Please wait a minute...
Frontiers in Biology

ISSN 1674-7984

ISSN 1674-7992(Online)

CN 11-5892/Q

Front. Biol.    2014, Vol. 9 Issue (6) : 489-503    https://doi.org/10.1007/s11515-014-1337-8
REVIEW
Mitochondrial dysfunction in Parkinson’s disease: a possible target for neuroprotection
Jacqueline A. GLEAVE,Peter D. PERRI,Joanne E. NASH()
Centre for Neurobiology of Stress, Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, M1C1A4, Canada
 Download: PDF(264 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Mitochondria are dynamic organelles which are required for maintaining cellular homeostasis. Thus, it is not surprising that irregularities in mitochondrial function result in cellular damage and are linked with neurodegenerative diseases, such as Parkinson’s disease. Evidence that mitochondrial dysfunction is key to the pathogenesis of Parkinson’s disease is founded in studies in post-mortem tissue from patients with Parkinson’s disease, and also from genetic studies stemming from patients with familial Parkinson’s disease. Whether triggered by environmental or genetic factors, mitochondrial dysfunction occurs early in the pathogenic process, and is central to Parkinson’s disease pathology. As such, targeting the mitochondria to slow or halt disease progression is an attractive strategy for disease-modifying agents in Parkinson’s disease. Indeed, several therapies which target the mitochondria have been investigated as neuroprotective treatments for Parkinson’s disease. This review will discuss the evidence supporting mitochondrial dysfunction in Parkinson’s disease pathology as well as treatment strategies that target the mitochondria.

Keywords Parkinson’s disease      mitochondria      oxidative stress      lysosome      UPS     
Corresponding Author(s): Joanne E. NASH   
Just Accepted Date: 24 October 2014   Issue Date: 13 January 2015
 Cite this article:   
Jacqueline A. GLEAVE,Peter D. PERRI,Joanne E. NASH. Mitochondrial dysfunction in Parkinson’s disease: a possible target for neuroprotection[J]. Front. Biol., 2014, 9(6): 489-503.
 URL:  
https://academic.hep.com.cn/fib/EN/10.1007/s11515-014-1337-8
https://academic.hep.com.cn/fib/EN/Y2014/V9/I6/489
1 Albani D, Polito L, Batelli S, De Mauro S, Fracasso C, Martelli G, Colombo L, Manzoni C, Salmona M, Caccia S, Negro A, Forloni G (2009). The SIRT1 activator resveratrol protects SK-N-BE cells from oxidative stress and against toxicity caused by alpha-synuclein or amyloid-beta (1-42) peptide. J Neurochem, 110(5): 1445–1456
https://doi.org/10.1111/j.1471-4159.2009.06228.x pmid: 19558452
2 Andres R H, Huber A W, Schlattner U, Pérez-Bouza A, Krebs S H, Seiler R W, Wallimann T, Widmer H R (2005). Effects of creatine treatment on the survival of dopaminergic neurons in cultured fetal ventral mesencephalic tissue. Neuroscience, 133(3): 701–713
https://doi.org/10.1016/j.neuroscience.2005.03.004 pmid: 15890457
3 Andres-Mateos E, Perier C, Zhang L, Blanchard-Fillion B, Greco T M, Thomas B, Ko H S, Sasaki M, Ischiropoulos H, Przedborski S, Dawson T M, Dawson V L (2007). DJ-1 gene deletion reveals that DJ-1 is an atypical peroxiredoxin-like peroxidase. Proc Natl Acad Sci USA, 104(37): 14807–14812
https://doi.org/10.1073/pnas.0703219104 pmid: 17766438
4 Bedford L, Hay D, Devoy A, Paine S, Powe D G, Seth R, Gray T, Topham I, Fone K, Rezvani N, Mee M, Soane T, Layfield R, Sheppard P W, Ebendal T, Usoskin D, Lowe J, Mayer R J(2008). Depletion of 26S proteasomes in mouse brain neurons causes neurodegeneration and Lewy-like inclusions resembling human pale bodies. J Neurosci, 28: 8189–8198
5 Beher D, Wu J, Cumine S, Kim K W, Lu S C, Atangan L, Wang M (2009). Resveratrol is not a direct activator of SIRT1 enzyme activity. Chem Biol Drug Des, 74(6): 619–624
https://doi.org/10.1111/j.1747-0285.2009.00901.x pmid: 19843076
6 Bender A, Krishnan K J, Morris C M, Taylor G A, Reeve A K, Perry R H, Jaros E, Hersheson J S, Betts J, Klopstock T, Taylor R W, Turnbull D M (2006). High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat Genet, 38(5): 515–517
https://doi.org/10.1038/ng1769 pmid: 16604074
7 Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F (1973). Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci, 20(4): 415–455
https://doi.org/10.1016/0022-510X(73)90175-5 pmid: 4272516
8 Blackinton J, Lakshminarasimhan M, Thomas K J, Ahmad R, Greggio E, Raza A S, Cookson M R, Wilson M A (2009). Formation of a stabilized cysteine sulfinic acid is critical for the mitochondrial function of the parkinsonism protein DJ-1. J Biol Chem, 284(10): 6476–6485
https://doi.org/10.1074/jbc.M806599200 pmid: 19124468
9 Bové J, Zhou C, Jackson-Lewis V, Taylor J, Chu Y, Rideout H J, Wu D C, Kordower J H, Petrucelli L, Przedborski S (2006). Proteasome inhibition and Parkinson’s disease modeling. Ann Neurol, 60(2): 260–264
https://doi.org/10.1002/ana.20937 pmid: 16862585
10 Brunet A, Sweeney L B, Sturgill J F, Chua K F, Greer P L, Lin Y, Tran H, Ross S E, Mostoslavsky R, Cohen H Y, Hu L S, Cheng H L, Jedrychowski M P, Gygi S P, Sinclair D A, Alt F W, Greenberg M E (2004). Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science, 303(5666): 2011–2015
https://doi.org/10.1126/science.1094637 pmid: 14976264
11 Canet-Avilés R M, Wilson M A, Miller D W, Ahmad R, McLendon C, Bandyopadhyay S, Baptista M J, Ringe D, Petsko G A, Cookson M R (2004). The Parkinson’s disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven mitochondrial localization. Proc Natl Acad Sci USA, 101(24): 9103–9108
https://doi.org/10.1073/pnas.0402959101 pmid: 15181200
12 Chan C S, Gertler T S, Surmeier D J(2010). A molecular basis for the increased vulnerability of substantia nigra dopamine neurons in aging and Parkinson's disease. Mov Disord, 25 (Suppl 1): S63–70
13 Chan C S, Guzman J N, Ilijic E, Mercer J N, Rick C, Tkatch T, Meredith G E, Surmeier D J (2007). ‘Rejuvenation’ protects neurons in mouse models of Parkinson’s disease. Nature, 447(7148): 1081–1086
https://doi.org/10.1038/nature05865 pmid: 17558391
14 Chao J, Yu M S, Ho Y S, Wang M, Chang R C (2008). Dietary oxyresveratrol prevents parkinsonian mimetic 6-hydroxydopamine neurotoxicity. Free Radic Biol Med, 45(7): 1019–1026
https://doi.org/10.1016/j.freeradbiomed.2008.07.002 pmid: 18675900
