Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-42gr6 Total loading time: 0 Render date: 2024-04-18T23:04:42.945Z Has data issue: false hasContentIssue false

24 - Animal models of Parkinson's disease

Published online by Cambridge University Press:  04 November 2009

Anumantha G. Kanthasamy
Affiliation:
Department of Biomedical Sciences 2062 Veterinary Medicine Building Iowa State University Ames, IA 50011 USA
Siddharth Kaul
Affiliation:
Department of Biomedical Sciences 2062 Veterinary Medicine Building Iowa State University Ames, IA 50011 USA
Turgut Tatlisumak
Affiliation:
Helsinki University Central Hospital
Marc Fisher
Affiliation:
University of Massachusetts Medical School
Get access

Summary

Introduction

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized primarily by the gradual dopaminergic loss in the substantia nigra of the midbrain region. Development of PD can be sporadic or can be associated with genetic mutations and deficiencies, or may result from the combination of these two precipitating factors. The pathogenesis of PD has been studied in numerous experimental models developed to replicate the salient features of the disease in a controlled environment. Although no single model exists today that mimics all the neurological and neuropathological features of PD, each model presents a particular aspect of the disease process induced either by natural or artificial toxic agents or by genetically induced deficiencies in experimental animals. Epidemiological and laboratory results suggest that environmental factors play a predominant role in the induction and propagation of dopaminergic degeneration. However, numerous familial cases indicate that development of PD might be aggravated by pre-existing genetic deficiencies that act as predisposing factors. This chapter describes in detail the extensive research conducted using animal and tissue-culture models of Parkinson's disease induced by both toxins and genetic manipulation. Furthermore, salient experimental findings are thoroughly described with regard to current perspectives on neurotoxic mechanisms of genetic variations and environmental toxins.

6-Hydroxydopamine model of PD

6-Hydroxydopamine (6-OHDA) was first demonstrated to effectively replicate Parkinsonian neurotoxic pathology in rats by stereotaxic nigral injection in rats as early as 1975.

Type
Chapter
Information
Handbook of Experimental Neurology
Methods and Techniques in Animal Research
, pp. 411 - 437
Publisher: Cambridge University Press
Print publication year: 2006

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Mendez, JS, Finn, BW. Use of 6-hydroxydopamine to create lesions in catecholamine neurons in rats. J. Neurosurg. 1975, 42: 166–173.CrossRefGoogle ScholarPubMed
Glinka, Y, Gassen, M, Youdim, MB. Mechanism of 6-hydroxydopamine neurotoxicity. J. Neural Transm. (Suppl.) 1997, 50: 55–66.CrossRefGoogle ScholarPubMed
Ungerstedt, U. 6-Hydroxydopamine induced degeneration of central monoamine neurons. Eur. J. Pharmacol. 1968, 5: 107–110.CrossRefGoogle Scholar
Ungerstedt, U. Adipsia and aphagia after 6-hydroxydopamine-induced degeneration of the nigro-striatal dopamine system. Acta Physiol. Scand. (Suppl.) 1971, 36 7: 95–122.CrossRefGoogle Scholar
Roedter, A, Winkler, C, Samii, M, et al. Comparison of unilateral and bilateral intrastriatal 6-hydroxydopamine-induced axon terminal lesions: evidence for interhemispheric functional coupling of the two nigrostriatal pathways. J. Comp. Neurol. 2001, 432: 217–229.CrossRefGoogle ScholarPubMed
Hefti, F, Melamed, E, Wurtman, RJ. Partial lesions of the dopaminergic nigrostriatal system in rat brain: biochemical characterization. Brain Res. 1980, 195: 123–137.CrossRefGoogle ScholarPubMed
Przedborski, S, Levivier, M, Jiang, H, et al. Dose-dependent lesions of the dopaminergic nigrostriatal pathway induced by intrastriatal injection of 6-hydroxydopamine. Neuroscience 1995, 67: 631–647.CrossRefGoogle ScholarPubMed
Srinivasan, J, Schmidt, WJ. Potentiation of Parkinsonian symptoms by depletion of locus coeruleus noradrenaline in 6-hydroxydopamine-induced partial degeneration of substantia nigra in rats. Eur. J. Neurosci. 2003, 17: 2586–2592.CrossRefGoogle ScholarPubMed
Whishaw, IQ, Pellis, SM, Gorny, BP. Skilled reaching in rats and humans: evidence for parallel development or homology. Behav. Brain Res. 1992, 47: 59–70.CrossRefGoogle ScholarPubMed
Kirik, D, Georgievska, B, Rosenblad, C, Bjorklund, A. Delayed infusion of GDNF promotes recovery of motor function in the partial lesion model of Parkinson's disease. Eur. J. Neurosci. 2001, 13: 1589–1599.CrossRefGoogle ScholarPubMed
Akerud, P, Canals, JM, Snyder, EY, Arenas, E. Neuroprotection through delivery of glial cell line-derived neurotrophic factor by neural stem cells in a mouse model of Parkinson's disease. J. Neurosci. 2001, 21: 8108–8118.CrossRefGoogle Scholar
Paul, G, Meissner, W, Rein, S, et al. Ablation of the subthalamic nucleus protects dopaminergic phenotype but not cell survival in a rat model of Parkinson's disease. Exp. Neurol. 2004, 185: 272–280.CrossRefGoogle ScholarPubMed
Carrasco, E, Werner, P. Selective destruction of dopaminergic neurons by low concentrations of 6-OHDA and MPP+: protection by acetylsalicylic acid aspirin. Parkinsonism Relat. Disord. 2002, 8: 407–411.CrossRefGoogle ScholarPubMed
