Classic and new animal models of Parkinson's disease

Javier Blesa, Sudarshan Phani, Vernice Jackson-Lewis, Serge Przedborski, Javier Blesa, Sudarshan Phani, Vernice Jackson-Lewis, Serge Przedborski

Abstract

Neurological disorders can be modeled in animals so as to recreate specific pathogenic events and behavioral outcomes. Parkinson's Disease (PD) is the second most common neurodegenerative disease of an aging population, and although there have been several significant findings about the PD disease process, much of this process still remains a mystery. Breakthroughs in the last two decades using animal models have offered insights into the understanding of the PD disease process, its etiology, pathology, and molecular mechanisms. Furthermore, while cellular models have helped to identify specific events, animal models, both toxic and genetic, have replicated almost all of the hallmarks of PD and are useful for testing new neuroprotective or neurorestorative strategies. Moreover, significant advances in the modeling of additional PD features have come to light in both classic and newer models. In this review, we try to provide an updated summary of the main characteristics of these models as well as the strengths and weaknesses of what we believe to be the most popular PD animal models. These models include those produced by 6-hydroxydopamine (6-OHDA), 1-methyl-1,2,3,6-tetrahydropiridine (MPTP), rotenone, and paraquat, as well as several genetic models like those related to alpha-synuclein, PINK1, Parkin and LRRK2 alterations.

Figures

Figure 1
Figure 1
Photomicrograph of a 6-OHDA lesioned rat striatum immunostained for tyrosine hydroxylase (TH). Densities of TH-immunoreactivity striatal fibers are clearly reduced after the 6-OHDA injection (right side) as compared to the densities of striatal TH-immunoreactivity fibers in control rat (left side).
Figure 2
Figure 2
Photomicrographs of nonhuman primate immunostained for tyrosine hydroxylase (TH). Dopaminergic neurons located in the substantia nigra compacta (SNc) project to the caudate (CD) and putamen (PUT). Note the markedly reduced TH immunoreactivity in the substantia nigra and striatum (CD and PUT) in the MPTP-treated monkey (b) compared to control (a).

References

    1. Lees AJ, Hardy J, Revesz T. Parkinson’s disease. The Lancet. 2009;373(9680):2055–2066.
    1. Spillantini MG, Schmidt ML, Lee VMY, Trojanowski JQ, Jakes R, Goedert M. α-synuclein in Lewy bodies [8] Nature. 1997;388(6645):839–840.
    1. Forno LS. Neuropathology of Parkinson’s disease. Journal of Neuropathology and Experimental Neurology. 1996;55(3):259–272.
    1. Braak H, Ghebremedhin E, Rüb U, Bratzke H, Del Tredici K. Stages in the development of Parkinson’s disease-related pathology. Cell and Tissue Research. 2004;318(1):121–134.
    1. Chaudhuri KR, Healy DG, Schapira AHV. Non-motor symptoms of Parkinson’s disease: diagnosis and management. Lancet Neurology. 2006;5(3):235–245.
    1. Dauer W, Przedborski S. Parkinson’s disease: mechanisms and models. Neuron. 2003;39(6):889–909.
    1. Carlsson A, Lindqvist M, Magnusson T. 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature. 1957;180(4596):p. 1200.
    1. Carlsson A. The occurrence, distribution and physiological role of catecholamines in the nervous system. Pharmacological reviews. 1959;11(2):490–493.
    1. Betarbet R, Sherer TB, Timothy Greenamyre J. Animal models of Parkinson’s disease. BioEssays. 2002;24(4):308–318.
    1. Dawson TM, Ko HS, Dawson VL. Genetic Animal Models of Parkinson’s Disease. Neuron. 2010;66(5):646–661.
    1. Schwarting RKW, Huston JP. Unilateral 6-hydroxydopamine lesions of meso-striatal dopamine neurons and their physiological sequelae. Progress in Neurobiology. 1996;49(3):215–266.