15 Chartier-Harlin M C, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J, Hulihan M, Waucquier N, Defebvre L, Amouyel P, Farrer M, Destée A (2004). Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet, 364(9440): 1167–1169
https://doi.org/10.1016/S0140-6736(04)17103-1 pmid: 15451224
16 Cherra S J 3rd, Steer E, Gusdon A M, Kiselyov K, Chu C T (2013). Mutant LRRK2 elicits calcium imbalance and depletion of dendritic mitochondria in neurons. Am J Pathol, 182(2): 474–484
https://doi.org/10.1016/j.ajpath.2012.10.027 pmid: 23231918
17 Chinta S J, Mallajosyula J K, Rane A, Andersen J K (2010). Mitochondrial α-synuclein accumulation impairs complex I function in dopaminergic neurons and results in increased mitophagy in vivo. Neurosci Lett, 486(3): 235–239
https://doi.org/10.1016/j.neulet.2010.09.061 pmid: 20887775
18 Ciron C, Lengacher S, Dusonchet J, Aebischer P, Schneider B L (2012). Sustained expression of PGC-1α in the rat nigrostriatal system selectively impairs dopaminergic function. Hum Mol Genet, 21(8): 1861–1876
https://doi.org/10.1093/hmg/ddr618 pmid: 22246294
19 Clark I E, Dodson M W, Jiang C, Cao J H, Huh J R, Seol J H, Yoo S J, Hay B A, Guo M (2006). Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin. Nature, 441(7097): 1162–1166
https://doi.org/10.1038/nature04779 pmid: 16672981
20 Cole N B, Murphy D D (2002). The cell biology of alpha-synuclein: a sticky problem? Neuromolecular Med, 1(2): 95–109
https://doi.org/10.1385/NMM:1:2:95 pmid: 12025860
21 Cookson M R (2003). Parkin’s substrates and the pathways leading to neuronal damage. Neuromolecular Med, 3(1): 1–13
https://doi.org/10.1385/NMM:3:1:1 pmid: 12665672
22 Couzin J (2007). Clinical research. Testing a novel strategy against Parkinson’s disease. Science, 315(5820): 1778
https://doi.org/10.1126/science.315.5820.1778 pmid: 17395800
23 Cuervo A M, Stefanis L, Fredenburg R, Lansbury P T, Sulzer D (2004). Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science, 305(5688): 1292–1295
https://doi.org/10.1126/science.1101738 pmid: 15333840
24 Dauer W, Przedborski S (2003). Parkinson’s disease: mechanisms and models. Neuron, 39(6): 889–909
https://doi.org/10.1016/S0896-6273(03)00568-3 pmid: 12971891
25 Devi L, Raghavendran V, Prabhu B M, Avadhani N G, Anandatheerthavarada H K (2008). Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J Biol Chem, 283(14): 9089–9100
https://doi.org/10.1074/jbc.M710012200 pmid: 18245082
26 Donmez G, Arun A, Chung C Y, McLean P J, Lindquist S, Guarente L(2012). SIRT1 protects against alpha-synuclein aggregation by activating molecular chaperones. J Neurosci, 32: 124–132
27 Dorsey E R, Constantinescu R, Thompson J P, Biglan K M, Holloway R G, Kieburtz K, Marshall F J, Ravina B M, Schifitto G, Siderowf A, Tanner C M (2007). Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030. Neurology, 68(5): 384–386
https://doi.org/10.1212/01.wnl.0000247740.47667.03 pmid: 17082464
28 Dryanovski D I, Guzman J N, Xie Z, Galteri D J, Volpicelli-Daley L A, Lee V M, Miller R J, Schumacker P T, Surmeier D J (2013). Calcium entry and alpha-synuclein inclusions elevate dendritic mitochondrial oxidant stress in dopaminergic neurons. J Neurosci, 33(24): 10154–10164
29 Ekstrand M I, Terzioglu M, Galter D, Zhu S, Hofstetter C, Lindqvist E, Thams S, Bergstrand A, Hansson F S, Trifunovic A, Hoffer B, Cullheim S, Mohammed A H, Olson L, Larsson N G (2007). Progressive parkinsonism in mice with respiratory-chain-deficient dopamine neurons. Proc Natl Acad Sci USA, 104(4): 1325–1330
https://doi.org/10.1073/pnas.0605208103 pmid: 17227870
30 Esteves A R, Lu J, Rodova M, Onyango I, Lezi E, Dubinsky R, Lyons K E, Pahwa R, Burns J M, Cardoso S M, Swerdlow R H (2010). Mitochondrial respiration and respiration-associated proteins in cell lines created through Parkinson’s subject mitochondrial transfer. J Neurochem, 113(3): 674–682
https://doi.org/10.1111/j.1471-4159.2010.06631.x pmid: 20132468
31 Exner N, Lutz A K, Haass C, Winklhofer K F (2012). Mitochondrial dysfunction in Parkinson’s disease: molecular mechanisms and pathophysiological consequences. EMBO J, 31(14): 3038–3062
https://doi.org/10.1038/emboj.2012.170 pmid: 22735187
32 Ferretta A, Gaballo A, Tanzarella P, Piccoli C, Capitanio N, Nico B, Annese T, Di Paola M, Dell’aquila C, De Mari M, Ferranini E, Bonifati V, Pacelli C, Cocco T (2014). Effect of resveratrol on mitochondrial function: implications in parkin-associated familiar Parkinson’s disease. Biochim Biophys Acta, 1842(7): 902–915
https://doi.org/10.1016/j.bbadis.2014.02.010 pmid: 24582596
33 Frye R A (2000). Phylogenetic classification of prokaryotic and eukaryotic Sir2-like proteins. Biochem Biophys Res Commun, 273(2): 793–798
https://doi.org/10.1006/bbrc.2000.3000 pmid: 10873683
34 Fuchs J, Nilsson C, Kachergus J, Munz M, Larsson E M, Schüle B, Langston J W, Middleton F A, Ross O A, Hulihan M, Gasser T, Farrer M J (2007). Phenotypic variation in a large Swedish pedigree due to SNCA duplication and triplication. Neurology, 68(12): 916–922
https://doi.org/10.1212/01.wnl.0000254458.17630.c5 pmid: 17251522
35 Gautier C A, Kitada T, Shen J (2008). Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress. Proc Natl Acad Sci USA, 105(32): 11364–11369
https://doi.org/10.1073/pnas.0802076105 pmid: 18687901
36 Gerhart-Hines Z, Rodgers J T, Bare O, Lerin C, Kim S H, Mostoslavsky R, Alt F W, Wu Z, Puigserver P (2007). Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha. EMBO J, 26(7): 1913–1923
https://doi.org/10.1038/sj.emboj.7601633 pmid: 17347648
37 Gispert S, Ricciardi F, Kurz A, Azizov M, Hoepken H H, Becker D, Voos W, Leuner K, Müller W E, Kudin A P, Kunz W S, Zimmermann A, Roeper J, Wenzel D, Jendrach M, García-Arencíbia M, Fernández-Ruiz J, Huber L, Rohrer H, Barrera M, Reichert A S, Rüb U, Chen A, Nussbaum R L, Auburger G (2009). Parkinson phenotype in aged PINK1-deficient mice is accompanied by progressive mitochondrial dysfunction in absence of neurodegeneration. PLoS ONE, 4(6): e5777
https://doi.org/10.1371/journal.pone.0005777 pmid: 19492057
38 Goedert M (2001). Alpha-synuclein and neurodegenerative diseases. Nat Rev Neurosci, 2(7): 492–501
https://doi.org/10.1038/35081564 pmid: 11433374