Yoshioka, M, Tanaka, K, Miyazaki, I, et al. The dopamine agonist cabergoline provides neuroprotection by activation of the glutathione system and scavenging free radicals. Neurosci. Res. 2002, 43: 259–267.CrossRefGoogle ScholarPubMed
Langston, JW. Mechanisms underlying neuronal degeneration in Parkinson's disease: an experimental and theoretical treatise. Mov. Disord. 1989, 4(Suppl. 1): S15–S25.CrossRefGoogle ScholarPubMed
Bradbury, AJ, Costall, B, Domeney, AM, et al. 1-methyl-4-phenylpyridine is neurotoxic to the nigrostriatal dopamine pathway. Nature 1986, 319: 56–57.CrossRefGoogle ScholarPubMed
Langston, JW, Ballard, P, Tetrud, JW, Irwin, I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science 1983, 219: 979–980.CrossRefGoogle ScholarPubMed
Chiba, K, Trevor, A, Castagnoli, N Jr. Metabolism of the neurotoxic tertiary amine, MPTP, by brain monoamine oxidase. Biochem. Biophys. Res. Commun. 1984, 120: 574–578.CrossRefGoogle ScholarPubMed
Zuddas, A, Fascetti, F, Corsini, GU, Piccardi, MP. In brown Norway rats, MPP+ is accumulated in the nigrostriatal dopaminergic terminals but it is not neurotoxic: a model of natural resistance to MPTP toxicity. Exp. Neurol. 1994, 127: 54–61.CrossRefGoogle Scholar
Da Cunha, C, Gevaerd, MS, Vital, MA, et al. Memory disruption in rats with nigral lesions induced by MPTP: a model for early Parkinson's disease amnesia. Behav. Brain Res. 2001, 124: 9–18.CrossRefGoogle ScholarPubMed
Miyoshi, E, Wietzikoski, S, Camplessei, M, et al. Impaired learning in a spatial working memory version and in a cued version of the water maze in rats with MPTP-induced mesencephalic dopaminergic lesions. Brain Res. Bull. 2002, 58: 41–47.CrossRefGoogle Scholar
Lau, YS, Meredith, GE. From drugs of abuse to Parkinsonism: the MPTP mouse model of Parkinson's disease. Methods Mol. Med. 2003, 79: 103–116.Google ScholarPubMed
Sedelis, M, Hofele, K, Auburger, GW, et al. MPTP susceptibility in the mouse: behavioral, neurochemical, and histological analysis of gender and strain differences. Behav. Genet. 2000, 30: 171–182.CrossRefGoogle ScholarPubMed
Langston, JW, Langston, EB, Irwin, I. MPTP-induced Parkinsonism in human and non-human primates: clinical and experimental aspects. Acta Neurol. Scand. (Suppl.) 1984, 100: 49–54.Google ScholarPubMed
Bezard, E, Gross, CE, Fournier, MC, et al. Absence of MPTP-induced neuronal death in mice lacking the dopamine transporter. Exp. Neurol. 1999, 155: 268–273.CrossRefGoogle ScholarPubMed
Przedborski, S, Jackson-Lewis, V. Mechanisms of MPTP toxicity. Mov. Disord. 1998, 13 (Suppl. 1): 35–38.Google ScholarPubMed
Cosi, C, Marien, M. Implication of poly (ADP-ribose) polymerase (PARP) in neurodegeneration and brain energy metabolism: decreases in mouse brain NAD+ and ATP caused by MPTP are prevented by the PARP inhibitor benzamide. Ann. NY Acad. Sci. 1999, 890: 227–239.CrossRefGoogle ScholarPubMed
Cleeter, MW, Cooper, JM, Schapira, AH. Irreversible inhibition of mitochondrial complex I by 1-methyl-4-phenylpyridinium: evidence for free radical involvement. J. Neurochem. 1992, 58: 786–789.CrossRefGoogle ScholarPubMed
Kaul, S, Kanthasamy, A, Kitazawa, M, Anantharam, V, Kanthasamy, AG. Caspase-3 dependent proteolytic activation of protein kinase Cdelta mediates and regulates 1-methyl-4-phenylpyridinium (MPP+)-induced apoptotic cell death in dopaminergic cells: relevance to oxidative stress in dopaminergic degeneration. Eur. J. Neurosci. 2003, 18: 1387–1401.CrossRefGoogle Scholar
Obata, T, Yamanaka, Y, Kinemuchi, H, Oreland, L. Release of dopamine by perfusion with 1-methyl-4-phenylpyridinium ion (MPP+) into the striatum is associated with hydroxyl free radical generation. Brain Res. 2001, 906: 170–175.CrossRefGoogle ScholarPubMed
Miyako, K, Irie, T, Muta, T, et al. 1-Methyl-4-phenylpyridinium ion (MPP+) selectively inhibits the replication of mitochondrial DNA. Eur. J. Biochem. 1999, 259: 412–418.CrossRefGoogle ScholarPubMed
Fiskum, G, Starkov, A, Polster, BM, Chinopoulos, C. Mitochondrial mechanisms of neural cell death and neuroprotective interventions in Parkinson's disease. Ann. NY Acad. Sci. 2003, 991: 111–119.CrossRefGoogle ScholarPubMed
Vila, M, Jackson-Lewis, V, Vukosavic, S, et al. Bax ablation prevents dopaminergic neurodegeneration in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease. Proc. Natl Acad. Sci. USA 2001, 98: 2837–2842.CrossRefGoogle ScholarPubMed
Cassarino, DS, Parks, JK, Parker, WD Jr, Bennett, JP Jr. The Parkinsonian neurotoxin MPP+ opens the mitochondrial permeability transition pore and releases cytochrome c in isolated mitochondria via an oxidative mechanism. Biochim. Biophys. Acta 1999, 1453: 49–62.CrossRefGoogle ScholarPubMed
Cheng, EH, Wei, MC, Weiler, S, et al. BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol. Cell 2001, 8: 705–711.CrossRefGoogle ScholarPubMed
Choi, WS, Lee, EH, Chung, CW, et al. Cleavage of Bax is mediated by caspase-dependent or -independent calpain activation in dopaminergic neuronal cells: protective role of Bcl-2. J. Neurochem. 2001, 77: 1531–1541.CrossRefGoogle ScholarPubMed
Oh, YJ, Wong, SC, Moffat, M, O'Malley, KL. Overexpression of Bcl-2 attenuates MPP+, but not 6-ODHA, induced cell death in a dopaminergic neuronal cell line. Neurobiol. Dis. 1995, 2: 157–167.CrossRefGoogle ScholarPubMed
Hassouna, I, Wickert, H, Zimmermann, M, Gillardon, F. Increase in bax expression in substantia nigra following 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment of mice. Neurosci. Lett. 1996, 204: 85–88.CrossRefGoogle Scholar
