Chesser, A. S., Tibbett, J., Grima, J. C., Duan, L., et al. (2011). Paraquat neurotoxicity is mediated by the dopamine transporter and organic cation transporter-3. Proc. Natl. Acad. Sci. U.S.A.108, 20766–20771. doi: 10.1073/pnas.1115141108
    • (2011) Proc. Natl. Acad. Sci. U.S.A. , vol.108 , pp. 20766-20771
    • Rappold, P.M.1    Cui, M.2    Chesser, A.S.3    Tibbett, J.4    Grima, J.C.5    Duan, L.6
  • 148
    • 0035017068 scopus 로고    scopus 로고
    • Sensitivity to MPTP is not increased in Parkinson’s disease-associated mutant alpha-synuclein transgenic mice
    • Rathke-Hartlieb, S., Kahle, P. J., Neumann, M., Ozmen, L., Haid, S., Okochi, M., et al. (2001). Sensitivity to MPTP is not increased in Parkinson’s disease-associated mutant alpha-synuclein transgenic mice. J. Neurochem.77, 1181–1184. doi: 10.1046/j.1471-4159.2001.00366.x
    • (2001) J. Neurochem. , vol.77 , pp. 1181-1184
    • Rathke-Hartlieb, S.1    Kahle, P.J.2    Neumann, M.3    Ozmen, L.4    Haid, S.5    Okochi, M.6
  • 149
    • 84897977607 scopus 로고    scopus 로고
    • Lewy body extracts from Parkinson disease brains trigger α-synuclein pathology and neurodegeneration in mice and monkeys
    • Recasens, A., Dehay, B., Bové, J., Carballo-Carbajal, I., Dovero, S., Pérez-Villalba, A., et al. (2014). Lewy body extracts from Parkinson disease brains trigger α-synuclein pathology and neurodegeneration in mice and monkeys. Ann. Neurol.75, 351–362. doi: 10.1002/ana.24066
    • (2014) Ann. Neurol. , vol.75 , pp. 351-362
    • Recasens, A.1    Dehay, B.2    Bové, J.3    Carballo-Carbajal, I.4    Dovero, S.5    Pérez-Villalba, A.6
  • 150
    • 0036605566 scopus 로고    scopus 로고
    • Differential neuropathological alterations in transgenic mice expressing alpha-synuclein from the platelet-derived growth factor and Thy-1 promoters
    • Rockenstein, E., Mallory, M., Hashimoto, M., Song, D., Shults, C. W., Lang, I., et al. (2002). Differential neuropathological alterations in transgenic mice expressing alpha-synuclein from the platelet-derived growth factor and Thy-1 promoters. J. Neurosci. Res.68, 568–578. doi: 10.1002/jnr.10231
    • (2002) J. Neurosci. Res. , vol.68 , pp. 568-578
    • Rockenstein, E.1    Mallory, M.2    Hashimoto, M.3    Song, D.4    Shults, C.W.5    Lang, I.6
  • 151
    • 0035077354 scopus 로고    scopus 로고
    • Motor behavioural changes after intracerebroventricular injection of 6-hydroxydopamine in the rat: An animal model of Parkinson’s disease
    • Rodríguez Díaz, M., Abdala, P., Barroso-Chinea, P., Obeso, J., and Gonzalez-Hernandez, T. (2001). Motor behavioural changes after intracerebroventricular injection of 6-hydroxydopamine in the rat: an animal model of Parkinson’s disease. Behav. Brain Res.122, 79–92. doi: 10.1016/S0166-4328(01)00168-1
    • (2001) Behav. Brain Res. , vol.122 , pp. 79-92
    • Rodríguez Díaz, M.1    Abdala, P.2    Barroso-Chinea, P.3    Obeso, J.4    Gonzalez-Hernandez, T.5
  • 152
    • 72149101615 scopus 로고    scopus 로고
    • Initial clinical manifestations of Parkinson’s disease: Features and pathophysiological mechanisms
    • Rodriguez-Oroz, M. C., Jahanshahi, M., Krack, P., Litvan, I., Macias, R., Bezard, E., et al. (2009). Initial clinical manifestations of Parkinson’s disease: features and pathophysiological mechanisms. Lancet Neurol. 8, 1128–1139. doi: 10.1016/S1474-4422(09)70293–70295
    • (2009) Lancet Neurol , vol.8 , pp. 1128-1139
    • Rodriguez-Oroz, M.C.1    Jahanshahi, M.2    Krack, P.3    Litvan, I.4    Macias, R.5    Bezard, E.6
  • 153
    • 84866850019 scopus 로고    scopus 로고
    • Progressive dopaminergic cell loss with unilateral-to-bilateral progression in a genetic model of Parkinson disease
    • Rousseaux, M. W. C., Marcogliese, P. C., Qu, D., Hewitt, S. J., Seang, S., Kim, R. H., et al. (2012). Progressive dopaminergic cell loss with unilateral-to-bilateral progression in a genetic model of Parkinson disease. Proc. Natl. Acad. Sci. U.S.A.109, 15918–15923. doi: 10.1073/pnas.1205102109
    • (2012) Proc. Natl. Acad. Sci. U.S.A. , vol.109 , pp. 15918-15923
    • Rousseaux, M.1    Marcogliese, P.C.2    Qu, D.3    Hewitt, S.J.4    Seang, S.5    Kim, R.H.6
  • 154
    • 84944865481 scopus 로고    scopus 로고
    • Heterogeneity of leucine-rich repeat kinase 2 mutations: Genetics, mechanisms and therapeutic implications
    • Rudenko, I. N., and Cookson, M. R. (2014). Heterogeneity of leucine-rich repeat kinase 2 mutations: genetics, mechanisms and therapeutic implications. Neurotherapeutics4, 738–750. doi: 10.1007/s13311-014-0284-z
    • (2014) Neurotherapeutics , vol.4 , pp. 738-750
    • Rudenko, I.N.1    Cookson, M.R.2
  • 155
    • 84904636970 scopus 로고    scopus 로고
    • Intramuscular injection of α-synuclein induces CNS α-synuclein pathology and a rapid-onset motor phenotype in transgenic mice
    • Sacino, A. N., Brooks, M., Thomas, M. A., McKinney, A. B., Lee, S., Regenhardt, R. W., et al. (2014). Intramuscular injection of α-synuclein induces CNS α-synuclein pathology and a rapid-onset motor phenotype in transgenic mice. Proc. Natl. Acad. Sci. U.S.A.111, 10732–10737. doi: 10.1073/pnas.1321785111
    • (2014) Proc. Natl. Acad. Sci. U.S.A. , vol.111 , pp. 10732-10737
    • Sacino, A.N.1    Brooks, M.2    Thomas, M.A.3    McKinney, A.B.4    Lee, S.5    Regenhardt, R.W.6
  • 156
    • 84899630517 scopus 로고    scopus 로고
    • Unaltered striatal dopamine release levels in young Parkin knockout, Pink1 knockout, DJ-1 knockout and LRRK2 R1441G transgenic mice
    • Sanchez, G., Varaschin, R. K., Büeler, H., Marcogliese, P. C., Park, D. S., and Trudeau, L.-E. (2014). Unaltered striatal dopamine release levels in young Parkin knockout, Pink1 knockout, DJ-1 knockout and LRRK2 R1441G transgenic mice. PLoS ONE9:e94826. doi: 10.1371/journal.pone.0094826
    • (2014) Plos ONE , vol.9
    • Sanchez, G.1    Varaschin, R.K.2    Büeler, H.3    Marcogliese, P.C.4    Park, D.S.5    Trudeau, L.-E.6
  • 157
    • 0028220834 scopus 로고
    • Progressive degeneration of nigros-triatal dopamine neurons following intrastriatal terminal lesions with 6-hydroxydopamine: A combined retrograde tracing and immunocytochemical study in the rat
    • Sauer, H., and Oertel, W. H. (1994). Progressive degeneration of nigros-triatal dopamine neurons following intrastriatal terminal lesions with 6-hydroxydopamine: a combined retrograde tracing and immunocytochemical study in the rat. Neuroscience59, 401–415. doi: 10.1016/0306-4522(94)90605-X
    • (1994) Neuroscience , vol.59 , pp. 401-415
    • Sauer, H.1    Oertel, W.H.2
  • 158
    • 84901639801 scopus 로고    scopus 로고
    • Parkin and PINK1: Much more than mitophagy
    • Scarffe, L. A., Stevens, D. A., Dawson, V. L., and Dawson, T. M. (2014). Parkin and PINK1: much more than mitophagy. Trends Neurosci. 37, 315–324. doi: 10.1016/j.tins.2014.03.004