39 Goldberg M S, Fleming S M, Palacino J J, Cepeda C, Lam H A, Bhatnagar A, Meloni E G, Wu N, Ackerson L C, Klapstein G J, Gajendiran M, Roth B L, Chesselet M F, Maidment N T, Levine M S, Shen J (2003). Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons. J Biol Chem, 278(44): 43628–43635
https://doi.org/10.1074/jbc.M308947200 pmid: 12930822
40 Gómez-Sánchez R, Gegg M E, Bravo-San Pedro J M, Niso-Santano M, Alvarez-Erviti L, Pizarro-Estrella E, Gutiérrez-Martín Y, Alvarez-Barrientos A, Fuentes J M, González-Polo R A, Schapira A H (2014). Mitochondrial impairment increases FL-PINK1 levels by calcium-dependent gene expression. Neurobiol Dis, 62: 426–440
https://doi.org/10.1016/j.nbd.2013.10.021 pmid: 24184327
41 González-Polo R, Niso-Santano M, Morán J M, Ortiz-Ortiz M A, Bravo-San Pedro J M, Soler G, Fuentes J M (2009). Silencing DJ-1 reveals its contribution in paraquat-induced autophagy. J Neurochem, 109(3): 889–898
https://doi.org/10.1111/j.1471-4159.2009.06020.x pmid: 19425177
42 Good C H, Hoffman A F, Hoffer B J, Chefer V I, Shippenberg T S, Backman C M, Larsson N G, Olson L, Gellhaar S, Galter D, Lupica C R(2011). Impaired nigrostriatal function precedes behavioral deficits in a genetic mitochondrial model of Parkinson's disease. FASEB J, 25:1333–1344
43 Greenamyre J T, Betarbet R, Sherer T B (2003). The rotenone model of Parkinson’s disease: genes, environment and mitochondria. Parkinsonism Relat Disord, 9(Suppl 2): S59–S64
https://doi.org/10.1016/S1353-8020(03)00023-3 pmid: 12915069
44 Greene J C, Whitworth A J, Kuo I, Andrews L A, Feany M B, Pallanck L J (2003). Mitochondrial pathology and apoptotic muscle degeneration in Drosophila parkin mutants. Proc Natl Acad Sci USA, 100(7): 4078–4083
https://doi.org/10.1073/pnas.0737556100 pmid: 12642658
45 Greggio E, Jain S, Kingsbury A, Bandopadhyay R, Lewis P, Kaganovich A, van der Brug M P, Beilina A, Blackinton J, Thomas K J, Ahmad R, Miller D W, Kesavapany S, Singleton A, Lees A, Harvey R J, Harvey K, Cookson M R (2006). Kinase activity is required for the toxic effects of mutant LRRK2/dardarin. Neurobiol Dis, 23(2): 329–341
https://doi.org/10.1016/j.nbd.2006.04.001 pmid: 16750377
46 Gu G, Reyes P E, Golden G T, Woltjer R L, Hulette C, Montine T J, Zhang J (2002). Mitochondrial DNA deletions/rearrangements in parkinson disease and related neurodegenerative disorders. J Neuropathol Exp Neurol, 61(7): 634–639
pmid: 12125742
47 Guardia-Laguarta C, Area-Gomez E, Rub C, Liu Y, Magrane J, Becker D, Voos W, Schon E A, Przedborski S(2014). alpha-Synuclein is localized to mitochondria-associated ER membranes. J Neurosc, 34: 249–259
48 Guzman J N, Sanchez-Padilla J, Wokosin D, Kondapalli J, Ilijic E, Schumacker P T, Surmeier D J (2010). Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature, 468(7324): 696–700
https://doi.org/10.1038/nature09536 pmid: 21068725
49 Haas R H, Nasirian F, Nakano K, Ward D, Pay M, Hill R, Shults C W (1995). Low platelet mitochondrial complex I and complex II/III activity in early untreated Parkinson’s disease. Ann Neurol, 37(6): 714–722
https://doi.org/10.1002/ana.410370604 pmid: 7778844
50 Hayashi Y, Yoshida M, Yamato M, Ide T, Wu Z, Ochi-Shindou M, Kanki T, Kang D, Sunagawa K, Tsutsui H, Nakanishi H(2008). Reverse of age-dependent memory impairment and mitochondrial DNA damage in microglia by an overexpression of human mitochondrial transcription factor a in mice. J Neurosci, 28: 8624–8634
51 Healy D G, Abou-Sleiman P M, Casas J P, Ahmadi K R, Lynch T, Gandhi S, Muqit M M, Foltynie T, Barker R, Bhatia K P, Quinn N P, Lees A J, Gibson J M, Holton J L, Revesz T, Goldstein D B, Wood N W (2006). UCHL-1 is not a Parkinson’s disease susceptibility gene. Ann Neurol, 59(4): 627–633
https://doi.org/10.1002/ana.20757 pmid: 16450370
52 H?glinger G U, Carrard G, Michel P P, Medja F, Lombès A, Ruberg M, Friguet B, Hirsch E C (2003). Dysfunction of mitochondrial complex I and the proteasome: interactions between two biochemical deficits in a cellular model of Parkinson’s disease. J Neurochem, 86(5): 1297–1307
https://doi.org/10.1046/j.1471-4159.2003.01952.x pmid: 12911637
53 Howitz K T, Bitterman K J, Cohen H Y, Lamming D W, Lavu S, Wood J G, Zipkin R E, Chung P, Kisielewski A, Zhang L L, Scherer B, Sinclair D A (2003). Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature, 425(6954): 191–196
https://doi.org/10.1038/nature01960 pmid: 12939617
54 Ikebe S, Tanaka M, Ozawa T (1995). Point mutations of mitochondrial genome in Parkinson’s disease. Brain Res Mol Brain Res, 28(2): 281–295
https://doi.org/10.1016/0169-328X(94)00209-W pmid: 7723627
55 Imai Y, Soda M, Takahashi R (2000). Parkin suppresses unfolded protein stress-induced cell death through its E3 ubiquitin-protein ligase activity. J Biol Chem, 275(46): 35661–35664
https://doi.org/10.1074/jbc.C000447200 pmid: 10973942
56 Inden M, Taira T, Kitamura Y, Yanagida T, Tsuchiya D, Takata K, Yanagisawa D, Nishimura K, Taniguchi T, Kiso Y, Yoshimoto K, Agatsuma T, Koide-Yoshida S, Iguchi-Ariga S M, Shimohama S, Ariga H (2006). PARK7 DJ-1 protects against degeneration of nigral dopaminergic neurons in Parkinson’s disease rat model. Neurobiol Dis, 24(1): 144–158
https://doi.org/10.1016/j.nbd.2006.06.004 pmid: 16860563
57 Irrcher I, Aleyasin H, Seifert E L, Hewitt S J, Chhabra S, Phillips M, Lutz A K, Rousseaux M W, Bevilacqua L, Jahani-Asl A, Callaghan S, MacLaurin J G, Winklhofer K F, Rizzu P, Rippstein P, Kim R H, Chen C X, Fon E A, Slack R S, Harper M E, McBride H M, Mak T W, Park D S (2010). Loss of the Parkinson’s disease-linked gene DJ-1 perturbs mitochondrial dynamics. Hum Mol Genet, 19(19): 3734–3746
https://doi.org/10.1093/hmg/ddq288 pmid: 20639397
58 Itier J M, Ibanez P, Mena M A, Abbas N, Cohen-Salmon C, Bohme G A, Laville M, Pratt J, Corti O, Pradier L, Ret G, Joubert C, Periquet M, Araujo F, Negroni J, Casarejos M J, Canals S, Solano R, Serrano A, Gallego E, Sanchez M, Denefle P, Benavides J, Tremp G, Rooney T A, Brice A, Garcia de Yebenes J (2003). Parkin gene inactivation alters behaviour and dopamine neurotransmission in the mouse. Hum Mol Genet, 12(18): 2277–2291
https://doi.org/10.1093/hmg/ddg239 pmid: 12915482
59 Jin F, Wu Q, Lu Y F, Gong Q H, Shi J S (2008). Neuroprotective effect of resveratrol on 6-OHDA-induced Parkinson’s disease in rats. Eur J Pharmacol, 600(1-3): 78–82