    1. Schwarting RKW, Huston JP. The unilateral 6-hydroxydopamine lesion model in behavioral brain research. Analysis of functional deficits, recovery and treatments. Progress in Neurobiology. 1996;50(2-3):275–331.
    1. Schwarting RKW, Huston JP. Behavioral and neurochemical dynamics of neurotoxic meso-striatal dopamine lesions. NeuroToxicology. 1997;18(3):689–708.
    1. Senoh S, Witkop B. Non-enzymatic conversions of dopamine to norepinephrine and trihydroxyphenethylamines. Journal of the American Chemical Society. 1959;81(23):6222–6231.
    1. Ungerstedt U. 6-hydroxy-dopamine induced degeneration of central monoamine neurons. European Journal of Pharmacology. 1968;5(1):107–110.
    1. Roeling TAP, Docter GJ, Voorn P, Melchers BPC, Wolters EC, Groenewegen HJ. Effects of unilateral 6-hydroxydopamine lesions on neuropeptide immunoreactivity in the basal ganglia of the common marmoset, Callithrix jacchus, a quantitative immunohistochemical analysis. Journal of Chemical Neuroanatomy. 1995;9(3):155–164.
    1. Valette H, Deleuze P, Syrota A, et al. Canine myocardial beta-adrenergic, muscarinic receptor densities after denervation: a PET study. Journal of Nuclear Medicine. 1995;36(1):140–146.
    1. Annett LE, Torres EM, Clarke DJ, et al. Survival of nigral grafts within the striatum of marmosets with 6-OHDA lesions depends critically on donor embryo age. Cell Transplantation. 1997;6(6):557–569.
    1. Ruffy R, Leonard M. Chemical cardiac sympathetic denervation hampers defibrillation in the dog. Journal of Cardiovascular Electrophysiology. 1997;8(1):62–67.
    1. Sachs C, Jonsson G. Mechanisms of action of 6-hydroxydopamine. Biochemical Pharmacology. 1975;24(1):1–8.
    1. Martin GE, Myers RD, Newberg DC. Catecholamine release by intracerebral perfusion of 6 hydroxydopamine and desipramine. European Journal of Pharmacology. 1976;36(2):299–311.
    1. Perese DA, Ulman J, Viola J, Ewing SE, Bankiewicz KS. A 6-hydroxydopamine-induced selective parkinsonian rat model. Brain Research. 1989;494(2):285–293.
    1. 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(3):631–647.
    1. Cohen G. Oxy-radical toxicity in catecholamine neurons. NeuroToxicology. 1984;5(1):77–82.
    1. Blandini F, Armentero MT, Martignoni E. The 6-hydroxydopamine model: news from the past. Parkinsonism and Related Disorders. 2008;14(2):S124–S129.
    1. Sauer H, Oertel WH. Progressive degeneration of nigrostriatal dopamine neurons following intrastriatal terminal lesions with 6-hydroxydopamine: a combined retrograde tracing and immunocytochemical study in the rat. Neuroscience. 1994;59(2):401–415.
    1. Lee CS, Sauer H, Björklund A. Dopaminergic neuronal degeneration and motor impairments following axon terminal lesion by intrastriatal 6-hydroxydopamine in the rat. Neuroscience. 1996;72(3):641–653.
    1. Ungerstedt U. Adipsia and aphagia after 6-hydroxydopamine induced degeneration of the nigro-striatal dopamine system. Acta Physiologica Scandinavica, Supplement. 1971;367:95–122.
    1. Bourn WM, Chin L, Picchioni AL. Enhancement of audiogenic seizure by 6-hydroxydopamine. Journal of Pharmacy and Pharmacology. 1972;24(11):913–914.
