-synuclein in parkinson’s disease - cold spring...

24
a-Synuclein in Parkinson’s Disease Leonidas Stefanis Laboratory of Neurodegenerative Diseases, Biomedical Research Foundation of the Academy of Athens, and Second Department of Neurology, University of Athens Medical School, Athens 11527, Greece Correspondence: [email protected] a-Synuclein is a presynaptic neuronal protein that is linked genetically and neuropatholog- ically to Parkinson’s disease (PD). a-Synuclein may contribute to PD pathogenesis in a number of ways, but it is generally thought that its aberrant soluble oligomeric confor- mations, termed protofibrils, are the toxic species that mediate disruption of cellular homeo- stasis and neuronal death, through effects on various intracellular targets, including synaptic function. Furthermore, secreted a-synuclein may exert deleterious effects on neighboring cells, including seeding of aggregation, thus possibly contributing to disease propagation. Although the extent to which a-synuclein is involved in all cases of PD is not clear, targeting the toxic functions conferred by this protein when it is dysregulated may lead to novel thera- peutic strategies not only in PD, but also in other neurodegenerative conditions, termed syn- ucleinopathies. T he first link of Parkinson’s disease (PD) to a- synuclein was also the first conclusive demon- stration of a genetic defect leading to PD, and thus has historical and conceptual value. Although familial contribution to the disease was discussed by some, it was felt to be minor at best by most. The tangible discovery of a gene defect linked to PD in particular families was a ground-breaking discovery, as it opened the door to aflood of studies investigating the genetic base of the disorder, cul- minating in more recent genome-wide association studies (GWAS), that, remarkably, have made a full circle back to the origins of the molecular genetic era of PD; it turns out that one of the major genes identified as linked to sporadic PD is none other than SNCA, the gene encoding for a-synuclein. A parallel story that has developed over the years is the abundant abnormal a-synuclein pathology that characterizes neuropathological specimens derived not only from PD patients, but also patients with other neurodegenerative conditions, collectively termed “synucleinopa- thies.” The ability to model abnormal a-synu- clein accumulation in various cellular and animal models has led to the study of the conse- quences of such accumulation, to the decipher- ing of the molecular pathways involved, and to the identification of potential therapeutic tar- gets. Such targets may prove of general utility, and provide the basis for future therapeutics in most forms of PD, and, potentially, other syn- ucleinopathies. Editor: Serge Przedborski Additional Perspectives on Parkinson’s Disease available at www.perspectivesinmedicine.org Copyright # 2012 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a009399 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 1 www.perspectivesinmedicine.org Press on February 24, 2019 - Published by Cold Spring Harbor Laboratory http://perspectivesinmedicine.cshlp.org/ Downloaded from

Upload: lybao

Post on 24-Feb-2019

223 views

Category:

Documents


0 download

TRANSCRIPT

a-Synuclein in Parkinson’s Disease

Leonidas Stefanis

Laboratory of Neurodegenerative Diseases, Biomedical Research Foundation of the Academyof Athens, and Second Department of Neurology, University of Athens Medical School,Athens 11527, Greece

Correspondence: [email protected]

a-Synuclein is a presynaptic neuronal protein that is linked genetically and neuropatholog-ically to Parkinson’s disease (PD). a-Synuclein may contribute to PD pathogenesis in anumber of ways, but it is generally thought that its aberrant soluble oligomeric confor-mations, termed protofibrils, are the toxic species that mediate disruption of cellular homeo-stasis and neuronal death, through effects on various intracellular targets, including synapticfunction. Furthermore, secreted a-synuclein may exert deleterious effects on neighboringcells, including seeding of aggregation, thus possibly contributing to disease propagation.Although the extent to which a-synuclein is involved in all cases of PD is not clear, targetingthe toxic functions conferred by this protein when it is dysregulated may lead to novel thera-peutic strategies not only in PD, but also in other neurodegenerative conditions, termed syn-ucleinopathies.

The first link of Parkinson’s disease (PD) to a-synuclein was also the first conclusive demon-

stration of a genetic defect leading to PD, and thushas historical and conceptual value. Althoughfamilial contribution to the disease was discussedby some, it was felt to be minor at best by most.The tangible discovery of a gene defect linked toPD in particular families was a ground-breakingdiscovery,as itopenedthedoortoafloodofstudiesinvestigating the genetic base of the disorder, cul-minating in more recent genome-wide associationstudies (GWAS), that, remarkably, have made afullcircle back to the origins of the molecular geneticera of PD; it turns out that one of the major genesidentified as linked to sporadic PD is none otherthan SNCA, the gene encoding fora-synuclein.

A parallel story that has developed over theyears is the abundant abnormal a-synucleinpathology that characterizes neuropathologicalspecimens derived not only from PD patients,but also patients with other neurodegenerativeconditions, collectively termed “synucleinopa-thies.” The ability to model abnormal a-synu-clein accumulation in various cellular andanimal models has led to the study of the conse-quences of such accumulation, to the decipher-ing of the molecular pathways involved, and tothe identification of potential therapeutic tar-gets. Such targets may prove of general utility,and provide the basis for future therapeuticsin most forms of PD, and, potentially, other syn-ucleinopathies.

Editor: Serge Przedborski

Additional Perspectives on Parkinson’s Disease available at www.perspectivesinmedicine.org

Copyright # 2012 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a009399

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

1

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

GENETICS OF PD AND OTHERDISORDERS WITH LINKS TOSYNUCLEINOPATHIES

In 1997, Polymeropoulos et al. reported thefirst specific genetic aberration linked to PDin a large Italian kindred, and in some Greekfamilial cases. The mutation corresponded to aG209A substitution in the SNCA gene encodingfor a-synuclein, resulting in an A53T aminoacid change. The pattern of inheritance was au-tosomal-dominant, and the disease was early-onset, usually in the 40s. Although there wassome initial skepticism, based on the fact thatthe threonine was the amino acid in position53 in the rodent SNCA homolog, this was setaside following the identification of two morepoint mutations in SNCA, leading to A30Pand E46K amino acid changes, which were as-sociated with autosomal-dominant PD (Krugeret al. 1998; Zarranz et al. 2004). The next land-mark study that linked SNCA to PD through adifferent mechanism identified triplications ofthe SNCA locus in separate families with an au-tosomal-dominant inheritance pattern (Single-ton et al. 2003). This led to a potential totalof a duplication of the a-synuclein load insuch patients, and this was confirmed laterat the protein level (Miller et al. 2004). Otherfamilies were subsequently described that har-bored duplications of the gene, which wassimilarly associated with autosomal-dominantdisease (Chartier-Harlin et al. 2004). Interest-ingly, a gene dosage effect exists, in that triplica-tion cases are earlieronset and have a more severecourse compared to the duplication cases (Fuchset al. 2007).

When a specific genetic defect is identified inrare familial cases, it is almost a reflex reaction tolook at the same gene in the sporadic disease,with the hope of identifying polymorphismsthat may be linked to the disease in associationstudies. This has not been particularly successfulin other diseases, but in the case of PD and theSNCA gene this approach has borne fruit. Initialstudies focused on the Rep1 polymorphic regionlocated approximately 10 kB upstream of theSNCA transcriptional initiation site. Particularpolymorphisms in this region conferred a rela-

tive risk for sporadic PD in initial associationstudies, which were replicated and then con-firmed in a large metanalysis (Maraganoreet al. 2006). Others studies found associationsof PD with 30 regions of the gene (Mueller et al.2005; Mizuta et al. 2006). Targeted associationstudies are, however, prone to false positives;therefore, it is very important that the link ofSNCA to sporadic PD has been confirmedthrough large unbiased GWAS. SNCA, and, inparticular, the 30 region, has been a consistenthit in such studies, and represents, of the linksidentified, probably the most robust one (Nallset al. 2011).

The conclusion from these genetic studiesis that SNCA is linked to both rare familialand sporadic PD from a genetic standpoint. Itis notable that the association of SNCAvariantsto disease has now also been extended to multi-ple-system atrophy (MSA), another prototypi-cal synucleinopathy, which is invariably spo-radic (Al-Chalabi et al. 2009; Scholz et al. 2009).

THE INVOLVEMENT OF a-SYNUCLEIN INTHE NEUROPATHOLOGICAL PICTURE OFPD AND RELATED DISORDERS

The genetic link of SNCA to PD in 1997 ledinvestigators to rapidly develop appropriate an-tibodies against a-synuclein and use them inhistopathological sections of PD patients. a-Synuclein turned out to be robustly expressedwithin Lewy bodies (LBs), and, in particular,in the “halo” of the inclusions (Spillantini et al.1997, 1998; Baba et al. 1998). In fact,a-synucleinimmunohistochemistry is currently the goldstandard in the neuropathological evaluationof PD. With repeated studies, three major in-sights emerged: (1) a-synuclein pathology inPD is not confined to the cell soma, but is alsoprominent in neuritic processes, (2) it is wide-spread in various brain regions in PD, and (3)it is present in a number of other synucleinopa-thies, such as MSA, dementia with Lewy bodies(DLBs), many cases of Alzheimer’s disease (AD)(the so-called LB variant of AD), neurodegen-eration with brain iron accumulation (NBIA)type I, pure autonomic failure (PAF), and evena subtype of essential tremor (reviewed in Kahle

L. Stefanis

2 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

2008). The distribution of the pathology at thecellular and regional level is different in eachdisease.

Braak et al. (2003), in their seminal study ofthe staging of PD pathology, used a-synucleinimmunohistochemistry in a large number ofautopsy cases. Based essentially on a-synucleinneuritic pathology (“Lewy neurites”), they sug-gested a six-stage scheme in which the pathol-ogy begins first at the olfactory bulb and thedorsal vagal nucleus and gradually follows anascending course, culminating in widespreada-synuclein pathology at the later stages, in-volving associative cortical regions. Notably,substantia nigra is only affected in stage 3 ofthis scheme. Although there have been crit-icisms against this hypothesis (Burke et al.2008), and it does not explain the absence ofclinical symptoms in subjects who on autopsyhave widespread a-synuclein pathology, or theclinical picture of DLB, it appears to hold upfor the majority, but not all, of cases examinedin large cohorts of LB cases (Dickson et al.2010). What is important to note here is thatoverall in the PD brain the majority of ab-normally deposited a-synuclein is in neuriticprocesses. The presence of a-synuclein in LBsis likely the tip of the iceberg. In fact, Kramerand Shulz-Schaeffer (2007), using a modifiedprotocol termed protein aggregate filtration,showed in DLB cases a remarkable degree ofaberrant neuritic a-synuclein pathology, in thevirtual absence of LBs.

Overall, it is clear that abnormal a-synu-clein deposition occurs early in PD. Giventhe fact that such pathology may develop sec-ondarily, for example, owing to iron mishan-dling in NBIA type I, we cannot be certainthat it is a primary factor in the disease process.However, the generally quite defined pattern ofa-synuclein pathology in PD, the cellular andanimal models of a-synuclein overexpressionthat will be discussed, and, most importantly,the combination with the genetic data men-tioned above, suggest that this aberrant deposi-tion is the driving force in PD pathogenesis.Such deposition, as mentioned, may be subtleand difficult to detect in the absence of elabo-rate immunohistochemical techniques that, it

is hoped, will become more widely applicableand available.

THE STRUCTURE AND FUNCTION OFa-SYNUCLEIN

The SNCA gene encodes for a 140 amino acidprotein, which in aqueous solutions does nothave a defined structure, hence the term “na-tively unfolded protein.” a-Synuclein, however,does form a-helical structures on binding tonegatively charged lipids, such as phospholipidspresent on cellular membranes, and b-sheet-rich structures on prolonged periods of incu-bation. The protein is composed of three dis-tinct regions: (1) an amino terminus (residues1–60), containing apolipoprotein lipid-bind-ing motifs, which are predicted to form amphi-philic helices conferring the propensity to forma-helical structures on membrane binding, (2)a central hydrophobic region (61–95), so-calledNAC (non-Ab component), which confers theb-sheet potential, and (3) a carboxyl terminusthat is highly negatively charged, and is proneto be unstructured (Fig. 1). There are at leasttwo shorter alternatively spliced variants of theSNCA gene transcript, but their physiologicaland pathological roles have not been well char-acterized (Maroteaux and Scheller 1991; Uedaet al. 1993; Maroteaux et al. 1988).

a-Synuclein is a member of the synucleinfamily of proteins, which also include b- andg-synuclein. What largely sets aparta-synucleinfrom the other members structurally is the NACregion. All three members of the family arepredominantly neuronal proteins that underphysiological conditions localize preferentiallyto presynaptic terminals (George 2002). Thereare some reports of extranigral b- and g-synu-clein neuritic pathology in synucleinopathies,but, if it exists, it does not appear to be wide-spread (Galvin et al. 1999). Interestingly, pointmutations in b-synuclein have been describedin rare cases with DLB (Ohtake et al. 2004),and expression of one of these in a transgenicmouse model-induced neurodegeneration (Fu-jita et al. 2010). On the other hand, wild-type(WT) b-synuclein has been found to be pro-tective in various settings against a-synuclein-

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 3

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

mediated neurodegeneration (Hashimoto et al.2001; Park and Lansbury, 2003). It would ap-pear therefore that WT b-synuclein may haveneuroprotective properties, but that, under cer-tain situations, it may assume a neurotoxic po-tential.

a-Synuclein is abundantly expressed in thenervous system, comprising 1% of total cy-tosolic protein. On immunohistochemistry innormal brains a-synuclein antibodies confera punctuate neuropil staining pattern, consis-tent with presynaptic terminal staining, whereasneuronal soma staining is less apparent (Kahle2008). In presynaptic terminals a-synuclein ispresent in close proximity, but not within, syn-aptic vesicles. In fact a-synuclein is also veryabundant, for unclear reasons, in erythrocytesand platelets (Barbour et al. 2008). Whether itis expressed at all under physiological condi-tions in glia is somewhat controversial; it wouldappear that, if it exists, such expression is pre-sent at very low levels (Mori et al. 2002a). Theexpression of a-synuclein is induced duringneuronal development, following determina-tion of neuronal phenotype and establishmentof synaptic connections, and lags behind theinduction of proteins involved in synapticstructure (Withers et al. 1997; Kholodilov et al.1999; Murphy et al. 2000; Rideout et al. 2003).Moreover, a-synuclein expression levels are

modulated in conditions that alter plasticity orconfer injury (George et al. 1995; Kholodilovet al. 1999; Vila et al. 2000). Such data have ledto the notion that a-synuclein may be a modu-lator of synaptic transmission. Examinations ofneurophysiological properties of mice null fora-synuclein and of cellular systems in whichWT a-synuclein is overexpressed have been in-strumental in this regard, as they reveal alter-ations in neurotransmitter release, which arenot always consistent across neuronal systems(Abeliovich et al. 2000; Murphy et al. 2000;Cabin et al., 2002; Larsen et al. 2006). Overallthough, it would appear that a-synuclein actsas a subtle break for neurotransmitter releaseunder circumstances of repeated firing. An ele-gant study by Fortin et al. (2005) showed thatfluorescently labeled a-synuclein moves awayfrom the vesicles on neuronal firing, and thengradually returns. These effects may involvetransient binding of a-synuclein to the vesicles,whereas a-synuclein may also be involved in ve-sicular biogenesis, through effects on phospha-tidic acid metabolism, and in compartmenta-lization between resting and readily releasablepools (Murphy et al. 2000). Consistent with arole fora-synuclein at the level of synaptic trans-mission, a-synuclein modulates profoundly thephenotype of mice null for the presynaptic pro-tein CSP-a (cysteine string protein-a), which

9561

Repeat 6Repeat 5

Apolipoprotein motif

Repeat 2 Repeat 4Repeat 3 Acidic tail140

Repeat 1

1N C

A53T

NAC region necessary for aggregation

E46KA30P

Mutations geneticallylinked to familial

autosomal-dominantParkinson’s disease

Figure 1. Schemtic structure of a-synuclein.

L. Stefanis

4 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

show synaptic degeneration (Chandra et al.2005). It is believed that a-synuclein, througha chaperone-like function, may act in synergywith CSP-a in the assembly of the SNARE com-plex (Chandra et al. 2005), and this effect mayinvolve, in particular, binding to synaptotag-min-2 (Burree et al. 2010).

Therefore, the main function ofa-synucleinwould appear to be the control of neurotrans-mitter release, through effects on the SNAREcomplex. Recent evidence, to be discussed be-low, suggests that this physiological functionmay provide insight into the aberrant functionunleashed in disease states.

SYNUCLEINOPATHY MODELS

Based on the genetic and pathological data, at-tempts have been made to create cellular andanimal models in which various forms of a-synuclein are overexpressed. We have electedto not present such models with any great detailhere, but rather highlight the inferences gainedfrom such models regarding the potentialpathological (and sometimes physiological) ef-fects of a-synuclein. Suffice it to say here, thatthere are a number of transgenic mice available,in which expression of various forms of a-syn-uclein is achieved through different promoters,allowing in some cases regional and temporalcontrol of expression. In such mice, there is gen-erally some degree of neurodegneration and LB-like pathology, but it does not involve primarilythe dopaminergic system. Some mice developbehavioral phenotypes that could be akin topresymptomatic PD (reviewed by Chesselet2008). An alternative approach has been theviral-mediated transduction of dopaminergicneurons of the substantia nigra of rodentswith a-synuclein, and this has been more suc-cessful in modeling the dopaminergic cell deathaspect of the disease (Kirik et al. 2002). A num-ber of invertebrate models, including fly (Feanyand Bender 2000) and Caenorhabditis elegans(Lasko et al. 2003), have also been created, andhave been very useful in testing theories regard-ing the toxic pathways elicited by aberrant a-synuclein.

THE AGGREGATION POTENTIAL OFa-SYNUCLEIN

The ability of a-synuclein to generate b-sheetstructures under certain circumstances gener-ated a lot of enthusiasm, as it provided parallelsto the b-sheet structure of b-amyloid, and con-sequently, a unified pathogenetic basis for thetwo most common neurodegenerative diseases.Indeed, WTas well as disease-related mutants ofa-synuclein form amyloid-like fibrils on pro-longed incubation in solution (Conway et al.2000). It is widely agreed on that such fibrilsform the basis of the mature LBs and Lewy neu-rites (LNs) present in synucleinopathies. In theprocess of fibril formation various intermediateforms ofa-synuclein develop. These are initiallysoluble oligomeric forms ofa-synuclein that as-sume spherical, ring-, and stringlike characteris-tics when seen under the electron microscope.Such structures, collectively termed protofibrils,gradually become insoluble and coalesce into fi-brils. It is difficult to capture these intermediatestructures in cells or in vivo, but their biochemi-cal equivalent isthought to besolubleoligomericforms, some, but not all, of which are SDS-stableand can be detected on SDS/polyacrylamidegel electrophoresis (PAGE) gels. Other forms,however, can only be captured on native gelsor by size-exclusion chromatography (Emma-nouilidou et al. 2010a). This cascade of events,starting from the natively unfolded proteinand culminating in the mature fibril formationis collectively termed a-synuclein aggregation.

The general consensus in the field is thataggregation is a main pathogenic feature of a-synuclein. This is buttressed by the lack of tox-icity conferred by WT b-synuclein or by a-syn-uclein variants that do not contain the NAC do-main and do not form aggregates (Periquet et al.2007), by the beneficial effects of overxepressingHSPs, which assist in refolding of aggregation-prone proteins, in synucleinopathy models(Auluck and Bonini 2002; Auluck et al. 2002;Klucken et al. 2004), and by the protection con-ferred in cellular models of a-synuclein overex-pression by reagents that inhibit or neutralizeaggregated species (Vekrellis et al. 2009; Bieschkeet al. 2010). However, the protective effects of

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 5

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

HSP overexpression are not always apparent(Shimshek et al. 2010). There is less agreementas to which particular species are neurotoxic.A predominant idea is that some of the earlysoluble oligomeric species have such a potential.This is based on the following pieces of data:

(1) A53T and A30P a-synuclein form moreprotofibrils than the WT protein, whereasonly A53T forms more readily mature fi-brils; however, E46K a-synuclein formsless protofibrils than the WT, thereforethere is no simple correlation that can ex-plain everything using this scheme, whichis based on in vitro cell-free systems (Con-way et al. 2000; Fredenburg et al. 2007).

(2) Dopamine and its metabolites act as inhib-itors of the conversion of protofibrils tomature fibrils in vitro and in vivo, presum-ably through the formation of dopamine/a-synuclein adducts (Conway et al. 2001;Mazzulli et al. 2006; Tsika et al. 2010), sug-gesting that the preferential vulnerability ofdopaminergic neurons to the pathogenicprocess of PD may be attributable to theenhanced presence of soluble oligomers(although, surprisingly, mouse nigral neu-rons, which contain more oligomers, ap-pear relatively resistant to transgenic a-synuclein-mediated neurodegeneration).

(3) In cellular systems and in vivo, the presenceof soluble oligomers, and not frank inclu-sions, generally correlates best with neu-rotoxicity (Gosavi et al. 2002; Lo Biancoet al. 2002). Two major studies providedsupport to this idea, as they showed en-hanced neurotoxicity both in cell cultureand in vivo with artificial mutants of a-synuclein that favored the formation of sol-uble oligomers over mature fibrils (Karpinaret al. 2009; Winner et al. 2011). It shouldbe noted, however, that the issue is notsettled; the protofibrils are heterogeneous,“off pathway” oligomers that do not leadto toxicity may exist, and frank inclusionshave the potential of being neurotoxic them-selvesover time,especially ifpresent inaxons,where they can block neuronal trafficking

or “suck in” essential neuronal components.On the other hand, if the protofibril theoryis correct, enhancing inclusions could beviewed as a method to remove the toxic inter-mediate oligomeric species, and this has beenattempted with some success in cellularmodels (Bodner et al. 2006).

a-SYNUCLEIN POSTTRANSLATIONALMODIFICATIONS AND INTERACTIONS

a-Synuclein may be modified by phosphoryla-tion, oxidation, nitrosylation, glycation, or gly-cosylation. Of all such modifications, the beststudied is phosphorylation. Antibodies specificto S129-phosphorylated a-synuclein promi-nently decorate LBs in synucleinopathies (Fuji-wara et al. 2002). Although the initial idea,which appeared to be buttressed by experimentsin transgenic flies, was that such phosphoryla-tion enhanced a-synuclein-soluble oligomers(but actually decreased a-synuclein inclusions)and neurotoxicity (Chen et al. 2005, 2009);subsequent studies have cast doubt on this no-tion, providing contradictory results (Gorba-tyuk et al. 2008; Paleologou et al. 2008; Azeredoda Silveira et al. 2009). In fact, it may be thatS129 phosphorylation, which occurs minimallyunder physiological circumstances, occurs afterthe fact in already formed LBs (Paleologou et al.2008). Enhancing a-synuclein phosphatase ac-tivity was, however, protective against a-synu-clein-mediated neurotoxicity, suggesting a causaldetrimental role of a-synuclein phosphorylationin the disease process (Lee et al. 2011). Polo-likekinases appear to be the physiological kinasesfor a-synuclein S129 phosphorylation (Ingliset al. 2009; Mbefo et al. 2010). S87 is anotherphosphorylation site for a-synuclein, and S87-Pa-synuclein is also apparent within LBs, but itspathological significance is unclear (Paleologouet al. 2010). Phosphorylation on a-synuclein ty-rosine residues has also been reported and mayact in opposing fashion to S129 phosphorylation,at least in Drosophila (Chen et al. 2009).

A number of studies in cell-free systemsand in cell culture have shown that a-synucleincan be oxidized, and this seems to drive its oli-gomerization (Lee et al. 2002; Betarbet et al.

L. Stefanis

6 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

2006; Mirzaei et al. 2006; Qin et al. 2007). Ironand dopamine appear to exert a similar effect,although whether this is a direct interaction ormediated by oxidation is less clear (Ostrerova-Golts et al. 2000; Conway et al. 2001; Kostkaet al. 2008; Leong et al. 2009). The potent oxi-dant 4-hydroxy-2-nonenal (HNE) promotedstable soluble oligomers, inhibiting their con-version to fibrils; such stable soluble oligomerswere highly toxic, favoring again the solubleoligomer toxicity theory (Qin et al. 2007). In asimilar vein, nitration of a-synuclein has beenreported; in fact, an antibody specifically gener-ated against nitrosylated a-synucelin decoratesLB-like pathology (Giasson et al. 2000). Mono-meric and dimeric nitrated a-synuclein pro-moted fibril formation (Hodara et al. 2004),whereas oligomeric nitrated a-synuclein actu-ally inhibited it, stabilizing as soluble oligomers(Uversky et al. 2005), suggesting complex effectsof nitrosylation on a-synuclein aggregation. Aswith S129 phosphorylation, it is possible thatthe intense nitrosylation of a-synuclein withinLB-like structures occurs after the fact.

Given the fact that oxidative/nitrative stressinduced by environmental factors, includingmitochondrial toxins, has been heavily impli-cated in PD, oxidation- or nitration-induceda-synuclein oligomerization has attracted at-tention, as a point of potential convergence be-tween environmental and genetic factors lead-ing to the disease. It should be noted, however,that a-synuclein aggregation could be dissoci-ated from mitochondrial toxicity-induced death(Liu et al. 2008), arguing against simplistic lin-ear pathways to neurodegeneration, and raisingthe issue whether in certain instances the ab-undant a-synuclein-related pathology may bean epiphenomenon, and not causally related toneuronal dysfunction and death. Similar infer-ences can be taken from a study in proteasomalinhibitor-treated neurons, where the b-sheet-like pathology of a-synuclein was dissociatedfrom death (Rideout et al. 2004).

Another modification that may be impor-tant is carboxy-terminal truncation of a-synu-clein, leading to fragments lacking part or thewhole of the acidic tail (Li et al. 2005). Thesefragments appear to be more prone to fibrilliza-

tion (Kim et al. 2002; Murrayet al. 2003). Calpain,one of the enzymes thought to perform suchprocessing, is especially interesting given its cal-cium dependence and localization at presynapticterminals (Mishizen-Eberz et al. 2003, 2005).

a-Synuclein is quite promiscuous in itsbinding properties, and may bind various pro-teins in neuronal cells, including componentsof dopamine metabolism, such as tyrosine hy-droxylase (TH) and the dopamine transporter(DAT), modulating their function (Ostrerovaet al. 1999; Perez et al. 2002; Wersinger andSidhu 2003; Wersinger et al. 2003; Peng et al.2005). There are quite a few reports suggestingthat through such binding to various proteins,a-synuclein may exert pro- or antiapoptoticeffects (Ostrerova et al. 1999; da Costa et al.2000; Jin et al. 2011), although no effect on pro-grammed neuronal cell death has been shown inmice null for a-synuclein (Stefanis et al. 2004).Interestingly, however, these mice are resistantto the well-known mitochondrial neurotoxinMPTP, through mechanisms that are still notdeciphered, but may involve steps both up-stream of and downstream from mitochon-drial dysfunction (Dauer et al. 2002). Such miceare also resistant against neuroinflammation-induced nigral degeneration conferred by lipo-polysaccharide (LPS) (Gao et al. 2008), suggest-ing that endogenous a-synuclein is somehowinvolved in such death pathways.

A number of interactions of a-synuclein withother proteins have been implicated in its aggrega-tion potential; most notable amongst these is syn-philin-1, a protein identified through a yeast-two-hybrid screen as an AS-interacting protein,which promotes a-synuclein aggregation andinclusion formation (Engelenderet al. 1999). In-terestingly, transgenic expression of synphilin-1on the background of A53T a-synuclein trans-genic mice attenuates neurodegeneration, as as-sessed by indices of cell death and gliosis, whileenhancing inclusion formation, suggesting thatpromoting a-synuclein inclusion formation isneuroprotective (Smith et al. 2010).

Interactions of a-synuclein with lipids maybe important not only for their physiologicfunctions, but also for their aberrant effects. Re-garding the former, it is interesting to note that

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 7

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

sequestration of the fatty acid arachidonic acidaway from the SNARE complex by a-synucleinhas been suggested to underlie its inhibitoryeffect on neuronal transmission (Darios et al.2010), and that alterations in levels of key en-zymes in lipid signaling, such as PLD2 andPLA2, may underlie the phenotype in SNCAnull microglia, which are overreactive but lackphagocytic potential (Austin et al. 2006, 2011).Regarding the latter, polyunsaturated fatty acids(PUFAs) have been shown to enhance oligome-rization and neurotoxicity of a-synuclein (Per-rin et al. 2001; Sharon et al. 2003; Assayag et al.2007).

GAIN-OF-FUNCTION ANDACCUMULATION OF a-SYNUCLEIN

The notion that enhanceda-synuclein levels arecausative in PD pathogenesis is derived fromthe familial SNCA multiplication cases show-ing a dose-dependent correlation ofa-synucleinload to the PD phenotype, the autosomal-dom-inant pattern of inheritance for the point muta-tions, the accumulation of a-synuclein in synu-cleinopathy brains, and the demonstration thatfeatures of the disease can be mimicked by WTa-synuclein overexpression in various models.Consistent with this idea, a-synuclein proteinlevels are increased with aging in the substantianigra, and correlate with decreased TH immu-nostaining (Chu and Kordower 2007). How-ever, it should be noted that there is insufficientevidence that there is a generalized excess ofa-synuclein in PD brains. In fact, mRNA stud-ies have been quite contradictory in this regard,with some even showing a decrease of SNCA ex-pression in PD nigra (Dachsel et al. 2007). Pro-tein levels are not obviously increased overall inPD brains, although clearly there is induction ofinsoluble components, including monomericand oligomeric species. A comprehensive studyof multiple brain areas showed that membrane-associated monomeric a-synuclein levels wereonly modestly increased in the substantia nigra,and not in other brain regions, of PD patientscompared to controls; even so, the levels insome PD cases were within the spectrum of con-trol values. In contrast, in MSA, the membrane-

associated a-synuclein increase was robust invulnerable brain regions (Tong et al. 2010). Itmay of course be that neurons with the highestexpression levels ofa-synuclein are most vulner-able and succumb early in the disease process,giving their place to glia, thus confounding theresults. To partly circumvent this issue, Grunde-mann et al. (2008) performed laser-capture mi-crodissection studies, and reported a consider-able increase of SNCA expression in survivingnigral neurons derived from PD brains com-pared with controls. However, this increase didnot appear to be specific for SNCA (Grunde-mann et al. 2008). Therefore, the issue is stillopen.

An indirect way of approaching this is byassessing whether polymorphic SNCA variantsthat confer disease risk in association studiesalter a-synuclein levels in model systems. Ininitial studies, the Rep1 polymorphism associ-ated with the disease led to increased SNCAtranscriptional activity in neuronal cell lines(Chiba-Falek and Nussbaum 2001); this findinghas been confirmed in mice (Cronin et al.2009), and even in the human brain (Linnertzet al. 2009), although in another study an asso-ciation between Rep1 polymorphisms and a-synuclein mRNA and protein levels in the brainwas not apparent (Fuchs et al. 2008). Polymor-phisms within the 30 region may also correlatewith AS levels (Fuchs et al. 2008; Mata et al.2010). Intriguingly, it was reported that risk-conferring alleles at the 30 untranslated region(UTR) were associated with increased expres-sion of an alternatively spliced form of SNCA(McCarthy et al. 2011), suggesting another me-chanism through which gene variations may belinked to the disease.

It is generally assumed that a-synuclein leadsto disease through a toxic gain-of-function thatis likely inherent in the normal protein when it ex-ceeds a certain level. In apparent agreement withthis, a-synuclein null mice, in contrast to trans-genic overexpressors, display no overt neuropa-thological or behavioral phenotype (Abeliovichet al. 2000; Cabin et al. 2002). However, whenstudied more closely, such mice do have somedeficits in the dopaminergic system, in par-ticular, a slightly reduced number of postnatal

L. Stefanis

8 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

dopaminergic neurons (Robertson et al. 2004),and a drop of dopaminergic terminals with ag-ing (Al-Wandi et al. 2010). Thus, the theory thatpart of the PD phenotype might be attributableto the sequestering of physiological a-synucleinin inclusions, which could cause neuronal, and,in particular (in view of the normal function ofthe protein), synaptic damage, cannot be dis-carded.

Despite the fact that transcriptional regu-lation of a-synuclein is so important for PDpathogenesis, little is known about the mecha-nisms involved. Transcription factors GATA-1(Scherzer et al. 2008) and ZSCAN21 (Cloughand Stefanis 2007; Clough et al. 2009) withinIntron 1 have been proposed to play a role asinducers of transcription. In the latter case, asignaling pathway involving ERK/PI3 kinasesignaling mediated ZSCAN-induced SNCAtranscriptional activation (Clough and Stefanis2007; Clough et al. 2009). A similar ERK-de-pendent signaling pathway regulating SNCAproduction was apparent in enteric neurons(Paillusson et al. 2010). Methylation may beimportant in SNCA expression, as methylationwithin Intron 1 suppressed SNCA transcrip-tional activity, and brains of patients with LBdiseases showed decreased methylation withinthis region, suggesting epigenetic regulation ofSNCA expression (Jowaed et al. 2010; Matsu-moto et al. 2010). In one study, it was proposedthat sequestration of the methylation factorDnmt1 away from the nucleus by a-synucleinmay lead to reduced methylation and enhancedSNCA exression, thus creating a feed-forwardloop of SNCA overexpression (Desplats et al.2011). Posttranscriptional regulation of a-syn-uclein can also occur through endogenousmicro RNAs, which bind to the 30 end of thegene (Junn et al. 2009; Doxakis 2010). A partic-ularly interesting twist to the effects of familialpoint mutations in SNCA has been providedby a series of studies that, somewhat counterin-tuitively, suggest that the expression of the mu-tant alleles is suppressed, especially in cases withprolonged disease (Markopoulou et al. 1999;Kobayashi et al. 2003), through a mechanismthat may involve histone modifications (Vout-sinas et al. 2010). Remarkably, Voutsinas et al.

(2010) found that the WT allele is induced in acompensatory fashion, and that the total levelof SNCA messenger RNA (mRNA) exceeds thatof controls, suggesting that total SNCA levels,rather than the presence of the A53T mutant,are responsible for the disease in these cases.Although this study was based on a single pa-tient, it does bring up again the issue of totalSNCA levels as the major determinant of toxicity.

In view of the above, a reasonable avenue toattack synucleinopathies may be the loweringof a-synuclein levels. A note of caution is in or-der here, given the uncertainty about potentialneurotoxicity associated with severe reductionof a-synuclein levels, as mentioned above. Infact, Gorbatyuk et al. (2010) found that siRNA-mediated down-regulation of a-synuclein ledto quite considerable neurotoxicity within therat nigral dopaminergic system within a periodof 4 wk. In contrast, no toxicity was observedwhen small interfering RNAs (siRNAs) againsta-synuclein were injected directly into the mon-key nigra, achieving 50%–60% down-regula-tion of expression (McCormack et al. 2010).

Control of a-synuclein levels can also beachieved at the level of its degradation. a-Synu-clein degradation has been controversial, but itappears that the bulk of degradation of at leastmonomeric WT a-synuclein in neuronal cellsystems occurs through the lysosomal pathwaysof chaperone-mediated autophagy (CMA) andmacroautophagy (Paxinou et al. 2001; Webbet al. 2003; Cuervo et al. 2004; Lee et al. 2004;Vogiatzi et al. 2008; Mak et al. 2010). It hasbeen proposed that dysfunction of these degra-dation pathways may be a contributing factorto PD pathogenesis (Xilouri et al. 2008). a-Syn-uclein may also be turned over by the protea-some in other experimental settings (Tofariset al. 2001; Webb et al. 2003). A factor thatmay play a role is the exact species ofa-synucleinstudied. In any case, strategies directed towardpromoting such endogenous degradation sys-tems to enhance clearance of excess a-synucleinhave the advantage that they could also alleviatethe aberrant effects ofa-synuclein on their func-tion (see below). Indeed, stimulation of macro-autophagy through pharmacological or molecu-lar means leads to increased WT a-synuclein

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 9

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

clearance and neuroprotection (Webb et al.2003; Spencer et al. 2009). Another attractivemethod of enhancing clearance of a-synucleinthrough the lysosomal pathway may be throughimmunization (Masliah et al. 2005, 2011).

POTENTIAL PATHOGENIC EFFECTS OFa-SYNUCLEIN: a-SYNUCLEIN AT THESYNAPSE

Given the physiological role of a-synuclein atthe synapse, it is not surprising that studieshave identified synaptic effects as significant de-terminants of a-synuclein-induced neurotoxic-ity in cell culture and in vivo models based onoverexpression of the protein; in some of thesecases levels of overexpression are modest, ex-pected to be present in at least the brains ofthe SNCA multiplication cases. The effectsseen include loss of presynaptic proteins, de-crease of neurotransmitter release, redistribu-tion of SNARE proteins, enlargement of synap-tic vesicles, and inhibition of synaptic vesiclerecycling (Chung et al. 2009; Garcia-Reitbocket al. 2010; Nemani et al. 2010; Scott et al.2010). It is likely that such deficits precede overtneuropathology and are causal in synaptic andneuritic degeneration, although the sequenceof synaptic events and the identification of theexact point in the cascade in which aberranta-synuclein assumes its neurotoxic potentialare still elusive.

Given the role of calcium is presynaptic sig-naling events, it is tempting to speculate thatperturbations in calcium homeostasis mayplay a role in the neuritic degeneration inducedby a-synuclein. Indeed, there is some evidencefor this, albeit still somewhat fragmentary,implicating primarily a-synuclein oligomers,which could form pores on cellular membranesor alter the properties of voltage-gated recep-tors, leading to excess calcium influx (Vollesand Lansbury 2002; Danzer et al. 2007; Hettiar-achchi et al. 2009). Through a similar porelikemechanism, oligomeric AS may also attack syn-aptic vesicles, leading to leakage of neurotrans-mitter in the cytosol; in the case of dopaminevesicles, this would mean excess cytosolic dop-amine, which could lead to oxidative stress-

induced death (Mosharov et al. 2006). Excessintracellular calcium enhanced cytosolic dopa-mine, and AS was required for death inducedby excess dopamine, creating a possible scenarioin which intracellular calcium, cystolic dopa-mine, and a-synuclein interact with each otherin a self-feeding cascade leading to neurodegen-eration (Mosharov et al. 2009). Another factorthat could be important in this setting is cal-cium-mediated calpain activation that couldlead, as mentioned, to carboxy-terminal a-syn-uclein truncation, consequent oligomerization,and further calcium influx, creating a viciouscycle.

a-SYNUCLEIN AT THE CYTOSKELETON

a-Synuclein may impact cytoskeletal dynamics.Most of the evidence suggests an association ofa-synuclein with microtubules. Various investi-gators have detected an interaction of a-synu-clein with tubulin (Alim et al. 2002; Zhouet al. 2010), but the effects reported on tubulinpolymerization are conflicting, with others re-porting enhanced (Alim et al. 2004; Lin et al.2009), and others reduced (Lee et al. 2006;Zhou et al. 2010) tubulin polymerization as aresult of a-synuclein overexpression. Further-more, tubulin depolymerization may lead toimpaired maturation of a-synuclein protofi-brils into mature fibrils, which may lead toenhanced neurotoxicity (Lee and Lee 2002).Another factor that may be important is the re-lationship of a-synuclein to tau, which is likelyimportant in the pathogenesis of sporadic dis-ease, as in GWAS the two corresponding genesare the ones showing the highest associationto PD (Nalls et al. 2011). Although tauopathyis not a prominent feature in PD, it is appreci-ated in certain cases (Kotzbauer et al. 2004).In such cases, a-synuclein and tau pathologymay occur in the same cell, but the formed ag-gregates are separate. In fact, each one of thesetwo proteins has the tendency to seed the aggre-gation of the other (Giasson et al. 2003). a-Syn-uclein appears to enhance tau phosphorylation,and this may lead to secondary effects on themicrotubular network (Jensen et al. 1999; Hag-gerty et al. 2011; Qureshi and Paudel 2011).

L. Stefanis

10 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Furthermore, a-synuclein may bind and in-fluence polymerization of actin, and thus per-haps indirectly affect cellular trafficking andsynaptic function; disparate effects on actinwere observed between WT and A30P mutanta-synuclein (Sousa et al. 2009). Another aspectthat appears to be affected by aberrant a-synu-clein, and may be related to the effects on thecytoskeleton, is axonal transport. In an adeno-associated virus (AAV) model of nigral synu-cleinopathy, early changes before cell deathincluded a diminution at the level of the stria-tum of motor proteins involved in anterogradetransport, and an increase of those involved inretrograde transport (Chung et al. 2009).

a-SYNUCLEIN AND PROTEINDEGRADATION SYSTEMS

An issue that has attracted a lot of attention is thepossibility that aberrant a-synuclein may im-pact protein degradation systems. This notionis attractive, as a number of neurodegenerativeconditions are associated with impairments ofprotein clearance, and such impairments havethe potential to create a vicious loop involvingaberrant a-synuclein accumulation. Aberranta-synuclein, mutant, or under certain circum-stances, WT, can induce proteasomal dysfunc-tion in a number of cell-free, cellular, and invivo settings (Stefanis et al. 2001; Tanaka et al.2001; Petrucelli et al. 2002; Snyder et al. 2003),although this is not universally observed (Mar-tin-Clemente et al. 2004). It is thought that oli-gomeric a-synuclein, and, in particular, smallsoluble oligomeric forms that are themselves de-graded by the proteasome, may exert this effect(Lindersson et al. 2004; Emmanouilidou et al.2010a). This would create the potential for thevicious cycle mentioned above.

Mutant a-synuclein may also aberrantlyimpact the lysosomes, causing impairment ofCMA (Cuervo et al. 2004; Xilouri et al. 2009;Alvarez-Erviti et al. 2010). WTAS, in an adductwith dopamine, can cause a similar effect (Mar-tinez-Vicente et al. 2008). Interestingly, the mainproteins involved in CMA, Lamp2a and Hsc70,are diminished in PD brains (Alvarez-Ervitiet al. 2010). Such CMA impairment may cause

compensatory macroautophagy induction thatmay be detrimental to neuronal survival (Xi-louri et al. 2009). The issue is, however, contro-versial, as others find that WT a-synuclein ac-tually diminishes macroautophagy, through aninhibitory action at the level of phagophore for-mation (Winslow et al. 2010).

An interesting issue that relates to a-synu-clein degradation and its effects on protein deg-radation systems is the relationship of PD toGaucher disease and glucocerebrosidase (GBA).Heterozygote and homozygote carriers of GBAmutations are at an increased risk of develop-ing PD. It is controversial whether this repre-sents a loss-of-function of GBA or a gain-of-function conferred by the mutants. Accordingto some, loss-of-function of GBA is sufficientto induce a-synuclein accumulation and aggre-gation (Manning-Bog et al. 2010; Mazzulli et al.2011; Xu et al. 2011), which may occur second-ary to lysosomal dysfunction (Mazzulli et al.2011); others find that a-synuclein accumulatesonly on overexpression of GBA mutants, favor-ing a gain of toxic function mechanism (Cullenet al. 2011). The accumulation of substratessuch as glycosylceramide owing to GBA loss-of-function may also confer increased tendencyof a-synuclein to oligomerize (Mazzulli et al.2011; Xu et al. 2011); on the other hand, oligo-meric a-synuclein may impact on GBA, leadingto its dysfunction, creating a vicious cycle of ly-sosomal dysfunction and aberrant a-synucleinaccumulation (Mazzulli et al. 2011).

a-SYNUCLEIN AT THE MITOCHONDRIAAND RELATIONSHIP TO OXIDATIVESTRESS

Many studies now have confirmed that a pro-portion of a-synuclein, endogenous or overex-pressed, resides within mitochondria, and thatit causes down-regulation of complex I activity,thus linking a-synuclein with the effects of mi-tochondrial toxins, and potentially, sporadicPD (Li et al. 2007; Devi et al. 2008; Nakamuraet al. 2008; Liu et al. 2009; Loeb et al. 2010).Furthermore, brain mitochondria morphologyis disrupted in a-synuclein transgenic mice(Martin et al. 2006). a-Synuclein, potentially

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 11

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

in the form of small oligomers, appears toinduce mitochondrial fragmentation, whichmay be responsible for subsequent mitochon-drial dysfunction and death (Kamp et al. 2010,Nakamura et al. 2011). a-Synuclein, and espe-cially the mutant form, perhaps through thesame mechanism of fragmentation, also in-duces inappropriate excessive mitophagy, whichleads to mitochondrial removal and neuronaldeath (Choubey et al. 2011). Interestingly, en-hancing the expression of PGC-1a, the mastertranscriptional regulator of nuclearly encodedmitochondrial subunits, which is down-regu-lated in PD brains, leads to protection againsta-synuclein-induced neurodegeneration in cel-lular models (Zheng et al. 2010).

Through such effects on mitochondria, andespecially the complex I inhibition, a-synucleincould potentially induce reactive oxygen species(ROS) production, oxidative stress, and resultantneuronal death. There is in fact some evidencethat a-synuclein overexpression can induce ROS,and that suppressing these through molec-ular or pharmacological means may be neuro-protective in mammalian (Hsu et al. 2000; Clarket al. 2010; Liu et al. 2011) or insect systems(Wassef et al. 2007; Botella et al. 2008).

a-SYNUCLEIN AT THE ER/GOLGI

In a neuronal cell model based on adenoviralexpression of a-synuclein, Golgi fragmentationwas an early feature and correlated with the ap-pearance of soluble oligomeric species (Gosaviet al. 2002). Smith et al. (2005) observed endo-plasmic reticulum (ER) stress as an early eventin an inducible model of A53T a-synuclein-mediated neurotoxicity, and were able to inhibitdeath using a pharmacological inhibitor of ERstress, also suggesting that a primary target ofa-synuclein may be the ER/Golgi. In a blindedscreen in a yeast model, the major class of modu-lators ofa-synuclein toxicity was genes involvedin vesicular trafficking (Willingham et al. 2003).This was taken one step further by Cooper et al.(2006), who identified ER-to-Golgi traffickingas the major cellular perturbation in this yeastmodel, and Rab1, a protein involved in this tran-sition, as a critical determinant of a-synuclein-

mediated toxicity in a variety of cellular and invivo models. Thayanidhi et al. (2010) suggestedthat in mammalian systems the ER/Golgi transi-tion was delayed owing to an antagonistic effectof a-synuclein on ER/Golgi SNAREs. Over-expression of other Rabs, more closely linkedto synaptic vesicular function, such as Rab3A,also prevented dopaminergic toxicity in the a-synuclein C. elegans model, suggesting that thesephenomena may more broadly reflect the inter-action and effects of a-synuclein on vesiculardynamics (Gitler et al. 2008).

a-SYNUCLEIN IN THE NUCLEUS

Synuclein bears its name from its apparent lo-calization in the nucleus and presynatic nerveterminals (Maroteaux et al. 1988). However,nuclear localization of a-synuclein has onlybeen observed inconsistently (Mori et al.2002b; Yu et al. 2007); it actually appears that,at least in transgenic mice, a-synuclein phos-phorylated at S129 may be preferentially local-ized in the nucleus (Schell et al. 2009). a-Synu-clein has been found to interact with histoneson nuclear translocation in nigral neurons in re-sponse to the herbicide paraquat (Goers et al.2003), and to reduce histone acetylation (Kon-topoulos et al. 2006). Inhibitors of histone de-acetylase (HDAC) rescued a-synuclein-medi-ated neurotoxicity in cell culture and in vivoDrosophila models (Kontopoulos et al. 2006).Furthermore, inhibition of Sirtuin 2, a mamma-lian HDAC, which led to increased acetylation,was associated with neuroprotection in variousmodels of synucleinopathy, although whetherpotential acetylation effects on histone were in-volved was not ascertained (Outeiro et al. 2007).

a-SYNUCLEIN OUTSIDE THE CELL:THE THEORY OF PROPAGATION

Although initially a-synuclein was thought tobe a purely intracellular protein, as it lacks a sig-naling sequence that would direct it to the secre-tory pathway, it gradually became clear that itcould be detected in the conditioned mediumof cells and in extracellular fluids, such asplasma and CSF (El-Agnaf et al. 2003; Lee

L. Stefanis

12 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

et al. 2005). The mechanism of a-synuclein se-cretion is not well understood, but it appears tooccur through a nonclassical secretory pathway(Lee et al. 2005), possibly within exosomes, en-docytic vesicles that are contained within mul-tivesicular bodies and are released on calciuminflux (Emmanouilidou et al. 2010b). The exis-tence of extracellular a-synuclein in rodent andhuman brain interstitial fluid has been con-firmed by microdialysis (Emmanouilidou et al.2011). Importantly, secreted a-synuclein canimpact neuronal homeostasis and lead to neu-ronal death, even at concentrations close tothose identified in bodily fluids, and the respon-sible species, at least in part, appear to be solubleoligomeric (Emmanouilidou et al. 2010b; Dan-zer et al. 2011). Furthermore, various reportshave shown that extracellular a-synuclein caninduce inflammatory reactions in glial cells,thus further potentiating neurodegeneration(Zhang et al. 2005; Klegeris et al. 2008; Leeet al. 2010). It should be noted, however, thatsuch studies are often based on high concentra-tions of recombinant a-synuclein that may notbe physiologic. This issue is also touched on byGendelman and colleagues (Mosley et al. inpress).

A related aspect of a-synuclein pathobiol-ogy that has attracted a lot of attention is its ap-parent ability to be uptaken by cells, although,at least in cell culture, this is highly dependenton the cell type and the particular species ofa-synuclein and is not easily achieved in intactneuronal cells (Emmanouilidou et al. 2010b).Oligomeric-aggregated a-synuclein species ap-pear to be especially prone to uptake and havethe potential to “seed” fibrillization of endoge-nous a-synuclein (Danzer et al. 2007, 2009,2011; Luk et al. 2009; Nonaka et al. 2010; Wax-man and Giasson 2010; Hansen et al. 2011).This observation has created a lot of excitement,as it could potentially explain the propagationof pathology observed in PD, according to theBraak hypothesis of staging of the disease. Twokey in vivo observations support this “prionlikepropagation” theory. On the one hand, progen-itor cells implanted in the hippocampus of re-cipient mice transgenic for a-synuclein incor-porated host a-synuclein in their cell soma

(Desplats et al. 2009), and fetal dopaminergicneurons also showed uptake of hosta-synuclein(Hansen et al. 2011); on the other, fetal dopami-nergic grafts in humans showed LB pathologymany years after implantation, although wheth-er the a-synuclein within the grafted neuronswas host-derived could not be determined withcertainty (Kordower et al. 2008). An intriguingstudy has brought together the mitochondrialdysfunction with the a-synuclein propagationtheories. Intragastric administration of the com-plex I inhibitor rotenone led to enteric a-synu-clein pathology, which gradually spread to thecentral nervous system (CNS), including dopa-minergic neurons, and caused delayed neuro-toxicity, suggesting that, once initiated by mito-chondrial dysfunction, a-synuclein aberrantconformations assume an independent propa-gating seeding and neurotoxic potential (Pan-Montojo et al. 2010). This would be consistentwith the findings of early enteric a-synucleinpathology in PD (Braak et al. 2006) and in a-synuclein transgenic mice (Kuo et al. 2010). Anote of caution is indicated in the general as-sumption that seeding is equated with neuro-toxicity. In fact, seeding and toxicity of extracel-lular a-synuclein can be dissociated (Danzeret al. 2007).

a-SYNUCLEIN BIOMARKERS

In view of the importance of a-synuclein inPD pathogenesis, a number of studies have at-tempted to use measurements of a-synucleinas biomarkers for the disease and for othersynucleinopathies. In the cerebrospinal fluid(CSF), most studies appear to show a reductionof total a-synuclein levels in PD patients com-pared with controls (Tokuda et al. 2006; Mol-lenhauer et al. 2008, 2011; Hong et al. 2010; Ka-suga et al. 2010); in fact, a comprehensive studysuggested thata-synuclein levels, measured by asensitive ELISA technique, may help to differen-tiate synucleinopathies, where a-synuclein isreduced compared with other neurodegenera-tive conditions, such as tauopathies or amy-loidoses (Mollenhauer et al. 2011). In contrast,oligomeric a-synuclein, measured by a specificELISA method, was higher in PD compared

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 13

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

with control CSF (Tokuda et al. 2010), extend-ing previous findings of identification of solubleoligomeric a-synuclein in DLB brain tissue(Paleologou et al. 2009), but this intriguing re-sult awaits replication in a larger cohort. Datafrom a-synuclein measurements in peripheraltissues/fluids have been inconsistent, and over-all peripherala-synuclein levels, despite the easeof obtaining the material, do not appear to havepotential as biomarkers.

CONCLUDING REMARKS

It is clear from the above thata-synuclein repre-sents a valid therapeutic target in PD and possi-bly in related synucleinpathies. Possible thera-peutic strategies that could be envisioned aredepicted in Figure 2. Perhaps the most promis-ing strategy would involve small molecules thatwould have the potential to alter the confor-mation of protofibrillar forms of a-synucleinand render them nonpathogenic. An exampleof such natural products could be the greentea derivative EGCG (Bieschke et al. 2010) orthe natural product scyllo inositol (Vekrelliset al. 2009). It may be that multiple therapeuticapproaches will need to be used, in view of thefeed-forward amplification loops involved inthe pathogenic effects of a-synuclein.

ACKNOWLEDGMENTS

L.S. was funded for PD-related research by ECFP7 Programmes NEURASYNC and MEFOPA.

REFERENCES�Reference is also in this collection.

Abeliovich A, Schmitz Y, Farinas I, Choi-Lundberg D, HoWH, Castillo PE, Shinsky N, Verdugo JM, Armanini M,Ryan A, et al. 2000. Mice lacking a-synuclein displayfunctional deficits in the nigrostriatal dopamine system.Neuron 25: 239–252.

Al-Chalabi A, Durr A, Wood NW, Parkinson MH, CamuzatA, Hulot JS, Morrison KE, Renton A, Sussmuth SD,Landwehrmeyer BG, et al. 2009. Genetic variants of thea-synuclein gene SNCA are associated with multiple sys-tem atrophy. PLoS One 4: e7114.

Alim MA, Hossain MS, Arima K, Takeda K, Izumiyama Y,Nakamura M, Kaji H, Shinoda T, Hisanaga S, Ueda K.2002. Tubulin seeds a-synuclein fibril formation. J BiolChem 277: 2112–2117.

Alim MA, Ma QL, Takeda K, Aizawa T, Matsubara M, Naka-mura M, Asada A, Saito T, Kaji H, Yoshii M, et al. 2004.Demonstration of a role for a-synuclein as a functionalmicrotubule-associated protein. J Alzheimers Dis 6:435–442.

Alvarez-Erviti L, Rodriguez-Oroz MC, Cooper JM, Cabal-lero C, Ferrer I, Obeso JA, Schapira AH. 2010. Chaper-one-mediated autophagy markers in Parkinson diseasebrains. Arch Neurol 67: 1464–1472.

Al-Wandi A, Ninkina N, Millership S, Williamson SJ, JonesPA, Buchman VL. 2010. Absence of a-synuclein affectsdopamine metabolism and synaptic markers in the stria-tum of aging mice. Neurobiol Aging 31: 796–804.

Preventingspecific aberrant

functions

Synapticvesicles

Proteasomes

Lysosomes

ER-Golgi

Cell membrane/Calcium

Mitochondria

Axonal transport

Promoting fibrilformation

Promoting off-pathwayoligomers

Inhibitingoligomer/protofibril

formation

EnhancingdegradationModulating

transcription

Inhibitingproaggregationposttranslational

modifications

Figure 2. Possible targets for therapeutic intervention in PD and related synucleinopathies.

L. Stefanis

14 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Assayag K, Yakunin E, Loeb V, Selkoe DJ, Sharon R. 2007.Polyunsaturated fatty acids induce a-synuclein-relatedpathogenic changes in neuronal cells. Am J Pathol 171:2000–2011.

Auluck PK, Bonini NM. 2002. Pharmacological preventionof Parkinson disease in Drosophila. Nat Med 8: 1185–1186.

Auluck PK, Chan HY, Trojanowski JQ, Lee VM, Bonini NM.2002. Chaperone suppression of a-synuclein toxicity in aDrosophila model for Parkinson’s disease. Science 295:865–868.

Austin SA, Floden AM, Murphy EJ, Combs CK. 2006. a-Synuclein expression modulates microglial activationphenotype. J Neurosci 26: 10558–10563.

Austin SA, Rojanathammanee L, Golovko MY, Murphy EJ,Combs CK. 2011. Lack of a-synuclein modulates micro-glial phenotype in vitro. Neurochem Res 36: 994–1004.

Azeredo da Silveira S, Schneider BL, Cifuentes-Diaz C, SageD, Abbas-Terki T, Iwatsubo T, Unser M, Aebischer P. 2009.Phosphorylation does not prompt, nor prevent, the for-mation of a-synuclein toxic species in a rat model of Par-kinson’s disease. Hum Mol Genet 18: 872–887.

Baba M, Nakajo S, Tu PH, Tomita T, Nakaya K, Lee VM,Trojanowski JQ, Iwatsubo T. 1998. Aggregation of a-syn-uclein in Lewy bodies of sporadic Parkinson’s disease anddementia with Lewy bodies. Am J Pathol 152: 879–884.

Barbour R, Kling K, Anderson JP, Banducci K, Cole T, DiepL, Fox M, Goldstein JM, Soriano F, Seubert P, et al. 2008.Red blood cells are the major source of a-synuclein inblood. Neurodegener Dis 5: 55–59.

Betarbet R, Canet-Aviles RM, Sherer TB, MastroberardinoPG, McLendon C, Kim JH, Lund S, Na HM, Taylor G,Bence NF, et al. 2006. Intersecting pathways to neurode-generation in Parkinson’s disease: Effects of the pesticiderotenone on DJ-1, a-synuclein, and the ubiquitin-pro-teasome system. Neurobiol Dis 22: 404–420.

Bieschke J, Russ J, Friedrich RP, Ehrnhoefer DE, Wobst H,Neugebauer K, Wanker EE. 2010. EGCG remodels ma-ture a-synuclein and amyloid-bfibrils and reduces cellu-lar toxicity. Proc Natl Acad Sci 107: 7710–7715.

Bodner RA, Outeiro TF, Altmann S, Maxwell MM, Cho SH,Hyman BT, McLean PJ, Young AB, Housman DE, Ka-zantsev AG. 2006. Pharmacological promotion of inclu-sion formation: A therapeutic approach for Huntington’sand Parkinson’s diseases. Proc Natl Acad Sci 103: 4246–4251.

Botella JA, Bayersdorfer F, Schneuwly S. 2008. Superoxidedismutase overexpression protects dopaminergic neu-rons in a Drosophila model of Parkinson’s disease. Neuro-biol Dis 30: 65–73.

Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN,Braak E. 2003. Staging of brain pathology related tosporadic Parkinson’s disease. Neurobiol Aging 24: 197–211.

Braak H, de Vos RA, Bohl J, Del Tredici K. 2006. Gastrica-synuclein immunoreactive inclusions in Meissner’sand Auerbach’s plexuses in cases staged for Parkinson’sdisease-related brain pathology. Neurosci Lett 396: 67–72.

Burke RE, Dauer WT, Vonsattel JP. 2008. A critical evalua-tion of the Braak staging scheme for Parkinson’s disease.Ann Neurol 64: 485–491.

Burre J, Sharma M, Tsetsenis T, Buchman V, Etherton MR,Sudhof TC. 2010. a-Synuclein promotes SNARE-com-plex assembly in vivo and in vitro. Science 329: 1663–1667.

Cabin DE, Shimazu K, Murphy D, Cole NB, Gottschalk W,McIlwain KL, Chen A, Ellis CE, Paylor R, Lu B, et al. 2002.Synaptic vesicle depletion correlates with attenuated syn-aptic responses to prolonged repetitive stimulation inmice lacking a-synuclein. J Neurosci 22: 8797–8807.

Chandra S, Gallardo G, Fernandez-Chacon R, Schluter OM,Sudhof TC. 2005. Alpha-synuclein cooperates withCSPalpha in preventing neurodegeneration. Cell 123:383–396.

Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V,Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J,Hulihan M, et al. 2004. a-Synuclein locus duplicationas a cause of familial Parkinson’s disease. Lancet 364:1167–1169.

Chen L, Feany MB. 2005. a-Synuclein phosphorylationcontrols neurotoxicity and inclusion formation in a Dro-sophila model of Parkinson disease. Nat Neurosci 8:657–663.

Chen L, Periquet M, Wang X, Negro A, McLean PJ, HymanBT, Feany MB. 2009. Tyrosine and serine phosphoryla-tion of a-synuclein have opposing effects on neurotoxic-ity and soluble oligomer formation. J Clin Invest 119:3257–3265.

Chesselet MF. 2008. In vivo a-synuclein overexpression inrodents: A useful model of Parkinson’s disease? Exp Neu-rol 209: 22–27.

Chiba-Falek O, Nussbaum RL. 2001. Effect of allelic varia-tion at the NACP-Rep1 repeat upstream of the a-synu-clein gene (SNCA) on transcription in a cell culture luci-ferase reporter system. Hum Mol Genet 10: 3101–3109.

Choubey V, Safiulina D, Vaarmann A, Cagalinec M, WareskiP, Kuum M, Zharkovsky A, Kaasik A. 2011. Mutant A53Ta-synuclein induces neuronal death by increasing mito-chondrial autophagy. J Biol Chem 286: 10814–10824.

Chu Y, Kordower JH. 2007. Age-associated increases ofa-synuclein in monkeys and humans are associatedwith nigrostriatal dopamine depletion: Is this the targetfor Parkinson’s disease? Neurobiol Dis 25: 134–149.

Chung CY, Koprich JB, Siddiqi H, Isacson O. 2009. Dynamicchanges in presynaptic and axonal transport proteinscombined with striatal neuroinflammation precede dop-aminergic neuronal loss in a rat model of AAV a-synu-cleinopathy. J Neurosci 29: 3365–3373.

Clark J, Clore EL, Zheng K, Adame A, Masliah E, Simon DK.2010. Oral N-acetyl-cysteine attenuates loss of dopami-nergic terminals in a-synuclein overexpressing mice.PLoS One 5: e12333.

Clough RL, Dermentzaki G, Stefanis L. 2009. Functionaldissection of the a-synuclein promoter: Transcriptionalregulation by ZSCAN21 and ZNF219. J Neurochem 110:1479–1490.

Clough RL, Stefanis L. 2007. A novel pathway for transcrip-tional regulation of a-synuclein. FASEB J 21: 596–607.

Conway KA, Lee SJ, Rochet JC, Ding TT, Williamson RE,Lansbury PT Jr. 2000. Acceleration of oligomerization,not fibrillization, is a shared property of botha-synucleinmutations linked to early-onset Parkinson’s disease:

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 15

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Implications for pathogenesis and therapy. Proc NatlAcad Sci 97: 571–576.

Conway KA, Rochet JC, Bieganski RM, Lansbury PT Jr.2001. Kinetic stabilization of the a-synuclein protofibrilby a dopamine-a-synuclein adduct. Science 294: 1346–1349.

Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ,Bhullar B, Liu K, Xu K, Strathearn KE, Liu F, et al.2006. a-Synuclein blocks ER-Golgi traffic and Rab1 res-cues neuron loss in Parkinson’s models. Science 313:324–328.

Cronin KD, Ge D, Manninger P, Linnertz C, Rossoshek A,Orrison BM, Bernard DJ, El-Agnaf OM, SchlossmacherMG, Nussbaum RL, et al. 2009. Expansion of the Parkin-son disease-associated SNCA-Rep1 allele upregulates hu-man a-synuclein in transgenic mouse brain. Hum MolGenet 18: 3274–3285.

Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, SulzerD. 2004. Impaired degradation of mutant a-synuclein bychaperone-mediated autophagy. Science 305: 1292–1295.

Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, Kolod-ziej P, Kahn I, Saftig P, Woulfe J, et al. 2011. Acid b-gluco-sidase mutants linked to Gaucher disease, Parkinson dis-ease, and Lewy body dementia alter a-synucleinprocessing. Ann Neurol 69: 940–953.

Dachsel JC, Lincoln SJ, Gonzalez J, Ross OA, Dickson DW,Farrer MJ. 2007 The ups and downs of a-synucleinmRNA expression. Movement Disord 22: 293–295.

da Costa CA, Ancolio K, Checler F. 2000. Wild-type butnot Parkinson’s disease-related ala-53! Thr mutanta-synuclein protects neuronal cells from apoptotic stim-uli. J Biol Chem 275: 24065–24069.

Danzer KM, Haasen D, Karow AR, Moussaud S, Habeck M,Giese A, Kretzschmar H, Hengerer B, Kostka M. 2007.Different species of a-synuclein oligomers induce cal-cium influx and seeding. J Neurosci 27: 9220–9232.

Danzer KM, Krebs SK, Wolff M, Birk G, Hengerer B. 2009.Seeding induced by a-synuclein oligomers provides evi-dence for spreading of a-synuclein pathology. J Neuro-chem 111: 192–203.

Danzer KM, Ruf WP, Putcha P, Joyner D, Hashimoto T,Glabe C, Hyman BT, McLean PJ. 2011. Heat-shock pro-tein 70 modulates toxic extracellulara-synuclein oligom-ers and rescues trans-synaptic toxicity. FASEB J 25:326–336.

Darios F, Ruiperez V, Lopez I, Villanueva J, Gutierrez LM,Davletov B. 2010. a-Synuclein sequesters arachidonicacid to modulate SNARE-mediated exocytosis. EMBORep 11: 528–533.

Dauer W, Kholodilov N, Vila M, Trillat AC, Goodchild R,Larsen KE, Staal R, Tieu K, Schmitz Y, Yuan CA, et al.2002. Resistance of a-synuclein null mice to the parkin-sonian neurotoxin MPTP. Proc Natl Acad Sci 99: 14524–14529.

Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L,Spencer B, Masliah E, Lee SJ. 2009. Inclusion formationand neuronal cell death through neuron-to-neurontransmission of a-synuclein. Proc Natl Acad Sci 106:13010–13015.

Desplats P, Spencer B, Coffee E, Patel P, Michael S, Patrick C,Adame A, Rockenstein E, Masliah E. 2011. a-Synuclein

sequesters Dnmt1 from the nucleus: A novel mechanismfor epigenetic alterations in Lewy body diseases. J BiolChem 286: 9031–9037.

Devi L, Raghavendran V, Prabhu BM, Avadhani NG, Anan-datheerthavarada HK. 2008. Mitochondrial import andaccumulation of a-synuclein impair complex I in humandopaminergic neuronal cultures and Parkinson diseasebrain. J Biol Chem 283: 9089–9100.

Dickson DW, Uchikado H, Fujishiro H, Tsuboi Y. 2010. Evi-dence in favor of Braak staging of Parkinson’s disease.Mov Disord 25 (Suppl 1): S78–S82.

Doxakis E. 2010. Post-transcriptional regulation of a-synu-clein expression by mir-7 and mir-153. J Biol Chem 285:12726–12734.

El-Agnaf OM, Salem SA, Paleologou KE, Cooper LJ, Full-wood NJ, Gibson MJ, Curran MD, Court JA, MannDM, Ikeda S, et al. 2003. a-Synuclein implicated in Par-kinson’s disease is present in extracellular biological flu-ids, including human plasma. FASEB J 17: 1945–1947.

Eliezer P, Zweckstetter D, Masliah M, Lashuel EHA. 2010.Phosphorylation at S87 is enhanced in synucleinopathies,inhibits a-synuclein oligomerization, and influencessynuclein-membrane interactions. J Neurosci 30: 3184–3198.

Emmanoulidou E, Stefanis L, Vekrellis K. 2010a. Specificcell-derived soluble a-synuclein oligomers are degradedby the 26S proteasome and impair its function. NeurobiolAging 31: 953–968.

Emmanouilidou E, Melachroinou K, Roumeliotis T, GarbisSD, Ntzouni M, Margaritis LH, Stefanis L, Vekrellis K.2010b. Cell-produced a-synuclein is secreted in a cal-cium-dependent manner by exosomes and impacts neu-ronal survival. J Neurosci 30: 6838–6851.

Emmanouilidiou E, Elenis D, Papasilekas T, Stranjalis G,Gerozissis K, Ioannou P, Vekrellis K. 2011. In vivo assess-ment of a-synuclein secretion. PLoS One 6: e22225.

Engelender S, Kaminsky Z, Guo X, Sharp AH, Amaravi RK,Kleiderlein JJ, Margolis RL, Troncoso JC, Lanahan AA,Worley PF, et al. 1999. Synphilin-1 associates with a-syn-uclein and promotes the formation of cytosolic inclu-sions. Nat Genet 22: 110–114.

Esposito A, Dohm CP, Kermer P, Bahr M, Wouters FS. 2007.a-Synuclein and its disease-related mutants interact dif-ferentially with the microtubule protein tau and associatewith the actin cytoskeleton. Neurobiol Dis 26: 521–531.

Feany MB, Bender WW. 2000. A Drosophila model of Par-kinson’s disease. Nature 404: 394–398.

Fortin DL, Nemani VM, Voglmaier SM, Anthony MD, RyanTA, Edwards RH. 2005. Neural activity controls the syn-aptic accumulation ofa-synuclein. J Neurosci 25: 10913–10921.

Fredenburg RA, Rospigliosi C, Meray RK, Kessler JC, La-shuel HA, Eliezer D, Lansbury PT Jr. 2007. The impactof the E46K mutation on the properties of a-synucleinin its monomeric and oligomeric states. Biochemistry46: 7107–7118.

Fuchs J, Nilsson C, Kachergus J, Munz M, Larsson EM,Schule B, Langston JW, Middleton FA, Ross OA, HulihanM, et al. 2007. Phenotypic variation in a large Swedishpedigree due to SNCA duplication and triplication. Neu-rology 68: 916–922.

L. Stefanis

16 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Fuchs J, Tichopad A, Golub Y, Munz M, Schweitzer KJ, WolfB, Berg D, Mueller JC, Gasser T. 2008. Genetic variabilityin the SNCA gene influences a-synuclein levels in theblood and brain. FASEB J 22: 1327–1334.

Fujita M, Sugama S, Sekiyama K, Sekigawa A, Tsukui T, Na-kai M, Waragai M, Takenouchi T, Takamatsu Y, Wei J,et al. 2010 A b-synuclein mutation linked to dementiaproduces neurodegeneration when expressed in mousebrain. Nat Commun 1: 110.

Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Mas-liah E, Goldberg MS, Shen J, Takio K, Iwatsubo T. 2002.a-Synuclein is phosphorylated in synucleinopathy le-sions. Nat Cell Biol 4: 160–164.

Galvin JE, Uryu K, Lee VM, Trojanowski JQ. 1999. Axonpathology in Parkinson’s disease and Lewy body demen-tia hippocampus contains a-, b-, and g-synuclein. ProcNatl Acad Sci 96: 13450–13455.

Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ,Lee VM. 2008. Neuroinflammation and oxidation/nitra-tion of a-synuclein linked to dopaminergic neurodegen-eration. J Neurosci 28: 7687–7698.

Garcia-Reitbock P, Anichtchik O, Bellucci A, Iovino M, Bal-lini C, Fineberg E, Ghetti B, Della Corte L, Spano P, Tofa-ris GK, et al. 2010. SNARE protein redistribution andsynaptic failure in a transgenic mouse model of Parkin-son’s disease. Brain 133 (Pt 7): 2032–2044.

George JM. 2002. The synucleins. Genome Biol 3: RE-VIEWS3002.

George JM, Jin H, Woods WS, Clayton DF. 1995. Character-ization of a novel protein regulated during the critical pe-riod for song learning in the zebra finch. Neuron 15:361–372.

Giasson BI, Duda JE, Murray IV, Chen Q, Souza JM, HurtigHI, Ischiropoulos H, Trojanowski JQ, Lee VM. 2000. Ox-idative damage linked to neurodegeneration by selectivea-synuclein nitration in synucleinopathy lesions. Science290: 985–989.

Giasson BI, Forman MS, Higuchi M, Golbe LI, Graves CL,Kotzbauer PT, Trojanowski JQ, Lee VM. 2003. Initiationand synergistic fibrillization of t anda-synuclein. Science300: 636–640.

Gitler AD, Bevis BJ, Shorter J, Strathearn KE, Hamamichi S,Su LJ, Caldwell KA, Caldwell GA, Rochet JC, McCafferyJM, et al. 2008. The Parkinson’s disease proteina-synuclein disrupts cellular Rab homeostasis. ProcNatl Acad Sci 105: 145–150.

Goers J, Manning-Bog AB, McCormack AL, Millett IS, Do-niach S, Di Monte DA, Uversky VN, Fink AL. 2003. Nu-clear localization of a-synuclein and its interaction withhistones. Biochemistry 42: 8465–8471.

Gorbatyuk OS, Li S, Sullivan LF, Chen W, Kondrikova G,Manfredsson FP, Mandel RJ, Muzyczka N. 2008. Thephosphorylation state of Ser-129 in human a-synucleindetermines neurodegeneration in a rat model of Parkin-son disease. Proc Natl Acad Sci 105: 763–768.

Gorbatyuk OS, Li S, Nash K, Gorbatyuk M, Lewin AS, Sul-livan LF, Mandel RJ, Chen W, Meyers C, Manfredsson FP,et al. 2010. In vivo RNAi-mediated a-synuclein silencinginduces nigrostriatal degeneration. Mol Ther 18: 1450–1457.

Gosavi N, Lee HJ, Lee JS, Patel S, Lee SJ. 2002. Golgi frag-mentation occurs in the cells with prefibrillara-synuclein

aggregates and precedes the formation of fibrillar inclu-sion. J Biol Chem 277: 48984–48992.

Grundemann J, Schlaudraff F, Haeckel O, Liss B. 2008. Ele-vated a-synuclein mRNA levels in individual UV-laser-microdissected dopaminergic substantia nigra neuronsin idiopathic Parkinson’s disease. Nucleic Acids Res 36:e38.

Haggerty T, Credle J, Rodriguez O, Wills J, Oaks AW, MasliahE, Sidhu A. 2011. Hyperphosphorylated t in an a-synu-clein-overexpressing transgenic model of Parkinson’s dis-ease. Eur J Neurosci 33: 1598–1610.

Hansen C, Angot E, Bergstrom AL, Steiner JA, Pieri L, PaulG, Outeiro TF, Melki R, Kallunki P, Fog K, et al. 2011.a-Synuclein propagates from mouse brain to grafteddopaminergic neurons and seeds aggregation in culturedhuman cells. J Clin Invest 121: 715–725.

Hashimoto M, Rockenstein E, Mante M, Mallory M, Mas-liah E. 2001. b-Synuclein inhibits a-synuclein aggrega-tion: A possible role as an anti-parkinsonian factor. Neu-ron 32: 213–223.

Hettiarachchi NT, Parker A, Dallas ML, Pennington K, HungCC, Pearson HA, Boyle JP, Robinson P, Peers C. 2009.a-Synuclein modulation of Ca2þ signaling in humanneuroblastoma (SH-SY5Y) cells. J Neurochem 111: 1192–1201.

Hodara R, Norris EH, Giasson BI, Mishizen-Eberz AJ,Lynch DR, Lee VM, Ischiropoulos H. 2004. Functionalconsequences of a-synuclein tyrosine nitration: Dimin-ished binding to lipid vesicles and increased fibril forma-tion. J Biol Chem 279: 47746–47753.

Hong Z, Shi M, Chung KA, Quinn JF, Peskind ER, GalaskoD, Jankovic J, Zabetian CP, Leverenz JB, Baird G, et al.2010. DJ-1 anda-synuclein in human cerebrospinal fluidas biomarkers of Parkinson’s disease. Brain 133 (Pt 3):713–726.

Hsu LJ, Sagara Y, Arroyo A, Rockenstein E, Sisk A, MalloryM, Wong J, Takenouchi T, Hashimoto M, Masliah E.2000. a-Synuclein promotes mitochondrial deficit andoxidative stress. Am J Pathol 157: 401–410.

Inglis KJ, Chereau D, Brigham EF, Chiou SS, Schobel S,Frigon NL, Yu M, Caccavello RJ, Nelson S, Motter R,et al. 2009. Polo-like kinase 2 (PLK2) phosphorylatesa-synuclein at serine 129 in central nervous system.J Biol Chem 284: 2598–2602.

Jensen PH, Hager H, Nielsen MS, Hojrup P, Gliemann J,Jakes R. 1999. a-Synuclein binds to t and stimulatesthe protein kinase A-catalyzed tphosphorylation of ser-ine residues 262 and 356. J Biol Chem 274: 25481–25489.

Jin H, Kanthasamy A, Ghosh A, Yang Y, Anantharam V,Kanthasamy AG. 2011. a-Synuclein negatively regulatesprotein kinase Cd expression to suppress apoptosis indopaminergic neurons by reducing p300 histone acetyl-transferase activity. J Neurosci 31: 2035–2051.

Jowaed A, Schmitt I, Kaut O, Wullner U. 2010. Methylationregulates a-synuclein expression and is decreased in Par-kinson’s disease patients’ brains. J Neurosci 30: 6355–6359.

Junn E, Lee KW, Jeong BS, Chan TW, Im JY, MouradianMM. 2009. Repression of a-synuclein expression andtoxicity by microRNA-7. Proc Natl Acad Sci 106: 13052–13057.

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 17

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Kahle PJ. 2008.a-Synucleinopathy models and human neu-ropathology: Similarities and differences. Acta Neuropa-thol 115: 87–95.

Kamp F, Exner N, Lutz AK, Wender N, Hegermann J, Brun-ner B, Nuscher B, Bartels T, Giese A, Beyer K, et al. 2010.Inhibition of mitochondrial fusion by a-synuclein is res-cued by PINK1, Parkin and DJ-1. EMBO J 29: 3571–3589.

Karpinar DP, Balija MB, Kugler S, Opazo F, Rezaei-GhalehN, Wender N, Kim HY, Taschenberger G, FalkenburgerBH, Heise H, et al. 2009. Pre-fibrillar a-synuclein var-iants with impaired b-structure increase neurotoxicityin Parkinson’s disease models. EMBO J 28: 3256–3268.

Kasuga K, Tokutake T, Ishikawa A, Uchiyama T, Tokuda T,Onodera O, Nishizawa M, Ikeuchi T. 2010. Differentiallevels of a-synuclein, b-amyloid42 and tau in CSFbetween patients with dementia with Lewy bodies andAlzheimer’s disease. J Neurol Neurosurg Psychiatry 81:608–610.

Kholodilov NG, Neystat M, Oo TF, Lo SE, Larsen KE, SulzerD, Burke RE. 1999. Increased expression of rat synucleinin the substantia nigra pars compacta identified bymRNA differential display in a model of developmentaltarget injury. J Neurochem 73: 2586–2599.

Kim TD, Paik SR, Yang CH. 2002. Structural and functionalimplications of C-terminal regions of a-synuclein. Bio-chemistry 41: 13782–13790.

Kirik D, Rosenblad C, Burger C, Lundberg C, Johansen TE,Muzyczka N, Mandel RJ, Bjorklund A. 2002. Parkinson-like neurodegeneration induced by targeted overexpres-sion of a-synuclein in the nigrostriatal system. J Neurosci22: 2780–2791.

Klegeris A, Pelech S, Giasson BI, Maguire J, Zhang H,McGeer EG, McGeer PL. 2008. a-Synuclein activatesstress signaling protein kinases in THP-1 cells and micro-glia. Neurobiol Aging 29: 739–752.

Klucken J, Shin Y, Masliah E, Hyman BT, McLean PJ. 2004.Hsp70 reduces a-synuclein aggregation and toxicity. JBiol Chem 279: 25497–25502.

Kobayashi H, Kruger R, Markopoulou K, Wszolek Z, ChaseB, Taka H, Mineki R, Murayama K, Riess O, Mizuno Y,et al. 2003. Haploinsufficiency at the a-synuclein geneunderlies phenotypic severity in familial Parkinson’s dis-ease. Brain 126 (Pt 1): 32–42.

Kontopoulos E, Parvin JD, Feany MB. 2006. a-Synucleinacts in the nucleus to inhibit histone acetylation and pro-mote neurotoxicity. Hum Mol Genet 15: 3012–3023.

Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW.2008. Lewy body-like pathology in long-term embryonicnigral transplants in Parkinson’s disease. Nat Med 14:504–506.

Kostka M, Hogen T, Danzer KM, Levin J, Habeck M, WirthA, Wagner R, Glabe CG, Finger S, Heinzelmann U, et al.2008. Single particle characterization of iron-inducedpore-forming a-synuclein oligomers. J Biol Chem 283:10992–11003.

Kotzbauer PT, Giasson BI, Kravitz AV, Golbe LI, Mark MH,Trojanowski JQ, Lee VM. 2004. Fibrillization of a-synu-clein and tau in familial Parkinson’s disease caused by theA53T a-synuclein mutation. Exp Neurol 187: 279–288.

Kramer ML, Schulz-Schaeffer WJ. 2007. Presynaptic a-syn-uclein aggregates, not Lewy bodies, cause neurodegener-

ation in dementia with Lewy bodies. J Neurosci 27:1405–1410.

Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kosel S,Przuntek H, Epplen JT, Schols L, Riess O. 1998. Ala30Promutation in the gene encoding a-synuclein in Parkin-son’s disease. Nat Genet 18: 106–108.

Kuo YM, Li Z, Jiao Y, Gaborit N, Pani AK, Orrison BM, Bru-neau BG, Giasson BI, Smeyne RJ, Gershon MD, et al.2010. Extensive enteric nervous system abnormalities inmice transgenic for artificial chromosomes containingParkinson disease-associated a-synuclein gene muta-tions precede central nervous system changes. Hum MolGenet 19: 1633–1650.

Lakso M, Vartiainen S, Moilanen AM, Sirvio J, Thomas JH,Nass R, Blakely RD, Wong G. 2003. Dopaminergic neuro-nal loss and motor deficits in Caenorhabditis elegansoverexpressing human a-synuclein. J Neurochem 86:165–172.

Larsen KE, Schmitz Y, Troyer M, Mosharov E, Dietrich P, Sa-valle M, Quazi AZ, Chaudhry FA, Edwards R, Stefanis L,et al. 2006. a-Synuclein overexpression in PC12 andchromaffin cells impairs catecholamine release by inter-fering with a late step in exocytosis. J Neurosci 26:11915–11922.

Lee HJ, Shin SY, Choi C, Lee YH, Lee SJ. 2002. Formationand removal of a-synuclein aggregates in cells exposedto mitochondrial inhibitors. J Biol Chem 277: 5411–5417.

Lee HJ, Khoshaghideh F, Patel S, Lee SJ. 2004. Clearance ofa-synuclein oligomeric intermediates via the lysosomaldegradation pathway. J Neurosci 24: 1888–1896.

Lee HJ, Patel S, Lee SJ. 2005. Intravesicular localization andexocytosis of a-synuclein and its aggregates. J Neurosci25: 6016–6024.

Lee HJ, Khoshaghideh F, Lee S, Lee SJ. 2006. Impairment ofmicrotubule-dependent trafficking by overexpression ofa-synuclein. Eur J Neurosci 24: 3153–3162.

Lee HJ, Suk JE, Patrick C, Bae EJ, Cho JH, Rho S, Hwang D,Masliah E, Lee SJ. 2010. Direct transfer of a-synucleinfrom neuron to astroglia causes inflammatory responsesin synucleinopathies. J Biol Chem 285: 9262–9272.

Lee KW, Chen W, Junn E, Im JY, Grosso H, Sonsalla PK, FengX, Ray N, Fernandez JR, Chao Y, et al. 2011. Enhancedphosphatase activity attenuates a-synucleinopathy in amouse model. J Neurosci 31: 6963–6971.

Leong SL, Pham CL, Galatis D, Fodero-Tavoletti MT, PerezK, Hill AF, Masters CL, Ali FE, Barnham KJ, Cappai R.2009. Formation of dopamine-mediated a-synuclein-soluble oligomers requires methionine oxidation. FreeRadical Bio Med 46: 1328–1337.

Li W, West N, Colla E, Pletnikova O, Troncoso JC, Marsh L,Dawson TM, Jakala P, Hartmann T, Price DL, et al. 2005.Aggregation promoting C-terminal truncation of a-syn-uclein is a normal cellular process and is enhanced by thefamilial Parkinson’s disease-linked mutations. Proc NatlAcad Sci 102: 2162–2167.

Li WW, Yang R, Guo JC, Ren HM, Zha XL, Cheng JS, Cai DF.2007. Localization of a-synuclein to mitochondriawithin midbrain of mice. Neuroreport 18: 1543–1546.

Lin X, Parisiadou L, Gu XL, Wang L, Shim H, Sun L, Xie C,Long CX, Yang WJ, Ding J, et al. 2009. Leucine-rich re-peat kinase 2 regulates the progression of neuropathology

L. Stefanis

18 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

induced by Parkinson’s-disease-related mutant a-synu-clein. Neuron 64: 807–827.

Lindersson E, Beedholm R, Højrup P, Moos T, Gai W, HendilKB, Jensen PH. 2004. Proteasomal inhibition by alpha-synuclein filaments and oligomers. J Biol Chem 279:12924–12934.

Linnertz C, Saucier L, Ge D, Cronin KD, Burke JR, Brown-dyke JN, Hulette CM, Welsh-Bohmer KA, Chiba-Falek O.2009. Genetic regulation of a-synuclein mRNA expres-sion in various human brain tissues. PLoS One 4: e7480.

Liu F, Nguyen JL, Hulleman JD, Li L, Rochet JC. 2008. Mech-anisms of DJ-1 neuroprotection in a cellular model ofParkinson’s disease. J Neurochem 105: 2435–2453.

Liu G, Zhang C, Yin J, Li X, Cheng F, Li Y, Yang H, Ueda K,Chan P, Yu S. 2009.a-Synuclein is differentially expressedin mitochondria from different rat brain regions anddose-dependently down-regulates complex I activity.Neurosci Lett 454: 187–192.

Liu Z, Yu Y, Li X, Ross CA, Smith WW. 2011. Curcumin pro-tects against A53T a-synuclein-induced toxicity in aPC12 inducible cell model for Parkinsonism. PharmacolRes 63: 439–444.

Lo Bianco C, Ridet JL, Schneider BL, Deglon N, Aebischer P.2002. a-Synucleinopathy and selective dopaminergicneuron loss in a rat lentiviral-based model of Parkinson’sdisease. Proc Natl Acad Sci 99: 10813–10818.

Loeb V, Yakunin E, Saada A, Sharon R. 2010. The transgenicoverexpression of a-synuclein and not its related pathol-ogy associates with complex I inhibition. J Biol Chem 285:7334–7343.

Luk KC, Song C, O’Brien P, Stieber A, Branch JR, BrundenKR, Trojanowski JQ, Lee VM. 2009. Exogenous a-synu-clein fibrils seed the formation of Lewy body-like intra-cellular inclusions in cultured cells. Proc Natl Acad Sci106: 20051–20056.

Mak SK, McCormack AL, Manning-Bog AB, Cuervo AM,Di Monte DA. 2010. Lysosomal degradation of a-synu-clein in vivo. J Biol Chem 285: 13621–13629.

Manning-Bog AB, Schule B, Langston JW. 2009. a-Synu-clein-glucocerebrosidase interactions in pharmacologicalGaucher models: A biological link between Gaucher dis-ease and parkinsonism. Neurotoxicology 30: 1127–1132.

Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioan-nidis JP, Kruger R, Rocca WA, Schneider NK, Lesnick TG,Lincoln SJ, et al. 2006. (GEO-PD) Consortium. Collabo-rative analysis of a-synuclein gene promoter variabilityand Parkinson disease. JAMA 296: 661–670.

Markopoulou K, Wszolek ZK, Pfeiffer RF, Chase BA. 1999.Reduced expression of the G209A a-synuclein allele infamilial Parkinsonism. Ann Neurol 46: 374–381.

Maroteaux L, Scheller RH. 1991. The rat brain synucleins;family of proteins transiently associated with neuronalmembrane. Brain Res Mol Brain Res 11: 335–343.

Maroteaux L, Campanelli JT, Scheller RH. 1988. Synuclein:A neuron-specific protein localized to the nucleus andpresynaptic nerve terminal. J Neurosci 8: 2804–2815.

Martin LJ, Pan Y, Price AC, Sterling W, Copeland NG, Jen-kins NA, Price DL, Lee MK. 2006. Parkinson’s diseasea-synuclein transgenic mice develop neuronal mito-chondrial degeneration and cell death. J Neurosci 26:41–50.

Martın-Clemente B, Alvarez-Castelao B, Mayo I, Sierra AB,Dıaz V, Milan M, Farinas I, Gomez-Isla T, Ferrer I, Cas-tano JG. 2004. a-Synuclein expression levels do not sig-nificantly affect proteasome function and expression inmice and stably transfected PC12 cell lines. J Biol Chem279: 52984–52990.

Martinez-Vicente M, Talloczy Z, Kaushik S, Massey AC,Mazzulli J, Mosharov EV, Hodara R, Fredenburg R, WuDC, Follenzi A, et al. 2008. Dopamine-modified alpha-synuclein blocks chaperone-mediated autophagy. J ClinInvest 118: 777–788.

Masliah E, Rockenstein E, Adame A, Alford M, Crews L, Ha-shimoto M, Seubert P, Lee M, Goldstein J, Chilcote T,et al. 2005. Effects of a-synuclein immunization in amouse model of Parkinson’s disease. Neuron 46: 857–868.

Masliah E, Rockenstein E, Mante M, Crews L, Spencer B,Adame A, Patrick C, Trejo M, Ubhi K, Rohn TT, et al.2011. Passive immunization reduces behavioral andneuropathological deficits in an a-synuclein transgenicmodel of Lewy body disease. PLoS One 6: e19338.

Mata IF, Shi M, Agarwal P, Chung KA, Edwards KL, FactorSA, Galasko DR, Ginghina C, Griffith A, Higgins DS,et al. 2010. SNCA variant associated with Parkinson dis-ease and plasmaa-synuclein level. Arch Neurol 67: 1350–1356.

Matsumoto L, Takuma H, Tamaoka A, Kurisaki H, Date H,Tsuji S, Iwata A. 2010. CpG demethylation enhancesa-synuclein expression and affects the pathogenesis ofParkinson’s disease. PLoS One 5: e15522.

Mazzulli JR, Mishizen AJ, Giasson BI, Lynch DR, ThomasSA, Nakashima A, Nagatsu T, Ota A, Ischiropoulos H.2006. Cytosolic catechols inhibita-synuclein aggregationand facilitate the formation of intracellular soluble oligo-meric intermediates. J Neurosci 26: 10068–10078.

Mazzulli JR, Xu YH, Sun Y, Knight AL, McLean PJ, CaldwellGA, Sidransky E, Grabowski GA, Krainc D. 2011.Gaucher disease glucocerebrosidase and a-synucleinform a bidirectional pathogenic loop in synucleinopa-thies. Cell 146: 37–52.

Mbefo MK, Paleologou KE, Boucharaba A, Oueslati A,Schell H, Fournier M, Olschewski D, Yin G, ZweckstetterM, Masliah E, et al. 2010. Phosphorylation of synucleinsby members of the Polo-like kinase family. J Biol Chem285: 2807–2822.

McCarthy JJ, Linnertz C, Saucier L, Burke JR, Hulette CM,Welsh-Bohmer KA, Chiba-Falek O. 2011. The effect ofSNCA 30 region on the levels of SNCA-112 splicing var-iant. Neurogenetics 12: 59–64.

McCormack AL, Mak SK, Henderson JM, Bumcrot D, Far-rer MJ, Di Monte DA. 2010. a-Synuclein suppression bytargeted small interfering RNA in the primate substantianigra. PLoS One 5: e12122.

Miller DW, Hague SM, Clarimon J, Baptista M, Gwinn-Hardy K, Cookson MR, Singleton AB. 2004. a-Synucleinin blood and brain from familial Parkinson disease withSNCA locus triplication. Neurology 62: 1835–1838.

Mirzaei H, Schieler JL, Rochet JC, Regnier F. 2006. Identifi-cation of rotenone-induced modifications ina-synucleinusing affinity pull-down and tandem mass spectrometry.Anal Chem 78: 2422–2431.

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 19

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Mishizen-Eberz AJ, Guttmann RP, Giasson BI, Day GA, Ho-dara R, Ischiropoulos H, Lee VM, Trojanowski JQ, LynchDR. 2003. Distinct cleavage patterns of normal andpathologic forms of a-synuclein by calpain I in vitro. JNeurochem 86: 836–847.

Mishizen-Eberz AJ, Norris EH, Giasson BI, Hodara R, Is-chiropoulos H, Lee VM, Trojanowski JQ, Lynch DR.2005. Cleavage of a-synuclein by calpain: Potential rolein degradation of fibrillized and nitrated species ofa-synuclein. Biochemistry 44: 7818–7829.

Mizuta I, Satake W, Nakabayashi Y, Ito C, Suzuki S, MomoseY, Nagai Y, Oka A, Inoko H, Fukae J, et al. 2006. Multiplecandidate gene analysis identifies a-synuclein as a sus-ceptibility gene for sporadic Parkinson’s disease. HumMol Genet 15: 1151–1158.

Mollenhauer B, Cullen V, Kahn I, Krastins B, Outeiro TF, Pe-pivani I, Ng J, Schulz-Schaeffer W, Kretzschmar HA,McLean PJ, et al. 2008. Direct quantification of CSFa-synuclein by ELISA and first cross-sectional studyin patients with neurodegeneration. Exp Neurol 213:315–325.

Mollenhauer B, Locascio JJ, Schulz-Schaeffer W, Sixel-Doring F, Trenkwalder C, Schlossmacher MG. 2011.a-Synuclein and tau concentrations in cerebrospinalfluid of patients presenting with parkinsonism: A cohortstudy. Lancet Neurol 10: 230–240.

Mori F, Tanji K, Yoshimoto M, Takahashi H, Wakabayashi K.2002a. Demonstration of a-synuclein immunoreactivityin neuronal and glial cytoplasm in normal human braintissue using proteinase K and formic acid pretreatment.Exp Neurol 176: 98–104.

Mori F, Tanji K, Yoshimoto M, Takahashi H, Wakabayashi K.2002b. Immunohistochemical comparison of a- and b-synuclein in adult rat central nervous system. Brain Res941: 118–126.

Mosharov EV, Staal RG, Bove J, Prou D, Hananiya A, MarkovD, Poulsen N, Larsen KE, Moore CM, Troyer MD, et al.2006. a-Synuclein overexpression increases cytosolic cat-echolamine concentration. J Neurosci 26: 9304–9311.

Mosharov EV, Larsen KE, Kanter E, Phillips KA, Wilson K,Schmitz Y, Krantz DE, Kobayashi K, Edwards RH, SulzerD. 2009. Interplay between cytosolic dopamine, calcium,anda-synuclein causes selective death of substantia nigraneurons. Neuron 62: 218–229.

� Mosley RL, Hutter-Saunders JA, Stone DK, Gendelman HE.2012. Inflammation and adaptive immunity in Parkin-son’s disease. Cold Spring Harb Perspect Med (in press).

Mueller JC, Fuchs J, Hofer A, Zimprich A, Lichtner P, Illig T,Berg D, Wullner U, Meitinger T, Gasser T. 2005. Multipleregions of a-synuclein are associated with Parkinson’sdisease. Ann Neurol 57: 535–541.

Murphy DD, Rueter SM, Trojanowski JQ, Lee VM. 2000.Synucleins are developmentally expressed, and a-synu-clein regulates the size of the presynaptic vesicular poolin primary hippocampal neurons. J Neurosci 20: 3214–3220.

Murray IV, Giasson BI, Quinn SM, Koppaka V, Axelsen PH,Ischiropoulos H, Trojanowski JQ, Lee VM. 2003. Role ofa-synuclein carboxy-terminus on fibril formation in vi-tro. Biochemistry 42: 8530–8540.

Nakamura K, Nemani VM, Wallender EK, Kaehlcke K, OttM, Edwards RH. 2008. Optical reporters for the confor-

mation of alpha-synuclein reveal a specific interactionwith mitochondria. J Neurosci 28: 12305–12317.

Nakamura K, Nemani VM, Azarbal F, Skibinski G, Levy JM,Egami K, Munishkina L, Zhang J, Gardner B, Wakabaya-shi J, et al. 2011. Direct membrane association drives mi-tochondrial fission by the Parkinson disease-associatedprotein a-synuclein. J Biol Chem 286: 20710–20726.

Nakayama K, Suzuki Y, Yazawa I. 2009. Microtubule depoly-merization suppresses a-synuclein accumulation in amouse model of multiple system atrophy. Am J Pathol174: 1471–1480.

Nalls MA, Plagnol V, Hernandez DG, Sharma M, SheerinUM, Saad M, Simon-Sanchez J, Schulte C, Lesage S,Sveinbjornsdottir S, et al. 2011. Imputation of sequencevariants for identification of genetic risks for Parkinson’sdisease: A meta-analysis of genome-wide associationstudies. Lancet 377: 641–649.

Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK,Chaudhry FA, Nicoll RA, Edwards RH. 2010. Increasedexpression of a-synuclein reduces neurotransmitter re-lease by inhibiting synaptic vesicle reclustering after en-docytosis. Neuron 65: 66–79.

Nonaka T, Watanabe ST, Iwatsubo T, Hasegawa M. 2010.Seeded aggregation and toxicity of a-synuclein and tau:Cellular models of neurodegenerative diseases. J BiolChem 285: 34885–34898.

Ohtake H, Limprasert P, Fan Y, Onodera O, Kakita A, Taka-hashi H, Bonner LT, Tsuang DW, Murray IV, Lee VM, et al.2004. b-Synuclein gene alterations in dementia withLewy bodies. Neurology 63: 805–811.

Ostrerova N, Petrucelli L, Farrer M, Mehta N, Choi P, HardyJ, Wolozin B. 1999.a-Synuclein shares physical and func-tional homology with 14–3–3 proteins. J Neurosci 19:5782–5791.

Ostrerova-Golts N, Petrucelli L, Hardy J, Lee JM, Farer M,Wolozin B. 2000. The A53T a-synuclein mutation in-creases iron-dependent aggregation and toxicity. J Neuro-sci 20: 6048–6054.

Outeiro TF, Kontopoulos E, Altmann SM, Kufareva I,Strathearn KE, Amore AM, Volk CB, Maxwell MM, Ro-chet JC, McLean PJ, et al. 2007. Sirtuin 2 inhibitors rescuea-synuclein-mediated toxicity in models of Parkinson’sdisease. Science 317: 516–519.

Paillusson S, Tasselli M, Lebouvier T, Mahe M, Chevalier J,Biraud M, Cario-Toumaniantz C, Neunlist M, Derkinde-ren P. 2010.a-Synuclein expression is induced by depola-rization and cyclic AMP in enteric neurons. J Neurochem115: 694–706.

Paleologou KE, Schmid AW, Rospigliosi CC, Kim HY, Lam-berto GR, Fredenburg RA, Lansbury PT, Fernandez CO,Eliezer D, Zweckstetter M, et al. 2008. Phosphorylationat Ser-129 but not the phosphomimics S129E/D inhibitsthe fibrillation of a-synuclein. J Biol Chem 283: 16895–16905.

Paleologou KE, Kragh CL, Mann DM, Salem SA, Al-ShamiR, Allsop D, Hassan AH, Jensen PH, El-Agnaf OM. 2009.Detection of elevated levels of soluble a-synucleinoligomers in post-mortem brain extracts from patientswith dementia with Lewy bodies. Brain 132 (Pt 4):1093–1101.

Paleologou KE, Oueslati A, Shakked G, Rospigliosi CC, KimHY, Lamberto GR, Fernandez CO, Schmid A, Chegini

L. Stefanis

20 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

F, Gai WP, et al. 2010. Phsophorylation at S87 is enhancedin synucleinopathies, nhibitsa-synuclein oligomerizationand influences synuclein-membrane interactions. J Neu-rosci 30: 3184–3198.

Pan-Montojo F, Anichtchik O, Dening Y, Knels L, Pursche S,Jung R, Jackson S, Gille G, Spillantini MG, Reichmann H,et al. 2010. Progression of Parkinson’s disease pathologyis reproduced by intragastric administration of rotenonein mice. PLoS One 5: e8762.

Park JY, Lansbury PT Jr. 2003. b-Synuclein inhibits forma-tion of a-synuclein protofibrils: A possible therapeuticstrategy against Parkinson’s disease. Biochemistry 42:3696–3700.

Paxinou E, Chen Q, Weisse M, Giasson BI, Norris EH, Rue-ter SM, Trojanowski JQ, Lee VM, Ischiropoulos H. 2001.Induction of a-synuclein aggregation by intracellular ni-trative insult. J Neurosci 21: 8053–8061.

Peng XM, Tehranian R, Dietrich P, Stefanis L, Perez R. 2005.a-Synuclein activation of protein phosphatase 2A re-duces tyrosine hydroxylase phosphorylation in dopami-nergic cells. J Cell Sci 118 (Pt 15): 3523–3530.

Perez RG, Waymire JC, Lin E, Liu JJ, Guo F, Zigmond MJ.2002. A role for a-synuclein in the regulation of dopa-mine biosynthesis. J Neurosci 22: 3090–3099.

Periquet M, Fulga T, Myllykangas L, Schlossmacher MG,Feany MB. 2007. Aggregated a-synuclein mediates dop-aminergic neurotoxicity in vivo. J Neurosci 27: 3338–3346.

Perrin RJ, Woods WS, Clayton DF, George JM. 2001. Expo-sure to long chain polyunsaturated fatty acids triggersrapid multimerization of synucleins. J Biol Chem 276:41958–41962.

Petrucelli L, O’Farrell C, Lockhart PJ, Baptista M, Kehoe K,Vink L, Choi P, Wolozin B, Farrer M, Hardy J, et al. 2002.Parkin protects against the toxicity associated with mu-tant a-synuclein: Proteasome dysfunction selectively af-fects catecholaminergic neurons. Neuron 36: 1007–1019.

Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, DehejiaA, Dutra A, Pike B, Root H, Rubenstein J, Boyer R,et al. 1997. Mutation in the a-synuclein gene identifiedin families with Parkinson’s disease. Science 276: 2045–2047.

Qin Z, Hu D, Han S, Reaney SH, Di Monte DA, Fink AL.2007. Effect of 4-hydroxy-2-nonenal modification ona-synuclein aggregation. J Biol Chem 282: 5862–5870.

Qureshi HY, Paudel HK. 2011. Parkinsonian neurotoxin1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)and a-synuclein mutations promote tprotein phosphor-ylation at Ser262 and destabilize microtubule cytoskele-ton in vitro. J Biol Chem 286: 5055–5068.

Rideout HJ, Dietrich P, Savalle M, Dauer WT, Stefanis L.2003. Regulation ofa-synuclein by bFGF in cultured ven-tral midbrain dopaminergic neurons. J Neurochem 84:803–813.

Rideout HJ, Dietrich P, Wang QH, Dauer WT, Stefanis L.2004. a-Synuclein is required for the fibrillar nature ofubiquitinated inclusions induced by proteasomal inhi-bition in primary neurons. J Biol Chem 279: 46915–46920.

Robertson DC, Schmidt O, Ninkina N, Jones PA, Sharkey J,Buchman VL. 2004. Developmental loss and resistance toMPTP toxicity of dopaminergic neurones in substantianigra pars compacta of g-synuclein, a-synuclein and

double a/g-synuclein null mutant mice. J Neurochem89: 1126–1136.

Schell H, Hasegawa T, Neumann M, Kahle PJ. 2009. Nuclearand neritic distribution of serine-129 phosphorylateda-synuclein in transgenic mice. Neuroscience 160: 796–804.

Scherzer CR, Grass JA, Liao Z, Pepivani I, Zheng B, EklundAC, Ney PA, Ng J, McGoldrick M, Mollenhauer B, et al.2008. GATA transcription factors directly regulate theParkinson’s disease-linked gene a-synuclein. Proc NatlAcad Sci 105: 10907–10912.

Scholz SW, Houlden H, Schulte C, Sharma M, Li A, Berg D,Melchers A, Paudel R, Gibbs JR, Simon-Sanchez J,et al. 2009. SNCA variants are associated with increasedrisk for multiple system atrophy. Ann Neurol 65:610–614.

Scott DA, Tabarean I, Tang Y, Cartier A, Masliah E, Roy S.2010. A pathologic cascade leading to synaptic dysfunc-tion in a-synuclein-induced neurodegeneration. J Neu-rosci 30: 8083–8095.

Sharon R, Bar-Joseph I, Frosch MP, Walsh DM, HamiltonJA, Selkoe DJ. 2003. The formation of highly solubleoligomers of a-synuclein is regulated by fatty acidsand enhanced in Parkinson’s disease. Neuron 37: 583–595.

Shimshek DR, Mueller M, Wiessner C, Schweizer T, van derPutten PH. 2010. The HSP70 molecular chaperone is notbeneficial in a mouse model of a-synucleinopathy. PLoSOne 5: e10014.

Singleton AB, Farrer M, Johnson J, Singleton A, Hague S,Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nuss-baum R, et al. 2003. a-Synuclein locus triplication causesParkinson’s disease. Science 302: 841.

Smith WW, Jiang H, Pei Z, Tanaka Y, Morita H, Sawa A,Dawson VL, Dawson TM, Ross CA. 2005. Endoplasmicreticulum stress and mitochondrial cell death pathwaysmediate A53T mutant alpha-synuclein-induced toxicity.Hum Mol Genet 14: 3801–3811.

Smith WW, Liu Z, Liang Y, Masuda N, Swing DA, JenkinsNA, Copeland NG, Troncoso JC, Pletnikov M, DawsonTM, et al. 2010. Synphilin-1 attenuates neuronal degen-eration in the A53T a-synuclein transgenic mousemodel. Hum Mol Genet 19: 2087–2098.

Snyder H, Mensah K, Theisler C, Lee J, Matouschek A, Wo-lozin B. 2003. Aggregated and monomeric a-synucleinbind to the S6’ proteasomal protein and inhibit proteaso-mal function. J Biol Chem 278: 11753–11759.

Sousa VL, Bellani S, Giannandrea M, Yousuf M, Valtorta F,Meldolesi J, Chieregatti E. 2009. a-Synuclein and itsA30P mutant affect actin cytoskeletal structure and dy-namics. Mol Biol Cell 20: 3725–3739.

Spencer B, Potkar R, Trejo M, Rockenstein E, Patrick C,Gindi R, Adame A, Wyss-Coray T, Masliah E. 2009. Beclin1 gene transfer activates autophagy and amelioratesthe neurodegenerative pathology in a-synuclein modelsof Parkinson’s and Lewy body diseases. J Neurosci 29:13578–13588.

Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ,Jakes R, Goedert M. 1997. a-Synuclein in Lewy bodies.Nature 388: 839–840.

Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goe-dert M. 1998. a-Synuclein in filamentous inclusions of

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 21

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

Lewy bodies from Parkinson’s disease and dementia withLewy bodies. Proc Natl Acad Sci 95: 6469–6473.

Stefanis L, Larsen KE, Rideout HJ, Sulzer D, Greene LA.2001. Expression of A53T mutant but not wild-typea-synuclein in PC12 cells induces alterations of theubiquitin-dependent degradation system, loss of dopa-mine release, and autophagic cell death. J Neurosci 21:9549–9560.

Stefanis L, Wang Q, Oo T, Lang-Rollin I, Burke RE, DauerWT. 2004. Lack of a-synuclein does not alter apoptosisof neonatal catecholaminergic neurons. Eur J Neurosci20: 1969–1972.

Tanaka Y, Engelender S, Igarashi S, Rao RK, Wanner T, TanziRE, Sawa A, Dawson V, Dawson TM, Ross CA. 2001.Inducible expression of mutant a-synuclein decreasesproteasome activity and increases sensitivity to mito-chondria-dependent apoptosis. Hum Mol Genet 10:919–926.

Thayanidhi N, Helm JR, Nycz DC, Bentely M, Liang Y, HayJC. 2010. a-Synuclein delays endoplasmic reticulum(ER)-to-Golgi transport in mammalian cells byantagonizing ER/Golgi SNAREs. Mol Biol Cell 21:1850–1863.

Tofaris GK, Layfield R, Spillantini MG. 2001. alpha-synu-clein metabolism and aggregation is linked to ubiqui-tin-independent degradation by the proteasome. FEBSLett 509: 22–26.

Tokuda T, Salem SA, Allsop D, Mizuno T, Nakagawa M, Qur-eshi MM, Locascio JJ, Schlossmacher MG, El-Agnaf OM.2006. Decreased a-synuclein in cerebrospinal fluid ofaged individuals and subjects with Parkinson’s disease.Biochem Biophys Res Commun 349: 162–166.

Tokuda T, Qureshi MM, Ardah MT, Varghese S, Shehab SA,Kasai T, Ishigami N, Tamaoka A, Nakagawa M, El-AgnafOM. 2010. Detection of elevated levels of a-synucleinoligomers in CSF from patients with Parkinson disease.Neurology 75: 1766–1772.

Tong J, Wong H, Guttman M, Ang LC, Forno LS, ShimadzuM, Rajput AH, Muenter MD, Kish SJ, Hornykiewicz O,et al. 2010. Brain a-synuclein accumulation in multiplesystem atrophy, Parkinson’s disease and progressivesupranuclear palsy: A comparative investigation. Brain133 (Pt 1): 172–188.

Tsika E, Moysidou M, Guo J, Cushman M, Gannon P, San-daltzopoulos R, Giasson BI, Krainc D, Ischiropoulos H,Mazzulli JR. 2010. Distinct region-specific a-synucleinoligomers in A53T transgenic mice: Implications forneurodegeneration. J Neurosci 30: 3409–3418.

Ueda K, Fukushima H, Masliah E, Xia Y, Iwai A, YoshimotoM, Otero DA, Kondo J, Ihara Y, Saitoh T. 1993. Molecularcloning of cDNA encoding an unrecognized componentof amyloid in Alzheimer disease. Proc Natl Acad Sci 90:11282–11286.

Uversky VN, Yamin G, Munishkina LA, Karymov MA, Mil-lett IS, Doniach S, Lyubchenko YL, Fink AL. 2005. Effectsof nitration on the structure and aggregation ofa-synuclein. Brain Res Mol Brain Res 134: 84–102.

Vekrellis K, Xilouri M, Emmanouilidou E, Stefanis L. 2009.Inducible over-expression of wild typea-synuclein in hu-man neuronal cells leads to caspase-dependent non-apoptotic death. J Neurochem 109: 1348–1362.

Vila M, Vukosavic S, Jackson-Lewis V, Neystat M, JakowecM, Przedborski S. 2000. a-Synuclein up-regulation insubstantia nigra dopaminergic neurons following ad-ministration of the parkinsonian toxin MPTP. J Neuro-chem 74: 721–729.

Vogiatzi T, Xilouri M, Vekrellis K, Stefanis L. 2008. Wild typea-synuclein is degraded by chaperone-mediated autoph-agy and macroautophagy in neuronal cells. J Biol Chem283: 23542–23556.

Volles MJ, Lansbury PT Jr. 2002. Vesicle permeabilization byprotofibrillar a-synuclein is sensitive to Parkinson’sdisease-linked mutations and occurs by a pore-like mech-anism. Biochemistry 41: 4595–4602.

Voutsinas GE, Stavrou EF, Karousos G, Dasoula A, Papa-chatzopoulou A, Syrrou M, Verkerk AJ, van der Spek P,Patrinos GP, Stoger R, et al. 2010. Allelic imbalance ofexpression and epigenetic regulation within the a-synu-clein wild-type and p.Ala53Thr alleles in Parkinson dis-ease. Hum Mutat 31: 685–691.

Wassef R, Haenold R, Hansel A, Brot N, HeinemannSH, Hoshi T. 2007. Methionine sulfoxide reductase Aand a dietary supplement S-methyl-L-cysteine pre-vent Parkinson’s-like symptoms. J Neurosci 27: 12808–12816.

Waxman EA, Giasson BI. 2010. A novel, high-efficiency cel-lular model of fibrillara-synuclein inclusions and the ex-amination of mutations that inhibit amyloid formation.J Neurochem 113: 374–388.

Webb JL, Ravikumar B, Atkins J, Skepper JN, RubinszteinDC. 2003. a-Synuclein is degraded by both autophagyand the proteasome. J Biol Chem 278: 25009–25013.

Wersinger C, Sidhu A. 2003. Attenuation of dopaminetransporter activity by a-synuclein. Neurosci Lett 340:189–192.

Wersinger C, Prou D, Vernier P, Sidhu A. 2003. Modulationof dopamine transporter function by a-synuclein is al-tered by impairment of cell adhesion and by inductionof oxidative stress. FASEB J 17: 2151–2153.

Willingham S, Outeiro TF, DeVit MJ, Lindquist SL, Mu-chowski PJ. 2003. Yeast genes that enhance the toxicityof a mutant huntingtin fragment or alpha-synuclein. Sci-ence 302: 1769–1772.

Winner B, Jappelli R, Maji SK, Desplats PA, Boyer L, AignerS, Hetzer C, Loher T, Vilar M, Campioni S, et al. 2011. Invivo demonstration that a-synuclein oligomers are toxic.Proc Natl Acad Sci 108: 4194–4199.

Winslow AR, Chen CW, Corrochano S, Acevedo-Arozena A,Gordon DE, Peden AA, Lichtenberg M, Menzies FM,Ravikumar B, Imarisio S, et al. 2010.a-Synuclein impairsmacroautophagy: Implications for Parkinson’s disease. JCell Biol 190: 1023–1037.

Withers GS, George JM, Banker GA, Clayton DF. 1997. De-layed localization of synelfin (synuclein, NACP) to pre-synaptic terminals in cultured rat hippocampal neurons.Brain Res Dev Brain Res 99: 87–94.

Xilouri M, Vogiatzi T, Vekrellis K, Stefanis L. 2008.a-Synuclein degradation by autophagic pathways: A po-tential key to Parkinson’s disease pathogenesis. Autoph-agy 4: 917–919.

Xilouri M, Vogiatzi T, Vekrellis K, Park D, Stefanis L. 2009.Abberant a-synuclein confers toxicity to neurons in

L. Stefanis

22 Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

part through inhibition of chaperone-mediated autoph-agy. PLoS One 4: e5515.

Xu YH, Sun Y, Ran H, Quinn B, Witte D, Grabowski GA.2011. Accumulation and distribution of a-synucleinand ubiquitin in the CNS of Gaucher disease mousemodels. Mol Genet Metab 102: 436–447.

Yu S, Li X, Liu G, Han J, Zhang C, Li Y, Xu S, Liu C, Gao Y,Yang H, et al. 2007. Extensive nuclear localization ofa-synuclein in normal rat brain neurons revealed by anovel monoclonal antibody. Neuroscience 145: 539–555.

Zarranz JJ, Alegre J, Gomez-Esteban JC, Lezcano E, Ros R,Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atares B,et al. 2004. The new mutation, E46K, of a-synucleincauses Parkinson and Lewy body dementia. Ann Neurol55: 164–173.

Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, WilsonB, Zhang W, Zhou Y, Hong JS, et al. 2005. Aggregateda-synuclein activates microglia: A process leading to dis-ease progression in Parkinson’s disease. FASEB J 19:533–542.

Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, WattML, Eklund AC, Zhang-James Y, Kim PD, Hauser MA,et al. 2010. PGC-1a, a potential therapeutic target forearly intervention in Parkinson’s disease. Sci Transl Med2: 52ra73.

Zhou RM, Huang YX, Li XL, Chen C, Shi Q, Wang GR, TianC, Wang ZY, Jing YY, Gao C, et al. 2010. Molecular inter-action of a-synuclein with tubulin influences on thepolymerization of microtubule in vitro and structure ofmicrotubule in cells. Mol Biol Rep 37: 3183–3192.

a-Synuclein in Parkinson’s Disease

Cite this article as Cold Spring Harb Perspect Med 2012;4:a009399 23

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from

December 13, 20112012; doi: 10.1101/cshperspect.a009399 originally published onlineCold Spring Harb Perspect Med 

 Leonidas Stefanis 

-Synuclein in Parkinson's Diseaseα

Subject Collection Parkinson's Disease

Functional Neuroanatomy of the Basal Ganglia

ObesoJosé L. Lanciego, Natasha Luquin and José A. Dysfunction in Parkinson's Disease

as a Model to Study MitochondrialDrosophila

Ming Guo

GeneticsAnimal Models of Parkinson's Disease: Vertebrate

DawsonYunjong Lee, Valina L. Dawson and Ted M. Pathways

and DJ-1 and Oxidative Stress and Mitochondrial Parkinsonism Due to Mutations in PINK1, Parkin,

Mark R. CooksonInnate Inflammation in Parkinson's Disease

V. Hugh PerryProgrammed Cell Death in Parkinson's Disease

Katerina Venderova and David S. Park

NeuropathologyParkinson's Disease and Parkinsonism:

Dennis W. DicksonDiseaseGenomics and Bioinformatics of Parkinson's

al.Sonja W. Scholz, Tim Mhyre, Habtom Ressom, et

Parkinson's DiseasePhysiological Phenotype and Vulnerability in

Sanchez, et al.D. James Surmeier, Jaime N. Guzman, Javier

DiseaseMotor Control Abnormalities in Parkinson's

CortésPietro Mazzoni, Britne Shabbott and Juan Camilo

ManagementFeatures, Diagnosis, and Principles of Clinical Approach to Parkinson's Disease:

João Massano and Kailash P. Bhatia

Parkinson's Disease: Gene Therapies

Patrick AebischerPhilippe G. Coune, Bernard L. Schneider and

The Role of Autophagy in Parkinson's DiseaseMelinda A. Lynch-Day, Kai Mao, Ke Wang, et al.

Functional Neuroimaging in Parkinson's Disease

EidelbergMartin Niethammer, Andrew Feigin and David

Parkinson's DiseaseDisruption of Protein Quality Control in

PetrucelliCasey Cook, Caroline Stetler and Leonard

Key QuestionsLeucine-Rich Repeat Kinase 2 for Beginners: Six

Lauren R. Kett and William T. Dauer

http://perspectivesinmedicine.cshlp.org/cgi/collection/ For additional articles in this collection, see

Copyright © 2012 Cold Spring Harbor Laboratory Press; all rights reserved

Press on February 24, 2019 - Published by Cold Spring Harbor Laboratoryhttp://perspectivesinmedicine.cshlp.org/Downloaded from