Stefanis - 2012 - α-Synuclein in Parkinson's Disease-annotated

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

Downloaded from http://perspectivesinmedicine.cshlp.

org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

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: lstefanis@bioacademy.gr

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.
www.perspectivesinmedicine.org

he first link of Parkinson’s disease (PD) to a- A parallel story that has developed over the
T synuclein was also the first conclusive demon-
stration of a genetic defect leading to PD, and thus
years is the abundant abnormal a-synuclein
pathology that characterizes neuropathological
has historical and conceptual value. Although specimens derived not only from PD patients,
familial contribution to the disease was discussed but also patients with other neurodegenerative
by some, it was felt to be minor at best by most. conditions, collectively termed “synucleinopa-
The tangible discovery of a gene defect linked to thies.” The ability to model abnormal a-synu-
PD in particular families was a ground-breaking clein accumulation in various cellular and
discovery,asitopenedthedoor toafloodof studies animal models has led to the study of the conse-
investigating the genetic base of the disorder, cul- quences of such accumulation, to the decipher-
minating in more recent genome-wide association ing of the molecular pathways involved, and to
studies (GWAS), that, remarkably, have made a full the identification of potential therapeutic tar-
circle back to the origins of the molecular genetic gets. Such targets may prove of general utility,
era of PD; it turns out that one of the major genes and provide the basis for future therapeutics
identified as linked to sporadic PD is none other in most forms of PD, and, potentially, other syn-
than SNCA, the gene encoding for a-synuclein. 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
Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

GENETICS OF PD AND OTHER tive risk for sporadic PD in initial association


DISORDERS WITH LINKS TO studies, which were replicated and then con-
SYNUCLEINOPATHIES firmed in a large metanalysis (Maraganore
et al. 2006). Others studies found associations
In 1997, Polymeropoulos et al. reported the of PD with 30 regions of the gene (Mueller et al.
first specific genetic aberration linked to PD 2005; Mizuta et al. 2006). Targeted association
in a large Italian kindred, and in some Greek studies are, however, prone to false positives;
familial cases. The mutation corresponded to a therefore, it is very important that the link of
G209A substitution in the SNCA gene encoding SNCA to sporadic PD has been confirmed
for a-synuclein, resulting in an A53T amino through large unbiased GWAS. SNCA, and, in
acid change. The pattern of inheritance was au- particular, the 30 region, has been a consistent
tosomal-dominant, and the disease was early- hit in such studies, and represents, of the links
onset, usually in the 40s. Although there was identified, probably the most robust one (Nalls
some initial skepticism, based on the fact that et al. 2011).
the threonine was the amino acid in position The conclusion from these genetic studies
53 in the rodent SNCA homolog, this was set is that SNCA is linked to both rare familial
aside following the identification of two more and sporadic PD from a genetic standpoint. It
point mutations in SNCA, leading to A30P is notable that the association of SNCAvariants
and E46K amino acid changes, which were as- to disease has now also been extended to multi-
sociated with autosomal-dominant PD (Kruger ple-system atrophy (MSA), another prototypi-
et al. 1998; Zarranz et al. 2004). The next land- cal synucleinopathy, which is invariably spo-
mark study that linked SNCA to PD through a radic (Al-Chalabi et al. 2009; Scholz et al. 2009).
different mechanism identified triplications of
www.perspectivesinmedicine.org

the SNCA locus in separate families with an au-


tosomal-dominant inheritance pattern (Single- THE INVOLVEMENT OF a-SYNUCLEIN IN
ton et al. 2003). This led to a potential total THE NEUROPATHOLOGICAL PICTURE OF
PD AND RELATED DISORDERS
of a duplication of the a-synuclein load in
such patients, and this was confirmed later The genetic link of SNCA to PD in 1997 led
at the protein level (Miller et al. 2004). Other investigators to rapidly develop appropriate an-
families were subsequently described that har- tibodies against a-synuclein and use them in
bored duplications of the gene, which was histopathological sections of PD patients. a-
similarly associated with autosomal-dominant Synuclein turned out to be robustly expressed
disease (Chartier-Harlin et al. 2004). Interest- within Lewy bodies (LBs), and, in particular,
ingly, a gene dosage effect exists, in that triplica- in the “halo” of the inclusions (Spillantini et al.
tion cases are earlier onset and have a more severe 1997, 1998; Baba et al. 1998). In fact, a-synuclein
course compared to the duplication cases (Fuchs immunohistochemistry is currently the gold
et al. 2007). standard in the neuropathological evaluation
When a specific genetic defect is identified in of PD. With repeated studies, three major in-
rare familial cases, it is almost a reflex reaction to sights emerged: (1) a-synuclein pathology in
look at the same gene in the sporadic disease, PD is not confined to the cell soma, but is also
with the hope of identifying polymorphisms prominent in neuritic processes, (2) it is wide-
that may be linked to the disease in association spread in various brain regions in PD, and (3)
studies. This has not been particularly successful it is present in a number of other synucleinopa-
in other diseases, but in the case of PD and the thies, such as MSA, dementia with Lewy bodies
SNCA gene this approach has borne fruit. Initial (DLBs), many cases of Alzheimer’s disease (AD)
studies focused on the Rep1 polymorphic region (the so-called LB variant of AD), neurodegen-
located approximately 10 kB upstream of the eration with brain iron accumulation (NBIA)
SNCA transcriptional initiation site. Particular type I, pure autonomic failure (PAF), and even
polymorphisms in this region conferred a rela- a subtype of essential tremor (reviewed in Kahle

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

2008). The distribution of the pathology at the is hoped, will become more widely applicable
cellular and regional level is different in each and available.
disease.
Braak et al. (2003), in their seminal study of
THE STRUCTURE AND FUNCTION OF
the staging of PD pathology, used a-synuclein
a-SYNUCLEIN
immunohistochemistry in a large number of
autopsy cases. Based essentially on a-synuclein The SNCA gene encodes for a 140 amino acid
neuritic pathology (“Lewy neurites”), they sug- protein, which in aqueous solutions does not
gested a six-stage scheme in which the pathol- have a defined structure, hence the term “na-
ogy begins first at the olfactory bulb and the tively unfolded protein.” a-Synuclein, however,
dorsal vagal nucleus and gradually follows an does form a-helical structures on binding to
ascending course, culminating in widespread negatively charged lipids, such as phospholipids
a-synuclein pathology at the later stages, in- present on cellular membranes, and b-sheet-
volving associative cortical regions. Notably, rich structures on prolonged periods of incu-
substantia nigra is only affected in stage 3 of bation. The protein is composed of three dis-
this scheme. Although there have been crit- tinct regions: (1) an amino terminus (residues
icisms against this hypothesis (Burke et al. 1 – 60), containing apolipoprotein lipid-bind-
2008), and it does not explain the absence of ing motifs, which are predicted to form amphi-
clinical symptoms in subjects who on autopsy philic helices conferring the propensity to form
have widespread a-synuclein pathology, or the a-helical structures on membrane binding, (2)
clinical picture of DLB, it appears to hold up a central hydrophobic region (61 –95), so-called
for the majority, but not all, of cases examined NAC (non-Ab component), which confers the
in large cohorts of LB cases (Dickson et al. b-sheet potential, and (3) a carboxyl terminus
www.perspectivesinmedicine.org

2010). What is important to note here is that that is highly negatively charged, and is prone
overall in the PD brain the majority of ab- to be unstructured (Fig. 1). There are at least
normally deposited a-synuclein is in neuritic two shorter alternatively spliced variants of the
processes. The presence of a-synuclein in LBs SNCA gene transcript, but their physiological
is likely the tip of the iceberg. In fact, Kramer and pathological roles have not been well char-
and Shulz-Schaeffer (2007), using a modified acterized (Maroteaux and Scheller 1991; Ueda
protocol termed protein aggregate filtration, et al. 1993; Maroteaux et al. 1988).
showed in DLB cases a remarkable degree of a-Synuclein is a member of the synuclein
aberrant neuritic a-synuclein pathology, in the family of proteins, which also include b- and
virtual absence of LBs. g-synuclein. What largely sets apart a-synuclein
Overall, it is clear that abnormal a-synu- from the other members structurally is the NAC
clein deposition occurs early in PD. Given region. All three members of the family are
the fact that such pathology may develop sec- predominantly neuronal proteins that under
ondarily, for example, owing to iron mishan- physiological conditions localize preferentially
dling in NBIA type I, we cannot be certain to presynaptic terminals (George 2002). There
that it is a primary factor in the disease process. are some reports of extranigral b- and g-synu-
However, the generally quite defined pattern of clein neuritic pathology in synucleinopathies,
a-synuclein pathology in PD, the cellular and but, if it exists, it does not appear to be wide-
animal models of a-synuclein overexpression spread (Galvin et al. 1999). Interestingly, point
that will be discussed, and, most importantly, mutations in b-synuclein have been described
the combination with the genetic data men- in rare cases with DLB (Ohtake et al. 2004),
tioned above, suggest that this aberrant deposi- and expression of one of these in a transgenic
tion is the driving force in PD pathogenesis. mouse model-induced neurodegeneration (Fu-
Such deposition, as mentioned, may be subtle jita et al. 2010). On the other hand, wild-type
and difficult to detect in the absence of elabo- (WT) b-synuclein has been found to be pro-
rate immunohistochemical techniques that, it tective in various settings against a-synuclein-

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

Apolipoprotein motif

Repeat 1 Repeat 2 Repeat 3 Repeat 4 Repeat 5 Repeat 6


Acidic tail
1 140
N C
61 95

A30P A53T
E46K

Mutations genetically
linked to familial
autosomal-dominant
Parkinson’s disease

NAC region necessary for aggregation

Figure 1. Schemtic structure of a-synuclein.

mediated neurodegeneration (Hashimoto et al. modulated in conditions that alter plasticity or


2001; Park and Lansbury, 2003). It would ap- confer injury (George et al. 1995; Kholodilov
pear therefore that WT b-synuclein may have et al. 1999; Vila et al. 2000). Such data have led
neuroprotective properties, but that, under cer- to the notion that a-synuclein may be a modu-
www.perspectivesinmedicine.org

tain situations, it may assume a neurotoxic po- lator of synaptic transmission. Examinations of
tential. neurophysiological properties of mice null for
a-Synuclein is abundantly expressed in the a-synuclein and of cellular systems in which
nervous system, comprising 1% of total cy- WT a-synuclein is overexpressed have been in-
tosolic protein. On immunohistochemistry in strumental in this regard, as they reveal alter-
normal brains a-synuclein antibodies confer ations in neurotransmitter release, which are
a punctuate neuropil staining pattern, consis- not always consistent across neuronal systems
tent with presynaptic terminal staining, whereas (Abeliovich et al. 2000; Murphy et al. 2000;
neuronal soma staining is less apparent (Kahle Cabin et al., 2002; Larsen et al. 2006). Overall
2008). In presynaptic terminals a-synuclein is though, it would appear that a-synuclein acts
present in close proximity, but not within, syn- as a subtle break for neurotransmitter release
aptic vesicles. In fact a-synuclein is also very under circumstances of repeated firing. An ele-
abundant, for unclear reasons, in erythrocytes gant study by Fortin et al. (2005) showed that
and platelets (Barbour et al. 2008). Whether it fluorescently labeled a-synuclein moves away
is expressed at all under physiological condi- from the vesicles on neuronal firing, and then
tions in glia is somewhat controversial; it would gradually returns. These effects may involve
appear that, if it exists, such expression is pre- transient binding of a-synuclein to the vesicles,
sent at very low levels (Mori et al. 2002a). The whereas a-synuclein may also be involved in ve-
expression of a-synuclein is induced during sicular biogenesis, through effects on phospha-
neuronal development, following determina- tidic acid metabolism, and in compartmenta-
tion of neuronal phenotype and establishment lization between resting and readily releasable
of synaptic connections, and lags behind the pools (Murphy et al. 2000). Consistent with a
induction of proteins involved in synaptic role for a-synuclein at the level of synaptic trans-
structure (Withers et al. 1997; Kholodilov et al. mission, a-synuclein modulates profoundly the
1999; Murphy et al. 2000; Rideout et al. 2003). phenotype of mice null for the presynaptic pro-
Moreover, a-synuclein expression levels are tein CSP-a (cysteine string protein-a), which

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

show synaptic degeneration (Chandra et al. THE AGGREGATION POTENTIAL OF


2005). It is believed that a-synuclein, through a-SYNUCLEIN
a chaperone-like function, may act in synergy
with CSP-a in the assembly of the SNARE com- The ability of a-synuclein to generate b-sheet
plex (Chandra et al. 2005), and this effect may structures under certain circumstances gener-
involve, in particular, binding to synaptotag- ated a lot of enthusiasm, as it provided parallels
min-2 (Burree et al. 2010). to the b-sheet structure of b-amyloid, and con-
Therefore, the main function of a-synuclein sequently, a unified pathogenetic basis for the
would appear to be the control of neurotrans- two most common neurodegenerative diseases.
mitter release, through effects on the SNARE Indeed, WTas well as disease-related mutants of
complex. Recent evidence, to be discussed be- a-synuclein form amyloid-like fibrils on pro-
low, suggests that this physiological function longed incubation in solution (Conway et al.
may provide insight into the aberrant function 2000). It is widely agreed on that such fibrils
unleashed in disease states. form the basis of the mature LBs and Lewy neu-
rites (LNs) present in synucleinopathies. In the
process of fibril formation various intermediate
forms of a-synuclein develop. These are initially
SYNUCLEINOPATHY MODELS
soluble oligomeric forms of a-synuclein that as-
Based on the genetic and pathological data, at- sume spherical, ring-, and stringlike characteris-
tempts have been made to create cellular and tics when seen under the electron microscope.
animal models in which various forms of a- Such structures, collectively termed protofibrils,
synuclein are overexpressed. We have elected gradually become insoluble and coalesce into fi-
to not present such models with any great detail brils. It is difficult to capture these intermediate
www.perspectivesinmedicine.org

here, but rather highlight the inferences gained structures in cells or in vivo, but their biochemi-
from such models regarding the potential cal equivalent isthought to be soluble oligomeric
pathological (and sometimes physiological) ef- forms, some, but not all, of which are SDS-stable
fects of a-synuclein. Suffice it to say here, that and can be detected on SDS/polyacrylamide
there are a number of transgenic mice available, gel electrophoresis (PAGE) gels. Other forms,
in which expression of various forms of a-syn- however, can only be captured on native gels
uclein is achieved through different promoters, or by size-exclusion chromatography (Emma-
allowing in some cases regional and temporal nouilidou et al. 2010a). This cascade of events,
control of expression. In such mice, there is gen- starting from the natively unfolded protein
erally some degree of neurodegneration and LB- and culminating in the mature fibril formation
like pathology, but it does not involve primarily is collectively termed a-synuclein aggregation.
the dopaminergic system. Some mice develop The general consensus in the field is that
behavioral phenotypes that could be akin to aggregation is a main pathogenic feature of a-
presymptomatic PD (reviewed by Chesselet synuclein. This is buttressed by the lack of tox-
2008). An alternative approach has been the icity conferred by WT b-synuclein or by a-syn-
viral-mediated transduction of dopaminergic uclein variants that do not contain the NAC do-
neurons of the substantia nigra of rodents main and do not form aggregates (Periquet et al.
with a-synuclein, and this has been more suc- 2007), by the beneficial effects of overxepressing
cessful in modeling the dopaminergic cell death HSPs, which assist in refolding of aggregation-
aspect of the disease (Kirik et al. 2002). A num- prone proteins, in synucleinopathy models
ber of invertebrate models, including fly (Feany (Auluck and Bonini 2002; Auluck et al. 2002;
and Bender 2000) and Caenorhabditis elegans Klucken et al. 2004), and by the protection con-
(Lasko et al. 2003), have also been created, and ferred in cellular models of a-synuclein overex-
have been very useful in testing theories regard- pression by reagents that inhibit or neutralize
ing the toxic pathways elicited by aberrant a- aggregated species (Vekrellis et al. 2009; Bieschke
synuclein. et al. 2010). However, the protective effects of