    1. Knoll J. The pharmacology of (-)deprenyl. Journal of Neural Transmission. Supplementum. 1986;22:75–89.
    1. Graham JD, Lewis MJ, Williams J. Proceedings: The effect of delta-1-tetrahydrocannabinol on the noradrenaline and dopamine content of the brain and heart of the rat. British Journal of Pharmacology. 1974;52(3):p. 446.
    1. Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic parkinsonism in humans due to a product of meperidine-analog synthesis. Science. 1983;219(4587):979–980.
    1. Forno LS, DeLanney LE, Irwin I, Langston JW. Similarities and differences between MPTP-induced parkinsonsim and Parkinson’s disease. Neuropathologic considerations. Advances in neurology. 1993;60:600–608.
    1. Halliday G, Herrero MT, Murphy K, et al. No Lewy pathology in monkeys with over 10 years of severe MPTP parkinsonism. Movement Disorders. 2009;24(10):1519–1523.
    1. Kowall NW, Hantraye P, Brouillet E, Beal MF, McKee AC, Ferrante RJ. MPTP induces alpha-synuclein aggregation in the substantia nigra of baboons. NeuroReport. 2000;11(1):211–213.
    1. Fornai F, Schlüter OM, Lenzi P, et al. Parkinson-like syndrome induced by continuous MPTP infusion: convergent roles of the ubiquitin-proteasome system and α-synuclein. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(9):3413–3418.
    1. Cui M, Aras R, Christian WV, et al. The organic cation transporter-3 is a pivotal modulator of neurodegeneration in the nigrostriatal dopaminergic pathway. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(19):8043–8048.
    1. Javitch JA, D’Amato RJ, Strittmatter SM, Snyder SH. Parkinsonism-inducing neurotoxin, N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine: Uptake of the metabolite N-methyl-4-phenylpyridine by dopamine neurons explains selective toxicity. Proceedings of the National Academy of Sciences of the United States of America. 1985;82(7):2173–2177.
    1. Bezard E, Gross CE, Fournier MC, Dovero S, Bloch B, Jaber M. Absence of MPTP-induced neuronal death in mice lacking the dopamine transporter. Experimental Neurology. 1999;155(2):268–273.
    1. Gainetdinov RR, Fumagalli F, Wang YM, et al. Increased MPTP neurotoxicity in vesicular monoamine transporter 2 heterozygote knockout mice. Journal of Neurochemistry. 1998;70(5):1973–1978.
    1. Guillot TS, Shepherd KR, Richardson JR, et al. Reduced vesicular storage of dopamine exacerbates methamphetamine-induced neurodegeneration and astrogliosis. Journal of Neurochemistry. 2008;106(5):2205–2217.
    1. Guillot TS, Miller GW. Protective actions of the vesicular monoamine transporter 2 (VMAT2) in monoaminergic neurons. Molecular Neurobiology. 2009;39(2):149–170.
    1. Burke WJ, Kumar VB, Pandey N, et al. Aggregation of α-synuclein by DOPAL, the monoamine oxidase metabolite of dopamine. Acta Neuropathologica. 2008;115(2):193–203.
    1. Panneton WM, Kumar VB, Gan Q, Burke WJ, Galvin JE. The neurotoxicity of DOPAL: behavioral and stereological evidence for its role in Parkinson disease pathogenesis. PLoS One. 2010;5(12) Article ID e15251.
    1. Zigmond MJ, Berger TW, Grace AA, Stricker EM. Compensatory responses to nigrostriatal bundle injury. Studies with 6-hydroxydopamine in an animal model of parkinsonism. Molecular and Chemical Neuropathology. 1989;10(3):185–200.
    1. Chiueh CC, Markey SP, Burns RS. Neurochemical and behavioral effects of systemic and intranigral administration of N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in the rat. European Journal of Pharmacology. 1984;100(2):189–194.
    1. Przedborski S, Jackson-Lewis V, Naini AB, et al. The parkinsonian toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): a technical review of its utility and safety. Journal of Neurochemistry. 2001;76(5):1265–1274.