Zou, H, Li, Y, Liu, X, Wang, X. An APAF-1 cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. J. Biol. Chem. 1999, 274: 11549–11556.CrossRefGoogle ScholarPubMed
Du, Y, Dodel, RC, Bales, KR, et al. Involvement of a caspase-3-like cysteine protease in 1-methyl-4-phenylpyridinium-mediated apoptosis of cultured cerebellar granule neurons. J. Neurochem. 1997, 69: 1382–1388.CrossRefGoogle ScholarPubMed
Hartmann, A, Hunot, S, Michel, PP, et al. Caspase-3: a vulnerability factor and final effector in apoptotic death of dopaminergic neurons in Parkinson's disease. Proc. Natl Acad. Sci. USA. 2000, 97: 2875–2880.CrossRefGoogle ScholarPubMed
Turmel, H, Hartmann, A, Parain, K, et al. Caspase-3 activation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. Mov. Disord. 2001, 16: 185–189.CrossRefGoogle ScholarPubMed
Dodel, RC, Du, Y, Bales, KR, et al. Peptide inhibitors of caspase-3-like proteases attenuate 1-methyl-4-phenylpyridinum-induced toxicity of cultured fetal rat mesencephalic dopamine neurons. Neuroscience 1998, 86: 701–707.CrossRefGoogle ScholarPubMed
Viswanath, V, Wu, Y, Boonplueang, R, et al. Caspase-9 activation results in downstream caspase-8 activation and bid cleavage in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease. J. Neurosci. 2001, 21: 9519–9528.CrossRefGoogle ScholarPubMed
Mandal, M, Adam, L, Kumar, R. Redistribution of activated caspase-3 to the nucleus during butyric acid-induced apoptosis. Biochem. Biophys. Res. Commun. 1999, 260: 775–780.CrossRefGoogle ScholarPubMed
Casciola-Rosen, L, Nicholson, DW, Chong, T, et al. Apopain/CPP32 cleaves proteins that are essential for cellular repair: a fundamental principle of apoptotic death. J. Exp. Med. 1996, 183: 1957–1964.CrossRefGoogle ScholarPubMed
Mandir, AS, Przedborski, S, Jackson-Lewis, V, et al. Poly(ADP-ribose) polymerase activation mediates 1-methyl-4-phenyl-1, 2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism. Proc. Natl Acad. Sci. USA 1999, 96: 5774–5779.CrossRefGoogle ScholarPubMed
Cosi, C, Colpaert, F, Koek, W, Degryse, A, Marien, M. Poly(ADP-ribose) polymerase inhibitors protect against MPTP-induced depletions of striatal dopamine and cortical noradrenaline in C57B1/6 mice. Brain Res. 1996, 729: 264–269.Google ScholarPubMed
Han, Z, Malik, N, Carter, T, et al. DNA-dependent protein kinase is a target for a CPP32-like apoptotic protease. J. Biol. Chem. 1996, 271: 25035–25040.CrossRefGoogle ScholarPubMed
Tatton, NA, Kish, SJ. In situ detection of apoptotic nuclei in the substantia nigra compacta of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice using terminal deoxynucleotidyl transferase labelling and acridine orange staining. Neuroscience 1997, 77: 1037–1048.CrossRefGoogle ScholarPubMed
Hartmann, A, Troadec, JD, Hunot, S, et al. Caspase-8 is an effector in apoptotic death of dopaminergic neurons in Parkinson's disease, but pathway inhibition results in neuronal necrosis. J. Neurosci. 2001, 21: 2247–2255.CrossRefGoogle ScholarPubMed
Duan, W, Zhu, X, Ladenheim, B, et al. p53 inhibitors preserve dopamine neurons and motor function in experimental parkinsonism. Ann. Neurol. 2002, 52: 597–606.CrossRefGoogle ScholarPubMed
Wang, H, Shimoji, M, Yu, SW, Dawson, TM, Dawson, VL. Apoptosis inducing factor and PARP-mediated injury in the MPTP mouse model of Parkinson's disease. Ann. NY Acad. Sci. 2003, 991: 132–139.CrossRefGoogle ScholarPubMed
Saporito, MS, Brown, EM, Miller, MS, Carswell, S. CEP-1347/KT-7515, an inhibitor of c-jun N-terminal kinase activation, attenuates the 1-methyl-4-phenyl tetrahydropyridine-mediated loss of nigrostriatal dopaminergic neurons in vivo. J. Pharmacol. Exp. Ther. 1999, 288: 421–427.Google ScholarPubMed
Saporito, MS, Thomas, BA, Scott, RW. MPTP activates c-Jun NH(2)-terminal kinase (JNK) and its upstream regulatory kinase MKK4 in nigrostriatal neurons in vivo. J. Neurochem. 2000, 75: 1200–1208.CrossRefGoogle ScholarPubMed
Chen, JY, Hsu, PC, Hsu, IL, Yeh, GC. Sequential up-regulation of the c-fos, c-jun and bax genes in the cortex, striatum and cerebellum induced by a single injection of a low dose of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in C57Bl/6 mice. Neurosci. Lett. 2001, 314: 49–52.CrossRefGoogle Scholar
Offen, D, Beart, PM, Cheung, NS, et al. Transgenic mice expressing human Bcl-2 in their neurons are resistant to 6-hydroxydopamine and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity. Proc. Natl Acad. Sci. USA 1998, 95: 5789–5794.CrossRefGoogle ScholarPubMed
Forno, LS, DeLanney, , Irwin, I, Langston, JW. Evolution of nerve fiber degeneration in the striatum in the MPTP-treated squirrel monkey. Mol. Neurobiol. 1994, 9: 163–170.CrossRefGoogle ScholarPubMed
Langston, JW, Forno, LS, Rebert, CS, Irwin, I. Selective nigral toxicity after systemic administration of 1-methyl-4- phenyl-1,2,5,6-tetrahydropyrine (MPTP) in the squirrel monkey. Brain Res. 1984, 292: 390–394.CrossRefGoogle ScholarPubMed
Imai, H, Nakamura, T, Endo, K, Narabayashi, H. Hemiparkinsonism in monkeys after unilateral caudate nucleus infusion of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): behavior and histology. Brain Res. 1988, 474: 327–332.CrossRefGoogle ScholarPubMed
Russ, H, Mihatsch, W, Gerlach, M, Riederer, P, Przuntek, H. Neurochemical and behavioural features induced by chronic low dose treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in the common marmoset: implications for Parkinson's disease? Neurosci. Lett. 1991, 123: 115–118.CrossRefGoogle ScholarPubMed