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

HSP overexpression are not always apparent or “suck in” essential neuronal components.
(Shimshek et al. 2010). There is less agreement On the other hand, if the protofibril theory
as to which particular species are neurotoxic. is correct, enhancing inclusions could be
A predominant idea is that some of the early viewed as a method to remove the toxic inter-
soluble oligomeric species have such a potential. mediate oligomeric species, and this has been
This is based on the following pieces of data: attempted with some success in cellular
models (Bodner et al. 2006).
(1) A53T and A30P a-synuclein form more
protofibrils than the WT protein, whereas
a-SYNUCLEIN POSTTRANSLATIONAL
only A53T forms more readily mature fi-
MODIFICATIONS AND INTERACTIONS
brils; however, E46K a-synuclein forms
less protofibrils than the WT, therefore a-Synuclein may be modified by phosphoryla-
there is no simple correlation that can ex- tion, oxidation, nitrosylation, glycation, or gly-
plain everything using this scheme, which cosylation. Of all such modifications, the best
is based on in vitro cell-free systems (Con- studied is phosphorylation. Antibodies specific
way et al. 2000; Fredenburg et al. 2007). to S129-phosphorylated a-synuclein promi-
nently decorate LBs in synucleinopathies (Fuji-
(2) Dopamine and its metabolites act as inhib-
wara et al. 2002). Although the initial idea,
itors of the conversion of protofibrils to
which appeared to be buttressed by experiments
mature fibrils in vitro and in vivo, presum-
in transgenic flies, was that such phosphoryla-
ably through the formation of dopamine/
tion enhanced a-synuclein-soluble oligomers
a-synuclein adducts (Conway et al. 2001;
(but actually decreased a-synuclein inclusions)
Mazzulli et al. 2006; Tsika et al. 2010), sug-
and neurotoxicity (Chen et al. 2005, 2009);
www.perspectivesinmedicine.org

gesting that the preferential vulnerability of


subsequent studies have cast doubt on this no-
dopaminergic neurons to the pathogenic
tion, providing contradictory results (Gorba-
process of PD may be attributable to the
tyuk et al. 2008; Paleologou et al. 2008; Azeredo
enhanced presence of soluble oligomers
da Silveira et al. 2009). In fact, it may be that
(although, surprisingly, mouse nigral neu-
S129 phosphorylation, which occurs minimally
rons, which contain more oligomers, ap-
under physiological circumstances, occurs after
pear relatively resistant to transgenic a-
the fact in already formed LBs (Paleologou et al.
synuclein-mediated neurodegeneration).
2008). Enhancing a-synuclein phosphatase ac-
(3) In cellular systems and in vivo, the presence tivity was, however, protective against a-synu-
of soluble oligomers, and not frank inclu- clein-mediated neurotoxicity, suggesting a causal
sions, generally correlates best with neu- detrimental role of a-synuclein phosphorylation
rotoxicity (Gosavi et al. 2002; Lo Bianco in the disease process (Lee et al. 2011). Polo-like
et al. 2002). Two major studies provided kinases appear to be the physiological kinases
support to this idea, as they showed en- for a-synuclein S129 phosphorylation (Inglis
hanced neurotoxicity both in cell culture et al. 2009; Mbefo et al. 2010). S87 is another
and in vivo with artificial mutants of a- phosphorylation site for a-synuclein, and S87-P
synuclein that favored the formation of sol- a-synuclein is also apparent within LBs, but its
uble oligomers over mature fibrils (Karpinar pathological significance is unclear (Paleologou
et al. 2009; Winner et al. 2011). It should et al. 2010). Phosphorylation on a-synuclein ty-
be noted, however, that the issue is not rosine residues has also been reported and may
settled; the protofibrils are heterogeneous, act in opposing fashion to S129 phosphorylation,
“off pathway” oligomers that do not lead at least in Drosophila (Chen et al. 2009).
to toxicity may exist, and frank inclusions A number of studies in cell-free systems
have the potential of being neurotoxic them- and in cell culture have shown that a-synuclein
selves over time, especially if present in axons, can be oxidized, and this seems to drive its oli-
where they can block neuronal trafficking gomerization (Lee et al. 2002; Betarbet et al.

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

2006; Mirzaei et al. 2006; Qin et al. 2007). Iron tion (Kim et al. 2002; Murray et al. 2003). Calpain,
and dopamine appear to exert a similar effect, one of the enzymes thought to perform such
although whether this is a direct interaction or processing, is especially interesting given its cal-
mediated by oxidation is less clear (Ostrerova- cium dependence and localization at presynaptic
Golts et al. 2000; Conway et al. 2001; Kostka terminals (Mishizen-Eberz et al. 2003, 2005).
et al. 2008; Leong et al. 2009). The potent oxi- a-Synuclein is quite promiscuous in its
dant 4-hydroxy-2-nonenal (HNE) promoted binding properties, and may bind various pro-
stable soluble oligomers, inhibiting their con- teins in neuronal cells, including components
version to fibrils; such stable soluble oligomers of dopamine metabolism, such as tyrosine hy-
were highly toxic, favoring again the soluble droxylase (TH) and the dopamine transporter
oligomer toxicity theory (Qin et al. 2007). In a (DAT), modulating their function (Ostrerova
similar vein, nitration of a-synuclein has been et al. 1999; Perez et al. 2002; Wersinger and
reported; in fact, an antibody specifically gener- Sidhu 2003; Wersinger et al. 2003; Peng et al.
ated against nitrosylated a-synucelin decorates 2005). There are quite a few reports suggesting
LB-like pathology (Giasson et al. 2000). Mono- that through such binding to various proteins,
meric and dimeric nitrated a-synuclein pro- a-synuclein may exert pro- or antiapoptotic
moted fibril formation (Hodara et al. 2004), effects (Ostrerova et al. 1999; da Costa et al.
whereas oligomeric nitrated a-synuclein actu- 2000; Jin et al. 2011), although no effect on pro-
ally inhibited it, stabilizing as soluble oligomers grammed neuronal cell death has been shown in
(Uversky et al. 2005), suggesting complex effects mice null for a-synuclein (Stefanis et al. 2004).
of nitrosylation on a-synuclein aggregation. As Interestingly, however, these mice are resistant
with S129 phosphorylation, it is possible that to the well-known mitochondrial neurotoxin
the intense nitrosylation of a-synuclein within MPTP, through mechanisms that are still not
www.perspectivesinmedicine.org

LB-like structures occurs after the fact. deciphered, but may involve steps both up-
Given the fact that oxidative/nitrative stress stream of and downstream from mitochon-
induced by environmental factors, including drial dysfunction (Dauer et al. 2002). Such mice
mitochondrial toxins, has been heavily impli- are also resistant against neuroinflammation-
cated in PD, oxidation- or nitration-induced induced nigral degeneration conferred by lipo-
a-synuclein oligomerization has attracted at- polysaccharide (LPS) (Gao et al. 2008), suggest-
tention, as a point of potential convergence be- ing that endogenous a-synuclein is somehow
tween environmental and genetic factors lead- involved in such death pathways.
ing to the disease. It should be noted, however, A number of interactions of a-synuclein with
that a-synuclein aggregation could be dissoci- other proteins have been implicated in its aggrega-
ated from mitochondrial toxicity-induced death tion potential; most notable amongst these is syn-
(Liu et al. 2008), arguing against simplistic lin- philin-1, a protein identified through a yeast-
ear pathways to neurodegeneration, and raising two-hybrid screen as an AS-interacting protein,
the issue whether in certain instances the ab- which promotes a-synuclein aggregation and
undant a-synuclein-related pathology may be inclusion formation (Engelender et al. 1999). In-
an epiphenomenon, and not causally related to terestingly, transgenic expression of synphilin-1
neuronal dysfunction and death. Similar infer- on the background of A53T a-synuclein trans-
ences can be taken from a study in proteasomal genic mice attenuates neurodegeneration, as as-
inhibitor-treated neurons, where the b-sheet- sessed by indices of cell death and gliosis, while
like pathology of a-synuclein was dissociated enhancing inclusion formation, suggesting that
from death (Rideout et al. 2004). promoting a-synuclein inclusion formation is
Another modification that may be impor- neuroprotective (Smith et al. 2010).
tant is carboxy-terminal truncation of a-synu- Interactions of a-synuclein with lipids may
clein, leading to fragments lacking part or the be important not only for their physiologic
whole of the acidic tail (Li et al. 2005). These functions, but also for their aberrant effects. Re-
fragments appear to be more prone to fibrilliza- garding the former, it is interesting to note that

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

sequestration of the fatty acid arachidonic acid associated a-synuclein increase was robust in
away from the SNARE complex by a-synuclein vulnerable brain regions (Tong et al. 2010). It
has been suggested to underlie its inhibitory may of course be that neurons with the highest
effect on neuronal transmission (Darios et al. expression levels of a-synuclein are most vulner-
2010), and that alterations in levels of key en- able and succumb early in the disease process,
zymes in lipid signaling, such as PLD2 and giving their place to glia, thus confounding the
PLA2, may underlie the phenotype in SNCA results. To partly circumvent this issue, Gründe-
null microglia, which are overreactive but lack mann et al. (2008) performed laser-capture mi-
phagocytic potential (Austin et al. 2006, 2011). crodissection studies, and reported a consider-
Regarding the latter, polyunsaturated fatty acids able increase of SNCA expression in surviving
(PUFAs) have been shown to enhance oligome- nigral neurons derived from PD brains com-
rization and neurotoxicity of a-synuclein (Per- pared with controls. However, this increase did
rin et al. 2001; Sharon et al. 2003; Assayag et al. not appear to be specific for SNCA (Gründe-
2007). mann et al. 2008). Therefore, the issue is still
open.
An indirect way of approaching this is by
GAIN-OF-FUNCTION AND
assessing whether polymorphic SNCA variants
ACCUMULATION OF a-SYNUCLEIN
that confer disease risk in association studies
The notion that enhanced a-synuclein levels are alter a-synuclein levels in model systems. In
causative in PD pathogenesis is derived from initial studies, the Rep1 polymorphism associ-
the familial SNCA multiplication cases show- ated with the disease led to increased SNCA
ing a dose-dependent correlation of a-synuclein transcriptional activity in neuronal cell lines
load to the PD phenotype, the autosomal-dom- (Chiba-Falek and Nussbaum 2001); this finding
www.perspectivesinmedicine.org

inant pattern of inheritance for the point muta- has been confirmed in mice (Cronin et al.
tions, the accumulation of a-synuclein in synu- 2009), and even in the human brain (Linnertz
cleinopathy brains, and the demonstration that et al. 2009), although in another study an asso-
features of the disease can be mimicked by WT ciation between Rep1 polymorphisms and a-
a-synuclein overexpression in various models. synuclein mRNA and protein levels in the brain
Consistent with this idea, a-synuclein protein was not apparent (Fuchs et al. 2008). Polymor-
levels are increased with aging in the substantia phisms within the 30 region may also correlate
nigra, and correlate with decreased TH immu- with AS levels (Fuchs et al. 2008; Mata et al.
nostaining (Chu and Kordower 2007). How- 2010). Intriguingly, it was reported that risk-
ever, it should be noted that there is insufficient conferring alleles at the 30 untranslated region
evidence that there is a generalized excess of (UTR) were associated with increased expres-
a-synuclein in PD brains. In fact, mRNA stud- sion of an alternatively spliced form of SNCA
ies have been quite contradictory in this regard, (McCarthy et al. 2011), suggesting another me-
with some even showing a decrease of SNCA ex- chanism through which gene variations may be
pression in PD nigra (Dachsel et al. 2007). Pro- linked to the disease.
tein levels are not obviously increased overall in It is generally assumed that a-synuclein leads
PD brains, although clearly there is induction of to disease through a toxic gain-of-function that
insoluble components, including monomeric is likely inherent in the normal protein when it ex-
and oligomeric species. A comprehensive study ceeds a certain level. In apparent agreement with
of multiple brain areas showed that membrane- this, a-synuclein null mice, in contrast to trans-
associated monomeric a-synuclein levels were genic overexpressors, display no overt neuropa-
only modestly increased in the substantia nigra, thological or behavioral phenotype (Abeliovich
and not in other brain regions, of PD patients et al. 2000; Cabin et al. 2002). However, when
compared to controls; even so, the levels in studied more closely, such mice do have some
some PD cases were within the spectrum of con- deficits in the dopaminergic system, in par-
trol values. In contrast, in MSA, the membrane- ticular, a slightly reduced number of postnatal