https://doi.org/10.1016/j.ejphar.2008.10.005 pmid: 18940189
60 Juhn M S, Tarnopolsky M(1998). Oral creatine supplementation and athletic performance: a critical review. Clin J Sport Med, 8: 286–297
61 Kakefuda K, Fujita Y, Oyagi A, Hyakkoku K, Kojima T, Umemura K, Tsuruma K, Shimazawa M, Ito M, Nozawa Y, Hara H (2009). Sirtuin 1 overexpression mice show a reference memory deficit, but not neuroprotection. Biochem Biophys Res Commun, 387(4): 784–788
https://doi.org/10.1016/j.bbrc.2009.07.119 pmid: 19643082
62 Katzenschlager R, Lees A J (2002). Treatment of Parkinson’s disease: levodopa as the first choice. J Neurol, 249(Suppl 2): II19–II24
https://doi.org/10.1007/s00415-002-1204-4 pmid: 12375059
63 Keeney P M, Quigley C K, Dunham L D, Papageorge C M, Iyer S, Thomas R R, Schwarz K M, Trimmer P A, Khan S M, Portell F R, Bergquist K E, Bennett J P Jr (2009). Mitochondrial gene therapy augments mitochondrial physiology in a Parkinson’s disease cell model. Hum Gene Ther, 20(8): 897–907
https://doi.org/10.1089/hum.2009.023 pmid: 19374590
64 Kim R H, Smith P D, Aleyasin H, Hayley S, Mount M P, Pownall S, Wakeham A, You-Ten A J, Kalia S K, Horne P, Westaway D, Lozano A M, Anisman H, Park D S, Mak T W (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 USA, 102(14): 5215–5220
https://doi.org/10.1073/pnas.0501282102 pmid: 15784737
65 Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N (1998). Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature, 392(6676): 605–608
https://doi.org/10.1038/33416 pmid: 9560156
66 Kitada T, Pisani A, Porter D R, Yamaguchi H, Tscherter A, Martella G, Bonsi P, Zhang C, Pothos E N, Shen J (2007). Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice. Proc Natl Acad Sci USA, 104(27): 11441–11446
https://doi.org/10.1073/pnas.0702717104 pmid: 17563363
67 Klivenyi P, Gardian G, Calingasan N Y, Yang L, Beal M F(2003). Additive neuroprotective effects of creatine and a cyclooxygenase 2 inhibitor against dopamine depletion in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. J Mol Neurosci, MN 21: 191–198
68 Klivenyi P, Calingasan N Y, Starkov A, Stavrovskaya I G, Kristal B S, Yang L, Wieringa B, Beal M F (2004). Neuroprotective mechanisms of creatine occur in the absence of mitochondrial creatine kinase. Neurobiol Dis, 15(3): 610–617
https://doi.org/10.1016/j.nbd.2003.12.014 pmid: 15056469
69 Klivenyi P, Siwek D, Gardian G, Yang L, Starkov A, Cleren C, Ferrante R J, Kowall N W, Abeliovich A, Beal M F (2006). Mice lacking alpha-synuclein are resistant to mitochondrial toxins. Neurobiol Dis, 21(3): 541–548
https://doi.org/10.1016/j.nbd.2005.08.018 pmid: 16298531
70 Kones R(2010). Mitochondrial therapy for Parkinson's disease: neuroprotective pharmaconutrition may be disease-modifying. Clin pharmacol, 2: 185–198
71 Kordower J H, Kanaan N M, Chu Y, Suresh Babu R, Stansell J 3rd, Terpstra B T, Sortwell C E, Steece-Collier K, Collier T J (2006). Failure of proteasome inhibitor administration to provide a model of Parkinson’s disease in rats and monkeys. Ann Neurol, 60(2): 264–268
https://doi.org/10.1002/ana.20935 pmid: 16862579
72 Kraytsberg Y, Kudryavtseva E, McKee A C, Geula C, Kowall N W, Khrapko K (2006). Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat Genet, 38(5): 518–520
https://doi.org/10.1038/ng1778 pmid: 16604072
73 Krebiehl G, Ruckerbauer S, Burbulla L F, Kieper N, Maurer B, Waak J, Wolburg H, Gizatullina Z, Gellerich F N, Woitalla D, Riess O, Kahle P J, Proikas-Cezanne T, Krüger R (2010). Reduced basal autophagy and impaired mitochondrial dynamics due to loss of Parkinson’s disease-associated protein DJ-1. PLoS ONE, 5(2): e9367
https://doi.org/10.1371/journal.pone.0009367 pmid: 20186336
74 Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, K?sel S, Przuntek H, Epplen J T, Sch?ls L, Riess O (1998). Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet, 18(2): 106–108
https://doi.org/10.1038/ng0298-106 pmid: 9462735
75 Lakshminarasimhan M, Rauh D, Schutkowski M, Steegborn C (2013). Sirt1 activation by resveratrol is substrate sequence-selective. Aging (Albany NY), 5(3): 151–154
pmid: 23524286
76 Langston J W, Ballard P, Tetrud J W, Irwin I (1983). Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science, 219(4587): 979–980
https://doi.org/10.1126/science.6823561 pmid: 6823561
77 Leroy E, Boyer R, Auburger G, Leube B, Ulm G, Mezey E, Harta G, Brownstein M J, Jonnalagada S, Chernova T, Dehejia A, Lavedan C, Gasser T, Steinbach P J, Wilkinson K D, Polymeropoulos M H (1998). The ubiquitin pathway in Parkinson’s disease. Nature, 395(6701): 451–452
https://doi.org/10.1038/26652 pmid: 9774100
78 Li C, Beal M F (2005). Leucine-rich repeat kinase 2: a new player with a familiar theme for Parkinson’s disease pathogenesis. Proc Natl Acad Sci USA, 102(46): 16535–16536
https://doi.org/10.1073/pnas.0508350102 pmid: 16275903
79 Li X, Kazgan N (2011). Mammalian sirtuins and energy metabolism. Int J Biol Sci, 7(5): 575–587
https://doi.org/10.7150/ijbs.7.575 pmid: 21614150
80 Lim K L (2007). Ubiquitin-proteasome system dysfunction in Parkinson’s disease: current evidence and controversies. Expert Rev Proteomics, 4(6): 769–781
https://doi.org/10.1586/14789450.4.6.769 pmid: 18067415
81 Lin T K, Chen S D, Chuang Y C, Lin H Y, Huang C R, Chuang J H, Wang P W, Huang S T, Tiao M M, Chen J B, Liou C W (2014). Resveratrol partially prevents rotenone-induced neurotoxicity in dopaminergic SH-SY5Y cells through induction of heme oxygenase-1 dependent autophagy. Int J Mol Sci, 15(1): 1625–1646
https://doi.org/10.3390/ijms15011625 pmid: 24451142
82 Liu D, Gharavi R, Pitta M, Gleichmann M, Mattson M P (2009). Nicotinamide prevents NAD+ depletion and protects neurons against excitotoxicity and cerebral ischemia: NAD+ consumption by SIRT1 may endanger energetically compromised neurons. Neuromolecular Med, 11(1): 28–42
https://doi.org/10.1007/s12017-009-8058-1 pmid: 19288225
83 Liu G, Zhang C, Yin J, Li X, Cheng F, Li Y, Yang H, Uéda K, Chan P, Yu S (2009). alpha-Synuclein is differentially expressed in mitochondria from different rat brain regions and dose-dependently down-regulates complex I activity. Neurosci Lett, 454(3): 187–192