    1. Langston JW, Langston EB, Irwin I. MPTP-induced parkinsonism in human and non-human primates—clinical and experimental aspects. Acta Neurologica Scandinavica. 1984;70(supplement 100):49–54.
    1. Bankiewicz KS, Oldfield EH, Chiueh CC. Hemiparkinsonism in monkeys after unilateral internal carotid artery infusion of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Life Sciences. 1986;39(1):7–16.
    1. Iravani MM, Syed E, Jackson MJ, Johnston LC, Smith LA, Jenner P. A modified MPTP treatment regime produces reproducible partial nigrostriatal lesions in common marmosets. European Journal of Neuroscience. 2005;21(4):841–854.
    1. Hantraye P, Varastet M, Peschanski M, et al. Stable parkinsonian syndrome and uneven loss of striatal dopamine fibres following chronic MPTP administration in baboons. Neuroscience. 1993;53(1):169–178.
    1. Bezard E, Imbert C, Deloire X, Bioulac B, Gross CE. A chronic MPTP model reproducing the slow evolution of Parkinson’s disease: evolution of motor symptoms in the monkey. Brain Research. 1997;766(1-2):107–112.
    1. Mounayar S, Boulet S, Tandé D, et al. A new model to study compensatory mechanisms in MPTP-treated monkeys exhibiting recovery. Brain. 2007;130(11):2898–2914.
    1. Blesa J, Juri C, Collantes M, et al. Progression of dopaminergic depletion in a model of MPTP-induced Parkinsonism in non-human primates. An 18F-DOPA and 11C-DTBZ PET study. Neurobiology of Disease. 2010;38(3):456–463.
    1. Ovadia A, Zhang Z, Gash DM. Increased susceptibility to MPTP toxicity in middle-aged rhesus monkeys. Neurobiology of Aging. 1995;16(6):931–937.
    1. Pessiglione M, Guehl D, Hirsch EC, Féger J, Tremblay L. Disruption of self-organized actions in monkeys with progressive MPTP-induced parkinsonism. I. Effects of task complexity. European Journal of Neuroscience. 2004;19(2):426–436.
    1. Schneider JS. Modeling Cognitive Deficits Associated with Parkinsonism in the Chronic-Low-Dose MPTP-Treated Monkey. Boca Raton, Fla, USA: CRC Press; 2006.
    1. Anderson G, Noorian AR, Taylor G, et al. Loss of enteric dopaminergic neurons and associated changes in colon motility in an MPTP mouse model of Parkinson’s disease. Experimental Neurology. 2007;207(1):4–12.
    1. Fox SH, Chuang R, Brotchie JM. Serotonin and Parkinson’s disease: on movement, mood, and madness. Movement Disorders. 2009;24(9):1255–1266.
    1. Barraud Q, Obeid I, Aubert I, et al. Neuroanatomical study of the A11 diencephalospinal pathway in the non-human primate. PLoS One. 2010;5(10) Article ID e13306.
    1. Vezoli J, Fifel K, Leviel V, et al. Early presymptomatic and long-term changes of rest activity cycles and cognitive behavior in a MPTP-monkey model of Parkinson's disease. PLoS One. 2011;6(8) Article ID e23952.
    1. Bergman H, Wichmann T, DeLong MR. Reversal of experimental Parkinsonism by lesions of the subthalamic nucleus. Science. 1990;249(4975):1436–1438.
    1. Guridi J, Herrero MT, Luquin MR, et al. Subthalamotomy in parkinsonian monkeys. Behavioural and biochemical analysis. Brain. 1996;119(5):1717–1727.
    1. Bezard E, Przedborski S. A tale on animal models of Parkinson's Disease. Movement Disorders. 2011;26(6):993–1002.
    1. 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. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(5):2837–2842.
    1. Dauer W, Kholodilov N, Vila M, et al. Resistance of α-synuclein null mice to the parkinsonian neurotoxin MPTP. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(22):14524–14529.