Albanese, A, Granata, R, Gregori, B, et al. Chronic administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine to monkeys: behavioural, morphological and biochemical correlates. Neuroscience 1993, 55: 823–832.CrossRefGoogle ScholarPubMed
Kupsch, A, Sautter, J, Schwarz, J, et al. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity in non-human primates is antagonized by pretreatment with nimodipine at the nigral, but not at the striatal level. Brain Res. 1996, 741: 185–196.CrossRefGoogle Scholar
Hansard, MJ, Smith, , Jackson, MJ, Cheetham, SC, Jenner, P. Dopamine, but not norepinephrine or serotonin, reuptake inhibition reverses motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated primates. J. Pharmacol. Exp. Ther. 2002, 303: 952–958.CrossRefGoogle ScholarPubMed
Jakowec, MW, Donaldson, DM, Barba, J, Petzinger, GM. Postnatal expression of alpha-synuclein protein in the rodent substantia nigra and striatum. Devel. Neurosci. 2001, 23: 91–99.CrossRefGoogle ScholarPubMed
Jenner, P. The contribution of the MPTP-treated primate model to the development of new treatment strategies for Parkinson's disease. Parkinsonism Relat. Disord. 2003, 9: 131–137.CrossRefGoogle ScholarPubMed
Liou, HH, Tsai, MC, Chen, CJ, et al. Environmental risk factors and Parkinson's disease: a case-control study in Taiwan. Neurology 1997, 48: 1583–1588.CrossRefGoogle ScholarPubMed
Grant, H, Lantos, PL, Parkinson, C. Cerebral damage in paraquat poisoning. Histopathology 1980, 4: 185–195.CrossRefGoogle ScholarPubMed
Barbeau, A, Dallaire, L, Buu, NT, et al. New amphibian models for the study of 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine (MPTP). Life Sci. 1985, 36: 1125–1134.CrossRefGoogle Scholar
Di Monte, D, Sandy, MS, Ekstrom, G, Smith, MT. Comparative studies on the mechanisms of paraquat and 1-methyl-4-phenylpyridine (MPP+) cytotoxicity. Biochem. Biophys. Res. Commun. 1986, 137: 303–309.CrossRefGoogle ScholarPubMed
Koller, WC. Paraquat and Parkinson's disease. Neurology 1986, 36: 1147.CrossRefGoogle ScholarPubMed
Rajput, AH, Uitti, RJ. Paraquat and Parkinson's disease. Neurology 1987, 37: 1820–1821.CrossRefGoogle ScholarPubMed
Sanchez-Ramos, JR, Hefti, F, Weiner, WJ. Paraquat and Parkinson's disease. Neurology 1987, 37: 728.CrossRefGoogle ScholarPubMed
Tanner, CM, Ottman, R, Goldman, SM, et al. Parkinson disease in twins: an etiologic study. J. Am. Med. Ass. 1999, 281: 341–346.CrossRefGoogle Scholar
Di Monte, DA. The environment and Parkinson's disease: is the nigrostriatal system preferentially targeted by neurotoxins? Lancet Neurol. 2003, 2: 531–538.CrossRefGoogle ScholarPubMed
Wesseling, C, Joode, Wendel B, Ruepert, C, et al. Paraquat in developing countries. Int. J. Occup. Environ. Health 2001, 7: 275–286.CrossRefGoogle ScholarPubMed
Markey, SP, Weisz, A, Bacon, JP. Reduced paraquat does not exhibit MPTP-like neurotoxicity. J. Anal. Toxicol. 1986, 10: 257.CrossRefGoogle Scholar
Li, X, Sun, AY. Paraquat induced activation of transcription factor AP-1 and apoptosis in PC12 cells. J. Neural Transm. 1999, 106: 1–21.CrossRefGoogle ScholarPubMed
Thiruchelvam, M, Brockel, BJ, Richfield, EK, Baggs, RB, Cory-Slechta, DA. Potentiated and preferential effects of combined paraquat and maneb on nigrostriatal dopamine systems: environmental risk factors for Parkinson's disease? Brain Res. 2000, 873: 225–234.CrossRefGoogle ScholarPubMed
Brooks, AI, Chadwick, CA, Gelbard, HA, Cory-Slechta, DA, Federoff, HJ. Paraquat-elicited neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Res. 1999, 823: 1–10.CrossRefGoogle ScholarPubMed
Chun, HS, Gibson, GE, DeGiorgio, , et al. Dopaminergic cell death induced by MPP(+), oxidant and specific neurotoxicants shares the common molecular mechanism. J. Neurochem. 2001, 76: 1010–1021.CrossRefGoogle Scholar
Shimizu, K, Ohtaki, K, Matsubara, K, et al. Carrier-mediated processes in blood–brain barrier penetration and neural uptake of paraquat. Brain Res. 2001, 906: 135–142.CrossRefGoogle ScholarPubMed
McCormack, AL, Thiruchelvam, M, Manning-Bog, AB, et al. Environmental risk factors and Parkinson's disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol. Dis. 2002, 10: 119–127.CrossRefGoogle ScholarPubMed
Thiruchelvam, M, Richfield, EK, Goodman, BM, Baggs, RB, Cory-Slechta, DA. Developmental exposure to the pesticides paraquat and maneb and the Parkinson's disease phenotype. Neurotoxicology 2002, 23: 621–633.CrossRefGoogle Scholar
Shimizu, K, Matsubara, K, Ohtaki, K, Shiono, H. Paraquat leads to dopaminergic neural vulnerability in organotypic midbrain culture. Neurosci. Res. 2003, 46: 523–532.CrossRefGoogle ScholarPubMed
Andersen, JK. Paraquat and iron exposure as possible synergistic environmental risk factors in Parkinson's disease. Neurotox. Res. 2003, 5: 307–313.CrossRefGoogle ScholarPubMed
Thiruchelvam, M, Richfield, EK, Baggs, RB, Tank, AW, Cory-Slechta, DA. The nigrostriatal dopaminergic system as a preferential target of repeated exposures to combined paraquat and maneb: implications for Parkinson's disease. J. Neurosci. 2000, 20: 9207–9214.CrossRefGoogle ScholarPubMed
Uversky, VN, Li, J, Fink, AL. Metal-triggered structural transformations, aggregation, and fibrillation of human alpha-synuclein: a possible molecular NK between Parkinson's disease and heavy metal exposure. J. Biol. Chem. 2001, 276: 44284–44296.CrossRefGoogle ScholarPubMed