https://doi.org/10.1016/j.neulet.2009.02.056 pmid: 19429081
84 Lu K T, Ko M C, Chen B Y, Huang J C, Hsieh C W, Lee M C, Chiou R Y, Wung B S, Peng C H, Yang Y L (2008). Neuroprotective effects of resveratrol on MPTP-induced neuron loss mediated by free radical scavenging. J Agric Food Chem, 56(16): 6910–6913
https://doi.org/10.1021/jf8007212 pmid: 18616261
85 Lutz A K, Exner N, Fett M E, Schlehe J S, Kloos K, L?mmermann K, Brunner B, Kurz-Drexler A, Vogel F, Reichert A S, Bouman L, Vogt-Weisenhorn D, Wurst W, Tatzelt J, Haass C, Winklhofer K F (2009). Loss of parkin or PINK1 function increases Drp1-dependent mitochondrial fragmentation. J Biol Chem, 284(34): 22938–22951
https://doi.org/10.1074/jbc.M109.035774 pmid: 19546216
86 Marques O, Outeiro T F (2012). Alpha-synuclein: from secretion to dysfunction and death. Cell Death Dis, 3(7): e350
https://doi.org/10.1038/cddis.2012.94 pmid: 22825468
87 Matthews R T, Ferrante R J, Klivenyi P, Yang L, Klein A M, Mueller G, Kaddurah-Daouk R, Beal M F (1999). Creatine and cyclocreatine attenuate MPTP neurotoxicity. Exp Neurol, 157(1): 142–149
https://doi.org/10.1006/exnr.1999.7049 pmid: 10222117
88 McLelland G L, Soubannier V, Chen C X, McBride H M, Fon E A (2014). Parkin and PINK1 function in a vesicular trafficking pathway regulating mitochondrial quality control. EMBO J, 33(4): 282–295
pmid: 24446486
89 McNaught K S, Perl D P, Brownell A L, Olanow C W (2004). Systemic exposure to proteasome inhibitors causes a progressive model of Parkinson’s disease. Ann Neurol, 56(1): 149–162
https://doi.org/10.1002/ana.20186 pmid: 15236415
90 Minakawa E N, Yamakado H, Tanaka A, Uemura K, Takeda S, Takahashi R (2013). Chicken DT40 cell line lacking DJ-1, the gene responsible for familial Parkinson’s disease, displays mitochondrial dysfunction. Neurosci Res, 77(4): 228–233
https://doi.org/10.1016/j.neures.2013.09.006 pmid: 24064392
91 Moisoi N, Fedele V, Edwards J, Martins L M (2014). Loss of PINK1 enhances neurodegeneration in a mouse model of Parkinson’s disease triggered by mitochondrial stress. Neuropharmacology, 77: 350–357
https://doi.org/10.1016/j.neuropharm.2013.10.009 pmid: 24161480
92 Morais V A, Haddad D, Craessaerts K, De Bock P J, Swerts J, Vilain S, Aerts L, Overbergh L, Grünewald A, Seibler P, Klein C, Gevaert K, Verstreken P, De Strooper B (2014). PINK1 loss-of-function mutations affect mitochondrial complex I activity via NdufA10 ubiquinone uncoupling. Science, 344(6180): 203–207
https://doi.org/10.1126/science.1249161 pmid: 24652937
93 Mortiboys H, Johansen K K, Aasly J O, Bandmann O (2010). Mitochondrial impairment in patients with Parkinson disease with the G2019S mutation in LRRK2. Neurology, 75(22): 2017–2020
https://doi.org/10.1212/WNL.0b013e3181ff9685 pmid: 21115957
94 Mortiboys H, Thomas K J, Koopman W J, Klaffke S, Abou-Sleiman P, Olpin S, Wood N W, Willems P H, Smeitink J A, Cookson M R, Bandmann O (2008). Mitochondrial function and morphology are impaired in parkin-mutant fibroblasts. Ann Neurol, 64(5): 555–565
https://doi.org/10.1002/ana.21492 pmid: 19067348
95 Mudò G, M?kel? J, Di Liberto V, Tselykh T V, Olivieri M, Piepponen P, Eriksson O, M?lki? A, Bonomo A, Kairisalo M, Aguirre J A, Korhonen L, Belluardo N, Lindholm D (2012). Transgenic expression and activation of PGC-1α protect dopaminergic neurons in the MPTP mouse model of Parkinson’s disease. Cell Mol Life Sci, 69(7): 1153–1165
https://doi.org/10.1007/s00018-011-0850-z pmid: 21984601
96 Murray A M, Weihmueller F B, Marshall J F, Hurtig H I, Gottleib G L, Joyce J N (1995). Damage to dopamine systems differs between Parkinson’s disease and Alzheimer’s disease with parkinsonism. Ann Neurol, 37(3): 300–312
https://doi.org/10.1002/ana.410370306 pmid: 7695230
97 Narendra D, Tanaka A, Suen D F, Youle R J (2008). Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol, 183(5): 795–803
https://doi.org/10.1083/jcb.200809125 pmid: 19029340
98 Neuspiel M, Schauss A C, Braschi E, Zunino R, Rippstein P, Rachubinski R A, Andrade-Navarro M A, McBride H M(2008). Cargo-selected transport from the mitochondria to peroxisomes is mediated by vesicular carriers. Curr Biol, CB 18: 102–108
99 Ng C H, Mok S Z, Koh C, Ouyang X, Fivaz M L, Tan E K, Dawson V L, Dawson T M, Yu F, Lim K L(2009). Parkin protects against LRRK2 G2019S mutant-induced dopaminergic neurodegeneration in Drosophila. J Neurosci, 29: 11257–11262
100 NINDS NET-PD Investigators (2006). A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology, 66(5): 664–671
https://doi.org/10.1212/01.wnl.0000201252.57661.e1 pmid: 16481597
101 Nishiyama S, Shitara H, Nakada K, Ono T, Sato A, Suzuki H, Ogawa T, Masaki H, Hayashi J, Yonekawa H (2010). Over-expression of Tfam improves the mitochondrial disease phenotypes in a mouse model system. Biochem Biophys Res Commun, 401(1): 26–31
https://doi.org/10.1016/j.bbrc.2010.08.143 pmid: 20816751
102 Niu J, Yu M, Wang C, Xu Z (2012). Leucine-rich repeat kinase 2 disturbs mitochondrial dynamics via Dynamin-like protein. J Neurochem, 122(3): 650–658
https://doi.org/10.1111/j.1471-4159.2012.07809.x pmid: 22639965
103 Noack H, Bednarek T, Heidler J, Ladig R, Holtz J, Szibor M (2006). TFAM-dependent and independent dynamics of mtDNA levels in C2C12 myoblasts caused by redox stress. Biochim Biophys Acta, 1760(2): 141–150
https://doi.org/10.1016/j.bbagen.2005.12.007 pmid: 16439064
104 O’Donnell K C, Lulla A, Stahl M C, Wheat N D, Bronstein J M, Sagasti A (2014). Axon degeneration and PGC-1α-mediated protection in a zebrafish model of α-synuclein toxicity. Dis Model Mech, 7(5): 571–582
https://doi.org/10.1242/dmm.013185 pmid: 24626988
105 Orenstein S J, Kuo S H, Tasset I, Arias E, Koga H, Fernandez-Carasa I, Cortes E, Honig L S, Dauer W, Consiglio A, Raya A, Sulzer D, Cuervo A M (2013). Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci, 16(4): 394–406
https://doi.org/10.1038/nn.3350 pmid: 23455607
106 Pacholec M, Bleasdale J E, Chrunyk B, Cunningham D, Flynn D, Garofalo R S, Griffith D, Griffor M, Loulakis P, Pabst B, Qiu X, Stockman B, Thanabal V, Varghese A, Ward J, Withka J, Ahn K (2010). SRT1720, SRT2183, SRT1460, and resveratrol are not direct activators of SIRT1. J Biol Chem, 285(11): 8340–8351