    • (2014) Trends Neurosci , vol.37 , pp. 315-324
    • Scarffe, L.A.1    Stevens, D.A.2    Dawson, V.L.3    Dawson, T.M.4
  • 159
    • 0033950675 scopus 로고    scopus 로고
    • CNS plasticity and assessment of forelimb sensorimotor outcome in unilateral rat models of stroke, cortical ablation, parkinsonism and spinal cord injury
    • Schallert, T., Fleming, S. M., Leasure, J. L., Tillerson, J. L., and Bland, S. T. (2000). CNS plasticity and assessment of forelimb sensorimotor outcome in unilateral rat models of stroke, cortical ablation, parkinsonism and spinal cord injury. Neuropharmacology39, 777–787. doi: 10.1016/S0028-3908(00)00005-8
    • (2000) Neuropharmacology , vol.39 , pp. 777-787
    • Schallert, T.1    Fleming, S.M.2    Leasure, J.L.3    Tillerson, J.L.4    Bland, S.T.5
  • 160
    • 84906306620 scopus 로고    scopus 로고
    • Slowing of neurodegeneration in Parkinson’s disease and Huntington’s disease: Future therapeutic perspectives
    • Schapira, A. H. V., Olanow, C. W., Greenamyre, J. T., and Bezard, E. (2014). Slowing of neurodegeneration in Parkinson’s disease and Huntington’s disease: future therapeutic perspectives. Lancet 384, 545–555. doi: 10.1016/S0140-6736(14)61010–61012
    • (2014) Lancet , vol.384 , pp. 545-555
    • Schapira, A.1    Olanow, C.W.2    Greenamyre, J.T.3    Bezard, E.4
  • 161
    • 84877010484 scopus 로고    scopus 로고
    • Atp13a2-deficient mice exhibit neuronal ceroid lipofuscinosis, limited α-synuclein accumulation and age-dependent sensorimotor deficits
    • Schultheis, P. J., Fleming, S. M., Clippinger, A. K., Lewis, J., Tsunemi, T., Giasson, B., et al. (2013). Atp13a2-deficient mice exhibit neuronal ceroid lipofuscinosis, limited α-synuclein accumulation and age-dependent sensorimotor deficits. Hum. Mol. Genet.22, 2067–2082. doi: 10.1093/hmg/ddt057
    • (2013) Hum. Mol. Genet. , vol.22 , pp. 2067-2082
    • Schultheis, P.J.1    Fleming, S.M.2    Clippinger, A.K.3    Lewis, J.4    Tsunemi, T.5    Giasson, B.6
  • 162
    • 0025074518 scopus 로고
    • Dose-dependent destruction of the coeruleus-cortical and nigral-striatal projections by MPTP
    • Seniuk, N. A., Tatton, W. G., and Greenwood, C. E. (1990). Dose-dependent destruction of the coeruleus-cortical and nigral-striatal projections by MPTP. Brain Res. 527, 7–20. doi: 10.1016/0006-8993(90)91055-L
    • (1990) Brain Res , vol.527 , pp. 7-20
    • Seniuk, N.A.1    Tatton, W.G.2    Greenwood, C.E.3
  • 163
    • 0037229425 scopus 로고    scopus 로고
    • Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation
    • Sherer, T. B., Kim, J. H., Betarbet, R., and Greenamyre, J. T. (2003). Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp. Neurol.179, 9–16. doi: 10.1006/exnr.2002.8072
    • (2003) Exp. Neurol. , vol.179 , pp. 9-16
    • Sherer, T.B.1    Kim, J.H.2    Betarbet, R.3    Greenamyre, J.T.4
  • 164
    • 15544366886 scopus 로고    scopus 로고
    • Absence of inclusion body formation in the MPTP mouse model of Parkinson’s disease
    • Shimoji, M., Zhang, L., Mandir, A. S., Dawson, V. L., and Dawson, T. M. (2005). Absence of inclusion body formation in the MPTP mouse model of Parkinson’s disease. Brain Res. Mol. Brain Res.134, 103–108. doi: 10.1016/j.molbrainres.2005.01.012
    • (2005) Brain Res. Mol. Brain Res. , vol.134 , pp. 103-108
    • Shimoji, M.1    Zhang, L.2    Mandir, A.S.3    Dawson, V.L.4    Dawson, T.M.5
  • 165
    • 0242300619 scopus 로고    scopus 로고
    • Alpha-Synuclein locus triplication causes Parkinson’s disease
    • Singleton, A. B., Farrer, M., Johnson, J., Singleton, A., Hague, S., Kachergus, J., et al. (2003). alpha-Synuclein locus triplication causes Parkinson’s disease. Science 302, 841. doi: 10.1126/science.1090278
    • (2003) Science , vol.302 , pp. 841
    • Singleton, A.B.1    Farrer, M.2    Johnson, J.3    Singleton, A.4    Hague, S.5    Kachergus, J.6
  • 166
    • 0034060717 scopus 로고    scopus 로고
    • Pattern of dopaminergic loss in the striatum of humans with MPTP induced parkinsonism
    • Snow, B. J., Vingerhoets, F. J., Langston, J. W., Tetrud, J. W., Sossi, V., and Calne, D. B. (2000). Pattern of dopaminergic loss in the striatum of humans with MPTP induced parkinsonism. J. Neurol. Neurosurg. Psychiatry68, 313–316. doi: 10.1136/jnnp.68.3.313
    • (2000) J. Neurol. Neurosurg , vol.68 , pp. 313-316
    • Snow, B.J.1    Vingerhoets, F.J.2    Langston, J.W.3    Tetrud, J.W.4    Sossi, V.5    Calne, D.B.6
  • 167
    • 0030605073 scopus 로고    scopus 로고
    • Treatment of mice with methamphetamine produces cell loss in the substantia nigra
    • Sonsalla, P. K., Jochnowitz, N. D., Zeevalk, G. D., Oostveen, J. A., and Hall, E. D. (1996). Treatment of mice with methamphetamine produces cell loss in the substantia nigra. Brain Res. 738, 172–175. doi: 10.1016/0006-8993(96)00995-X
    • (1996) Brain Res , vol.738 , pp. 172-175
    • Sonsalla, P.K.1    Jochnowitz, N.D.2    Zeevalk, G.D.3    Oostveen, J.A.4    Hall, E.D.5
  • 168
    • 78650170744 scopus 로고    scopus 로고
    • Selective noradrenergic vulnerability in α-synuclein transgenic mice
    • Sotiriou, E., Vassilatis, D. K., Vila, M., and Stefanis, L. (2010). Selective noradrenergic vulnerability in α-synuclein transgenic mice. Neurobiol. Aging 31, 2103–2114. doi: 10.1016/j.neurobiolaging.2008.11.010
    • (2010) Neurobiol. Aging , vol.31 , pp. 2103-2114
    • Sotiriou, E.1    Vassilatis, D.K.2    Vila, M.3    Stefanis, L.4
  • 169
    • 70349917814 scopus 로고    scopus 로고
    • Mutant alpha-synuclein overexpression mediates early proinflammatory activity
    • Su, X., Federoff, H. J., and Maguire-Zeiss, K. A. (2009). Mutant alpha-synuclein overexpression mediates early proinflammatory activity. Neurotox. Res.16, 238–254. doi: 10.1007/s12640-009-9053-x
    • (2009) Neurotox. Res. , vol.16 , pp. 238-254
    • Su, X.1    Federoff, H.J.2    Maguire-Zeiss, K.A.3
  • 170
    • 0027954983 scopus 로고
    • Methamphetamine protects against MPTP neurotoxicity in C57BL mice
    • Sziráki, I., Kardos, V., Patthy, M., Pátfalusi, M., and Budai, G. (1994). Methamphetamine protects against MPTP neurotoxicity in C57BL mice. Eur. J. Pharmacol. 251, 311–314. doi: 10.1016/0014-2999(94)90416-2
    • (1994) Eur. J. Pharmacol , vol.251 , pp. 311-314
    • Sziráki, I.1    Kardos, V.2    Patthy, M.3    Pátfalusi, M.4    Budai, G.5
  • 171
    • 0030931721 scopus 로고    scopus 로고
    • VMAT2 knockout mice: heterozygotes display reduced amphetamine-conditioned reward, enhanced amphetamine locomotion, and enhanced MPTP toxicity
    • Takahashi, N., Miner, L. L., Sora, I., Ujike, H., Revay, R. S., Kostic, V., et al. (1997). VMAT2 knockout mice: heterozygotes display reduced amphetamine-conditioned reward, enhanced amphetamine locomotion, and enhanced MPTP toxicity. Proc. Natl. Acad. Sci. U.S.A. 94, 9938–9943.
    • (1997) Proc. Natl. Acad. Sci. U.S.A , vol.94 , pp. 9938-9943
    • Takahashi, N.1    Miner, L.L.2    Sora, I.3    Ujike, H.4    Revay, R.S.5    Kostic, V.6
  • 173
    • 84886587153 scopus 로고    scopus 로고
    • Reduced vesicular storage of catecholamines causes progressive degeneration in the locus ceruleus
    • Taylor, T. N., Alter, S. P., Wang, M., Goldstein, D. S., and Miller, G. W. (2014). Reduced vesicular storage of catecholamines causes progressive degeneration in the locus ceruleus. Neuropharmacology76(Pt A), 97–105. doi: 10.1016/j.neuropharm.2013.08.033
    • (2014) Neuropharmacology , vol.76 , pp. 97-105
    • Taylor, T.N.1    Alter, S.P.2    Wang, M.3    Goldstein, D.S.4    Miller, G.W.5
  • 174
    • 79955067247 scopus 로고    scopus 로고
    • VMAT2-deficient mice display nigral and extranigral pathology and motor and nonmotor symptoms of Parkinson’s disease
    • 124165
    • Taylor, T. N., Caudle, W. M., and Miller, G. W. (2011). VMAT2-deficient mice display nigral and extranigral pathology and motor and nonmotor symptoms of Parkinson’s disease. Parkinsons Dis. 2011, 124165. doi: 10.4061/2011/124165
    • (2011) Parkinsons Dis , vol.2011
    • Taylor, T.N.1    Caudle, W.M.2    Miller, G.W.3
  • 175
    • 77952319464 scopus 로고    scopus 로고
    • Behavioral phenotyp-ing of mouse models of Parkinson’s disease
    • Taylor, T. N., Greene, J. G., and Miller, G. W. (2010). Behavioral phenotyp-ing of mouse models of Parkinson’s disease. Behav. Brain Res.211, 1–10. doi: 10.1016/j.bbr.2010.03.004
    • (2010) Behav. Brain Res. , vol.211 , pp. 1-10
    • Taylor, T.N.1    Greene, J.G.2    Miller, G.W.3
  • 176
    • 0034637267 scopus 로고    scopus 로고
    • Potentiated and preferential effects of combined paraquat and maneb on nigrostriatal dopamine systems: Environmental risk factors for Parkinson’s disease?
    • Thiruchelvam, M., Brockel, B. J., Richfield, E. K., Baggs, R. B., and Cory-Slechta, D. A. (2000). Potentiated and preferential effects of combined paraquat and maneb on nigrostriatal dopamine systems: environmental risk factors for Parkinson’s disease? Brain Res. 873, 225–234. doi: 10.1016/S0006-8993(00) 02496-3
    • (2000) Brain Res , vol.873 , pp. 225-234
    • Thiruchelvam, M.1    Brockel, B.J.2    Richfield, E.K.3    Baggs, R.B.4    Cory-Slechta, D.A.5
  • 177
    • 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, E. K., Baggs, R. B., Tank, A. W., Di Monte, D. A., et al. (2003). Age-related irreversible progressive nigrostriatal dopaminergic neurotoxicity in the paraquat and maneb model of the Parkinson’s disease phenotype. Eur. J. Neurosci. 18, 589–600. doi: 10.1046/j.1460-9568.2003.02781.x
    • (2003) Eur. J. Neurosci , vol.18 , pp. 589-600
    • Thiruchelvam, M.1    McCormack, A.2    Richfield, E.K.3    Baggs, R.B.4    Tank, A.W.5    Di Monte, D.A.6
  • 178
    • 1542359628 scopus 로고    scopus 로고
    • Risk factors for dopaminergic neuron loss in human alpha-synuclein transgenic mice
    • Thiruchelvam, M. J., Powers, J. M., Cory-Slechta, D. A., and Richfield, E. K. (2004). Risk factors for dopaminergic neuron loss in human alpha-synuclein transgenic mice. Eur. J. Neurosci. 19, 845–854. doi: 10.1111/j.0953-816X.2004.03139.x
    • (2004) Eur. J. Neurosci , vol.19 , pp. 845-854
    • Thiruchelvam, M.J.1    Powers, J.M.2    Cory-Slechta, D.A.3    Richfield, E.K.4
  • 179
    • 76049120240 scopus 로고    scopus 로고
    • Methamphetamine-induced neurotoxicity: The road to Parkinson’s disease. Pharmacol
    • Thrash, B., Thiruchelvan, K., Ahuja, M., Suppiramaniam, V., and Dhanasekaran, M. (2009). Methamphetamine-induced neurotoxicity: the road to Parkinson’s disease. Pharmacol. Rep. 61, 966–977. doi: 10.1016/S1734-1140(09)70158-6
    • (2009) Rep , vol.61 , pp. 966-977
    • Thrash, B.1    Thiruchelvan, K.2    Ahuja, M.3    Suppiramaniam, V.4    Dhanasekaran, M.5
  • 180
    • 84884907135 scopus 로고    scopus 로고
    • A guide to neurotoxic animal models of Parkinson’s disease
    • Tieu, K. (2011). A guide to neurotoxic animal models of Parkinson’s disease. Cold Spring Harb. Perspect. Med. 1, a009316. doi: 10.1101/cshperspect.a009316
    • (2011) Cold Spring Harb. Perspect. Med , vol.1
    • Tieu, K.1
  • 182
    • 77953090478 scopus 로고    scopus 로고
    • Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of alpha-synuclein, and apoptotic cell death inaged mice
    • Tong, Y., Yamaguchi, H., Giaime, E., Boyle, S., Kopan, R., Kelleher, R. J., et al. (2010). Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of alpha-synuclein, and apoptotic cell death inaged mice. Proc. Natl. Acad. Sci. U.S.A. 107, 9879–9884. doi: 10.1073/pnas.1004676107
    • (2010) Proc. Natl. Acad. Sci. U.S.A , vol.107 , pp. 9879-9884
    • Tong, Y.1    Yamaguchi, H.2    Giaime, E.3    Boyle, S.4    Kopan, R.5    Kelleher, R.J.6
  • 183
    • 0022330533 scopus 로고
    • Effects of chronic methamphetamine on the nigral-striatal dopamine system in rat brain: Tyrosine hydroxylase immunochemistry and quantitative light microscopic studies
    • Trulson, M. E., Cannon, M. S., Faegg, T. S., and Raese, J. D. (1985). Effects of chronic methamphetamine on the nigral-striatal dopamine system in rat brain: tyrosine hydroxylase immunochemistry and quantitative light microscopic studies. Brain Res. Bull. 15, 569–577. doi: 10.1016/0361-9230(85)90206–90200
    • (1985) Brain Res. Bull , vol.15 , pp. 569-577
    • Trulson, M.E.1    Cannon, M.S.2    Faegg, T.S.3    Raese, J.D.4
  • 184
    • 84949117317 scopus 로고    scopus 로고
    • Conditional expression of Parkinson’s disease-related R1441C LRRK2 in midbrain dopaminergic neurons of mice causes nuclear abnormalities without neurodegeneration
    • Tsika, E., Kannan, M., Foo, C. S.-Y., Dikeman, D., Glauser, L., Gellhaar, S., et al. (2014). Conditional expression of Parkinson’s disease-related R1441C LRRK2 in midbrain dopaminergic neurons of mice causes nuclear abnormalities without neurodegeneration. Neurobiol. Dis. 71, 345–358. doi: 10.1016/j.nbd.2014.08.027
    • (2014) Neurobiol. Dis , vol.71 , pp. 345-358
    • Tsika, E.1    Kannan, M.2    Foo, C.S.3    Dikeman, D.4    Glauser, L.5    Gellhaar, S.6
  • 185
    • 77955625476 scopus 로고    scopus 로고
    • Presynaptic dopaminergic compartment determines the susceptibility to L-DOPA-induced dyskinesia in rats
    • Ulusoy, A., Sahin, G., and Kirik, D. (2010). Presynaptic dopaminergic compartment determines the susceptibility to L-DOPA-induced dyskinesia in rats. Proc. Natl. Acad. Sci. U.S.A.107, 13159–13164. doi: 10.1073/pnas.1003432107
    • (2010) Proc. Natl. Acad. Sci. U.S.A. , vol.107 , pp. 13159-13164
    • Ulusoy, A.1    Sahin, G.2    Kirik, D.3
  • 186
    • 0014376793 scopus 로고
    • 6-Hydroxy-dopamine induced degeneration of central monoamine neurons
    • Ungerstedt, U. (1968). 6-Hydroxy-dopamine induced degeneration of central monoamine neurons. Eur. J. Pharmacol. 5, 107–110. doi: 10.1016/0014-2999(68)90164-7
    • (1968) Eur. J. Pharmacol , vol.5 , pp. 107-110
    • Ungerstedt, U.1
  • 187
    • 0038614833 scopus 로고    scopus 로고
    • Pitx3 is required for motor activity and for survival of a subset of midbrain dopaminergic neurons
    • van den Munckhof, P., Luk, K. C., Ste-Marie, L., Montgomery, J., Blanchet, P. J., Sadikot, A. F., et al. (2003). Pitx3 is required for motor activity and for survival of a subset of midbrain dopaminergic neurons. Development130, 2535–2542. doi: 10.1242/dev.00464
    • (2003) Development , vol.130 , pp. 2535-2542
    • Van Den Munckhof, P.1    Luk, K.C.2    Ste-Marie, L.3    Montgomery, J.4    Blanchet, P.J.5    Sadikot, A.F.6
  • 189
    • 84897018170 scopus 로고    scopus 로고
    • Long-term overexpression of human wild-type and T240R mutant Parkin in rat substantia nigra induces progressive dopamin-ergic neurodegeneration
    • Van Rompuy, A.-S., Lobbestael, E., Van der Perren, A., Van den Haute, C., and Baekelandt, V. (2014). Long-term overexpression of human wild-type and T240R mutant Parkin in rat substantia nigra induces progressive dopamin-ergic neurodegeneration. J. Neuropathol. Exp. Neurol.73, 159–174. doi: 10.1097/NEN.0000000000000039
    • (2014) J. Neuropathol. Exp. Neurol. , vol.73 , pp. 159-174
    • Van Rompuy, A.-S.1    Lobbestael, E.2    Van Der Perren, A.3    Van Den Haute, C.4    Baekelandt, V.5
  • 190
    • 80054755695 scopus 로고    scopus 로고
    • Pathological roles of α-synuclein in neurological disorders
    • Vekrellis, K., Xilouri, M., Emmanouilidou, E., Rideout, H. J., and Stefanis, L. (2011). Pathological roles of α-synuclein in neurological disorders. Lancet Neurol. 10, 1015–1025. doi: 10.1016/S1474-4422(11)70213–70217
    • (2011) Lancet Neurol , vol.10 , pp. 1015-1025
    • Vekrellis, K.1    Xilouri, M.2    Emmanouilidou, E.3    Rideout, H.J.4    Stefanis, L.5
  • 191
    • 0033966487 scopus 로고    scopus 로고
    • Alpha-synuclein up-regulation in substantia nigra dopaminergic neurons following administration of the parkinsonian toxin MPTP
    • Vila, M., Vukosavic, S., Jackson-Lewis, V., Neystat, M., Jakowec, M., and Przedborski, S. (2000). Alpha-synuclein up-regulation in substantia nigra dopaminergic neurons following administration of the parkinsonian toxin MPTP. J. Neurochem. 74, 721–729. doi: 10.1046/j.1471-4159.2000.740721.x
    • (2000) J. Neurochem , vol.74 , pp. 721-729
    • Vila, M.1    Vukosavic, S.2    Jackson-Lewis, V.3    Neystat, M.4    Jakowec, M.5    Przedborski, S.6
  • 192
    • 84876213313 scopus 로고    scopus 로고
    • The PINK1-Parkin pathway promotes both mitophagy and selective respiratory chain turnover in vivo
    • Vincow, E. S., Merrihew, G., Thomas, R. E., Shulman, N. J., Beyer, R. P., MacCoss, M. J., et al. (2013). The PINK1-Parkin pathway promotes both mitophagy and selective respiratory chain turnover in vivo. Proc. Natl. Acad. Sci. U.S.A.110, 6400–6405. doi: 10.1073/pnas.1221132110
    • (2013) Proc. Natl. Acad. Sci. U.S.A. , vol.110 , pp. 6400-6405
    • Vincow, E.S.1    Merrihew, G.2    Thomas, R.E.3    Shulman, N.J.4    Beyer, R.P.5    Maccoss, M.J.6
  • 193
  • 194
    • 0018770389 scopus 로고
    • Methamphetamine-induced changes in brain catecholamines in rats and guinea pigs
    • Wagner, G. C., Seiden, L. S., and Schuster, C. R. (1979). Methamphetamine-induced changes in brain catecholamines in rats and guinea pigs. Drug Alcohol Depend. 4, 435–438. doi: 10.1016/0376-8716(79)90076–90070
    • (1979) Drug Alcohol Depend , vol.4 , pp. 435-438
    • Wagner, G.C.1    Seiden, L.S.2    Schuster, C.R.3
  • 195
    • 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., Sakagami, J., Ukai, Y., Ono, M., et al. (2008). Selective loss of nigral dopamine neurons induced by overexpression of truncated human alpha-synuclein in mice. Neurobiol. Aging29, 574–585. doi: 10.1016/j.neurobiolaging.2006.11.017
    • (2008) Neurobiol. Aging , vol.29 , pp. 574-585
    • Wakamatsu, M.1    Ishii, A.2    Iwata, S.3    Sakagami, J.4    Ukai, Y.5    Ono, M.6
  • 196
    • 79151473945 scopus 로고    scopus 로고
    • Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice
    • Winner, B., Melrose, H. L., Zhao, C., Hinkle, K. M., Yue, M., Kent, C., et al. (2011). Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol. Dis.41, 706–716. doi: 10.1016/j.nbd.2010.12.008
    • (2011) Neurobiol. Dis. , vol.41 , pp. 706-716
    • Winner, B.1    Melrose, H.L.2    Zhao, C.3    Hinkle, K.M.4    Yue, M.5    Kent, C.6
  • 197
    • 70849084551 scopus 로고    scopus 로고
    • Stereo-taxical infusion of rotenone: A reliable rodent model for Parkinson’s disease
    • Xiong, N., Huang, J., Zhang, Z., Zhang, Z., Xiong, J., Liu, X., et al. (2009). Stereo-taxical infusion of rotenone: a reliable rodent model for Parkinson’s disease. PLoS ONE4:e7878. doi: 10.1371/journal.pone.0007878
    • (2009) Plos ONE , vol.4
    • Xiong, N.1    Huang, J.2    Zhang, Z.3    Zhang, Z.4    Xiong, J.5    Liu, X.6
  • 198
    • 84907484591 scopus 로고    scopus 로고
    • Mutations in the ATP13A2 gene and parkinsonism: A preliminary review
    • Yang, X., and Xu, Y. (2014). Mutations in the ATP13A2 gene and parkinsonism: a preliminary review. Biomed Res. Int.2014, 371256. doi: 10.1155/2014/371256
    • (2014) Biomed Res. Int. , vol.2014 , pp. 371256
    • Yang, X.1    Xu, Y.2
  • 199
    • 84930668368 scopus 로고    scopus 로고
    • Defective autophagy in Parkinson’s disease: Lessons from genetics
    • [Epub ahead of print]
    • Zhang, H., Duan, C., and Yang, H. (2014). Defective autophagy in Parkinson’s disease: lessons from genetics. Mol. Neurobiol. doi: 10.1007/s12035-014-8787–8785 [Epub ahead of print].
    • (2014) Mol. Neurobiol
    • Zhang, H.1    Duan, C.2    Yang, H.3
  • 200
    • 84858342244 scopus 로고    scopus 로고
    • Age-related changes in dopamine signaling in Nurr1 deficient mice as a model of Parkinson’s disease
    • Zhang, L., Le, W., Xie, W., and Dani, J. A. (2012). Age-related changes in dopamine signaling in Nurr1 deficient mice as a model of Parkinson’s disease. Neurobiol. Aging 33, 1001.e7–1001.e16. doi: 10.1016/j.neurobiolaging.2011.03.022
    • (2012) Neurobiol. Aging , vol.33
    • Zhang, L.1    Le, W.2    Xie, W.3    Dani, J.A.4
  • 201
    • 79960972765 scopus 로고    scopus 로고
    • Temporal expression of mutant LRRK2 in adult rats impairs dopamine reuptake
    • Zhou, H., Huang, C., Tong, J., Hong, W. C., Liu, Y.-J., and Xia, X.-G. (2011). Temporal expression of mutant LRRK2 in adult rats impairs dopamine reuptake. Int. J. Biol. Sci. 7, 753–761. doi: 10.7150/ijbs.7.753
    • (2011) Int. J. Biol. Sci , vol.7 , pp. 753-761
    • Zhou, H.1    Huang, C.2    Tong, J.3    Hong, W.C.4    Liu, Y.-J.5    Xia, X.-G.6
  • 202


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.