Uversky, VN, Li, J, Fink, AL. Pesticides directly accelerate the rate of alpha-synuclein fibril formation: a possible factor in Parkinson's disease. FEBS Lett. 2001, 500: 105–108.CrossRefGoogle ScholarPubMed
Corasaniti, MT, Strongoli, MC, Rotiroti, D, Bagetta, G, Nistico, G. Paraquat: a useful tool for the in vivo study of mechanisms of neuronal cell death. Pharmacol. Toxicol. 1998, 83: 1–7.CrossRefGoogle ScholarPubMed
Liou, HH, Chen, RC, Tsai, YF, et al. Effects of Paraquat on the substantia nigra of the Wistar rats: neurochemical, histological, and behavioral studies. Toxicol. Appl. Pharmacol. 1996, 137: 34–41.CrossRefGoogle ScholarPubMed
Peng, J, Mao, XO, Stevenson, FF, Hsu, M, Andersen, JK. The herbicide Paraquat induces dopaminergic nigral apoptosis through sustained activation of the JNK pathway. J. Biol. Chem. 2004.CrossRefGoogle ScholarPubMed
Schuler, F, Casida, JE. Functional coupling of PSST and ND1 subunits in NADH: ubiquinone oxidoreductase established by photoaffinity labeling. Biochim. Biophys. Acta 2001, 1506: 79–87.CrossRefGoogle ScholarPubMed
Greenamyre, JT, Sherer, TB, Betarbet, R, Panov, AV. Complex I and Parkinson's disease. IUBMB Life 2001, 52: 135–141.CrossRefGoogle ScholarPubMed
Greenamyre, JT, MacKenzie, G, Peng, TI, Stephans, SE. Mitochondrial dysfunction in Parkinson's disease. Biochem. Soc. Symp. 1999, 66: 85–97.CrossRefGoogle ScholarPubMed
Giasson, BI, Duda, JE, Murray, IV, et al. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science 2000, 290: 985–989.CrossRefGoogle ScholarPubMed
Trojanowski, JQ. Rotenone neurotoxicity: a new window on environmental causes of Parkinson's disease and related brain amyloidoses. Exp. Neurol. 2003, 179: 6–8.CrossRefGoogle ScholarPubMed
Orth, M, Tabrizi, SJ. Models of Parkinson's disease. Mov. Disord. 2003, 18: 729–737.CrossRefGoogle ScholarPubMed
Heikkila, RE, Nicklas, WJ, Vyas, I, Duvoisin, RC. Dopaminergic toxicity of rotenone and the 1-methyl-4-phenylpyridinium ion after their stereotaxic administration to rats: implication for the mechanism of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity. Neurosci. Lett. 1985, 62: 389–394.CrossRefGoogle ScholarPubMed
Ferrante, RJ, Schulz, JB, Kowall, NW, Beal, MF. Systemic administration of rotenone produces selective damage in the striatum and globus pallidus, but not in the substantia nigra. Brain Res. 1997, 753: 157–162.CrossRefGoogle Scholar
Thiffault, C, Langston, JW, Di Monte, DA. Increased striatal dopamine turnover following acute administration of rotenone to mice. Brain Res. 2000, 885: 283–288.CrossRefGoogle ScholarPubMed
Betarbet, R, Sherer, TB, MacKenzie, G, et al. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nature Neurosci. 2000, 3: 1301–1306.CrossRefGoogle ScholarPubMed
Sherer, TB, Betarbet, R, Stout, AK, et al. An in vitro model of Parkinson's disease: linking mitochondrial impairment to altered alpha-synuclein metabolism and oxidative damage. J. Neurosci. 2002, 22: 7006–7015.CrossRefGoogle Scholar
Alam, M, Schmidt, WJ. Rotenone destroys dopaminergic neurons and induces Parkinsonian symptoms in rats. Behav. Brain Res. 2002, 136: 317–324.CrossRefGoogle ScholarPubMed
Betarbet, R, Sherer, TB, Di Monte, DA, Greenamyre, JT. Mechanistic approaches to Parkinson's disease pathogenesis. Brain Pathol. 2002, 12: 499–510.CrossRefGoogle ScholarPubMed
Tada-Oikawa, S, Hiraku, Y, Kawanishi, M, Kawanishi, S. Mechanism for generation of hydrogen peroxide and change of mitochondrial membrane potential during rotenone-induced apoptosis. Life Sci. 2003, 73: 3277–3288.CrossRefGoogle ScholarPubMed
Sherer, TB, Kim, JH, Betarbet, R, Greenamyre, JT. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp. Neurol. 2003, 179: 9–16.CrossRefGoogle ScholarPubMed
Alam, M, Mayerhofer, A, Schmidt, WJ. The neurobehavioral changes induced by bilateral rotenone lesion in medial forebrain bundle of rats are reversed by l-DOPA. Behav. Brain Res. 2004, 151: 117–124.CrossRefGoogle ScholarPubMed
Hoglinger, GU, Feger, J, Prigent, A, et al. Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. J. Neurochem. 2003, 84: 491–502.CrossRefGoogle ScholarPubMed
Braak, H, Del Tredici, K, Bratzke, H, et al. Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson's disease (preclinical and clinical stages). J. Neurol. 2002, 249 (Suppl. 3): III/1–5.CrossRefGoogle Scholar
Forno, LS. Neuropathology of Parkinson's disease. J. Neuropathol. Exp. Neurol. 1996, 55: 259–272.CrossRefGoogle ScholarPubMed
Lang-Rollin, I, Vekrellis, K, Wang, Q, Rideout, HJ, Stefanis, L. Application of proteasomal inhibitors to mouse sympathetic neurons activates the intrinsic apoptotic pathway. J. Neurochem. 2004, 90: 1511–1520.CrossRefGoogle ScholarPubMed
Ardley, HC, Scott, GB, Rose, SA, Tan, NG, Robinson, PA. UCH-L1 aggresome formation in response to proteasome impairment indicates a role in inclusion formation in Parkinson's disease. J. Neurochem. 2004, 90: 379–391.CrossRefGoogle ScholarPubMed
McNaught, KS, Bjorklund, LM, Belizaire, R, et al. Proteasome inhibition causes nigral degeneration with inclusion bodies in rats. NeuroReport 2002, 13: 1437–1441.CrossRefGoogle ScholarPubMed
McNaught, KS, Jenner, P. Proteasomal function is impaired in substantia nigra in Parkinson's disease. Neurosci. Lett. 2001, 297: 191–194.CrossRefGoogle ScholarPubMed
McNaught, KS, Perl, DP, Brownell, AL, Olanow, CW. Systemic exposure to proteasome inhibitors causes a progressive model of Parkinson's disease. Ann. Neurol. 2004, 56: 149–162.CrossRefGoogle ScholarPubMed
Moilanen, JS, Autere, JM, Myllyla, VV, Majamaa, K. Complex segregation analysis of Parkinson's disease in the Finnish population. Hum. Genet. 2001, 108: 184–189.Google ScholarPubMed
Polymeropoulos, MH, Lavedan, C, Leroy, E, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science 1997, 276: 2045–2047.CrossRefGoogle ScholarPubMed
Marder, K, Logroscino, G, Alfaro, B, et al. Environmental risk factors for Parkinson's disease in an urban multiethnic community. Neurology 1998, 50: 279–281.CrossRefGoogle Scholar
Athanassiadou, A, Voutsinas, G, Psiouri, L, et al. Genetic analysis of families with Parkinson disease that carry the Ala53Thr mutation in the gene encoding alpha-synuclein. Am. J. Hum. Genet. 1999, 65: 555–558.CrossRefGoogle ScholarPubMed
Papapetropoulos, S, Paschalis, C, Athanassiadou, A, et al. Clinical phenotype in patients with alpha-synuclein Parkinson's disease living in Greece in comparison with patients with sporadic Parkinson's disease. J. Neurol. Neurosurg. Psychiat. 2001, 70: 662–665.CrossRefGoogle ScholarPubMed
Farrer, M, Wavrant-De Vrieze, F, Crook, R, et al. Low frequency of alpha-synuclein mutations in familial Parkinson's disease. Ann. Neurol. 1998, 43: 394–397.CrossRefGoogle ScholarPubMed
Vaughan, JR, Farrer, MJ, Wszolek, ZK, et al. Sequencing of the alpha-synuclein gene in a large series of cases of familial Parkinson's disease fails to reveal any further mutations: the European Consortium on Genetic Susceptibility in Parkinson's Disease (GSPD). Hum. Mol. Genet. 1998, 7: 751–753.CrossRefGoogle Scholar
Langston, JW, Sastry, S, Chan, P, et al. Novel alpha-synuclein-immunoreactive proteins in brain samples from the Contursi kindred, Parkinson's, and Alzheimer's disease. Exp. Neurol. 1998, 154: 684–690.CrossRefGoogle ScholarPubMed
Narhi, L, Wood, SJ, Stevenson, S, et al. Both familial Parkinson's disease mutations accelerate alpha-synuclein aggregation. J. Biol. Chem. 1999, 274: 9843–9846.CrossRefGoogle ScholarPubMed
Conway, KA, Harper, JD, Lansbury, PT Jr. Fibrils formed in vitro from alpha-synuclein and two mutant forms linked to Parkinson's disease are typical amyloid. Biochemistry 2000, 39: 2552–2563.CrossRefGoogle ScholarPubMed
Conway, KA, Lee, SJ, Rochet, JC, et al. Accelerated oligomerization by Parkinson's disease linked alpha-synuclein mutants. Ann. NY Acad. Sci. 2000, 920: 42–45.CrossRefGoogle ScholarPubMed
Li, J, Uversky, VN, Fink, AL. Effect of familial Parkinson's disease point mutations A30P and A53T on the structural properties, aggregation, and fibrillation of human alpha-synuclein. Biochemistry 2001, 40: 11604–11613.CrossRefGoogle ScholarPubMed
Sommer, B, Barbieri, S, Hofele, K, et al. Mouse models of alpha-synucleinopathy and Lewy pathology. Exp. Gerontol. 2000, 35: 1389–1403.CrossRefGoogle ScholarPubMed
Lo Bianco, C, Ridet, JL, Schneider, BL, Deglon, N, Aebischer, P. Alpha-synucleinopathy and selective dopaminergic neuron loss in a rat lentiviral-based model of Parkinson's disease. Proc. Natl Acad. Sci. USA 2002, 99: 10813–10818.CrossRefGoogle Scholar
Lee, MK, Stirling, W, Xu, Y, et al. Human alpha-synuclein-harboring familial Parkinson's disease-linked Ala-53 → Thr mutation causes neurodegenerative disease with alpha-synuclein aggregation in transgenic mice. Proc. Natl Acad. Sci. USA 2002, 99: 8968–8973.CrossRefGoogle ScholarPubMed
Kotzbauer, PT, Trojanowsk, JQ, Lee, VM. Lewy body pathology in Alzheimer's disease. J. Mol. Neurosci. 2001, 17: 225–232.CrossRefGoogle ScholarPubMed
Jo, E, McLaurin, J, Yip, CM, St George-Hyslop, P, Fraser, PE. Alpha-synuclein membrane interactions and lipid specificity. J. Biol. Chem. 2000, 275: 34328–34334.CrossRefGoogle ScholarPubMed
Davidson, WS, Jonas, A, Clayton, DF, George, JM. Stabilization of alpha-synuclein secondary structure upon binding to synthetic membranes. J. Biol. Chem. 1998, 273: 9443–9449.CrossRefGoogle ScholarPubMed
Zhu, M, Li, J, Fink, AL. The association of alpha-synuclein with membranes affects bilayer structure, stability and fibril formation. J. Biol. Chem. 2003.Google ScholarPubMed
Bussell, R Jr, Eliezer, D. Residual structure and dynamics in Parkinson's disease-associated mutants of alpha-synuclein. J. Biol. Chem. 2001, 276: 45996–46003.CrossRefGoogle ScholarPubMed
Perrin, RJ, Woods, WS, Clayton, DF, George, JM. Interaction of human alpha-synuclein and Parkinson's disease variants with phospholipids: structural analysis using site-directed mutagenesis. J. Biol. Chem. 2000, 275: 34393–34398.CrossRefGoogle ScholarPubMed
Cole, NB, Murphy, DD. The cell biology of alpha-synuclein: a sticky problem? Neuromol. Med. 2002, 1: 95–109.CrossRefGoogle ScholarPubMed
Park, JY, Lansbury, PT Jr. Beta-synuclein inhibits formation of alpha-synuclein protofibrils: a possible therapeutic strategy against Parkinson's disease. Biochemistry 2003, 42: 3696–3700.CrossRefGoogle ScholarPubMed
Windisch, M, Hutter-Paier, B, Rockenstein, E, et al. Development of a new treatment for Alzheimer's disease and Parkinson's disease using anti-aggregatory beta-synuclein-derived peptides. J. Mol. Neurosci. 2002, 19: 63–69.CrossRefGoogle ScholarPubMed
Hashimoto, M, Bar-On, P, Ho, G, et al. Beta-synuclein regulates Akt activity in neuronal cells: a possible mechanism for neuroprotection in Parkinson's disease. J. Biol. Chem. 2004, 279: 23622–23629.CrossRefGoogle ScholarPubMed
da Costa, CA, Masliah, E, Checler, F. Beta-synuclein displays an antiapoptotic p53-dependent phenotype and protects neurons from 6-hydroxydopamine-induced caspase 3 activation: cross-talk with alpha-synuclein and implication for Parkinson's disease. J. Biol. Chem. 2003, 278: 37330–37335.CrossRefGoogle ScholarPubMed
Neystat, M, Lynch, T, Przedborski, S, et al. Alpha-synuclein expression in substantia nigra and cortex in Parkinson's disease. Mov. Disord. 1999, 14: 417–422.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Kholodilov, NG, Oo, TF, Burke, RE. Synuclein expression is decreased in rat substantia nigra following induction of apoptosis by intrastriatal 6-hydroxydopamine. Neurosci. Lett. 1999, 275: 105–108.CrossRefGoogle ScholarPubMed
Petersen, K, Olesen, OF, Mikkelsen, JD. Developmental expression of alpha-synuclein in rat hippocampus and cerebral cortex. Neuroscience 1999, 91: 651–659.CrossRefGoogle ScholarPubMed
Orth, M, Tabrizi, SJ, Tomlinson, C, et al. G209A mutant alpha synuclein expression specifically enhances dopamine induced oxidative damage. Neurochem. Int. 2004, 45: 669–676.CrossRefGoogle ScholarPubMed
Wersinger, C, Sidhu, A. Attenuation of dopamine transporter activity by alpha-synuclein. Neurosci. Lett. 2003, 340: 189–192.CrossRefGoogle ScholarPubMed
Hashimoto, M, Hsu, LJ, Rockenstein, E, et al. Alpha-synuclein protects against oxidative stress via inactivation of the c-Jun N-terminal kinase stress-signaling pathway in neuronal cells. J. Biol. Chem. 2002, 277: 11465–11472.CrossRefGoogle ScholarPubMed
Baptista, MJ, O'Farrell, C, Daya, S, et al. Co-ordinate transcriptional regulation of dopamine synthesis genes by alpha-synuclein in human neuroblastoma cell lines. J. Neurochem. 2003, 85: 957–968.CrossRefGoogle ScholarPubMed
Manning-Bog, AB, McCormack, AL, Purisai, MG, Bolin, LM, Di Monte, DA. Alpha-synuclein overexpression protects against paraquat-induced neurodegeneration. J. Neurosci. 2003, 23: 3095–3099.CrossRefGoogle ScholarPubMed
Seo, JH, Rah, JC, Choi, SH, et al. Alpha-synuclein regulates neuronal survival via Bcl-2 family expression and PI3/Akt kinase pathway. FASEB J. 2002, 16: 1826–1828.CrossRefGoogle ScholarPubMed
Hattori, N, Kitada, T, Matsumine, H, et al. Molecular genetic analysis of a novel Parkin gene in Japanese families with autosomal recessive juvenile Parkinsonism: evidence for variable homozygous deletions in the Parkin gene in affected individuals. Ann. Neurol. 1998, 44: 935–941.CrossRefGoogle Scholar
Leroy, E, Anastasopoulos, D, Konitsiotis, S, Lavedan, C, Polymeropoulos, MH. Deletions in the Parkin gene and genetic heterogeneity in a Greek family with early onset Parkinson's disease. Hum. Genet. 1998, 103: 424–427.CrossRefGoogle Scholar
Abbas, N, Lucking, CB, Ricard, S, et al. A wide variety of mutations in the parkin gene are responsible for autosomal recessive parkinsonism in Europe: French Parkinson's Disease Genetics Study Group and the European Consortium on Genetic Susceptibility in Parkinson's Disease. Hum. Mol. Genet. 1999, 8: 567–574.CrossRefGoogle Scholar
Satoh, J, Kuroda, Y. Association of codon 167 Ser/Asn heterozygosity in the Parkin gene with sporadic Parkinson's disease. NeuroReport 1999, 10: 2735–2739.CrossRefGoogle Scholar
Hayashi, S, Wakabayashi, K, Ishikawa, A, et al. An autopsy case of autosomal-recessive juvenile Parkinsonism with a homozygous exon 4 deletion in the parkin gene. Mov. Disord. 2000, 15: 884–888.3.0.CO;2-8>CrossRefGoogle ScholarPubMed
Warrenburg, BP, Lammens, M, Lucking, CB, et al. Clinical and pathologic abnormalities in a family with parkinsonism and Parkin gene mutations. Neurology 2001, 56: 555–557.CrossRefGoogle Scholar
Farrer, M, Chan, P, Chen, R, et al. Lewy bodies and Parkinsonism in families with Parkin mutations. Ann. Neurol. 2001, 50: 293–300.CrossRefGoogle ScholarPubMed
Dawson, TM, Dawson, VL. Rare genetic mutations shed light on the pathogenesis of Parkinson disease. J. Clin. Invest. 2003, 111: 145–151.CrossRefGoogle ScholarPubMed
Forloni, G, Terreni, L, Bertani, I, et al. Protein misfolding in Alzheimer's and Parkinson's disease: genetics and molecular mechanisms. Neurobiol. Aging. 2002, 23: 957–976.CrossRefGoogle ScholarPubMed
Takahashi, H. [Juvenile parkinsonism: its neuropathological aspects.] No To Shinkei. 1994, 46: 523–529. (In Japanese)Google ScholarPubMed
Schlossmacher, MG, Frosch, MP, Gai, WP, et al. Parkin localizes to the Lewy bodies of Parkinson disease and dementia with Lewy bodies. Am. J. Pathol. 2002, 160: 1655–1667.CrossRefGoogle ScholarPubMed
Shimura, H, Schlossmacher, MG, Hattori, N, et al. Ubiquitination of a new form of alpha-synuclein by Parkin from human brain: implications for Parkinson's disease. Science 2001, 293: 263–269.CrossRefGoogle ScholarPubMed
Kahle, PJ, Neumann, M, Ozmen, L, et al. Subcellular localization of wild-type and Parkinson's disease-associated mutant alpha-synuclein in human and transgenic mouse brain. J. Neurosci. 2000, 20: 6365–6373.CrossRefGoogle ScholarPubMed
Imai, Y, Soda, M, Murakami, T, et al. A product of the human gene adjacent to parkin is a component of Lewy bodies and suppresses Pael receptor-induced cell death. J. Biol. Chem. 2003, 278: 51901–51910.CrossRefGoogle ScholarPubMed
Zhang, Y, Gao, J, Chung, KK, et al. Parkin functions as an E2-dependent ubiquitin-protein ligase and promotes the degradation of the synaptic vesicle-associated protein, CDCrel-1. Proc. Natl Acad. Sci. USA 2000, 97: 13354–13359.CrossRefGoogle ScholarPubMed