https://doi.org/10.1074/jbc.M109.088682 pmid: 20061378
107 Papkovskaia T D, Chau K Y, Inesta-Vaquera F, Papkovsky D B, Healy D G, Nishio K, Staddon J, Duchen M R, Hardy J, Schapira A H, Cooper J M (2012). G2019S leucine-rich repeat kinase 2 causes uncoupling protein-mediated mitochondrial depolarization. Hum Mol Genet, 21(19): 4201–4213
https://doi.org/10.1093/hmg/dds244 pmid: 22736029
108 Pardo P S, Mohamed J S, Lopez M A, Boriek A M (2011). Induction of Sirt1 by mechanical stretch of skeletal muscle through the early response factor EGR1 triggers an antioxidative response. J Biol Chem, 286(4): 2559–2566
https://doi.org/10.1074/jbc.M110.149153 pmid: 20971845
109 Parihar M S, Parihar A, Fujita M, Hashimoto M, Ghafourifar P (2008). Mitochondrial association of alpha-synuclein causes oxidative stress. Cell Mol Life Sci, 65(7-8): 1272–1284
https://doi.org/10.1007/s00018-008-7589-1 pmid: 18322646
110 Park J, Kim S Y, Cha G H, Lee S B, Kim S, Chung J (2005). Drosophila DJ-1 mutants show oxidative stress-sensitive locomotive dysfunction. Gene, 361: 133–139
https://doi.org/10.1016/j.gene.2005.06.040 pmid: 16203113
111 Park J, Lee S B, Lee S, Kim Y, Song S, Kim S, Bae E, Kim J, Shong M, Kim J M, Chung J (2006). Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin. Nature, 441(7097): 1157–1161
https://doi.org/10.1038/nature04788 pmid: 16672980
112 Parker W D Jr, Boyson S J, Parks J K (1989). Abnormalities of the electron transport chain in idiopathic Parkinson’s disease. Ann Neurol, 26(6): 719–723
https://doi.org/10.1002/ana.410260606 pmid: 2557792
113 Pesah Y, Pham T, Burgess H, Middlebrooks B, Verstreken P, Zhou Y, Harding M, Bellen H, Mardon G (2004). Drosophila parkin mutants have decreased mass and cell size and increased sensitivity to oxygen radical stress. Development, 131(9): 2183–2194
https://doi.org/10.1242/dev.01095 pmid: 15073152
114 Ping H X, Shepard P D (1999). Blockade of SK-type Ca2+-activated K+ channels uncovers a Ca2+-dependent slow afterdepolarization in nigral dopamine neurons. J Neurophysiol, 81(3): 977–984
pmid: 10085326
115 Polymeropoulos M H, Lavedan C, Leroy E, Ide S E, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos E S, Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Johnson W G, Lazzarini A M, Duvoisin R C, Di Iorio G, Golbe L I, Nussbaum R L (1997). Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science, 276(5321): 2045–2047
https://doi.org/10.1126/science.276.5321.2045 pmid: 9197268
116 Poole A C, Thomas R E, Andrews L A, McBride H M, Whitworth A J, Pallanck L J (2008). The PINK1/Parkin pathway regulates mitochondrial morphology. Proc Natl Acad Sci USA, 105(5): 1638–1643
https://doi.org/10.1073/pnas.0709336105 pmid: 18230723
117 Ramirez A, Heimbach A, Gründemann J, Stiller B, Hampshire D, Cid L P, Goebel I, Mubaidin A F, Wriekat A L, Roeper J, Al-Din A, Hillmer A M, Karsak M, Liss B, Woods C G, Behrens M I, Kubisch C (2006). Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat Genet, 38(10): 1184–1191
https://doi.org/10.1038/ng1884 pmid: 16964263
118 Ramonet D, Daher J P, Lin B M, Stafa K, Kim J, Banerjee R, Westerlund M, Pletnikova O, Glauser L, Yang L, Liu Y, Swing D A, Beal M F, Troncoso J C, McCaffery J M, Jenkins N A, Copeland N G, Galter D, Thomas B, Lee M K, Dawson T M, Dawson V L, Moore D J (2011). Dopaminergic neuronal loss, reduced neurite complexity and autophagic abnormalities in transgenic mice expressing G2019S mutant LRRK2. PLoS ONE, 6(4): e18568
https://doi.org/10.1371/journal.pone.0018568 pmid: 21494637
119 Richfield E K, Thiruchelvam M J, Cory-Slechta D A, Wuertzer C, Gainetdinov R R, Caron M G, Di Monte D A, Federoff H J (2002). Behavioral and neurochemical effects of wild-type and mutated human alpha-synuclein in transgenic mice. Exp Neurol, 175(1): 35–48
https://doi.org/10.1006/exnr.2002.7882 pmid: 12009758
120 Saha S, Guillily M D, Ferree A, Lanceta J, Chan D, Ghosh J, Hsu C H, Segal L, Raghavan K, Matsumoto K, Hisamoto N, Kuwahara T, Iwatsubo T, Moore L, Goldstein L, Cookson M, Wolozin B(2009). LRRK2 modulates vulnerability to mitochondrial dysfunction in Caenorhabditis elegans. J Neurosci, 29: 9210–9218
121 Sakata E, Yamaguchi Y, Kurimoto E, Kikuchi J, Yokoyama S, Yamada S, Kawahara H, Yokosawa H, Hattori N, Mizuno Y, Tanaka K, Kato K (2003). Parkin binds the Rpn10 subunit of 26S proteasomes through its ubiquitin-like domain. EMBO Rep, 4(3): 301–306
https://doi.org/10.1038/sj.embor.embor764 pmid: 12634850
122 Sarafian T A, Ryan C M, Souda P, Masliah E, Kar U K, Vinters H V, Mathern G W, Faull K F, Whitelegge J P, Watson J B (2013). Impairment of mitochondria in adult mouse brain overexpressing predominantly full-length, N-terminally acetylated human α-synuclein. PLoS ONE, 8(5): e63557 PMID:23667637
https://doi.org/10.1371/journal.pone.0063557
123 Scarffe L A, Stevens D A, Dawson V L, Dawson T M (2014). Parkin and PINK1: much more than mitophagy. Trends Neurosci, 37(6): 315–324
https://doi.org/10.1016/j.tins.2014.03.004 pmid: 24735649
124 Schapira A H, Cooper J M, Dexter D, Jenner P, Clark J B, Marsden C D (1989). Mitochondrial complex I deficiency in Parkinson’s disease. Lancet, 1(8649): 1269
https://doi.org/10.1016/S0140-6736(89)92366-0 pmid: 2566813
125 Shavali S, Brown-Borg H M, Ebadi M, Porter J (2008). Mitochondrial localization of alpha-synuclein protein in alpha-synuclein overexpressing cells. Neurosci Lett, 439(2): 125–128
https://doi.org/10.1016/j.neulet.2008.05.005 pmid: 18514418
126 Shimura H, Hattori N, Kubo S, Mizuno Y, Asakawa S, Minoshima S, Shimizu N, Iwai K, Chiba T, Tanaka K, Suzuki T (2000). Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat Genet, 25(3): 302–305
https://doi.org/10.1038/77060 pmid: 10888878
127 Shin J H, Ko H S, Kang H, Lee Y, Lee Y I, Pletinkova O, Troconso J C, Dawson V L, Dawson T M (2011). PARIS (ZNF746) repression of PGC-1α contributes to neurodegeneration in Parkinson’s disease. Cell, 144(5): 689–702
https://doi.org/10.1016/j.cell.2011.02.010 pmid: 21376232
128 Simon D K, Lin M T, Zheng L, Liu G J, Ahn C H, Kim L M, Mauck W M, Twu F, Beal M F, Johns D R (2004). Somatic mitochondrial DNA mutations in cortex and substantia nigra in aging and Parkinson’s disease. Neurobiol Aging, 25(1): 71–81
https://doi.org/10.1016/S0197-4580(03)00037-X pmid: 14675733