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

dopaminergic neurons (Robertson et al. 2004), (2010) found that the WT allele is induced in a
and a drop of dopaminergic terminals with ag- compensatory fashion, and that the total level
ing (Al-Wandi et al. 2010). Thus, the theory that of SNCA messenger RNA (mRNA) exceeds that
part of the PD phenotype might be attributable of controls, suggesting that total SNCA levels,
to the sequestering of physiological a-synuclein rather than the presence of the A53T mutant,
in inclusions, which could cause neuronal, and, are responsible for the disease in these cases.
in particular (in view of the normal function of Although this study was based on a single pa-
the protein), synaptic damage, cannot be dis- tient, it does bring up again the issue of total
carded. SNCA levels as the major determinant of toxicity.
Despite the fact that transcriptional regu- In view of the above, a reasonable avenue to
lation of a-synuclein is so important for PD attack synucleinopathies may be the lowering
pathogenesis, little is known about the mecha- of a-synuclein levels. A note of caution is in or-
nisms involved. Transcription factors GATA-1 der here, given the uncertainty about potential
(Scherzer et al. 2008) and ZSCAN21 (Clough neurotoxicity associated with severe reduction
and Stefanis 2007; Clough et al. 2009) within of a-synuclein levels, as mentioned above. In
Intron 1 have been proposed to play a role as fact, Gorbatyuk et al. (2010) found that siRNA-
inducers of transcription. In the latter case, a mediated down-regulation of a-synuclein led
signaling pathway involving ERK/PI3 kinase to quite considerable neurotoxicity within the
signaling mediated ZSCAN-induced SNCA rat nigral dopaminergic system within a period
transcriptional activation (Clough and Stefanis of 4 wk. In contrast, no toxicity was observed
2007; Clough et al. 2009). A similar ERK-de- when small interfering RNAs (siRNAs) against
pendent signaling pathway regulating SNCA a-synuclein were injected directly into the mon-
production was apparent in enteric neurons key nigra, achieving 50% – 60% down-regula-
www.perspectivesinmedicine.org

(Paillusson et al. 2010). Methylation may be tion of expression (McCormack et al. 2010).
important in SNCA expression, as methylation Control of a-synuclein levels can also be
within Intron 1 suppressed SNCA transcrip- achieved at the level of its degradation. a-Synu-
tional activity, and brains of patients with LB clein degradation has been controversial, but it
diseases showed decreased methylation within appears that the bulk of degradation of at least
this region, suggesting epigenetic regulation of monomeric WT a-synuclein in neuronal cell
SNCA expression (Jowaed et al. 2010; Matsu- systems occurs through the lysosomal pathways
moto et al. 2010). In one study, it was proposed of chaperone-mediated autophagy (CMA) and
that sequestration of the methylation factor macroautophagy (Paxinou et al. 2001; Webb
Dnmt1 away from the nucleus by a-synuclein et al. 2003; Cuervo et al. 2004; Lee et al. 2004;
may lead to reduced methylation and enhanced Vogiatzi et al. 2008; Mak et al. 2010). It has
SNCA exression, thus creating a feed-forward been proposed that dysfunction of these degra-
loop of SNCA overexpression (Desplats et al. dation pathways may be a contributing factor
2011). Posttranscriptional regulation of a-syn- to PD pathogenesis (Xilouri et al. 2008). a-Syn-
uclein can also occur through endogenous uclein may also be turned over by the protea-
micro RNAs, which bind to the 30 end of the some in other experimental settings (Tofaris
gene (Junn et al. 2009; Doxakis 2010). A partic- et al. 2001; Webb et al. 2003). A factor that
ularly interesting twist to the effects of familial may play a role is the exact species of a-synuclein
point mutations in SNCA has been provided studied. In any case, strategies directed toward
by a series of studies that, somewhat counterin- promoting such endogenous degradation sys-
tuitively, suggest that the expression of the mu- tems to enhance clearance of excess a-synuclein
tant alleles is suppressed, especially in cases with have the advantage that they could also alleviate
prolonged disease (Markopoulou et al. 1999; the aberrant effects of a-synuclein on their func-
Kobayashi et al. 2003), through a mechanism tion (see below). Indeed, stimulation of macro-
that may involve histone modifications (Vout- autophagy through pharmacological or molecu-
sinas et al. 2010). Remarkably, Voutsinas et al. lar means leads to increased WT a-synuclein

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

clearance and neuroprotection (Webb et al. induced death (Mosharov et al. 2006). Excess
2003; Spencer et al. 2009). Another attractive intracellular calcium enhanced cytosolic dopa-
method of enhancing clearance of a-synuclein mine, and AS was required for death induced
through the lysosomal pathway may be through by excess dopamine, creating a possible scenario
immunization (Masliah et al. 2005, 2011). in which intracellular calcium, cystolic dopa-
mine, and a-synuclein interact with each other
in a self-feeding cascade leading to neurodegen-
POTENTIAL PATHOGENIC EFFECTS OF
eration (Mosharov et al. 2009). Another factor
a-SYNUCLEIN: a-SYNUCLEIN AT THE
that could be important in this setting is cal-
SYNAPSE
cium-mediated calpain activation that could
Given the physiological role of a-synuclein at lead, as mentioned, to carboxy-terminal a-syn-
the synapse, it is not surprising that studies uclein truncation, consequent oligomerization,
have identified synaptic effects as significant de- and further calcium influx, creating a vicious
terminants of a-synuclein-induced neurotoxic- cycle.
ity in cell culture and in vivo models based on
overexpression of the protein; in some of these
a-SYNUCLEIN AT THE CYTOSKELETON
cases levels of overexpression are modest, ex-
pected to be present in at least the brains of a-Synuclein may impact cytoskeletal dynamics.
the SNCA multiplication cases. The effects Most of the evidence suggests an association of
seen include loss of presynaptic proteins, de- a-synuclein with microtubules. Various investi-
crease of neurotransmitter release, redistribu- gators have detected an interaction of a-synu-
tion of SNARE proteins, enlargement of synap- clein with tubulin (Alim et al. 2002; Zhou
tic vesicles, and inhibition of synaptic vesicle et al. 2010), but the effects reported on tubulin
www.perspectivesinmedicine.org

recycling (Chung et al. 2009; Garcia-Reitböck polymerization are conflicting, with others re-
et al. 2010; Nemani et al. 2010; Scott et al. porting enhanced (Alim et al. 2004; Lin et al.
2010). It is likely that such deficits precede overt 2009), and others reduced (Lee et al. 2006;
neuropathology and are causal in synaptic and Zhou et al. 2010) tubulin polymerization as a
neuritic degeneration, although the sequence result of a-synuclein overexpression. Further-
of synaptic events and the identification of the more, tubulin depolymerization may lead to
exact point in the cascade in which aberrant impaired maturation of a-synuclein protofi-
a-synuclein assumes its neurotoxic potential brils into mature fibrils, which may lead to
are still elusive. enhanced neurotoxicity (Lee and Lee 2002).
Given the role of calcium is presynaptic sig- Another factor that may be important is the re-
naling events, it is tempting to speculate that lationship of a-synuclein to tau, which is likely
perturbations in calcium homeostasis may important in the pathogenesis of sporadic dis-
play a role in the neuritic degeneration induced ease, as in GWAS the two corresponding genes
by a-synuclein. Indeed, there is some evidence are the ones showing the highest association
for this, albeit still somewhat fragmentary, to PD (Nalls et al. 2011). Although tauopathy
implicating primarily a-synuclein oligomers, is not a prominent feature in PD, it is appreci-
which could form pores on cellular membranes ated in certain cases (Kotzbauer et al. 2004).
or alter the properties of voltage-gated recep- In such cases, a-synuclein and tau pathology
tors, leading to excess calcium influx (Volles may occur in the same cell, but the formed ag-
and Lansbury 2002; Danzer et al. 2007; Hettiar- gregates are separate. In fact, each one of these
achchi et al. 2009). Through a similar porelike two proteins has the tendency to seed the aggre-
mechanism, oligomeric AS may also attack syn- gation of the other (Giasson et al. 2003). a-Syn-
aptic vesicles, leading to leakage of neurotrans- uclein appears to enhance tau phosphorylation,
mitter in the cytosol; in the case of dopamine and this may lead to secondary effects on the
vesicles, this would mean excess cytosolic dop- microtubular network (Jensen et al. 1999; Hag-
amine, which could lead to oxidative stress- gerty et al. 2011; Qureshi and Paudel 2011).

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

Furthermore, a-synuclein may bind and in- compensatory macroautophagy induction that
fluence polymerization of actin, and thus per- may be detrimental to neuronal survival (Xi-
haps indirectly affect cellular trafficking and louri et al. 2009). The issue is, however, contro-
synaptic function; disparate effects on actin versial, as others find that WT a-synuclein ac-
were observed between WT and A30P mutant tually diminishes macroautophagy, through an
a-synuclein (Sousa et al. 2009). Another aspect inhibitory action at the level of phagophore for-
that appears to be affected by aberrant a-synu- mation (Winslow et al. 2010).
clein, and may be related to the effects on the An interesting issue that relates to a-synu-
cytoskeleton, is axonal transport. In an adeno- clein degradation and its effects on protein deg-
associated virus (AAV) model of nigral synu- radation systems is the relationship of PD to
cleinopathy, early changes before cell death Gaucher disease and glucocerebrosidase (GBA).
included a diminution at the level of the stria- Heterozygote and homozygote carriers of GBA
tum of motor proteins involved in anterograde mutations are at an increased risk of develop-
transport, and an increase of those involved in ing PD. It is controversial whether this repre-
retrograde transport (Chung et al. 2009). sents a loss-of-function of GBA or a gain-of-
function conferred by the mutants. According
to some, loss-of-function of GBA is sufficient
a-SYNUCLEIN AND PROTEIN
to induce a-synuclein accumulation and aggre-
DEGRADATION SYSTEMS
gation (Manning-Bog et al. 2010; Mazzulli et al.
An issue that has attracted a lot of attention is the 2011; Xu et al. 2011), which may occur second-
possibility that aberrant a-synuclein may im- ary to lysosomal dysfunction (Mazzulli et al.
pact protein degradation systems. This notion 2011); others find that a-synuclein accumulates
is attractive, as a number of neurodegenerative only on overexpression of GBA mutants, favor-
www.perspectivesinmedicine.org

conditions are associated with impairments of ing a gain of toxic function mechanism (Cullen
protein clearance, and such impairments have et al. 2011). The accumulation of substrates
the potential to create a vicious loop involving such as glycosylceramide owing to GBA loss-
aberrant a-synuclein accumulation. Aberrant of-function may also confer increased tendency
a-synuclein, mutant, or under certain circum- of a-synuclein to oligomerize (Mazzulli et al.
stances, WT, can induce proteasomal dysfunc- 2011; Xu et al. 2011); on the other hand, oligo-
tion in a number of cell-free, cellular, and in meric a-synuclein may impact on GBA, leading
vivo settings (Stefanis et al. 2001; Tanaka et al. to its dysfunction, creating a vicious cycle of ly-
2001; Petrucelli et al. 2002; Snyder et al. 2003), sosomal dysfunction and aberrant a-synuclein
although this is not universally observed (Mar- accumulation (Mazzulli et al. 2011).
tin-Clemente et al. 2004). It is thought that oli-
gomeric a-synuclein, and, in particular, small
a-SYNUCLEIN AT THE MITOCHONDRIA
soluble oligomeric forms that are themselves de-
AND RELATIONSHIP TO OXIDATIVE
graded by the proteasome, may exert this effect
STRESS
(Lindersson et al. 2004; Emmanouilidou et al.
2010a). This would create the potential for the Many studies now have confirmed that a pro-
vicious cycle mentioned above. portion of a-synuclein, endogenous or overex-
Mutant a-synuclein may also aberrantly pressed, resides within mitochondria, and that
impact the lysosomes, causing impairment of it causes down-regulation of complex I activity,
CMA (Cuervo et al. 2004; Xilouri et al. 2009; thus linking a-synuclein with the effects of mi-
Alvarez-Erviti et al. 2010). WTAS, in an adduct tochondrial toxins, and potentially, sporadic
with dopamine, can cause a similar effect (Mar- PD (Li et al. 2007; Devi et al. 2008; Nakamura
tinez-Vicente et al. 2008). Interestingly, the main et al. 2008; Liu et al. 2009; Loeb et al. 2010).
proteins involved in CMA, Lamp2a and Hsc70, Furthermore, brain mitochondria morphology
are diminished in PD brains (Alvarez-Erviti is disrupted in a-synuclein transgenic mice
et al. 2010). Such CMA impairment may cause (Martin et al. 2006). a-Synuclein, potentially

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

in the form of small oligomers, appears to mediated toxicity in a variety of cellular and in
induce mitochondrial fragmentation, which vivo models. Thayanidhi et al. (2010) suggested
may be responsible for subsequent mitochon- that in mammalian systems the ER/Golgi transi-
drial dysfunction and death (Kamp et al. 2010, tion was delayed owing to an antagonistic effect
Nakamura et al. 2011). a-Synuclein, and espe- of a-synuclein on ER/Golgi SNAREs. Over-
cially the mutant form, perhaps through the expression of other Rabs, more closely linked
same mechanism of fragmentation, also in- to synaptic vesicular function, such as Rab3A,
duces inappropriate excessive mitophagy, which also prevented dopaminergic toxicity in the a-
leads to mitochondrial removal and neuronal synuclein C. elegans model, suggesting that these
death (Choubey et al. 2011). Interestingly, en- phenomena may more broadly reflect the inter-
hancing the expression of PGC-1a, the master action and effects of a-synuclein on vesicular
transcriptional regulator of nuclearly encoded dynamics (Gitler et al. 2008).
mitochondrial subunits, which is down-regu-
lated in PD brains, leads to protection against
a-SYNUCLEIN IN THE NUCLEUS
a-synuclein-induced neurodegeneration in cel-
lular models (Zheng et al. 2010). Synuclein bears its name from its apparent lo-
Through such effects on mitochondria, and calization in the nucleus and presynatic nerve
especially the complex I inhibition, a-synuclein terminals (Maroteaux et al. 1988). However,
could potentially induce reactive oxygen species nuclear localization of a-synuclein has only
(ROS) production, oxidative stress, and resultant been observed inconsistently (Mori et al.
neuronal death. There is in fact some evidence 2002b; Yu et al. 2007); it actually appears that,
that a-synuclein overexpression can induce ROS, at least in transgenic mice, a-synuclein phos-
and that suppressing these through molec- phorylated at S129 may be preferentially local-
www.perspectivesinmedicine.org

ular or pharmacological means may be neuro- ized in the nucleus (Schell et al. 2009). a-Synu-
protective in mammalian (Hsu et al. 2000; Clark clein has been found to interact with histones
et al. 2010; Liu et al. 2011) or insect systems on nuclear translocation in nigral neurons in re-
(Wassef et al. 2007; Botella et al. 2008). sponse to the herbicide paraquat (Goers et al.
2003), and to reduce histone acetylation (Kon-
topoulos et al. 2006). Inhibitors of histone de-
a-SYNUCLEIN AT THE ER/GOLGI
acetylase (HDAC) rescued a-synuclein-medi-
In a neuronal cell model based on adenoviral ated neurotoxicity in cell culture and in vivo
expression of a-synuclein, Golgi fragmentation Drosophila models (Kontopoulos et al. 2006).
was an early feature and correlated with the ap- Furthermore, inhibition of Sirtuin 2, a mamma-
pearance of soluble oligomeric species (Gosavi lian HDAC, which led to increased acetylation,
et al. 2002). Smith et al. (2005) observed endo- was associated with neuroprotection in various
plasmic reticulum (ER) stress as an early event models of synucleinopathy, although whether
in an inducible model of A53T a-synuclein- potential acetylation effects on histone were in-
mediated neurotoxicity, and were able to inhibit volved was not ascertained (Outeiro et al. 2007).
death using a pharmacological inhibitor of ER
stress, also suggesting that a primary target of
a-SYNUCLEIN OUTSIDE THE CELL:
a-synuclein may be the ER/Golgi. In a blinded
THE THEORY OF PROPAGATION
screen in a yeast model, the major class of modu-
lators of a-synuclein toxicity was genes involved Although initially a-synuclein was thought to
in vesicular trafficking (Willingham et al. 2003). be a purely intracellular protein, as it lacks a sig-
This was taken one step further by Cooper et al. naling sequence that would direct it to the secre-
(2006), who identified ER-to-Golgi trafficking tory pathway, it gradually became clear that it
as the major cellular perturbation in this yeast could be detected in the conditioned medium
model, and Rab1, a protein involved in this tran- of cells and in extracellular fluids, such as
sition, as a critical determinant of a-synuclein- plasma and CSF (El-Agnaf et al. 2003; Lee