Shimura, H, Hattori, N, Kubo, S, et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nature Genet. 2000, 25: 302–305.CrossRefGoogle ScholarPubMed
Jiang, H, Ren, Y, Zhao, J, Feng, J. Parkin protects human dopaminergic neuroblastoma cells against dopamine-induced apoptosis. Hum. Mol. Genet. 2004, 13: 1745–1754.CrossRefGoogle ScholarPubMed
Petrucelli, L, O'Farrell, C, Lockhart, PJ, et al. Parkin protects against the toxicity associated with mutant alpha-synuclein: proteasome dysfunction selectively affects catecholaminergic neurons. Neuron 2002, 36: 1007–1019.CrossRefGoogle ScholarPubMed
Ledesma, MD, Galvan, C, Hellias, B, Dotti, C, Jensen, PH. Astrocytic but not neuronal increased expression and redistribution of parkin during unfolded protein stress. J. Neurochem. 2002, 83: 1431–1440.CrossRefGoogle Scholar
Mengesdorf, T, Jensen, PH, Mies, G, Aufenberg, C, Paschen, W. Down-regulation of parkin protein in transient focal cerebral ischemia: a link between stroke and degenerative disease? Proc. Natl Acad. Sci. USA 2002, 99: 15042–15047.CrossRefGoogle ScholarPubMed
Higashi, Y, Asanuma, M, Miyazaki, I, et al. Parkin attenuates manganese-induced dopaminergic cell death. J. Neurochem. 2004, 89: 1490–1497.CrossRefGoogle ScholarPubMed
Takahashi, R, Imai, Y. Pael receptor, endoplasmic reticulum stress, and Parkinson's disease. J. Neurol. 2003, 250 (Suppl. 3): III25–9.CrossRefGoogle ScholarPubMed
Baptista, MJ, Cookson, MR, Miller, DW. Parkin and alpha-synuclein: opponent actions in the pathogenesis of Parkinson's disease. Neuroscientist 2004, 10: 63–72.CrossRefGoogle ScholarPubMed
Darios, F, Corti, O, Lucking, CB, et al. Parkin prevents mitochondrial swelling and cytochrome c release in mitochondria-dependent cell death. Hum. Mol. Genet. 2003, 12: 517–526.CrossRefGoogle ScholarPubMed
Briggs, MD, Mortier, GR, Cole, WG, et al. Diverse mutations in the gene for cartilage oligomeric matrix protein in the pseudoachondroplasia–multiple epiphyseal dysplasia disease spectrum. Am. J. Hum. Genet. 1998, 62: 311–319.CrossRefGoogle ScholarPubMed
Liu, Y, Fallon, L, Lashuel, HA, Liu, Z, Lansbury, PT Jr. The UCH-L1 gene encodes two opposing enzymatic activities that affect alpha-synuclein degradation and Parkinson's disease susceptibility. Cell 2002, 111: 209–218.CrossRefGoogle ScholarPubMed
Valente, EM, Bentivoglio, AR, Dixon, PH, et al. Localization of a novel locus for autosomal recessive early-onset parkinsonism, PARK6, on human chromosome 1p35–p36. Am. J. Hum. Genet. 2001, 68: 895–900.CrossRefGoogle ScholarPubMed
Bonifati, V, Dekker, MC, Vanacore, N, et al. Autosomal recessive early-onset Parkinsonism is linked to three loci: PARK2, PARK6, and PARK7. Neurol. Sci. 2002, 23 (Suppl. 2): S59–S60.CrossRefGoogle ScholarPubMed
Baik, JH, Picetti, R, Saiardi, A, et al. Parkinsonian-like locomotor impairment in mice lacking dopamine D2 receptors. Nature 1995, 377: 424–428.CrossRefGoogle ScholarPubMed
Dracheva, S, Haroutunian, V. Locomotor behavior of dopamine D1 receptor transgenic/D2 receptor deficient hybrid mice. Brain Res. 2001, 905: 142–151.CrossRefGoogle ScholarPubMed
Jaber, M, Jones, S, Giros, B, Caron, MG. The dopamine transporter: a crucial component regulating dopamine transmission. Mov. Disord. 1997, 12: 629–633.CrossRefGoogle ScholarPubMed
Grunewald, T, Beal, MF. NOS knockouts and neuroprotection. Nature Med. 1999, 5: 1354–1355.CrossRefGoogle ScholarPubMed
Andreassen, OA, Ferrante, RJ, Dedeoglu, A, et al. Mice with a partial deficiency of manganese superoxide dismutase show increased vulnerability to the mitochondrial toxins malonate, 3- nitropropionic acid, and MPTP. Exp. Neurol. 2001, 167: 189–195.CrossRefGoogle ScholarPubMed
Maier, CM, Chan, PH. Role of superoxide dismutases in oxidative damage and neurodegenerative disorders. Neuroscientist 2002, 8: 323–334.CrossRefGoogle ScholarPubMed
Wu, DC, Teismann, P, Tieu, K, et al. NADPH oxidase mediates oxidative stress in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson's disease. Proc. Natl Acad. Sci. USA. 2003, 100: 6145–6150.CrossRefGoogle ScholarPubMed
Gao, HM, Liu, B, Hong, JS. Critical role for microglial NADPH oxidase in rotenone-induced degeneration of dopaminergic neurons. J. Neurosci. 2003, 23: 6181–6187.CrossRefGoogle ScholarPubMed
Sriram, K, Matheson, JM, Benkovic, SA, et al. Mice deficient in TNF receptors are protected against dopaminergic neurotoxicity: implications for Parkinson's disease. FASEB J. 2002, 16: 1474–1476.CrossRefGoogle ScholarPubMed
Dauer, W, Kholodilov, N, Vila, M, et al. Resistance of alpha-synuclein null mice to the Parkinsonian neurotoxin MPTP. Proc. Natl Acad. Sci. USA 2002, 99: 14524–14529.CrossRefGoogle ScholarPubMed
Gerlai, R, McNamara, A, Choi-Lundberg, DL, et al. Impaired water maze learning performance without altered dopaminergic function in mice heterozygous for the GDNF mutation. Eur. J. Neurosci. 2001, 14: 1153–1163.CrossRefGoogle ScholarPubMed
Chen, JF, Xu, K, Petzer, JP, et al. Neuroprotection by caffeine and A(2A) adenosine receptor inactivation in a model of Parkinson's disease. J. Neurosci. 2001, 21: RC143.CrossRefGoogle Scholar
Witta, J, Baffi, JS, Palkovits, M, et al. Nigrostriatal innervation is preserved in Nurr1-null mice, although dopaminergic neuron precursors are arrested from terminal differentiation. Brain Res. Mol. Brain Res. 2000, 84: 67–78.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×