    1. Berry C, La Vecchia C, Nicotera P. Paraquat and parkinson’s disease. Cell Death and Differentiation. 2010;17(7):1115–1125.
    1. Day BJ, Patel M, Calavetta L, Chang LY, Stamler JS. A mechanism of paraquat toxicity involving nitric oxide synthase. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(22):12760–12765.
    1. Przedborski S, Ischiropoulos H. Reactive oxygen and nitrogen species: weapons of neuronal destruction in models of Parkinson’s disease. Antioxidants and Redox Signaling. 2005;7(5-6):685–693.
    1. Brooks AI, Chadwick CA, Gelbard HA, Cory-Slechta DA, Federoff HJ. Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Research. 1999;823(1-2):1–10.
    1. 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. Neurobiology of Disease. 2002;10(2):119–127.
    1. Thiffault C, Langston JW, Di Monte DA. Increased striatal dopamine turnover following acute administration of rotenone to mice. Brain Research. 2000;885(2):283–288.
    1. 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 Research. 2000;873(2):225–234.
    1. Rappold PM, Cui M, Chesser AS, et al. Paraquat neurotoxicity is mediated by the dopamine transporter and organic cation transporter-3. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(51):20766–20771.
    1. Manning-Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Di Monte DA. The herbicide paraquat causes up-regulation and aggregation of α-synuclein in mice: paraquat and α-synuclein. Journal of Biological Chemistry. 2002;277(3):1641–1644.
    1. Takahashi RN, Rogerio R, Zanin M. Maneb enhances MPTP neurotoxicity in mice. Research Communications in Chemical Pathology and Pharmacology. 1989;66(1):167–170.
    1. Kachroo A, Irizarry MC, Schwarzschild MA. Caffeine protects against combined paraquat and maneb-induced dopaminergic neuron degeneration. Experimental Neurology. 2010;223(2):657–661.
    1. Butterfield PG, Valanis BG, Spencer PS, Lindeman CA, Nutt JG. Environmental antecedents of young-onset Parkinson’s disease. Neurology. 1993;43(6):1150–1158.
    1. Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Richardson RJ. The risk of Parkinson’s disease with exposure to pesticides, farming, well water, and rural living. Neurology. 1998;50(5):1346–1350.
    1. Barlow BK, Richfield EK, Cory-Slechta DA, Thiruchelvam M. A fetal risk factor for Parkinson’s disease. Developmental Neuroscience. 2004;26(1):11–23.
    1. Tang CC, Poston KL, Dhawan V, Eidelberg D. Abnormalities in metabolic network activity precede the onset of motor symptoms in Parkinson’s disease. Journal of Neuroscience. 2010;30(3):1049–1056.
    1. Tanner CM, Kame F, Ross GW, et al. Rotenone, paraquat, and Parkinson's disease. Environmental Health Perspectives. 2011;119(6):866–872.
    1. Hisata J. Lake and stream rehabilitation: rotenone use and health risks. Final supplemental environmental impact statement. Washington Satet Deparment of Fish and Wildlife; 2002.
    1. Inden M, Kitamura Y, Takeuchi H, et al. Neurodegeneration of mouse nigrostriatal dopaminergic system induced by repeated oral administration of rotenone is prevented by 4-phenylbutyrate, a chemical chaperone. Journal of Neurochemistry. 2007;101(6):1491–1504.
    1. Inden M, Kitamura Y, Abe M, Tamaki A, Takata K, Taniguchi T. Parkinsonian rotenone mouse model: reevaluation of long-term administration of rotenone in C57BL/6 mice. Biological and Pharmaceutical Bulletin. 2011;34(1):92–96.
    1. Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nature Neuroscience. 2000;3(12):1301–1306.
    1. 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. Behavioural Brain Research. 2004;151(1-2):117–124.
    1. Cannon JR, Tapias V, Na HM, Honick AS, Drolet RE, Greenamyre JT. A highly reproducible rotenone model of Parkinson’s disease. Neurobiology of Disease. 2009;34(2):279–290.
    1. Höglinger GU, Féger J, Prigent A, et al. Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. Journal of Neurochemistry. 2003;84(3):491–502.
    1. Sherer TB, Kim JH, Betarbet R, Greenamyre JT. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and α-synuclein aggregation. Experimental Neurology. 2003;179(1):9–16.
    1. Wu Y-N, Johnson SW. Dopamine oxidation facilitates rotenone-dependent potentiation of N-methyl-d-aspartate currents in rat substantia nigra dopamine neurons. Neuroscience. 2011;195:138–144.
    1. 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 Research. 1997;753(1):157–162.
    1. Takeuchi H, Yanagida T, Inden M, et al. Nicotinic receptor stimulation protects nigral dopaminergic neurons in rotenone-induced Parkinson’s disease models. Journal of Neuroscience Research. 2009;87(2):576–585.
    1. Pan-Montojo F, Anichtchik O, Dening Y, et al. Progression of Parkinson’s disease pathology is reproduced by intragastric administration of rotenone in mice. PLoS One. 2010;5(1) Article ID e8762.
    1. Pan-Montojo FJ, Funk RHW. Oral administration of rotenone using a gavage and image analysis of alpha-synuclein inclusions in the enteric nervous system. Journal of Visualized Experiments. 2010;(44) Article ID e2123.
    1. Krüger R, Kuhn W, Müller T, et al. Ala30Pro mutation in the gene encoding α-synuclein in Parkinson's disease. Nature Genetics. 1998;18(2):106–108.
    1. Giasson BI, Duda JE, Quinn SM, Zhang B, Trojanowski JQ, Lee VMY. Neuronal α-synucleinopathy with severe movement disorder in mice expressing A53T human α-synuclein. Neuron. 2002;34(4):521–533.
    1. Masliah E, Rockenstein E, Veinbergs I, et al. Dopaminergic loss and inclusion body formation in α-synuclein mice: Implications for neurodegenerative disorders. Science. 2000;287(5456):1265–1269.
    1. Abeliovich A, Schmitz Y, Fariñas I, et al. Mice lacking α-synuclein display functional deficits in the nigrostriatal dopamine system. Neuron. 2000;25(1):239–252.
    1. Thomas B, Mandir AS, West N, et al. Resistance to MPTP-Neurotoxicity in α-synuclein knockout mice is complemented by human α-synuclein and associated with increased β-synuclein and Akt activation. PLoS One. 2011;6(1) Article ID e16706.
    1. Feany MB, Bender WW. A Drosophila model of Parkinson’s disease. Nature. 2000;404(6776):394–398.
    1. Wang D, Tang B, Zhao G, et al. Dispensable role of Drosophila ortholog of LRRK2 kinase activity in survival of dopaminergic neurons. Molecular Neurodegeneration. 2008;3(1, article no. 3)
    1. Zimprich A, Biskup S, Leitner P, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44(4):601–607.
    1. Li Y, Liu W, Oo TF, et al. Mutant LRRK2R1441G BAC transgenic mice recapitulate cardinal features of Parkinson’s disease. Nature Neuroscience. 2009;12(7):826–828.
    1. Moore DJ, Dawson TM. Value of genetic models in understanding the cause and mechanisms of Parkinson’s disease. Current Neurology and Neuroscience Reports. 2008;8(4):288–296.
    1. Shin J-H, Ko HS, Kang H, et al. PARIS (ZNF746) repression of PGC-1α contributes to neurodegeneration in parkinson's disease. Cell. 2011;144(5):689–702.
    1. Peng J, Oo ML, Andersen JK. Synergistic effects of environmental risk factors and gene mutations in Parkinson’s disease accelerate age-related neurodegeneration. Journal of Neurochemistry. 2010;115(6):1363–1373.

Source: PubMed

3
Iratkozz fel