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

et al. 2005). The mechanism of a-synuclein se- (Desplats et al. 2009), and fetal dopaminergic
cretion is not well understood, but it appears to neurons also showed uptake of host a-synuclein
occur through a nonclassical secretory pathway (Hansen et al. 2011); on the other, fetal dopami-
(Lee et al. 2005), possibly within exosomes, en- nergic grafts in humans showed LB pathology
docytic vesicles that are contained within mul- many years after implantation, although wheth-
tivesicular bodies and are released on calcium er the a-synuclein within the grafted neurons
influx (Emmanouilidou et al. 2010b). The exis- was host-derived could not be determined with
tence of extracellular a-synuclein in rodent and certainty (Kordower et al. 2008). An intriguing
human brain interstitial fluid has been con- study has brought together the mitochondrial
firmed by microdialysis (Emmanouilidou et al. dysfunction with the a-synuclein propagation
2011). Importantly, secreted a-synuclein can theories. Intragastric administration of the com-
impact neuronal homeostasis and lead to neu- plex I inhibitor rotenone led to enteric a-synu-
ronal death, even at concentrations close to clein pathology, which gradually spread to the
those identified in bodily fluids, and the respon- central nervous system (CNS), including dopa-
sible species, at least in part, appear to be soluble minergic neurons, and caused delayed neuro-
oligomeric (Emmanouilidou et al. 2010b; Dan- toxicity, suggesting that, once initiated by mito-
zer et al. 2011). Furthermore, various reports chondrial dysfunction, a-synuclein aberrant
have shown that extracellular a-synuclein can conformations assume an independent propa-
induce inflammatory reactions in glial cells, gating seeding and neurotoxic potential (Pan-
thus further potentiating neurodegeneration Montojo et al. 2010). This would be consistent
(Zhang et al. 2005; Klegeris et al. 2008; Lee with the findings of early enteric a-synuclein
et al. 2010). It should be noted, however, that pathology in PD (Braak et al. 2006) and in a-
such studies are often based on high concentra- synuclein transgenic mice (Kuo et al. 2010). A
www.perspectivesinmedicine.org

tions of recombinant a-synuclein that may not note of caution is indicated in the general as-
be physiologic. This issue is also touched on by sumption that seeding is equated with neuro-
Gendelman and colleagues (Mosley et al. in toxicity. In fact, seeding and toxicity of extracel-
press). lular a-synuclein can be dissociated (Danzer
A related aspect of a-synuclein pathobiol- et al. 2007).
ogy that has attracted a lot of attention is its ap-
parent ability to be uptaken by cells, although,
a-SYNUCLEIN BIOMARKERS
at least in cell culture, this is highly dependent
on the cell type and the particular species of In view of the importance of a-synuclein in
a-synuclein and is not easily achieved in intact PD pathogenesis, a number of studies have at-
neuronal cells (Emmanouilidou et al. 2010b). tempted to use measurements of a-synuclein
Oligomeric-aggregated a-synuclein species ap- as biomarkers for the disease and for other
pear to be especially prone to uptake and have synucleinopathies. In the cerebrospinal fluid
the potential to “seed” fibrillization of endoge- (CSF), most studies appear to show a reduction
nous a-synuclein (Danzer et al. 2007, 2009, of total a-synuclein levels in PD patients com-
2011; Luk et al. 2009; Nonaka et al. 2010; Wax- pared with controls (Tokuda et al. 2006; Mol-
man and Giasson 2010; Hansen et al. 2011). lenhauer et al. 2008, 2011; Hong et al. 2010; Ka-
This observation has created a lot of excitement, suga et al. 2010); in fact, a comprehensive study
as it could potentially explain the propagation suggested that a-synuclein levels, measured by a
of pathology observed in PD, according to the sensitive ELISA technique, may help to differen-
Braak hypothesis of staging of the disease. Two tiate synucleinopathies, where a-synuclein is
key in vivo observations support this “prionlike reduced compared with other neurodegenera-
propagation” theory. On the one hand, progen- tive conditions, such as tauopathies or amy-
itor cells implanted in the hippocampus of re- loidoses (Mollenhauer et al. 2011). In contrast,
cipient mice transgenic for a-synuclein incor- oligomeric a-synuclein, measured by a specific
porated host a-synuclein in their cell soma ELISA method, was higher in PD compared

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

Preventing
specific aberrant
functions
Synaptic
Enhancing
Modulating vesicles
degradation
transcription Proteasomes
Lysosomes
ER-Golgi
Cell membrane/Calcium
Mitochondria

Axonal transport

Inhibiting Promoting fibril


proaggregation Inhibiting
oligomer/protofibril formation
posttranslational
modifications formation

Promoting off-pathway
oligomers

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

with control CSF (Tokuda et al. 2010), extend- ACKNOWLEDGMENTS


ing previous findings of identification of soluble
L.S. was funded for PD-related research by EC
www.perspectivesinmedicine.org

oligomeric a-synuclein in DLB brain tissue


FP7 Programmes NEURASYNC and MEFOPA.
(Paleologou et al. 2009), but this intriguing re-
sult awaits replication in a larger cohort. Data
from a-synuclein measurements in peripheral REFERENCES
 Reference is also in this collection.
tissues/fluids have been inconsistent, and over-
all peripheral a-synuclein levels, despite the ease
Abeliovich A, Schmitz Y, Farinas I, Choi-Lundberg D, Ho
of obtaining the material, do not appear to have WH, Castillo PE, Shinsky N, Verdugo JM, Armanini M,
potential as biomarkers. Ryan A, et al. 2000. Mice lacking a-synuclein display
functional deficits in the nigrostriatal dopamine system.
Neuron 25: 239– 252.
CONCLUDING REMARKS Al-Chalabi A, Dürr A, Wood NW, Parkinson MH, Camuzat
A, Hulot JS, Morrison KE, Renton A, Sussmuth SD,
It is clear from the above that a-synuclein repre- Landwehrmeyer BG, et al. 2009. Genetic variants of the
sents a valid therapeutic target in PD and possi- a-synuclein gene SNCA are associated with multiple sys-
tem atrophy. PLoS One 4: e7114.
bly in related synucleinpathies. Possible thera-
Alim MA, Hossain MS, Arima K, Takeda K, Izumiyama Y,
peutic strategies that could be envisioned are Nakamura M, Kaji H, Shinoda T, Hisanaga S, Ueda K.
depicted in Figure 2. Perhaps the most promis- 2002. Tubulin seeds a-synuclein fibril formation. J Biol
ing strategy would involve small molecules that Chem 277: 2112– 2117.
Alim MA, Ma QL, Takeda K, Aizawa T, Matsubara M, Naka-
would have the potential to alter the confor- mura M, Asada A, Saito T, Kaji H, Yoshii M, et al. 2004.
mation of protofibrillar forms of a-synuclein Demonstration of a role for a-synuclein as a functional
and render them nonpathogenic. An example microtubule-associated protein. J Alzheimers Dis 6:
435 –442.
of such natural products could be the green
Alvarez-Erviti L, Rodriguez-Oroz MC, Cooper JM, Cabal-
tea derivative EGCG (Bieschke et al. 2010) or lero C, Ferrer I, Obeso JA, Schapira AH. 2010. Chaper-
the natural product scyllo inositol (Vekrellis one-mediated autophagy markers in Parkinson disease
et al. 2009). It may be that multiple therapeutic brains. Arch Neurol 67: 1464–1472.
approaches will need to be used, in view of the Al-Wandi A, Ninkina N, Millership S, Williamson SJ, Jones
PA, Buchman VL. 2010. Absence of a-synuclein affects
feed-forward amplification loops involved in dopamine metabolism and synaptic markers in the stria-
the pathogenic effects of a-synuclein. tum of aging mice. Neurobiol Aging 31: 796 –804.

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

Assayag K, Yakunin E, Loeb V, Selkoe DJ, Sharon R. 2007. Burré J, Sharma M, Tsetsenis T, Buchman V, Etherton MR,
Polyunsaturated fatty acids induce a-synuclein-related Südhof TC. 2010. a-Synuclein promotes SNARE-com-
pathogenic changes in neuronal cells. Am J Pathol 171: plex assembly in vivo and in vitro. Science 329: 1663–
2000– 2011. 1667.
Auluck PK, Bonini NM. 2002. Pharmacological prevention Cabin DE, Shimazu K, Murphy D, Cole NB, Gottschalk W,
of Parkinson disease in Drosophila. Nat Med 8: 1185– McIlwain KL, Chen A, Ellis CE, Paylor R, Lu B, et al. 2002.
1186. Synaptic vesicle depletion correlates with attenuated syn-
Auluck PK, Chan HY, Trojanowski JQ, Lee VM, Bonini NM. aptic responses to prolonged repetitive stimulation in
2002. Chaperone suppression of a-synuclein toxicity in a mice lacking a-synuclein. J Neurosci 22: 8797–8807.
Drosophila model for Parkinson’s disease. Science 295: Chandra S, Gallardo G, Fernández-Chacón R, Schlüter OM,
865–868. Südhof TC. 2005. Alpha-synuclein cooperates with
Austin SA, Floden AM, Murphy EJ, Combs CK. 2006. a- CSPalpha in preventing neurodegeneration. Cell 123:
Synuclein expression modulates microglial activation 383 –396.
phenotype. J Neurosci 26: 10558– 10563. Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V,
Austin SA, Rojanathammanee L, Golovko MY, Murphy EJ, Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J,
Combs CK. 2011. Lack of a-synuclein modulates micro- Hulihan M, et al. 2004. a-Synuclein locus duplication
glial phenotype in vitro. Neurochem Res 36: 994– 1004. as a cause of familial Parkinson’s disease. Lancet 364:
Azeredo da Silveira S, Schneider BL, Cifuentes-Diaz C, Sage 1167–1169.
D, Abbas-Terki T, Iwatsubo T, Unser M, Aebischer P. 2009. Chen L, Feany MB. 2005. a-Synuclein phosphorylation
Phosphorylation does not prompt, nor prevent, the for- controls neurotoxicity and inclusion formation in a Dro-
mation of a-synuclein toxic species in a rat model of Par- sophila model of Parkinson disease. Nat Neurosci 8:
kinson’s disease. Hum Mol Genet 18: 872– 887. 657 –663.
Baba M, Nakajo S, Tu PH, Tomita T, Nakaya K, Lee VM, Chen L, Periquet M, Wang X, Negro A, McLean PJ, Hyman
Trojanowski JQ, Iwatsubo T. 1998. Aggregation of a-syn- BT, Feany MB. 2009. Tyrosine and serine phosphoryla-
uclein in Lewy bodies of sporadic Parkinson’s disease and tion of a-synuclein have opposing effects on neurotoxic-
dementia with Lewy bodies. Am J Pathol 152: 879 –884. ity and soluble oligomer formation. J Clin Invest 119:
Barbour R, Kling K, Anderson JP, Banducci K, Cole T, Diep 3257–3265.
L, Fox M, Goldstein JM, Soriano F, Seubert P, et al. 2008. Chesselet MF. 2008. In vivo a-synuclein overexpression in
www.perspectivesinmedicine.org

Red blood cells are the major source of a-synuclein in rodents: A useful model of Parkinson’s disease? Exp Neu-
blood. Neurodegener Dis 5: 55–59. rol 209: 22– 27.
Betarbet R, Canet-Aviles RM, Sherer TB, Mastroberardino Chiba-Falek O, Nussbaum RL. 2001. Effect of allelic varia-
PG, McLendon C, Kim JH, Lund S, Na HM, Taylor G, tion at the NACP-Rep1 repeat upstream of the a-synu-
Bence NF, et al. 2006. Intersecting pathways to neurode- clein gene (SNCA) on transcription in a cell culture luci-
generation in Parkinson’s disease: Effects of the pesticide ferase reporter system. Hum Mol Genet 10: 3101–3109.
rotenone on DJ-1, a-synuclein, and the ubiquitin-pro-
teasome system. Neurobiol Dis 22: 404 –420. Choubey V, Safiulina D, Vaarmann A, Cagalinec M, Wareski
P, Kuum M, Zharkovsky A, Kaasik A. 2011. Mutant A53T
Bieschke J, Russ J, Friedrich RP, Ehrnhoefer DE, Wobst H, a-synuclein induces neuronal death by increasing mito-
Neugebauer K, Wanker EE. 2010. EGCG remodels ma- chondrial autophagy. J Biol Chem 286: 10814–10824.
ture a-synuclein and amyloid-bfibrils and reduces cellu-
lar toxicity. Proc Natl Acad Sci 107: 7710–7715. Chu Y, Kordower JH. 2007. Age-associated increases of
a-synuclein in monkeys and humans are associated
Bodner RA, Outeiro TF, Altmann S, Maxwell MM, Cho SH, with nigrostriatal dopamine depletion: Is this the target
Hyman BT, McLean PJ, Young AB, Housman DE, Ka-
for Parkinson’s disease? Neurobiol Dis 25: 134– 149.
zantsev AG. 2006. Pharmacological promotion of inclu-
sion formation: A therapeutic approach for Huntington’s Chung CY, Koprich JB, Siddiqi H, Isacson O. 2009. Dynamic
and Parkinson’s diseases. Proc Natl Acad Sci 103: 4246– changes in presynaptic and axonal transport proteins
4251. combined with striatal neuroinflammation precede dop-
aminergic neuronal loss in a rat model of AAV a-synu-
Botella JA, Bayersdorfer F, Schneuwly S. 2008. Superoxide
cleinopathy. J Neurosci 29: 3365– 3373.
dismutase overexpression protects dopaminergic neu-
rons in a Drosophila model of Parkinson’s disease. Neuro- Clark J, Clore EL, Zheng K, Adame A, Masliah E, Simon DK.
biol Dis 30: 65–73. 2010. Oral N-acetyl-cysteine attenuates loss of dopami-
Braak H, Del Tredici K, Rüb U, de Vos RA, Jansen Steur EN, nergic terminals in a-synuclein overexpressing mice.
Braak E. 2003. Staging of brain pathology related to PLoS One 5: e12333.
sporadic Parkinson’s disease. Neurobiol Aging 24: 197– Clough RL, Dermentzaki G, Stefanis L. 2009. Functional
211. dissection of the a-synuclein promoter: Transcriptional
Braak H, de Vos RA, Bohl J, Del Tredici K. 2006. Gastric regulation by ZSCAN21 and ZNF219. J Neurochem 110:
a-synuclein immunoreactive inclusions in Meissner’s 1479–1490.
and Auerbach’s plexuses in cases staged for Parkinson’s Clough RL, Stefanis L. 2007. A novel pathway for transcrip-
disease-related brain pathology. Neurosci Lett 396: 67– tional regulation of a-synuclein. FASEB J 21: 596 –607.
72. Conway KA, Lee SJ, Rochet JC, Ding TT, Williamson RE,
Burke RE, Dauer WT, Vonsattel JP. 2008. A critical evalua- Lansbury PT Jr. 2000. Acceleration of oligomerization,
tion of the Braak staging scheme for Parkinson’s disease. not fibrillization, is a shared property of both a-synuclein
Ann Neurol 64: 485– 491. mutations linked to early-onset Parkinson’s disease:

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

Implications for pathogenesis and therapy. Proc Natl sequesters Dnmt1 from the nucleus: A novel mechanism
Acad Sci 97: 571 –576. for epigenetic alterations in Lewy body diseases. J Biol
Conway KA, Rochet JC, Bieganski RM, Lansbury PT Jr. Chem 286: 9031– 9037.
2001. Kinetic stabilization of the a-synuclein protofibril Devi L, Raghavendran V, Prabhu BM, Avadhani NG, Anan-
by a dopamine-a-synuclein adduct. Science 294: 1346– datheerthavarada HK. 2008. Mitochondrial import and
1349. accumulation of a-synuclein impair complex I in human
Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, dopaminergic neuronal cultures and Parkinson disease
Bhullar B, Liu K, Xu K, Strathearn KE, Liu F, et al. brain. J Biol Chem 283: 9089– 9100.
2006. a-Synuclein blocks ER-Golgi traffic and Rab1 res- Dickson DW, Uchikado H, Fujishiro H, Tsuboi Y. 2010. Evi-
cues neuron loss in Parkinson’s models. Science 313: dence in favor of Braak staging of Parkinson’s disease.
324–328. Mov Disord 25 (Suppl 1): S78 –S82.
Cronin KD, Ge D, Manninger P, Linnertz C, Rossoshek A, Doxakis E. 2010. Post-transcriptional regulation of a-synu-
Orrison BM, Bernard DJ, El-Agnaf OM, Schlossmacher clein expression by mir-7 and mir-153. J Biol Chem 285:
MG, Nussbaum RL, et al. 2009. Expansion of the Parkin- 12726– 12734.
son disease-associated SNCA-Rep1 allele upregulates hu-
El-Agnaf OM, Salem SA, Paleologou KE, Cooper LJ, Full-
man a-synuclein in transgenic mouse brain. Hum Mol
wood NJ, Gibson MJ, Curran MD, Court JA, Mann
Genet 18: 3274–3285.
DM, Ikeda S, et al. 2003. a-Synuclein implicated in Par-
Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer kinson’s disease is present in extracellular biological flu-
D. 2004. Impaired degradation of mutant a-synuclein by ids, including human plasma. FASEB J 17: 1945–1947.
chaperone-mediated autophagy. Science 305: 1292–
Eliezer P, Zweckstetter D, Masliah M, Lashuel EHA. 2010.
1295.
Phosphorylation at S87 is enhanced in synucleinopathies,
Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, Kolod- inhibits a-synuclein oligomerization, and influences
ziej P, Kahn I, Saftig P, Woulfe J, et al. 2011. Acid b-gluco- synuclein-membrane interactions. J Neurosci 30: 3184–
sidase mutants linked to Gaucher disease, Parkinson dis- 3198.
ease, and Lewy body dementia alter a-synuclein
processing. Ann Neurol 69: 940–953. Emmanoulidou E, Stefanis L, Vekrellis K. 2010a. Specific
cell-derived soluble a-synuclein oligomers are degraded
Dachsel JC, Lincoln SJ, Gonzalez J, Ross OA, Dickson DW, by the 26S proteasome and impair its function. Neurobiol
Farrer MJ. 2007 The ups and downs of a-synuclein
Aging 31: 953– 968.
mRNA expression. Movement Disord 22: 293– 295.
www.perspectivesinmedicine.org

Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis


da Costa CA, Ancolio K, Checler F. 2000. Wild-type but
SD, Ntzouni M, Margaritis LH, Stefanis L, Vekrellis K.
not Parkinson’s disease-related ala-53 ! Thr mutant
2010b. Cell-produced a-synuclein is secreted in a cal-
a-synuclein protects neuronal cells from apoptotic stim-
cium-dependent manner by exosomes and impacts neu-
uli. J Biol Chem 275: 24065–24069.
ronal survival. J Neurosci 30: 6838– 6851.
Danzer KM, Haasen D, Karow AR, Moussaud S, Habeck M,
Emmanouilidiou E, Elenis D, Papasilekas T, Stranjalis G,
Giese A, Kretzschmar H, Hengerer B, Kostka M. 2007.
Different species of a-synuclein oligomers induce cal- Gerozissis K, Ioannou P, Vekrellis K. 2011. In vivo assess-
cium influx and seeding. J Neurosci 27: 9220– 9232. ment of a-synuclein secretion. PLoS One 6: e22225.
Danzer KM, Krebs SK, Wolff M, Birk G, Hengerer B. 2009. Engelender S, Kaminsky Z, Guo X, Sharp AH, Amaravi RK,
Seeding induced by a-synuclein oligomers provides evi- Kleiderlein JJ, Margolis RL, Troncoso JC, Lanahan AA,
dence for spreading of a-synuclein pathology. J Neuro- Worley PF, et al. 1999. Synphilin-1 associates with a-syn-
chem 111: 192 –203. uclein and promotes the formation of cytosolic inclu-
sions. Nat Genet 22: 110– 114.
Danzer KM, Ruf WP, Putcha P, Joyner D, Hashimoto T,
Glabe C, Hyman BT, McLean PJ. 2011. Heat-shock pro- Esposito A, Dohm CP, Kermer P, Bähr M, Wouters FS. 2007.
tein 70 modulates toxic extracellular a-synuclein oligom- a-Synuclein and its disease-related mutants interact dif-
ers and rescues trans-synaptic toxicity. FASEB J 25: ferentially with the microtubule protein tau and associate
326–336. with the actin cytoskeleton. Neurobiol Dis 26: 521 –531.
Darios F, Ruipérez V, López I, Villanueva J, Gutierrez LM, Feany MB, Bender WW. 2000. A Drosophila model of Par-
Davletov B. 2010. a-Synuclein sequesters arachidonic kinson’s disease. Nature 404: 394– 398.
acid to modulate SNARE-mediated exocytosis. EMBO Fortin DL, Nemani VM, Voglmaier SM, Anthony MD, Ryan
Rep 11: 528– 533. TA, Edwards RH. 2005. Neural activity controls the syn-
Dauer W, Kholodilov N, Vila M, Trillat AC, Goodchild R, aptic accumulation of a-synuclein. J Neurosci 25: 10913–
Larsen KE, Staal R, Tieu K, Schmitz Y, Yuan CA, et al. 10921.
2002. Resistance of a-synuclein null mice to the parkin- Fredenburg RA, Rospigliosi C, Meray RK, Kessler JC, La-
sonian neurotoxin MPTP. Proc Natl Acad Sci 99: 14524– shuel HA, Eliezer D, Lansbury PT Jr. 2007. The impact
14529. of the E46K mutation on the properties of a-synuclein
Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, in its monomeric and oligomeric states. Biochemistry
Spencer B, Masliah E, Lee SJ. 2009. Inclusion formation 46: 7107–7118.
and neuronal cell death through neuron-to-neuron Fuchs J, Nilsson C, Kachergus J, Munz M, Larsson EM,
transmission of a-synuclein. Proc Natl Acad Sci 106: Schüle B, Langston JW, Middleton FA, Ross OA, Hulihan
13010–13015. M, et al. 2007. Phenotypic variation in a large Swedish
Desplats P, Spencer B, Coffee E, Patel P, Michael S, Patrick C, pedigree due to SNCA duplication and triplication. Neu-
Adame A, Rockenstein E, Masliah E. 2011. a-Synuclein rology 68: 916– 922.

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

Fuchs J, Tichopad A, Golub Y, Munz M, Schweitzer KJ, Wolf aggregates and precedes the formation of fibrillar inclu-
B, Berg D, Mueller JC, Gasser T. 2008. Genetic variability sion. J Biol Chem 277: 48984– 48992.
in the SNCA gene influences a-synuclein levels in the Gründemann J, Schlaudraff F, Haeckel O, Liss B. 2008. Ele-
blood and brain. FASEB J 22: 1327–1334. vated a-synuclein mRNA levels in individual UV-laser-
Fujita M, Sugama S, Sekiyama K, Sekigawa A, Tsukui T, Na- microdissected dopaminergic substantia nigra neurons
kai M, Waragai M, Takenouchi T, Takamatsu Y, Wei J, in idiopathic Parkinson’s disease. Nucleic Acids Res 36:
et al. 2010 A b-synuclein mutation linked to dementia e38.
produces neurodegeneration when expressed in mouse Haggerty T, Credle J, Rodriguez O, Wills J, Oaks AW, Masliah
brain. Nat Commun 1: 110. E, Sidhu A. 2011. Hyperphosphorylated t in an a-synu-
Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Mas- clein-overexpressing transgenic model of Parkinson’s dis-
liah E, Goldberg MS, Shen J, Takio K, Iwatsubo T. 2002. ease. Eur J Neurosci 33: 1598–1610.
a-Synuclein is phosphorylated in synucleinopathy le-
Hansen C, Angot E, Bergström AL, Steiner JA, Pieri L, Paul
sions. Nat Cell Biol 4: 160– 164.
G, Outeiro TF, Melki R, Kallunki P, Fog K, et al. 2011.
Galvin JE, Uryu K, Lee VM, Trojanowski JQ. 1999. Axon a-Synuclein propagates from mouse brain to grafted
pathology in Parkinson’s disease and Lewy body demen- dopaminergic neurons and seeds aggregation in cultured
tia hippocampus contains a-, b-, and g-synuclein. Proc human cells. J Clin Invest 121: 715– 725.
Natl Acad Sci 96: 13450–13455.
Hashimoto M, Rockenstein E, Mante M, Mallory M, Mas-
Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, liah E. 2001. b-Synuclein inhibits a-synuclein aggrega-
Lee VM. 2008. Neuroinflammation and oxidation/nitra- tion: A possible role as an anti-parkinsonian factor. Neu-
tion of a-synuclein linked to dopaminergic neurodegen- ron 32: 213– 223.
eration. J Neurosci 28: 7687–7698.
Hettiarachchi NT, Parker A, Dallas ML, Pennington K, Hung
Garcia-Reitböck P, Anichtchik O, Bellucci A, Iovino M, Bal- CC, Pearson HA, Boyle JP, Robinson P, Peers C. 2009.
lini C, Fineberg E, Ghetti B, Della Corte L, Spano P, Tofa-
a-Synuclein modulation of Ca2þ signaling in human
ris GK, et al. 2010. SNARE protein redistribution and
neuroblastoma (SH-SY5Y) cells. J Neurochem 111: 1192–
synaptic failure in a transgenic mouse model of Parkin-
1201.
son’s disease. Brain 133 (Pt 7): 2032–2044.
Hodara R, Norris EH, Giasson BI, Mishizen-Eberz AJ,
George JM. 2002. The synucleins. Genome Biol 3: RE-
Lynch DR, Lee VM, Ischiropoulos H. 2004. Functional
VIEWS3002.
consequences of a-synuclein tyrosine nitration: Dimin-
www.perspectivesinmedicine.org

George JM, Jin H, Woods WS, Clayton DF. 1995. Character- ished binding to lipid vesicles and increased fibril forma-
ization of a novel protein regulated during the critical pe- tion. J Biol Chem 279: 47746– 47753.
riod for song learning in the zebra finch. Neuron 15:
361–372. Hong Z, Shi M, Chung KA, Quinn JF, Peskind ER, Galasko
D, Jankovic J, Zabetian CP, Leverenz JB, Baird G, et al.
Giasson BI, Duda JE, Murray IV, Chen Q, Souza JM, Hurtig 2010. DJ-1 and a-synuclein in human cerebrospinal fluid
HI, Ischiropoulos H, Trojanowski JQ, Lee VM. 2000. Ox-
as biomarkers of Parkinson’s disease. Brain 133 (Pt 3):
idative damage linked to neurodegeneration by selective
713 –726.
a-synuclein nitration in synucleinopathy lesions. Science
290: 985– 989. Hsu LJ, Sagara Y, Arroyo A, Rockenstein E, Sisk A, Mallory
M, Wong J, Takenouchi T, Hashimoto M, Masliah E.
Giasson BI, Forman MS, Higuchi M, Golbe LI, Graves CL,
2000. a-Synuclein promotes mitochondrial deficit and
Kotzbauer PT, Trojanowski JQ, Lee VM. 2003. Initiation
oxidative stress. Am J Pathol 157: 401– 410.
and synergistic fibrillization of t and a-synuclein. Science
300: 636– 640. Inglis KJ, Chereau D, Brigham EF, Chiou SS, Schöbel S,
Gitler AD, Bevis BJ, Shorter J, Strathearn KE, Hamamichi S, Frigon NL, Yu M, Caccavello RJ, Nelson S, Motter R,
Su LJ, Caldwell KA, Caldwell GA, Rochet JC, McCaffery et al. 2009. Polo-like kinase 2 (PLK2) phosphorylates
JM, et al. 2008. The Parkinson’s disease protein a-synuclein at serine 129 in central nervous system.
a-synuclein disrupts cellular Rab homeostasis. Proc J Biol Chem 284: 2598–2602.
Natl Acad Sci 105: 145–150. Jensen PH, Hager H, Nielsen MS, Hojrup P, Gliemann J,
Goers J, Manning-Bog AB, McCormack AL, Millett IS, Do- Jakes R. 1999. a-Synuclein binds to t and stimulates
niach S, Di Monte DA, Uversky VN, Fink AL. 2003. Nu- the protein kinase A-catalyzed tphosphorylation of ser-
clear localization of a-synuclein and its interaction with ine residues 262 and 356. J Biol Chem 274: 25481– 25489.
histones. Biochemistry 42: 8465–8471. Jin H, Kanthasamy A, Ghosh A, Yang Y, Anantharam V,
Gorbatyuk OS, Li S, Sullivan LF, Chen W, Kondrikova G, Kanthasamy AG. 2011. a-Synuclein negatively regulates
Manfredsson FP, Mandel RJ, Muzyczka N. 2008. The protein kinase Cd expression to suppress apoptosis in
phosphorylation state of Ser-129 in human a-synuclein dopaminergic neurons by reducing p300 histone acetyl-
determines neurodegeneration in a rat model of Parkin- transferase activity. J Neurosci 31: 2035–2051.
son disease. Proc Natl Acad Sci 105: 763–768. Jowaed A, Schmitt I, Kaut O, Wüllner U. 2010. Methylation
Gorbatyuk OS, Li S, Nash K, Gorbatyuk M, Lewin AS, Sul- regulates a-synuclein expression and is decreased in Par-
livan LF, Mandel RJ, Chen W, Meyers C, Manfredsson FP, kinson’s disease patients’ brains. J Neurosci 30: 6355–
et al. 2010. In vivo RNAi-mediated a-synuclein silencing 6359.
induces nigrostriatal degeneration. Mol Ther 18: 1450– Junn E, Lee KW, Jeong BS, Chan TW, Im JY, Mouradian
1457. MM. 2009. Repression of a-synuclein expression and
Gosavi N, Lee HJ, Lee JS, Patel S, Lee SJ. 2002. Golgi frag- toxicity by microRNA-7. Proc Natl Acad Sci 106: 13052–
mentation occurs in the cells with prefibrillar a-synuclein 13057.

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

Kahle PJ. 2008. a-Synucleinopathy models and human neu- ation in dementia with Lewy bodies. J Neurosci 27:
ropathology: Similarities and differences. Acta Neuropa- 1405–1410.
thol 115: 87–95. Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kosel S,
Kamp F, Exner N, Lutz AK, Wender N, Hegermann J, Brun- Przuntek H, Epplen JT, Schols L, Riess O. 1998. Ala30Pro
ner B, Nuscher B, Bartels T, Giese A, Beyer K, et al. 2010. mutation in the gene encoding a-synuclein in Parkin-
Inhibition of mitochondrial fusion by a-synuclein is res- son’s disease. Nat Genet 18: 106–108.
cued by PINK1, Parkin and DJ-1. EMBO J 29: 3571– Kuo YM, Li Z, Jiao Y, Gaborit N, Pani AK, Orrison BM, Bru-
3589. neau BG, Giasson BI, Smeyne RJ, Gershon MD, et al.
Karpinar DP, Balija MB, Kügler S, Opazo F, Rezaei-Ghaleh 2010. Extensive enteric nervous system abnormalities in
N, Wender N, Kim HY, Taschenberger G, Falkenburger mice transgenic for artificial chromosomes containing
BH, Heise H, et al. 2009. Pre-fibrillar a-synuclein var- Parkinson disease-associated a-synuclein gene muta-
iants with impaired b-structure increase neurotoxicity tions precede central nervous system changes. Hum Mol
in Parkinson’s disease models. EMBO J 28: 3256– 3268. Genet 19: 1633–1650.
Kasuga K, Tokutake T, Ishikawa A, Uchiyama T, Tokuda T, Lakso M, Vartiainen S, Moilanen AM, Sirvio J, Thomas JH,
Onodera O, Nishizawa M, Ikeuchi T. 2010. Differential Nass R, Blakely RD, Wong G. 2003. Dopaminergic neuro-
levels of a-synuclein, b-amyloid42 and tau in CSF nal loss and motor deficits in Caenorhabditis elegans
between patients with dementia with Lewy bodies and overexpressing human a-synuclein. J Neurochem 86:
Alzheimer’s disease. J Neurol Neurosurg Psychiatry 81: 165 –172.
608–610. Larsen KE, Schmitz Y, Troyer M, Mosharov E, Dietrich P, Sa-
Kholodilov NG, Neystat M, Oo TF, Lo SE, Larsen KE, Sulzer valle M, Quazi AZ, Chaudhry FA, Edwards R, Stefanis L,
D, Burke RE. 1999. Increased expression of rat synuclein et al. 2006. a-Synuclein overexpression in PC12 and
in the substantia nigra pars compacta identified by chromaffin cells impairs catecholamine release by inter-
mRNA differential display in a model of developmental fering with a late step in exocytosis. J Neurosci 26:
target injury. J Neurochem 73: 2586–2599. 11915– 11922.
Kim TD, Paik SR, Yang CH. 2002. Structural and functional Lee HJ, Shin SY, Choi C, Lee YH, Lee SJ. 2002. Formation
implications of C-terminal regions of a-synuclein. Bio- and removal of a-synuclein aggregates in cells exposed
chemistry 41: 13782– 13790. to mitochondrial inhibitors. J Biol Chem 277: 5411–
Kirik D, Rosenblad C, Burger C, Lundberg C, Johansen TE, 5417.
Muzyczka N, Mandel RJ, Bjorklund A. 2002. Parkinson- Lee HJ, Khoshaghideh F, Patel S, Lee SJ. 2004. Clearance of
www.perspectivesinmedicine.org

like neurodegeneration induced by targeted overexpres- a-synuclein oligomeric intermediates via the lysosomal
sion of a-synuclein in the nigrostriatal system. J Neurosci degradation pathway. J Neurosci 24: 1888– 1896.
22: 2780–2791. Lee HJ, Patel S, Lee SJ. 2005. Intravesicular localization and
Klegeris A, Pelech S, Giasson BI, Maguire J, Zhang H, exocytosis of a-synuclein and its aggregates. J Neurosci
McGeer EG, McGeer PL. 2008. a-Synuclein activates 25: 6016–6024.
stress signaling protein kinases in THP-1 cells and micro- Lee HJ, Khoshaghideh F, Lee S, Lee SJ. 2006. Impairment of
glia. Neurobiol Aging 29: 739– 752. microtubule-dependent trafficking by overexpression of
Klucken J, Shin Y, Masliah E, Hyman BT, McLean PJ. 2004. a-synuclein. Eur J Neurosci 24: 3153–3162.
Hsp70 reduces a-synuclein aggregation and toxicity. J Lee HJ, Suk JE, Patrick C, Bae EJ, Cho JH, Rho S, Hwang D,
Biol Chem 279: 25497–25502. Masliah E, Lee SJ. 2010. Direct transfer of a-synuclein
Kobayashi H, Krüger R, Markopoulou K, Wszolek Z, Chase from neuron to astroglia causes inflammatory responses
B, Taka H, Mineki R, Murayama K, Riess O, Mizuno Y, in synucleinopathies. J Biol Chem 285: 9262– 9272.
et al. 2003. Haploinsufficiency at the a-synuclein gene Lee KW, Chen W, Junn E, Im JY, Grosso H, Sonsalla PK, Feng
underlies phenotypic severity in familial Parkinson’s dis- X, Ray N, Fernandez JR, Chao Y, et al. 2011. Enhanced
ease. Brain 126 (Pt 1): 32–42. phosphatase activity attenuates a-synucleinopathy in a
Kontopoulos E, Parvin JD, Feany MB. 2006. a-Synuclein mouse model. J Neurosci 31: 6963– 6971.
acts in the nucleus to inhibit histone acetylation and pro- Leong SL, Pham CL, Galatis D, Fodero-Tavoletti MT, Perez
mote neurotoxicity. Hum Mol Genet 15: 3012–3023. K, Hill AF, Masters CL, Ali FE, Barnham KJ, Cappai R.
Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. 2009. Formation of dopamine-mediated a-synuclein-
2008. Lewy body-like pathology in long-term embryonic soluble oligomers requires methionine oxidation. Free
nigral transplants in Parkinson’s disease. Nat Med 14: Radical Bio Med 46: 1328– 1337.
504–506. Li W, West N, Colla E, Pletnikova O, Troncoso JC, Marsh L,
Kostka M, Högen T, Danzer KM, Levin J, Habeck M, Wirth Dawson TM, Jäkälä P, Hartmann T, Price DL, et al. 2005.
A, Wagner R, Glabe CG, Finger S, Heinzelmann U, et al. Aggregation promoting C-terminal truncation of a-syn-
2008. Single particle characterization of iron-induced uclein is a normal cellular process and is enhanced by the
pore-forming a-synuclein oligomers. J Biol Chem 283: familial Parkinson’s disease-linked mutations. Proc Natl
10992–11003. Acad Sci 102: 2162– 2167.
Kotzbauer PT, Giasson BI, Kravitz AV, Golbe LI, Mark MH, Li WW, Yang R, Guo JC, Ren HM, Zha XL, Cheng JS, Cai DF.
Trojanowski JQ, Lee VM. 2004. Fibrillization of a-synu- 2007. Localization of a-synuclein to mitochondria
clein and tau in familial Parkinson’s disease caused by the within midbrain of mice. Neuroreport 18: 1543– 1546.
A53T a-synuclein mutation. Exp Neurol 187: 279 –288. Lin X, Parisiadou L, Gu XL, Wang L, Shim H, Sun L, Xie C,
Kramer ML, Schulz-Schaeffer WJ. 2007. Presynaptic a-syn- Long CX, Yang WJ, Ding J, et al. 2009. Leucine-rich re-
uclein aggregates, not Lewy bodies, cause neurodegener- peat kinase 2 regulates the progression of neuropathology

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

induced by Parkinson’s-disease-related mutant a-synu- Martı̀n-Clemente B, Alvarez-Castelao B, Mayo I, Sierra AB,


clein. Neuron 64: 807– 827. Dı̀az V, Milán M, Fariñas I, Gómez-Isla T, Ferrer I, Cas-
Lindersson E, Beedholm R, Højrup P, Moos T, Gai W, Hendil taño JG. 2004. a-Synuclein expression levels do not sig-
KB, Jensen PH. 2004. Proteasomal inhibition by alpha- nificantly affect proteasome function and expression in
synuclein filaments and oligomers. J Biol Chem 279: mice and stably transfected PC12 cell lines. J Biol Chem
12924–12934. 279: 52984–52990.
Linnertz C, Saucier L, Ge D, Cronin KD, Burke JR, Brown- Martinez-Vicente M, Talloczy Z, Kaushik S, Massey AC,
dyke JN, Hulette CM, Welsh-Bohmer KA, Chiba-Falek O. Mazzulli J, Mosharov EV, Hodara R, Fredenburg R, Wu
2009. Genetic regulation of a-synuclein mRNA expres- DC, Follenzi A, et al. 2008. Dopamine-modified alpha-
sion in various human brain tissues. PLoS One 4: e7480. synuclein blocks chaperone-mediated autophagy. J Clin
Liu F, Nguyen JL, Hulleman JD, Li L, Rochet JC. 2008. Mech- Invest 118: 777 –788.
anisms of DJ-1 neuroprotection in a cellular model of Masliah E, Rockenstein E, Adame A, Alford M, Crews L, Ha-
Parkinson’s disease. J Neurochem 105: 2435–2453. shimoto M, Seubert P, Lee M, Goldstein J, Chilcote T,
Liu G, Zhang C, Yin J, Li X, Cheng F, Li Y, Yang H, Uéda K, et al. 2005. Effects of a-synuclein immunization in a
Chan P, Yu S. 2009. a-Synuclein is differentially expressed mouse model of Parkinson’s disease. Neuron 46: 857–
in mitochondria from different rat brain regions and 868.
dose-dependently down-regulates complex I activity. Masliah E, Rockenstein E, Mante M, Crews L, Spencer B,
Neurosci Lett 454: 187–192. Adame A, Patrick C, Trejo M, Ubhi K, Rohn TT, et al.
Liu Z, Yu Y, Li X, Ross CA, Smith WW. 2011. Curcumin pro- 2011. Passive immunization reduces behavioral and
tects against A53T a-synuclein-induced toxicity in a neuropathological deficits in an a-synuclein transgenic
PC12 inducible cell model for Parkinsonism. Pharmacol model of Lewy body disease. PLoS One 6: e19338.
Res 63: 439– 444. Mata IF, Shi M, Agarwal P, Chung KA, Edwards KL, Factor
Lo Bianco C, Ridet JL, Schneider BL, Deglon N, Aebischer P. SA, Galasko DR, Ginghina C, Griffith A, Higgins DS,
2002. a-Synucleinopathy and selective dopaminergic et al. 2010. SNCA variant associated with Parkinson dis-
neuron loss in a rat lentiviral-based model of Parkinson’s ease and plasma a-synuclein level. Arch Neurol 67: 1350–
disease. Proc Natl Acad Sci 99: 10813–10818. 1356.
Loeb V, Yakunin E, Saada A, Sharon R. 2010. The transgenic Matsumoto L, Takuma H, Tamaoka A, Kurisaki H, Date H,
overexpression of a-synuclein and not its related pathol- Tsuji S, Iwata A. 2010. CpG demethylation enhances
www.perspectivesinmedicine.org

ogy associates with complex I inhibition. J Biol Chem 285: a-synuclein expression and affects the pathogenesis of
7334– 7343. Parkinson’s disease. PLoS One 5: e15522.
Luk KC, Song C, O’Brien P, Stieber A, Branch JR, Brunden Mazzulli JR, Mishizen AJ, Giasson BI, Lynch DR, Thomas
KR, Trojanowski JQ, Lee VM. 2009. Exogenous a-synu- SA, Nakashima A, Nagatsu T, Ota A, Ischiropoulos H.
clein fibrils seed the formation of Lewy body-like intra- 2006. Cytosolic catechols inhibit a-synuclein aggregation
cellular inclusions in cultured cells. Proc Natl Acad Sci and facilitate the formation of intracellular soluble oligo-
106: 20051– 20056. meric intermediates. J Neurosci 26: 10068– 10078.
Mak SK, McCormack AL, Manning-Bog AB, Cuervo AM, Mazzulli JR, Xu YH, Sun Y, Knight AL, McLean PJ, Caldwell
Di Monte DA. 2010. Lysosomal degradation of a-synu- GA, Sidransky E, Grabowski GA, Krainc D. 2011.
clein in vivo. J Biol Chem 285: 13621–13629. Gaucher disease glucocerebrosidase and a-synuclein
Manning-Boğ AB, Schüle B, Langston JW. 2009. a-Synu- form a bidirectional pathogenic loop in synucleinopa-
clein-glucocerebrosidase interactions in pharmacological thies. Cell 146: 37–52.
Gaucher models: A biological link between Gaucher dis- Mbefo MK, Paleologou KE, Boucharaba A, Oueslati A,
ease and parkinsonism. Neurotoxicology 30: 1127– 1132. Schell H, Fournier M, Olschewski D, Yin G, Zweckstetter
Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioan- M, Masliah E, et al. 2010. Phosphorylation of synucleins
nidis JP, Kruger R, Rocca WA, Schneider NK, Lesnick TG, by members of the Polo-like kinase family. J Biol Chem
Lincoln SJ, et al. 2006. (GEO-PD) Consortium. Collabo- 285: 2807– 2822.
rative analysis of a-synuclein gene promoter variability McCarthy JJ, Linnertz C, Saucier L, Burke JR, Hulette CM,
and Parkinson disease. JAMA 296: 661–670. Welsh-Bohmer KA, Chiba-Falek O. 2011. The effect of
Markopoulou K, Wszolek ZK, Pfeiffer RF, Chase BA. 1999. SNCA 30 region on the levels of SNCA-112 splicing var-
Reduced expression of the G209A a-synuclein allele in iant. Neurogenetics 12: 59–64.
familial Parkinsonism. Ann Neurol 46: 374– 381. McCormack AL, Mak SK, Henderson JM, Bumcrot D, Far-
Maroteaux L, Scheller RH. 1991. The rat brain synucleins; rer MJ, Di Monte DA. 2010. a-Synuclein suppression by
family of proteins transiently associated with neuronal targeted small interfering RNA in the primate substantia
membrane. Brain Res Mol Brain Res 11: 335 –343. nigra. PLoS One 5: e12122.
Maroteaux L, Campanelli JT, Scheller RH. 1988. Synuclein: Miller DW, Hague SM, Clarimon J, Baptista M, Gwinn-
A neuron-specific protein localized to the nucleus and Hardy K, Cookson MR, Singleton AB. 2004. a-Synuclein
presynaptic nerve terminal. J Neurosci 8: 2804–2815. in blood and brain from familial Parkinson disease with
Martin LJ, Pan Y, Price AC, Sterling W, Copeland NG, Jen- SNCA locus triplication. Neurology 62: 1835– 1838.
kins NA, Price DL, Lee MK. 2006. Parkinson’s disease Mirzaei H, Schieler JL, Rochet JC, Regnier F. 2006. Identifi-
a-synuclein transgenic mice develop neuronal mito- cation of rotenone-induced modifications in a-synuclein
chondrial degeneration and cell death. J Neurosci 26: using affinity pull-down and tandem mass spectrometry.
41–50. Anal Chem 78: 2422–2431.

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

Mishizen-Eberz AJ, Guttmann RP, Giasson BI, Day GA, Ho- mation of alpha-synuclein reveal a specific interaction
dara R, Ischiropoulos H, Lee VM, Trojanowski JQ, Lynch with mitochondria. J Neurosci 28: 12305–12317.
DR. 2003. Distinct cleavage patterns of normal and Nakamura K, Nemani VM, Azarbal F, Skibinski G, Levy JM,
pathologic forms of a-synuclein by calpain I in vitro. J Egami K, Munishkina L, Zhang J, Gardner B, Wakabaya-
Neurochem 86: 836– 847. shi J, et al. 2011. Direct membrane association drives mi-
Mishizen-Eberz AJ, Norris EH, Giasson BI, Hodara R, Is- tochondrial fission by the Parkinson disease-associated
chiropoulos H, Lee VM, Trojanowski JQ, Lynch DR. protein a-synuclein. J Biol Chem 286: 20710–20726.
2005. Cleavage of a-synuclein by calpain: Potential role Nakayama K, Suzuki Y, Yazawa I. 2009. Microtubule depoly-
in degradation of fibrillized and nitrated species of merization suppresses a-synuclein accumulation in a
a-synuclein. Biochemistry 44: 7818– 7829. mouse model of multiple system atrophy. Am J Pathol
Mizuta I, Satake W, Nakabayashi Y, Ito C, Suzuki S, Momose 174: 1471– 1480.
Y, Nagai Y, Oka A, Inoko H, Fukae J, et al. 2006. Multiple Nalls MA, Plagnol V, Hernandez DG, Sharma M, Sheerin
candidate gene analysis identifies a-synuclein as a sus- UM, Saad M, Simón-Sánchez J, Schulte C, Lesage S,
ceptibility gene for sporadic Parkinson’s disease. Hum Sveinbjörnsdóttir S, et al. 2011. Imputation of sequence
Mol Genet 15: 1151–1158. variants for identification of genetic risks for Parkinson’s
Mollenhauer B, Cullen V, Kahn I, Krastins B, Outeiro TF, Pe- disease: A meta-analysis of genome-wide association
pivani I, Ng J, Schulz-Schaeffer W, Kretzschmar HA, studies. Lancet 377: 641– 649.
McLean PJ, et al. 2008. Direct quantification of CSF Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK,
a-synuclein by ELISA and first cross-sectional study Chaudhry FA, Nicoll RA, Edwards RH. 2010. Increased
in patients with neurodegeneration. Exp Neurol 213: expression of a-synuclein reduces neurotransmitter re-
315–325. lease by inhibiting synaptic vesicle reclustering after en-
Mollenhauer B, Locascio JJ, Schulz-Schaeffer W, Sixel- docytosis. Neuron 65: 66–79.
Döring F, Trenkwalder C, Schlossmacher MG. 2011. Nonaka T, Watanabe ST, Iwatsubo T, Hasegawa M. 2010.
a-Synuclein and tau concentrations in cerebrospinal Seeded aggregation and toxicity of a-synuclein and tau:
fluid of patients presenting with parkinsonism: A cohort Cellular models of neurodegenerative diseases. J Biol
study. Lancet Neurol 10: 230– 240. Chem 285: 34885–34898.
Mori F, Tanji K, Yoshimoto M, Takahashi H, Wakabayashi K. Ohtake H, Limprasert P, Fan Y, Onodera O, Kakita A, Taka-
2002a. Demonstration of a-synuclein immunoreactivity hashi H, Bonner LT, Tsuang DW, Murray IV, Lee VM, et al.
in neuronal and glial cytoplasm in normal human brain 2004. b-Synuclein gene alterations in dementia with
www.perspectivesinmedicine.org

tissue using proteinase K and formic acid pretreatment. Lewy bodies. Neurology 63: 805– 811.
Exp Neurol 176: 98– 104. Ostrerova N, Petrucelli L, Farrer M, Mehta N, Choi P, Hardy
Mori F, Tanji K, Yoshimoto M, Takahashi H, Wakabayashi K. J, Wolozin B. 1999. a-Synuclein shares physical and func-
2002b. Immunohistochemical comparison of a- and b- tional homology with 14–3 –3 proteins. J Neurosci 19:
synuclein in adult rat central nervous system. Brain Res 5782–5791.
941: 118– 126. Ostrerova-Golts N, Petrucelli L, Hardy J, Lee JM, Farer M,
Mosharov EV, Staal RG, Bové J, Prou D, Hananiya A, Markov Wolozin B. 2000. The A53T a-synuclein mutation in-
D, Poulsen N, Larsen KE, Moore CM, Troyer MD, et al. creases iron-dependent aggregation and toxicity. J Neuro-
2006. a-Synuclein overexpression increases cytosolic cat- sci 20: 6048– 6054.
echolamine concentration. J Neurosci 26: 9304– 9311. Outeiro TF, Kontopoulos E, Altmann SM, Kufareva I,
Mosharov EV, Larsen KE, Kanter E, Phillips KA, Wilson K, Strathearn KE, Amore AM, Volk CB, Maxwell MM, Ro-
Schmitz Y, Krantz DE, Kobayashi K, Edwards RH, Sulzer chet JC, McLean PJ, et al. 2007. Sirtuin 2 inhibitors rescue
D. 2009. Interplay between cytosolic dopamine, calcium, a-synuclein-mediated toxicity in models of Parkinson’s
and a-synuclein causes selective death of substantia nigra disease. Science 317: 516 –519.
neurons. Neuron 62: 218 –229.
Paillusson S, Tasselli M, Lebouvier T, Mahé M, Chevalier J,
 Mosley RL, Hutter-Saunders JA, Stone DK, Gendelman HE. Biraud M, Cario-Toumaniantz C, Neunlist M, Derkinde-
2012. Inflammation and adaptive immunity in Parkin- ren P. 2010. a-Synuclein expression is induced by depola-
son’s disease. Cold Spring Harb Perspect Med (in press). rization and cyclic AMP in enteric neurons. J Neurochem
Mueller JC, Fuchs J, Hofer A, Zimprich A, Lichtner P, Illig T, 115: 694–706.
Berg D, Wüllner U, Meitinger T, Gasser T. 2005. Multiple Paleologou KE, Schmid AW, Rospigliosi CC, Kim HY, Lam-
regions of a-synuclein are associated with Parkinson’s berto GR, Fredenburg RA, Lansbury PT, Fernandez CO,
disease. Ann Neurol 57: 535 –541. Eliezer D, Zweckstetter M, et al. 2008. Phosphorylation
Murphy DD, Rueter SM, Trojanowski JQ, Lee VM. 2000. at Ser-129 but not the phosphomimics S129E/D inhibits
Synucleins are developmentally expressed, and a-synu- the fibrillation of a-synuclein. J Biol Chem 283: 16895–
clein regulates the size of the presynaptic vesicular pool 16905.
in primary hippocampal neurons. J Neurosci 20: 3214– Paleologou KE, Kragh CL, Mann DM, Salem SA, Al-Shami
3220. R, Allsop D, Hassan AH, Jensen PH, El-Agnaf OM. 2009.
Murray IV, Giasson BI, Quinn SM, Koppaka V, Axelsen PH, Detection of elevated levels of soluble a-synuclein
Ischiropoulos H, Trojanowski JQ, Lee VM. 2003. Role of oligomers in post-mortem brain extracts from patients
a-synuclein carboxy-terminus on fibril formation in vi- with dementia with Lewy bodies. Brain 132 (Pt 4):
tro. Biochemistry 42: 8530–8540. 1093–1101.
Nakamura K, Nemani VM, Wallender EK, Kaehlcke K, Ott Paleologou KE, Oueslati A, Shakked G, Rospigliosi CC, Kim
M, Edwards RH. 2008. Optical reporters for the confor- HY, Lamberto GR, Fernandez CO, Schmid A, Chegini

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

F, Gai WP, et al. 2010. Phsophorylation at S87 is enhanced double a/g-synuclein null mutant mice. J Neurochem
in synucleinopathies, nhibits a-synuclein oligomerization 89: 1126–1136.
and influences synuclein-membrane interactions. J Neu- Schell H, Hasegawa T, Neumann M, Kahle PJ. 2009. Nuclear
rosci 30: 3184–3198. and neritic distribution of serine-129 phosphorylated
Pan-Montojo F, Anichtchik O, Dening Y, Knels L, Pursche S, a-synuclein in transgenic mice. Neuroscience 160: 796–
Jung R, Jackson S, Gille G, Spillantini MG, Reichmann H, 804.
et al. 2010. Progression of Parkinson’s disease pathology Scherzer CR, Grass JA, Liao Z, Pepivani I, Zheng B, Eklund
is reproduced by intragastric administration of rotenone AC, Ney PA, Ng J, McGoldrick M, Mollenhauer B, et al.
in mice. PLoS One 5: e8762. 2008. GATA transcription factors directly regulate the
Park JY, Lansbury PT Jr. 2003. b-Synuclein inhibits forma- Parkinson’s disease-linked gene a-synuclein. Proc Natl
tion of a-synuclein protofibrils: A possible therapeutic Acad Sci 105: 10907–10912.
strategy against Parkinson’s disease. Biochemistry 42: Scholz SW, Houlden H, Schulte C, Sharma M, Li A, Berg D,
3696– 3700. Melchers A, Paudel R, Gibbs JR, Simon-Sanchez J,
Paxinou E, Chen Q, Weisse M, Giasson BI, Norris EH, Rue- et al. 2009. SNCA variants are associated with increased
ter SM, Trojanowski JQ, Lee VM, Ischiropoulos H. 2001. risk for multiple system atrophy. Ann Neurol 65:
Induction of a-synuclein aggregation by intracellular ni- 610 –614.
trative insult. J Neurosci 21: 8053–8061. Scott DA, Tabarean I, Tang Y, Cartier A, Masliah E, Roy S.
Peng XM, Tehranian R, Dietrich P, Stefanis L, Perez R. 2005. 2010. A pathologic cascade leading to synaptic dysfunc-
a-Synuclein activation of protein phosphatase 2A re- tion in a-synuclein-induced neurodegeneration. J Neu-
duces tyrosine hydroxylase phosphorylation in dopami- rosci 30: 8083– 8095.
nergic cells. J Cell Sci 118 (Pt 15): 3523–3530. Sharon R, Bar-Joseph I, Frosch MP, Walsh DM, Hamilton
Perez RG, Waymire JC, Lin E, Liu JJ, Guo F, Zigmond MJ. JA, Selkoe DJ. 2003. The formation of highly soluble
2002. A role for a-synuclein in the regulation of dopa- oligomers of a-synuclein is regulated by fatty acids
mine biosynthesis. J Neurosci 22: 3090–3099. and enhanced in Parkinson’s disease. Neuron 37: 583–
Periquet M, Fulga T, Myllykangas L, Schlossmacher MG, 595.
Feany MB. 2007. Aggregated a-synuclein mediates dop- Shimshek DR, Mueller M, Wiessner C, Schweizer T, van der
aminergic neurotoxicity in vivo. J Neurosci 27: 3338– Putten PH. 2010. The HSP70 molecular chaperone is not
3346. beneficial in a mouse model of a-synucleinopathy. PLoS
One 5: e10014.
www.perspectivesinmedicine.org

Perrin RJ, Woods WS, Clayton DF, George JM. 2001. Expo-
sure to long chain polyunsaturated fatty acids triggers Singleton AB, Farrer M, Johnson J, Singleton A, Hague S,
rapid multimerization of synucleins. J Biol Chem 276: Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nuss-
41958–41962. baum R, et al. 2003. a-Synuclein locus triplication causes
Petrucelli L, O’Farrell C, Lockhart PJ, Baptista M, Kehoe K, Parkinson’s disease. Science 302: 841.
Vink L, Choi P, Wolozin B, Farrer M, Hardy J, et al. 2002. Smith WW, Jiang H, Pei Z, Tanaka Y, Morita H, Sawa A,
Parkin protects against the toxicity associated with mu- Dawson VL, Dawson TM, Ross CA. 2005. Endoplasmic
tant a-synuclein: Proteasome dysfunction selectively af- reticulum stress and mitochondrial cell death pathways
fects catecholaminergic neurons. Neuron 36: 1007– 1019. mediate A53T mutant alpha-synuclein-induced toxicity.
Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia Hum Mol Genet 14: 3801–3811.
A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Smith WW, Liu Z, Liang Y, Masuda N, Swing DA, Jenkins
et al. 1997. Mutation in the a-synuclein gene identified NA, Copeland NG, Troncoso JC, Pletnikov M, Dawson
in families with Parkinson’s disease. Science 276: 2045– TM, et al. 2010. Synphilin-1 attenuates neuronal degen-
2047. eration in the A53T a-synuclein transgenic mouse
Qin Z, Hu D, Han S, Reaney SH, Di Monte DA, Fink AL. model. Hum Mol Genet 19: 2087– 2098.
2007. Effect of 4-hydroxy-2-nonenal modification on Snyder H, Mensah K, Theisler C, Lee J, Matouschek A, Wo-
a-synuclein aggregation. J Biol Chem 282: 5862–5870. lozin B. 2003. Aggregated and monomeric a-synuclein
Qureshi HY, Paudel HK. 2011. Parkinsonian neurotoxin bind to the S6’ proteasomal protein and inhibit proteaso-
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mal function. J Biol Chem 278: 11753– 11759.
and a-synuclein mutations promote tprotein phosphor- Sousa VL, Bellani S, Giannandrea M, Yousuf M, Valtorta F,
ylation at Ser262 and destabilize microtubule cytoskele- Meldolesi J, Chieregatti E. 2009. a-Synuclein and its
ton in vitro. J Biol Chem 286: 5055– 5068. A30P mutant affect actin cytoskeletal structure and dy-
Rideout HJ, Dietrich P, Savalle M, Dauer WT, Stefanis L. namics. Mol Biol Cell 20: 3725–3739.
2003. Regulation of a-synuclein by bFGF in cultured ven- Spencer B, Potkar R, Trejo M, Rockenstein E, Patrick C,
tral midbrain dopaminergic neurons. J Neurochem 84: Gindi R, Adame A, Wyss-Coray T, Masliah E. 2009. Beclin
803–813. 1 gene transfer activates autophagy and ameliorates
Rideout HJ, Dietrich P, Wang QH, Dauer WT, Stefanis L. the neurodegenerative pathology in a-synuclein models
2004. a-Synuclein is required for the fibrillar nature of of Parkinson’s and Lewy body diseases. J Neurosci 29:
ubiquitinated inclusions induced by proteasomal inhi- 13578– 13588.
bition in primary neurons. J Biol Chem 279: 46915–46920. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ,
Robertson DC, Schmidt O, Ninkina N, Jones PA, Sharkey J, Jakes R, Goedert M. 1997. a-Synuclein in Lewy bodies.
Buchman VL. 2004. Developmental loss and resistance to Nature 388: 839 –840.
MPTP toxicity of dopaminergic neurones in substantia Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goe-
nigra pars compacta of g-synuclein, a-synuclein and dert M. 1998. a-Synuclein in filamentous inclusions of

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

L. Stefanis

Lewy bodies from Parkinson’s disease and dementia with Vila M, Vukosavic S, Jackson-Lewis V, Neystat M, Jakowec
Lewy bodies. Proc Natl Acad Sci 95: 6469–6473. M, Przedborski S. 2000. a-Synuclein up-regulation in
Stefanis L, Larsen KE, Rideout HJ, Sulzer D, Greene LA. substantia nigra dopaminergic neurons following ad-
2001. Expression of A53T mutant but not wild-type ministration of the parkinsonian toxin MPTP. J Neuro-
a-synuclein in PC12 cells induces alterations of the chem 74: 721 –729.
ubiquitin-dependent degradation system, loss of dopa- Vogiatzi T, Xilouri M, Vekrellis K, Stefanis L. 2008. Wild type
mine release, and autophagic cell death. J Neurosci 21: a-synuclein is degraded by chaperone-mediated autoph-
9549– 9560. agy and macroautophagy in neuronal cells. J Biol Chem
Stefanis L, Wang Q, Oo T, Lang-Rollin I, Burke RE, Dauer 283: 23542–23556.
WT. 2004. Lack of a-synuclein does not alter apoptosis Volles MJ, Lansbury PT Jr. 2002. Vesicle permeabilization by
of neonatal catecholaminergic neurons. Eur J Neurosci protofibrillar a-synuclein is sensitive to Parkinson’s
20: 1969–1972. disease-linked mutations and occurs by a pore-like mech-
Tanaka Y, Engelender S, Igarashi S, Rao RK, Wanner T, Tanzi anism. Biochemistry 41: 4595–4602.
RE, Sawa A, Dawson V, Dawson TM, Ross CA. 2001. Voutsinas GE, Stavrou EF, Karousos G, Dasoula A, Papa-
Inducible expression of mutant a-synuclein decreases chatzopoulou A, Syrrou M, Verkerk AJ, van der Spek P,
proteasome activity and increases sensitivity to mito- Patrinos GP, Stöger R, et al. 2010. Allelic imbalance of
chondria-dependent apoptosis. Hum Mol Genet 10: expression and epigenetic regulation within the a-synu-
919–926. clein wild-type and p.Ala53Thr alleles in Parkinson dis-
Thayanidhi N, Helm JR, Nycz DC, Bentely M, Liang Y, Hay ease. Hum Mutat 31: 685– 691.
JC. 2010. a-Synuclein delays endoplasmic reticulum Wassef R, Haenold R, Hansel A, Brot N, Heinemann
(ER)-to-Golgi transport in mammalian cells by SH, Hoshi T. 2007. Methionine sulfoxide reductase A
antagonizing ER/Golgi SNAREs. Mol Biol Cell 21: and a dietary supplement S-methyl-L-cysteine pre-
1850– 1863. vent Parkinson’s-like symptoms. J Neurosci 27: 12808–
Tofaris GK, Layfield R, Spillantini MG. 2001. alpha-synu- 12816.
clein metabolism and aggregation is linked to ubiqui- Waxman EA, Giasson BI. 2010. A novel, high-efficiency cel-
tin-independent degradation by the proteasome. FEBS lular model of fibrillar a-synuclein inclusions and the ex-
Lett 509: 22– 26. amination of mutations that inhibit amyloid formation.
Tokuda T, Salem SA, Allsop D, Mizuno T, Nakagawa M, Qur- J Neurochem 113: 374– 388.
www.perspectivesinmedicine.org

eshi MM, Locascio JJ, Schlossmacher MG, El-Agnaf OM. Webb JL, Ravikumar B, Atkins J, Skepper JN, Rubinsztein
2006. Decreased a-synuclein in cerebrospinal fluid of DC. 2003. a-Synuclein is degraded by both autophagy
aged individuals and subjects with Parkinson’s disease. and the proteasome. J Biol Chem 278: 25009– 25013.
Biochem Biophys Res Commun 349: 162– 166. Wersinger C, Sidhu A. 2003. Attenuation of dopamine
Tokuda T, Qureshi MM, Ardah MT, Varghese S, Shehab SA, transporter activity by a-synuclein. Neurosci Lett 340:
Kasai T, Ishigami N, Tamaoka A, Nakagawa M, El-Agnaf 189 –192.
OM. 2010. Detection of elevated levels of a-synuclein Wersinger C, Prou D, Vernier P, Sidhu A. 2003. Modulation
oligomers in CSF from patients with Parkinson disease. of dopamine transporter function by a-synuclein is al-
Neurology 75: 1766–1772. tered by impairment of cell adhesion and by induction
Tong J, Wong H, Guttman M, Ang LC, Forno LS, Shimadzu of oxidative stress. FASEB J 17: 2151– 2153.
M, Rajput AH, Muenter MD, Kish SJ, Hornykiewicz O, Willingham S, Outeiro TF, DeVit MJ, Lindquist SL, Mu-
et al. 2010. Brain a-synuclein accumulation in multiple chowski PJ. 2003. Yeast genes that enhance the toxicity
system atrophy, Parkinson’s disease and progressive of a mutant huntingtin fragment or alpha-synuclein. Sci-
supranuclear palsy: A comparative investigation. Brain ence 302: 1769– 1772.
133 (Pt 1): 172 –188. Winner B, Jappelli R, Maji SK, Desplats PA, Boyer L, Aigner
Tsika E, Moysidou M, Guo J, Cushman M, Gannon P, San- S, Hetzer C, Loher T, Vilar M, Campioni S, et al. 2011. In
daltzopoulos R, Giasson BI, Krainc D, Ischiropoulos H, vivo demonstration that a-synuclein oligomers are toxic.
Mazzulli JR. 2010. Distinct region-specific a-synuclein Proc Natl Acad Sci 108: 4194– 4199.
oligomers in A53T transgenic mice: Implications for Winslow AR, Chen CW, Corrochano S, Acevedo-Arozena A,
neurodegeneration. J Neurosci 30: 3409–3418. Gordon DE, Peden AA, Lichtenberg M, Menzies FM,
Ueda K, Fukushima H, Masliah E, Xia Y, Iwai A, Yoshimoto Ravikumar B, Imarisio S, et al. 2010. a-Synuclein impairs
M, Otero DA, Kondo J, Ihara Y, Saitoh T. 1993. Molecular macroautophagy: Implications for Parkinson’s disease. J
cloning of cDNA encoding an unrecognized component Cell Biol 190: 1023–1037.
of amyloid in Alzheimer disease. Proc Natl Acad Sci 90: Withers GS, George JM, Banker GA, Clayton DF. 1997. De-
11282–11286. layed localization of synelfin (synuclein, NACP) to pre-
Uversky VN, Yamin G, Munishkina LA, Karymov MA, Mil- synaptic terminals in cultured rat hippocampal neurons.
lett IS, Doniach S, Lyubchenko YL, Fink AL. 2005. Effects Brain Res Dev Brain Res 99: 87– 94.
of nitration on the structure and aggregation of Xilouri M, Vogiatzi T, Vekrellis K, Stefanis L. 2008.
a-synuclein. Brain Res Mol Brain Res 134: 84– 102. a-Synuclein degradation by autophagic pathways: A po-
Vekrellis K, Xilouri M, Emmanouilidou E, Stefanis L. 2009. tential key to Parkinson’s disease pathogenesis. Autoph-
Inducible over-expression of wild type a-synuclein in hu- agy 4: 917–919.
man neuronal cells leads to caspase-dependent non- Xilouri M, Vogiatzi T, Vekrellis K, Park D, Stefanis L. 2009.
apoptotic death. J Neurochem 109: 1348– 1362. Abberant a-synuclein confers toxicity to neurons in

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

a-Synuclein in Parkinson’s Disease

part through inhibition of chaperone-mediated autoph- Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, Wilson
agy. PLoS One 4: e5515. B, Zhang W, Zhou Y, Hong JS, et al. 2005. Aggregated
Xu YH, Sun Y, Ran H, Quinn B, Witte D, Grabowski GA. a-synuclein activates microglia: A process leading to dis-
2011. Accumulation and distribution of a-synuclein ease progression in Parkinson’s disease. FASEB J 19:
and ubiquitin in the CNS of Gaucher disease mouse 533 –542.
models. Mol Genet Metab 102: 436–447. Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt
Yu S, Li X, Liu G, Han J, Zhang C, Li Y, Xu S, Liu C, Gao Y, ML, Eklund AC, Zhang-James Y, Kim PD, Hauser MA,
Yang H, et al. 2007. Extensive nuclear localization of et al. 2010. PGC-1a, a potential therapeutic target for
a-synuclein in normal rat brain neurons revealed by a early intervention in Parkinson’s disease. Sci Transl Med
novel monoclonal antibody. Neuroscience 145: 539 –555. 2: 52ra73.
Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Zhou RM, Huang YX, Li XL, Chen C, Shi Q, Wang GR, Tian
Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atarés B, C, Wang ZY, Jing YY, Gao C, et al. 2010. Molecular inter-
et al. 2004. The new mutation, E46K, of a-synuclein action of a-synuclein with tubulin influences on the
causes Parkinson and Lewy body dementia. Ann Neurol polymerization of microtubule in vitro and structure of
55: 164 –173. microtubule in cells. Mol Biol Rep 37: 3183–3192.
www.perspectivesinmedicine.org

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


Downloaded from http://perspectivesinmedicine.cshlp.org/ on May 29, 2023 - Published by Cold Spring Harbor Laboratory Press

α-Synuclein in Parkinson's Disease


Leonidas Stefanis

Cold Spring Harb Perspect Med 2012; doi: 10.1101/cshperspect.a009399 originally published online
December 13, 2011

Subject Collection Parkinson's Disease

Functional Neuroanatomy of the Basal Ganglia Drosophila as a Model to Study Mitochondrial


José L. Lanciego, Natasha Luquin and José A. Dysfunction in Parkinson's Disease
Obeso Ming Guo
Animal Models of Parkinson's Disease: Vertebrate Parkinsonism Due to Mutations in PINK1, Parkin,
Genetics and DJ-1 and Oxidative Stress and Mitochondrial
Yunjong Lee, Valina L. Dawson and Ted M. Pathways
Dawson Mark R. Cookson
Innate Inflammation in Parkinson's Disease Programmed Cell Death in Parkinson's Disease
V. Hugh Perry Katerina Venderova and David S. Park
Parkinson's Disease and Parkinsonism: Genomics and Bioinformatics of Parkinson's
Neuropathology Disease
Dennis W. Dickson Sonja W. Scholz, Tim Mhyre, Habtom Ressom, et
al.
Physiological Phenotype and Vulnerability in Motor Control Abnormalities in Parkinson's
Parkinson's Disease Disease
D. James Surmeier, Jaime N. Guzman, Javier Pietro Mazzoni, Britne Shabbott and Juan Camilo
Sanchez, et al. Cortés
Clinical Approach to Parkinson's Disease: Parkinson's Disease: Gene Therapies
Features, Diagnosis, and Principles of Philippe G. Coune, Bernard L. Schneider and
Management Patrick Aebischer
João Massano and Kailash P. Bhatia
The Role of Autophagy in Parkinson's Disease Functional Neuroimaging in Parkinson's Disease
Melinda A. Lynch-Day, Kai Mao, Ke Wang, et al. Martin Niethammer, Andrew Feigin and David
Eidelberg
Disruption of Protein Quality Control in Leucine-Rich Repeat Kinase 2 for Beginners: Six
Parkinson's Disease Key Questions
Casey Cook, Caroline Stetler and Leonard Lauren R. Kett and William T. Dauer
Petrucelli

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

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

You might also like