129 Snyder H, Mensah K, Theisler C, Lee J, Matouschek A, Wolozin B (2003). Aggregated and monomeric alpha-synuclein bind to the S6′ proteasomal protein and inhibit proteasomal function. J Biol Chem, 278(14): 11753–11759
https://doi.org/10.1074/jbc.M208641200 pmid: 12551928
130 Song D D, Shults C W, Sisk A, Rockenstein E, Masliah E (2004). Enhanced substantia nigra mitochondrial pathology in human alpha-synuclein transgenic mice after treatment with MPTP. Exp Neurol, 186(2): 158–172
https://doi.org/10.1016/S0014-4886(03)00342-X pmid: 15026254
131 Soubannier V, McLelland G L, Zunino R, Braschi E, Rippstein P, Fon E A, McBride H M(2012). A vesicular transport pathway shuttles cargo from mitochondria to lysosomes. Curr Biol, CB 22: 135–141
132 Spillantini M G, Crowther R A, Jakes R, Hasegawa M, Goedert M (1998). alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies. Proc Natl Acad Sci USA, 95(11): 6469–6473
https://doi.org/10.1073/pnas.95.11.6469 pmid: 9600990
133 Spillantini M G, Schmidt M L, Lee V M, Trojanowski J Q, Jakes R, Goedert M (1997). Alpha-synuclein in Lewy bodies. Nature, 388(6645): 839–840
https://doi.org/10.1038/42166 pmid: 9278044
134 St-Pierre J, Drori S, Uldry M, Silvaggi J M, Rhee J, J?ger S, Handschin C, Zheng K, Lin J, Yang W, Simon D K, Bachoo R, Spiegelman B M (2006). Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell, 127(2): 397–408
https://doi.org/10.1016/j.cell.2006.09.024 pmid: 17055439
135 Su Y C, Qi X (2013). Inhibition of excessive mitochondrial fission reduced aberrant autophagy and neuronal damage caused by LRRK2 G2019S mutation. Hum Mol Genet, 22(22): 4545–4561
https://doi.org/10.1093/hmg/ddt301 pmid: 23813973
136 Sulzer D, Zecca L (2000). Intraneuronal dopamine-quinone synthesis: a review. Neurotox Res, 1(3): 181–195
https://doi.org/10.1007/BF03033289 pmid: 12835101
137 Surmeier D J (2007). Calcium, ageing, and neuronal vulnerability in Parkinson’s disease. Lancet Neurol, 6(10): 933–938
https://doi.org/10.1016/S1474-4422(07)70246-6 pmid: 17884683
138 Surmeier D J, Guzman J N, Sanchez-Padilla J (2010). Calcium, cellular aging, and selective neuronal vulnerability in Parkinson’s disease. Cell Calcium, 47(2): 175–182
https://doi.org/10.1016/j.ceca.2009.12.003 pmid: 20053445
139 Taira T, Saito Y, Niki T, Iguchi-Ariga S M, Takahashi K, Ariga H (2004). DJ-1 has a role in antioxidative stress to prevent cell death. EMBO Rep, 5(2): 213–218
https://doi.org/10.1038/sj.embor.7400074 pmid: 14749723
140 Tanaka M, Kim Y M, Lee G, Junn E, Iwatsubo T, Mouradian M M (2004). Aggresomes formed by alpha-synuclein and synphilin-1 are cytoprotective. J Biol Chem, 279(6): 4625–4631
https://doi.org/10.1074/jbc.M310994200 pmid: 14627698
141 Valente E M, Abou-Sleiman P M, Caputo V, Muqit M M, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio A R, Healy D G, Albanese A, Nussbaum R, González-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks W P, Latchman D S, Harvey R J, Dallapiccola B, Auburger G, Wood N W (2004). Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science, 304(5674): 1158–1160
https://doi.org/10.1126/science.1096284 pmid: 15087508
142 Valente E M, Salvi S, Ialongo T, Marongiu R, Elia A E, Caputo V, Romito L, Albanese A, Dallapiccola B, Bentivoglio A R (2004). PINK1 mutations are associated with sporadic early-onset parkinsonism. Ann Neurol, 56(3): 336–341
https://doi.org/10.1002/ana.20256 pmid: 15349860
143 van der Horst A, Tertoolen L G, de Vries-Smits L M, Frye R A, Medema R H, Burgering B M (2004). FOXO4 is acetylated upon peroxide stress and deacetylated by the longevity protein hSir2(SIRT1). J Biol Chem, 279(28): 28873–28879
https://doi.org/10.1074/jbc.M401138200 pmid: 15126506
144 Vaquero A, Scher M, Erdjument-Bromage H, Tempst P, Serrano L, Reinberg D (2007). SIRT1 regulates the histone methyl-transferase SUV39H1 during heterochromatin formation. Nature, 450(7168): 440–444
https://doi.org/10.1038/nature06268 pmid: 18004385
145 Vaquero A, Scher M, Lee D, Erdjument-Bromage H, Tempst P, Reinberg D (2004). Human SirT1 interacts with histone H1 and promotes formation of facultative heterochromatin. Mol Cell, 16(1): 93–105
https://doi.org/10.1016/j.molcel.2004.08.031 pmid: 15469825
146 Wang X, Winter D, Ashrafi G, Schlehe J, Wong Y L, Selkoe D, Rice S, Steen J, LaVoie M J, Schwarz T L (2011). PINK1 and Parkin target Miro for phosphorylation and degradation to arrest mitochondrial motility. Cell, 147(4): 893–906
https://doi.org/10.1016/j.cell.2011.10.018 pmid: 22078885
147 Wang X, Yan M H, Fujioka H, Liu J, Wilson-Delfosse A, Chen S G, Perry G, Casadesus G, Zhu X (2012). LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum Mol Genet, 21(9): 1931–1944
https://doi.org/10.1093/hmg/dds003 pmid: 22228096
148 Wareski P, Vaarmann A, Choubey V, Safiulina D, Liiv J, Kuum M, Kaasik A (2009). PGC-1alpha and PGC-1beta regulate mitochondrial density in neurons. J Biol Chem, 284(32): 21379–21385
https://doi.org/10.1074/jbc.M109.018911 pmid: 19542216
149 West A B, Moore D J, Biskup S, Bugayenko A, Smith W W, Ross C A, Dawson V L, Dawson T M (2005). Parkinson’s disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity. Proc Natl Acad Sci USA, 102(46): 16842–16847
https://doi.org/10.1073/pnas.0507360102 pmid: 16269541
150 Westerheide S D, Anckar J, Stevens S M Jr, Sistonen L, Morimoto R I (2009). Stress-inducible regulation of heat shock factor 1 by the deacetylase SIRT1. Science, 323(5917): 1063–1066
https://doi.org/10.1126/science.1165946 pmid: 19229036
151 Winslow A R, Chen C W, Corrochano S, Acevedo-Arozena A, Gordon D E, Peden A A, Lichtenberg M, Menzies F M, Ravikumar B, Imarisio S, Brown S, O’Kane C J, Rubinsztein D C (2010). α-Synuclein impairs macroautophagy: implications for Parkinson’s disease. J Cell Biol, 190(6): 1023–1037
https://doi.org/10.1083/jcb.201003122 pmid: 20855506
152 Wood-Kaczmar A, Gandhi S, Yao Z, Abramov A Y, Miljan E A, Keen G, Stanyer L, Hargreaves I, Klupsch K, Deas E, Downward J, Mansfield L, Jat P, Taylor J, Heales S, Duchen M R, Latchman D, Tabrizi S J, Wood N W (2008). PINK1 is necessary for long term survival and mitochondrial function in human dopaminergic neurons. PLoS ONE, 3(6): e2455
https://doi.org/10.1371/journal.pone.0002455 pmid: 18560593
153 Xilouri M, Vogiatzi T, Vekrellis K, Park D, Stefanis L (2009). Abberant alpha-synuclein confers toxicity to neurons in part through inhibition of chaperone-mediated autophagy. PLoS ONE, 4(5): e5515
https://doi.org/10.1371/journal.pone.0005515 pmid: 19436756
154 Yang S R, Wright J, Bauter M, Seweryniak K, Kode A, Rahman I (2007). Sirtuin regulates cigarette smoke-induced proinflammatory mediator release via RelA/p65 NF-kappaB in macrophages in vitro and in rat lungs in vivo: implications for chronic inflammation and aging. Am J Physiol Lung Cel l Mol Physiol, 292(2): L567–L576
https://doi.org/10.1152/ajplung.00308.2006 pmid: 17041012
155 Yeung F, Hoberg J E, Ramsey C S, Keller M D, Jones D R, Frye R A, Mayo M W (2004). Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J, 23(12): 2369–2380
https://doi.org/10.1038/sj.emboj.7600244 pmid: 15152190
156 Yong-Kee C J, Salomonczyk D, Nash J E (2011). Development and validation of a screening assay for the evaluation of putative neuroprotective agents in the treatment of Parkinson’s disease. Neurotox Res, 19(4): 519–526
https://doi.org/10.1007/s12640-010-9174-2 pmid: 20361292
157 Yong-Kee C J, Sidorova E, Hanif A, Perera G, Nash J E (2012). Mitochondrial dysfunction precedes other sub-cellular abnormalities in an in vitro model linked with cell death in Parkinson’s disease. Neurotox Res, 21(2): 185–194
https://doi.org/10.1007/s12640-011-9259-6 pmid: 21773851
158 Zarranz J J, Alegre J, Gómez-Esteban J C, Lezcano E, Ros R, Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atarés B, Llorens V, Gomez Tortosa E, del Ser T, Mu?oz D G, de Yebenes J G (2004). The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol, 55(2): 164–173
https://doi.org/10.1002/ana.10795 pmid: 14755719
159 Zhang L, Shimoji M, Thomas B, Moore D J, Yu S W, Marupudi N I, Torp R, Torgner I A, Ottersen O P, Dawson T M, Dawson V L (2005). Mitochondrial localization of the Parkinson’s disease related protein DJ-1: implications for pathogenesis. Hum Mol Genet, 14(14): 2063–2073
https://doi.org/10.1093/hmg/ddi211 pmid: 15944198
160 Zhang N Y, Tang Z, Liu C W (2008). alpha-Synuclein protofibrils inhibit 26 S proteasome-mediated protein degradation: understanding the cytotoxicity of protein protofibrils in neurodegenerative disease pathogenesis. J Biol Chem, 283(29): 20288–20298
https://doi.org/10.1074/jbc.M710560200 pmid: 18502751
161 Zheng B, Liao Z, Locascio J J, Lesniak K A, Roderick S S, Watt M L, Eklund A C, Zhang-James Y, Kim P D, Hauser M A, Grünblatt E, Moran L B, Mandel S A, Riederer P, Miller R M, Federoff H J, Wüllner U, Papapetropoulos S, Youdim M B, Cantuti-Castelvetri I, Young A B, Vance J M, Davis R L, Hedreen J C, Adler C H, Beach T G, Graeber M B, Middleton F A, Rochet J C, Scherzer C R, Global P D G E C, and the Global PD Gene Expression (GPEX) Consortium (2010). PGC-1α, a potential therapeutic target for early intervention in Parkinson’s disease. Sci Transl Med, 2(52): 52ra73
https://doi.org/10.1126/scitranslmed.3001059 pmid: 20926834
[1] Karimeh Haghani, Pouyan Asadi, Gholamreza Taheripak, Ali Noori-Zadeh, Shahram Darabi, Salar Bakhtiyari. Association of mitochondrial dysfunction and lipid metabolism with type 2 diabetes mellitus: A review of literature[J]. Front. Biol., 2018, 13(6): 406-417.
[2] Hanane Gourine, Hadria Grar, Wafaa Dib, Nabila Mehedi, Ahmed Boualga, Djamel Saidi, Omar Kheroua. Effect of a normal protein diet on oxidative stress and organ damage in malnourished rats[J]. Front. Biol., 2018, 13(5): 366-375.
[3] Volodymyr Padalko, Viktoriya Dzyuba, Olena Kozlova, Hanna Sheremet, Olena Protsenko. Zingiber officinale extends Drosophila melanogaster life span in xenobiotic-induced oxidative stress conditions[J]. Front. Biol., 2018, 13(2): 130-136.
[4] Vadim V. Davydov, Alexander V. Shestopalov, Evgenya R. Grabovetskaya. Physiological significance of oxidative stress and its role in adaptation of the human body to deleterious factors[J]. Front. Biol., 2018, 13(1): 19-27.
[5] Bharti Chaudhary, Sonam Agarwal, Renu Bist. Invulnerability of bromelain against oxidative degeneration and cholinergic deficits imposed by dichlorvos in mice brains[J]. Front. Biol., 2018, 13(1): 56-62.
[6] Mohammad Jodeiri Farshbaf. Succinate dehydrogenase in Parkinson’s disease[J]. Front. Biol., 2017, 12(3): 175-182.
[7] Xin-Min Qin,Xiao-Wen Yang,Li-Xia Hou,Hui-Min Li. Complete mitochondrial genome of Ampittia dioscorides (Lepidoptera: Hesperiidae) and its phylogenetic analysis[J]. Front. Biol., 2017, 12(1): 71-81.
[8] Jian Zou,Jinbo Yu,Yuqing Zhu,Jiali Zhu,Jing Du,Xu Yang. Application of glutathione to antagonize H2O2-induced oxidative stress in rat tracheal epithelial cells[J]. Front. Biol., 2016, 11(1): 59-63.
[9] Vadim V. Davydov,Evgenya R. Grabovetskaya,Amjad Hamdallah. Age-dependent peculiarities modulation of activity of aldehyde scavenger enzymes in mitochondria of rat thigh muscle during stress[J]. Front. Biol., 2016, 11(1): 28-31.
[10] Zhuo Wu,Jingquan Li,Ping Ma,Baizhan Li,Yang Xu. Long-term dermal exposure to diisononyl phthalate exacerbates atopic dermatitis through oxidative stress in an FITC-induced mouse model[J]. Front. Biol., 2015, 10(6): 537-545.
[11] Xin-Min Qin,Qing-Xin Guan,Hui-Min Li,Yu Zhang,Yu-Ji Liu,Dan-Ni Guo. The complete mitogenome of Lamproptera curia (Lepidoptera: Papilionidae) and phylogenetic analyses of Lepidoptera[J]. Front. Biol., 2015, 10(5): 458-472.
[12] Yu Lu,Biao Yan,Xudong Liu,Yuchao Zhang,Shibi Zeng,Hao Hu,Rong Xiang,Yu Xu,Ying Yu,Xu Yang. Comparative study of oxidative stress induced by sand flower and schistose nanosized layered double hydroxides in N2a cells[J]. Front. Biol., 2015, 10(3): 279-286.
[13] Ruth Beckervordersandforth,Benjamin M. Häberle,D. Chichung Lie. Metabolic regulation of adult stem cell-derived neurons[J]. Front. Biol., 2015, 10(2): 107-116.
[14] Massimo Bonora,Paolo Pinton,Keisuke Ito. Mitochondrial control of hematopoietic stem cell balance and hematopoiesis[J]. Front. Biol., 2015, 10(2): 117-124.
[15] Young Bong CHOI,Edward William HARHAJ. Functional implications of mitochondrial reactive oxygen species generated by oncogenic viruses[J]. Front. Biol., 2014, 9(6): 423-436.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed