You are on page 1of 11

Personal View

Are synucleinopathies prion-like disorders?


Elodie Angot*, Jennifer A Steiner*, Christian Hansen, Jia-Yi Li, Patrik Brundin

Lancet Neurol 2010; 9: 1128–38 A shared neuropathological feature of idiopathic Parkinson’s disease, dementia with Lewy bodies, and multiple system
Published Online atrophy is the development of intracellular aggregates of α-synuclein that gradually engage increasing parts of the nervous
September 15, 2010 system. The pathogenetic mechanisms underlying these neurodegenerative disorders, however, are unknown. Several
DOI:10.1016/S1474-
studies have highlighted similarities between classic prion diseases and these neurological proteinopathies. Specifically,
4422(10)70213-1
identification of Lewy bodies in fetal mesencephalic neurons transplanted in patients with Parkinson’s disease raised the
*These authors contributed
equally hypothesis that α-synuclein, the main component of Lewy bodies, could be transmitted from the host brain to a graft of
Neuronal Survival Unit,
healthy neurons. These results and others have led to the hypothesis that a prion-like mechanism might underlie
Wallenberg Neuroscience progression of synucleinopathy within the nervous system. We review experimental findings showing that misfolded
Centre, Lund University, Lund, α-synuclein can transfer between cells and, once transferred into a new cell, can act as a seed that recruits endogenous
Sweden (E Angot PhD, α-synuclein, leading to formation of larger aggregates. This model suggests that strategies aimed at prevention of cell-to-
J A Steiner PhD, C Hansen PhD,
J-Y Li MD, P Brundin MD)
cell transfer of α-synuclein could retard progression of symptoms in Parkinson’s disease and other synucleinopathies.
Correspondence to:
Patrik Brundin, Neuronal Survival Introduction In this Personal View, we first briefly introduce current
Unit, Wallenberg Neuroscience Protein misfolding is central to the pathogenesis of both knowledge on the mechanisms of spread of prion disease
Centre, Lund University, prion diseases and Parkinson’s disease and other related within and between animals. Next, we review the central
BMC A10, 221 84 Lund, Sweden
patrik.brundin@med.lu.se
neurodegenerative disorders. Some neurodegenerative role of α-synuclein in Parkinson’s disease and other Lewy
disorders, such as Parkinson’s disease, can be body-related disorders and discuss evidence suggesting
characterised by intracytoplasmic inclusions, named that α-synuclein could indeed be classified as a protein
Lewy bodies.1 A prion-like spread of misfolded α-synuclein with prion-like abilities. Furthermore, we identify gaps in
(the predominant protein in Lewy bodies)2 has been knowledge that must be filled in order to state definitively
proposed to contribute to the propagation of Lewy bodies that α-synuclein spreads within the nervous system in a
throughout the nervous system during progression of prion-like manner in synucleinopathies. Finally, we
Parkinson’s disease.3–9 discuss future research directions and their implications
How do synucleinopathies compare with prion diseases? for the development of novel therapeutic strategies.
The distinguishing feature of prion diseases resides in the
nature of the pathogen that spreads not only within the Prion propagation
affected organism but also between and within human Prion diseases in animals, such as scrapie and bovine
beings and animals. Unlike conventional pathogenic spongiform encephalopathy, are characterised by
organisms, such as viruses, bacteria, or yeast, a protein non-viral spread of diseased proteins from one animal to
was identified as the infectious and pathogenetic agent.10 another. The disease-causing pathogens can be
Although there is no evidence so far to support inter- transmitted infectiously within and between species.
individual or interspecies transmission of synucleino- Additionally, several familial and sporadic forms of
pathies, we discuss current evidence that α-synuclein human prion disease are well characterised, such as fatal
could act in a prion-like manner to transfer between cells familial insomnia and Creutzfeldt-Jakob disease.11 Once a
within an organism. Hence, we use the term prion-like to human being or animal acquires a prion disease,
indicate that α-synuclein might share some features with prognosis is poor; patients with Creutzfeldt-Jakob disease
prions, albeit without infectivity properties. typically die within a year after disease onset.12 Prion
diseases are progressive and incurable, and the prions
themselves are fairly resistant to methods that destroy
PrPSc viruses and bacteria.10,13
Genetic, dominantly inherited human prion diseases
PrPc are caused by mutations in the prion protein (PrP) gene
? (PRNP).14–16 PrP is expressed ubiquitously and its
functions are under active investigation;17,18 mice without
PrP have grossly normal development but are resistant to
scrapie.19–21 The typical cellular form of PrP is designated
PrPc, to distinguish it from the alternatively folded and
disease-related scrapie isoform (PrPSc). These two
isoforms share the same aminoacid sequence but differ
Figure 1: Model of templating and cell-to-cell transfer of prion proteins in their secondary structures: PrPc is rich in α-helices,
Exogenous PrPSc (red), acting as a seed, recruits intracellular PrPC (black) and
converts its three-dimensional structure into an elongated PrPSc, thus extending
whereas β-sheets dominate the structure of PrPSc.22,23
the amyloid aggregate. The aggregates transfer intercellularly via an unknown Additionally, PrPSc assumes different structures according
mechanism. to which prion disease it causes.11,24

1128 www.thelancet.com/neurology Vol 9 November 2010


Personal View

During sporadic, genetic, or infectious means of nanotubes have been suggested to participate in PrPSc
disease acquisition, once the change in conformation propagation not only within neurons in the CNS but
that creates PrPSc takes place, this protein isoform acts as also from the lymphoid system to the peripheral
a template (also termed a seed), recruiting PrPc into nervous system.30
aggregates and causing its conversion to PrPSc (figure 1).
Details of this transition from PrPc to PrPSc are currently Similarities of prion diseases and
unclear, but antibody and computational studies could neurodegenerative diseases
pave the way for ultrastructural determinations of The lesions that characterise Alzheimer’s disease
intermediate PrP structures.25,26 Accumulation of histopathologically are senile plaques (which consist of
aggregates of PrPSc and subsequent breakage of the extracellular aggregated amyloid β peptides) and
amyloid chains results in generation of more seeds, neurofibrillary tangles (composed of intracellular
which cause accelerated accumulation of PrPSc within polymers of tau protein). Echoing the disease-associated
cells and propagation between cells, leading to widespread conformations of PrPSc, aggregation-prone amyloid β
accumulation of PrPSc and, thus, disease progression.24,27 (derived from amyloid precursor protein) adopts β-sheet
The ability of the amyloid aggregates to break is vital; structures typically seen in all amyloid plaques.38 In view
brittle prion strains are most toxic, because they of these commonalities, several research groups have
contribute extra free ends for recruitment of PrPc and reported prion-like transmission for both amyloid β and
amyloid extension.24,27 tau.39–42 One group showed that brain extracts from a
Transfer of PrPSc from one cell to another can take place patient with Alzheimer’s disease—when injected into
via various processes. Mechanisms mediated by receptors, the brains of transgenic mice that expressed amyloid
exosomes, and tunnelling nanotubes have all been precursor protein—could induce aggregation
suggested to have a role.28–33 For example, neuronal and deposition of amyloid β.39 Implantation of
LDL-receptor-related protein 1 mediates endocytosis of amyloid-β-contaminated steel wires into the hippocampus
PrPSc from the extracellular space.31 and cortex of transgenic mice is sufficient to transmit
Active research into exosomes has provided new insight amyloid β.40 Similarly, healthy mice that express non-
into protein transfer mechanisms. Both PrPSc and PrPc mutant human tau develop pathogenic tau inclusions in
can be released from non-neuronal29 and neuronal their brains after injection with brain extracts from
cells28,32,33 and may be associated with exosomes. Exosomes mutant human tau-expressing mice.42 Findings of studies
are small secreted vesicles that derive from the endosomal also indicate that aggregates of misfolded tau can move
pathway.34 Thus, as a first step, proteins endocytosed at from the extracellular space into cultured cells and
the plasma membrane are internalised in endocytic between cells.41 Taken together, these results suggest that
vesicles that fuse with early endosomes in the cytoplasm. exogenous aggregates of amyloid β or tau can induce
During maturation of early endosomes, the molecules aggregation or formation of inclusions containing
they contain are sorted into smaller vesicles that bud endogenously expressed amyloid β or tau, possibly
from the endosomal membrane into the lumen. When leading to subsequent spreading of pathologies to
these multiple intralumenal vesicles are secreted neighbouring cells.
extracellularly through exocytosis, they are called
exosomes. Once in the extracellular space, exosomes can Symptoms and neuropathology of
be taken up by neighbouring cells, probably after fusion synucleinopathies
with the outer membrane of the recipient cell.34 Exosomes Similar to prion diseases and Alzheimer’s disease,
carrying PrPSc are infectious in vitro and in vivo29,33 and disorders characterised by Lewy body formation—such
have been proposed as a vehicle for propagation of PrPSc as Parkinson’s disease, dementia with Lewy bodies, and
from one cell to another.35 multiple system atrophy—involve aggregation of the
Tunnelling nanotubes constitute another non- misfolded protein α-synuclein. Although these
conventional intercellular communication pathway that synucleinopathies share the presence of Lewy bodies and
has been implicated in PrPSc propagation.30 These some symptoms, they also have many differences.43
nanotubes are thin extensions of surface membrane The most common synucleinopathy, Parkinson’s
that connect cells over long distances. They are formed disease, is characterised by loss of pigmented dopamine
either by actin-driven protrusion from one cell to neurons in the substantia nigra pars compacta and by the
another and subsequent membrane fusion or during cardinal symptoms of bradykinesia, rigidity, and resting
cell division.36 Tunnelling nanotubes mediate spread of tremor. Although traditional thinking emphasises motor
PrPSc between co-cultured neuronal cells and from deficits in Parkinson’s disease, many non-motor
bone-marrow-derived dendritic cells to primary symptoms have been highlighted, such as loss of
neurons.30 In bovine spongiform encephalopathy, olfaction, constipation, sleep disturbances, impotence,
ingested PrPSc transfers from the gut to the lymphoid and cognitive dysfunction.44 The average survival of an
system, then to the peripheral nervous system, and affected individual from the first signs of Parkinson’s
finally to the CNS.37 In this disease, tunnelling disease is two to three decades.

www.thelancet.com/neurology Vol 9 November 2010 1129


Personal View

Misfolded and post-translationally modified α-synuclein neurons. Multiple system atrophy has been classified
is the primary proteinaceous component of Lewy bodies into separate subtypes depending on the predominance
and Lewy neurites,2 the typical intracytoplasmic of symptoms.49 The disease course is shorter than that of
inclusions that develop in the cell body and neurites, people with idiopathic Parkinson’s disease, with most
respectively, of affected neurons. Whether Lewy bodies patients living less than a decade after diagnosis.
and Lewy neurites are toxic or neuroprotective is unclear
and the subject of much debate.45 Lewy bodies and Lewy In-vivo evidence for α-synuclein spread
neurites are found in patients with Parkinson’s disease, α-synuclein is an abundant protein in the brain, found in
dementia with Lewy bodies, and multiple system atrophy most cellular compartments and enriched at presynaptic
but are distributed according to different patterns.43 As terminals.50,51 It is believed to have a role in vesicular
described in detail below, Lewy bodies are present in transport and neurotransmitter release, although its
brainstem neurons early in Parkinson’s disease and then exact functions are unknown.2 The gene encoding human
in the cortex as disease progresses.46–48 α-synuclein (SNCA) can carry rare mutations or can be
Patients with dementia with Lewy bodies survive for duplicated or triplicated, changes which are all linked to
about 5–8 years and show early dementia, concurrent dominant forms of inherited Parkinson’s disease and
with akinetic and rigid motor symptoms.43 Dopaminergic parkinsonism.52–56 α-synuclein is present in the
neurons in the substantia nigra and cholinergic neurons cerebrospinal fluid and plasma of both controls and
in the nucleus basalis of Meynert both degenerate in patients with neurodegenerative diseases,57–59 suggestive
people with this disorder. Lewy bodies are more of exocytosis. Concentrations of α-synuclein in neuronal
widespread and abundant than in other synucleino- cell bodies increase during healthy ageing in human
pathies, and amyloid plaques are also present in the beings and monkeys.60 Thus, a raised cytoplasmic
brain of these patients.43 concentration of α-synuclein in neuronal cell bodies,
Individuals with multiple system atrophy are diagnosed both as a result of ageing and of gene duplications and
on the basis of a combination of ataxia, parkinsonism, triplications, seems to be a disease risk factor.
and autonomic dysfunction due to neuron loss in the
olivopontocerebellar, striatonigral, brainstem, and Dual-hit hypothesis
autonomic systems. The disease is characterised by Braak and co-workers46–48 have suggested that α-synuclein
cytoplasmic inclusions found in glia and occasionally in pathology spreads throughout the nervous system in

Neocortex
Cognitive decline

Midbrain
Motor symptoms
Olfactory bulb
Anosmia

Gut
Constipation Caudal brainstem
Autonomic symptoms

Figure 2: Dual-hit hypothesis of propagation of synucleinopathy during Parkinson’s disease


Parkinson’s disease-associated neuropathology originates in the gut (first hit) or the nose (olfactory bulb; second hit) and then propagates to the caudal brainstem
and the temporal lobe. Lewy body pathology then ascends to midbrain structures and cortical areas. Blue arrows depict the proposed ascending progression of
Parkinson’s disease pathology. Boxes indicate affected systems and main associated symptoms.

1130 www.thelancet.com/neurology Vol 9 November 2010


Personal View

Dissection of ventral Transplant preparation Grafting procedure Immunosuppression


mesencephalic tissue Fresh or hibernated tissue is Stereotactic injection in caudate Ciclosporin for 6 months or long-term
One to eight donor embryos homogenised into cell suspension and/or putamen triple drug therapy (ciclosporin, azathioprine,
used as a source of tissue or small tissue pieces Three to eight injection tracts per prednisolone) to prevent rejection
striatum

20 μm

Figure 3: Grafting of neurons into brains of patients with Parkinson’s disease


(A) Summary of the procedure used to prepare long-term grafts.65–67 (B) Immunostaining with α-synuclein antibodies of sections from a 16-year-old graft (left panel).
Lewy bodies (arrows) in the graft are similar to those seen in the substantia nigra of the host (right panel).

Parkinson’s disease according to a stereotypic pattern Lewy bodies and Lewy neurites have been detected in
following long unmyelinated axons of known anatomical tufted neurons47 and mitral cells47,63 in the olfactory bulb of
pathways (figure 2). According to this so-called dual-hit patients with Parkinson’s disease; mitral cells receive
hypothesis,61,62 Parkinson’s disease pathology originates direct input from neurons of the olfactory epithelium.
in the nose and foregut after inhalation of an unknown Lewy body pathology is also apparent all along the
neurotropic pathogen and subsequent swallowing of olfactory pathway (anterior olfactory nucleus, olfactory
nasal mucus in saliva. Among many theories and tubercle, and cortices),64 even though the olfactory
hypotheses, Braak and colleagues speculated that epithelium itself seems devoid of α-synuclein aggregates.65
“unconventional pathogens with prion-like properties” In the foregut, Lewy bodies and Lewy neurites have been
might induce spreading of Parkinson’s disease found in enteric nerve cell plexa in patients with
pathology.61,62 After crossing the epithelium, this Parkinson’s disease.66
pathogenic agent could get access to and be transported After reaching the CNS via nasal and gastric routes,
in an anterograde direction along axons of neurons Lewy pathology has been proposed to ascend from the
projecting from the olfactory epithelium to the temporal medulla oblongata to midbrain structures, including the
lobe, and retrogradely from the enteric epithelium via substantia nigra, and finally to cortical areas, along a
sympathetic fibres in the vagus nerve to the CNS network of neurons interconnecting all these regions
(figure 2).61,62 (figure 2).46–48 In view of the direct anatomical connection

www.thelancet.com/neurology Vol 9 November 2010 1131


Personal View

between the olfactory system and the temporal lobe of carrying Lewy bodies is functionally impaired. In one
the neocortex, temporal structures would be presumed to individual, Lewy bodies were noted in 1·9% and 5·0% of
display Lewy bodies and Lewy neurites earlier if inhalation pigmented neurons, which were grafted (into the right
were the predominant mode of pathogen entry. Thus, and left striatum) 12 and 16 years, respectively, before
oral ingestion might be the main method of pathogen death.77 40% of transplanted dopaminergic neurons
access to the body. Moreover, the topography of Lewy showed cytoplasmic α-synuclein staining in the
pathology could be correlated with the extent and severity 12-year-old grafts versus 80% in the 16-year-old grafts.75
of symptoms. The chronology of appearance of In other studies, cell bodies in 18-month-old grafts were
Parkinson’s disease symptoms can be described briefly devoid of any immunoreactivity to α-synuclein whereas
as follows: the presymptomatic phase occurs first, and it neurons in 4-year-old and 16-year-old transplants showed
is loosely defined by non-motor symptoms such as cytoplasmic α-synuclein staining and α-synuclein-positive
olfactory deficits, sleep disturbances, constipation, and spherical aggregates, respectively.76
erectile dysfunction, all of which can occasionally first Taken together, these observations suggest that Lewy
arise at later disease stages;67,68 the motor symptom phase, body development in the grafted neurons is a gradual
occurs later, and includes bradykinesia and rigidity; and process, and that whole Lewy bodies do not move between
finally, the late complication phase is characterised by cells.76,77 Prion-like spread of misfolded α-synuclein,
cognitive decline and psychiatric symptoms.69 During the which might act as a seed and lead to progressive
early non-motor phase of Parkinson’s disease, Lewy accumulation of α-synuclein, could result in formation of
bodies and Lewy neurites are restricted to the peripheral Lewy bodies in transplanted neurons.5,78
enteric system,66 the olfactory bulb, and the caudal
brainstem.47 Next, Lewy pathology appears in the mid- Animal models of synucleinopathy propagation
brain, especially in the substantia nigra pars compacta, Researchers have reported a mouse model79 that
within the period of the onset of motor symptoms, and recapitulates some aspects of the pathological staging
eventually it reaches the neocortex at later stages described by Braak and collaborators in patients with
characterised by cognitive impairment (figure 2).47 In the Parkinson’s disease.46,47 The model is based on
following sections, we discuss arguments for and against intragastric administration of rotenone, an inhibitor of
the Braak hypothesis. complex I of the mitochondrial respiratory chain.
Rotenone is a widely used pesticide that has been linked
Lewy pathology in neural grafts to Parkinson’s disease in epidemiological studies.80–82 It
In some patients with Parkinson’s disease, induces formation of large perinuclear α-synuclein
transplantation of embryonic dopamine neurons was inclusions in cultured cells expressing human
started more than two decades ago in an attempt to α-synuclein.83 Chronic intravenous84 or intraperitoneal85
restore dopaminergic neurotransmission (figure 3A).70–75 administration of rotenone in rats has been reported to
Findings of histological studies on post-mortem tissue trigger α-synuclein aggregation in nigral neurons,
revealed Lewy bodies within the transplanted probably directly, in view of the capacity of rotenone to
dopaminergic neurons in eight patients who were cross the blood–brain barrier. In the new mouse model,
operated on in four centres worldwide and who died there is progressive development of α-synuclein
more than a decade after surgery (figure 3B).73–75 The inclusions, starting in the enteric nervous system and
Lewy bodies in the grafts shared classic features with then arising sequentially in spinal cord, caudal
those in the substantia nigra of the host, including brainstem, and the substantia nigra79—ie, the
α-synuclein and ubiquitin immunoreactivity. same structures that show Lewy pathology in
Further studies characterised Lewy pathology in Parkinson’s disease. One possible explanation for
transplanted neurons, with detection of α-synuclein these results lies in the induction of α-synuclein
phosphorylated on serine 129,75,76 characteristic β-sheet aggregation in enteric nerves by intragastric
structures stained by thioflavine S,76,77 and α-synuclein administration of the toxin, and then propagation of
fibrils revealed by electron microscopy.77 Tyrosine the pathology to vulnerable CNS areas via an unknown
hydroxylase is rarely expressed in grafted neurons mechanism that might include intercellular transfer of
containing Lewy bodies, although the fact that Lewy α-synuclein aggregates.
bodies are seen in grafted cells containing neuromelanin The first experimental evidence for intercellular
suggests that they are present in catecholaminergic transfer of α-synuclein in vivo came from a study on
(presumed dopaminergic) neurons. These observations— mouse cortical neuronal stem cells grafted to the
together with a reported decrease of dopamine hippocampus of transgenic mice overexpressing human
transporter immunoreactivity,73,74,76 and a reduction in α-synuclein.86 At 1–4 weeks after grafting, 2·5–15% of
intensity of tyrosine hydroxylase immunostaining74,76 in transplanted cells showed human α-synuclein staining,
cells with Lewy pathology within a transplant from a indicating that α-synuclein can transfer from the host
patient who survived 14 years after transplantation— brain to a graft of proliferating neuronal progenitors.
could indicate that the subset of transplanted cells These results bear some resemblance to clinical

1132 www.thelancet.com/neurology Vol 9 November 2010


Personal View

findings with neural grafts described above, but differ


in the sense that no large aggregates resembling Lewy A B
Donor cell
bodies were seen and that proliferating rather than
post-mitotic cells were grafted.
Natively unfolded
Finally, in a report of transgenic mice that overexpress monomer
human α-synuclein under the control of a neuronal
Release
promoter, α-synuclein accumulation was detected not α-synuclein
only in neurons but also in glia.87 Because the promoter Self-aggregation
was neuron-specific, glial cells were devoid of human
α-synuclein mRNA. These results suggest that glial cells
that do not express the α-synuclein transgene take up
α-synuclein released by neurons. The low basal expression
level of α-synuclein in glia88 raises the question of the
origin of glial α-synuclein inclusions characterising Soluble oligomer

multiple system atrophy. Propagation of α-synuclein Recipient cell


from neurons to glial cells, perhaps relying on prion-like
properties of α-synuclein, could account for the
appearance of widespread glial α-synuclein inclusions Entry
during progression of multiple system atrophy.
Insoluble amyloid fibril Seeding
In-vitro studies
Structure of α-synuclein
During the oligomerisation of α-synuclein into fibrils,
the structure of α-synuclein undergoes major
modifications that could underlie its toxic effects.
Accumulating evidence suggests that α-synuclein
Figure 4: Putative mechanisms of α-synuclein prion-like propagation
oligomers, and possibly protofibrils, are the toxic species (A) The different species of α-synuclein assemblies coexist in a highly dynamic equilibrium. (B) According to the
that cause cell death.89 Therefore, deeper understanding prion-like hypothesis, a donor cell releases α-synuclein into the extracellular space via exocytosis or during its
of the structure of α-synuclein and its changes during death. Next, α-synuclein enters a recipient cell via passive membrane translocation or endocytosis. Alternative
mechanisms include exosomal release or transport along tunnelling nanotubes. Once in the recipient cell, the
oligomerisation could give fundamental insights into the transferred α-synuclein protein (blue) recruits the endogenous α-synuclein protein (red), induces misfolding, and
pathogenesis of synucleinopathies. α-synuclein is natively seeds aggregation. Green stars indicate possible targets for disease-modifying drugs to reduce, stop, or delay
unfolded at low concentrations but, on accumulation, it systematic spread of α-synuclein throughout the human body.
self-aggregates into soluble oligomers and can eventually
form insoluble fibrillar aggregates90–94 with a typical plasma in human beings,57,58 and in the medium of
amyloid nature (figure 4A).95 As with PrPSc and amyloid β, several neuronal culture models.57,98 The mechanism by
α-synuclein fibrils have unbranched morphology and an which α-synuclein is released from cells is unclear, but
antiparallel β-sheet structure,95 bind thioflavine S93 and exocytosis is likely to underlie this secretion (figure 4B),
Congo red, and are resistant to proteolysis.95 Similar to given that the process is inhibited at low temperature
PrPSc propagation, α-synuclein fibrillisation is a and α-synuclein can be detected in the lumen of vesicles
nucleation-dependent process starting with a lag-phase, isolated from rat brain or neuroblastoma cells.98,99
during which soluble α-synuclein oligomers assemble Furthermore, α-synuclein translocation to vesicles and
and form a nucleus.96 Aggregates of α-synuclein then subsequent vesicle release increase under conditions
grow rapidly around this nucleus during the growth or that promote its misfolding and could be part of a
elongation phase, until they reach a thermodynamic cellular quality-control system aimed at removal of
equilibrium with monomers in a steady-state phase.96 damaged proteins.99 Furthermore, researchers have
Moreover, aggregation of recombinant α-synuclein suggested that exosomes could have a role in secretion
monomers can be seeded by addition of α-synuclein of α-synuclein by cultured neuroblastoma cells.100
aggregates acting as exogenous nuclei.96 The three After release of α-synuclein, the next steps are crucial.
mutations of α-synuclein recorded in patients with What are the mechanisms by which cells take up
Parkinson’s disease—Ala53Thr, Ala30Pro, and extracellular α-synuclein? Similar to infection of new
Glu46Lys—accelerate this process, indicating a probable cells by PrPSc, extracellular α-synuclein can be internalised
role for α-synuclein misfolding and aggregation in by surrounding cells (figure 4B). Whereas recombinant
familial Parkinson’s disease.90,94,97 α-synuclein monomers have been suggested to
translocate passively across plasma membranes of
Possible mechanisms of release and uptake neuroblastoma cells,101,102 an endocytic process is probably
α-synuclein can access the extracellular space, consistent needed for internalisation of larger order α-synuclein
with its reported presence in cerebrospinal fluid and assemblages.102 Uptake of recombinant α-synuclein

www.thelancet.com/neurology Vol 9 November 2010 1133


Personal View

oligomers and fibrils is indeed inhibited at low neurodegenerative disorders. So far, in-vitro studies in
temperatures and by expression of a dominant negative which seeding has been shown have used highly
mutant of dynamin 1, a neuron-specific GTPase artificial techniques for introduction of α-synuclein into
important for endocytosis, suggesting that uptake takes cells,107,108 which are not compatible with physiological
place via an endocytic process.102 In co-cultured neuronal conditions. Similarly, studies that have shown seeding
cells, transmitted α-synuclein proteins colocalise with of α-synuclein in vivo—and even more importantly,
the endosomal GTPases Rab5a and Rab7,86 and in animal work on α-synuclein intercellular transfer—are
particular, Rab5a has been suggested to be vital for currently sparse. So far, published data are restricted to
α-synuclein endocytosis.103 In rat dopaminergic cells, the study mentioned above,86 in which proliferative
some internalised aggregates of α-synuclein are degraded neuronal stem cells are grafted into the hippocampus
by a Rab11a-dependent endosome-lysosome pathway of transgenic mice overexpressing human α-synuclein.
and others are resecreted through exocytosis.104 To date, no data are available to show that post-mitotic
In addition to α-synuclein transfer between neuronal dopaminergic neurons—the primary cell type affected
cells, glial cells might take up α-synuclein derived from in Parkinson’s disease—can take up α-synuclein
neurons via an endocytic mechanism that is blocked by released from donor neurons in an experimental animal
expression of a mutant form of dynamin 1.87 Moreover, model, or that transferred α-synuclein can trigger
Lee and colleagues87 showed formation of Lewy body-like formation of Lewy bodies.
aggregates after uptake of human α-synuclein into glial Even if future research establishes that α-synuclein
cells. Thus, primary astrocytes co-cultured with fulfils all criteria for a prion-like protein in culture and in
neuroblastoma cells overexpressing human α-synuclein animals, the relevance of these discoveries in relation to
develop inclusions of human α-synuclein, which stain the pathogenesis of synucleinopathies will still be
with thioflavine S and contain some proteins found in questionable. The hypothesis that misfolded α-synuclein
Lewy bodies, such as ubiquitin, the 20S proteasome accumulation is the central force driving the
α-subunit, and the chaperone protein HSP/HSC70.87 neurodegenerative process is not adopted unanimously
Taken together, the mechanism by which cells take up in the field, and the correlation proposed by Braak and
α-synuclein could be tightly linked with the putative next collaborators46–48 between the spread of Lewy pathology
steps of inclusion formation. and the temporal pattern of development of symptoms
has been questioned.109 Thus, α-synuclein misfolding and
Potential seeding effect of α-synuclein aggregation might alternatively be considered an
If intercellular transmission of aggregated α-synuclein is epiphenomenon of a pathological neuronal micro-
governed by a prion-like mechanism, the ultimate step environment. For example, differences in distribution of
would be the seeding effect—ie, recruitment of unfolded Lewy bodies in the synucleinopathies could be explained
α-synuclein proteins endogenously expressed by the by selective neuronal vulnerability in each disease,
acceptor cell and their conversion into misfolded forms underlying the characteristic temporal patterns of
that will aggregate around the nucleus of transmitted progression of pathology. Alternatively, neuro-
α-synuclein (figure 4B). A few studies have looked at the inflammation, which leads to formation of intraneuronal
seeding effect of recombinant α-synuclein. Oligomers of α-synuclein inclusions in animals110,111 and has been
α-synuclein, thought to be the toxic species,89 seed the detected in post-mortem analysis of brains of patients
aggregation of α-synuclein proteins endogenously with Parkinson’s disease,112 could lie upstream of
expressed by neuroblastoma cells105 and by primary α-synuclein aggregation in the cascade of pathogenic
cortical neurons105,106 in a time-dependent and events leading to progression of Parkinson’s disease.
concentration-dependent manner.106 In other studies,107,108 What makes prion-like propagation of α-synuclein an
artificial techniques (such as cationic liposome-based especially interesting hypothesis is that it can account for
transduction) to load cells with exogenous fibrillised the sequence of appearance of symptoms and the
α-synuclein proteins are needed for the seeding process findings on neuropathological changes in grafts of
to take place. patients with Parkinson’s disease.73–75 Thus, this
hypothesis remains a highly attractive and promising
Arguments against synucleinopathies being research axis.
prion-like disorders Could interindividual infectivity be another feature
As described in the previous section, evidence that shared by PrPSc and misfolded α-synuclein? A paucity of
α-synuclein can transfer between co-cultured neurons epidemiological evidence, or at least case reports,
in vitro is compelling, and studies have provided some suggesting that Parkinson’s disease can spread from one
insight into underlying mechanisms. Nonetheless, the person to another, however, argues against this notion.
subsequent seeding activity of transferred α-synuclein How do the prion-like properties of α-synuclein differ
is a crucial prerequisite for propagation of α-synuclein from those of PrPSc? Under what conditions might
pathology to take place during progression of α-synuclein act as an infectious agent? These questions
Parkinson’s disease and other Lewy body-related still need to be answered. By contrast with the instability

1134 www.thelancet.com/neurology Vol 9 November 2010


Personal View

of PrPSc assemblies, which readily break and generate


further free ends that act as seeds,23,24 the possible relative Search strategy and selection criteria
stability of α-synuclein fibrils might account for both We searched PubMed from January, 1994, to July, 2010, with
non-infectivity of α-synuclein and the typical disease the search terms: “alpha-synuclein”, “synuclein”, “Parkinson
course of the synucleinopathies (from several years to disease”, “prion”, “scrapie”, “Creutzfeldt-Jakob”, “dementia
decades), versus the fairly quick (within 1 year) duration of with lewy bodies”, “multiple systems atrophy”, “prion-like”,
prion disease. Alternatively, major differences in structure “Alzheimer disease”, “Braak hypothesis”, “non-motor
and cellular localisation between α-synuclein and PrP symptoms”, “neural grafting”, “rotenone”, “fibrillization”,
could account for the varying kinetics of conversion from “cell-to-cell transfer”, “exocytosis”, “endocytosis”, “seeding”,
normal to misfolded protein, and hence could affect the “amyloid”, “tau”, “nanotube”, “exosome”, “biomarker”, and
rate of disease progression. Similarly, variability in disease “English”. We largely targeted publications from the past
duration of the synucleinopathies (5–8 years for dementia 10 years but did not exclude commonly referenced, highly
with Lewy bodies after diagnosis, less than 10 years for regarded, and relevant older publications. Furthermore, we
multiple system atrophy, decades for Parkinson’s disease) examined the search results obtained above and selected the
might also rely on different forms of misfolded α-synuclein, most relevant for this Personal View.
with various stabilities or intermediate species along the
conversion process of normal into pathogenic protein,
even if no evidence for such heterogeneity in α-synuclein of α-synuclein are unlikely to enter the organism
proteins has been reported so far. through direct nasal inhalation. Instead, contact with
In summary, the hypothesis that progression of substances present in our environment—eg, specific
synucleinopathies relies on the same mechanisms as pesticides, which are known to be risk factors for
prion disorders is highly attractive. However, it has Parkinson’s disease80–82 and cause α-synuclein
potential drawbacks and alternatives, which are currently aggregation83—could be an initial event that triggers
under investigation. α-synuclein misfolding in the periphery. As a result, by
using exposure to a peripheral toxin and exploiting all
Conclusions and future perspectives the aforementioned knowledge about the putative
Connections between in-vitro studies and clinical prion-like mechanism of α-synuclein spread,
observations are important but in need of improvement development of animal models that faithfully reproduce
and extension. What are the next steps for basic science all the characteristics of Parkinson’s disease might be
research that will bring more insight to human disease possible. One alternative could be to manipulate
and, specifically, to synucleinopathies? selectively and transgenically the α-synuclein gene in
As described above, exocytosis and endocytosis seem the periphery and cause a regional synucleinopathy
to have important roles in cell-to-cell transfer of outside the CNS. Early gastrointestinal dysfunction and
misfolded α-synuclein. However, in view of the aggregates of α-synuclein in enteric nervous system
similarities between PrPSc and α-synuclein, other ganglia have been reported in transgenic mice carrying
mechanisms of cell-to-cell transfer reported to an artificial chromosome with Parkinson’s disease-linked
contribute to PrPSc propagation should also be human mutations (Ala53Thr or Ala30Pro) of the
considered. Both exosomes and tunnelling nanotubes α-synuclein gene.115 These models might mimic not
might contribute to intercellular transfer of PrPSc.28–30,32,33 only Parkinson’s disease-related neuropathological
A minute fraction of α-synuclein released from cultured changes and neurodegenerative events but also motor
neuroblastoma cells is associated with extracellular and non-motor symptoms, and, thus, they would be
vesicles99 and exosomal secretion has been reported.100 invaluable for assessment of the efficacy of new drugs
The relevance of α-synuclein-containing exosomes in for Parkinson’s disease at early preclinical stages.
other cell types, notably astrocytes (reported to secrete All current treatments for Parkinson’s disease, from
exosomes),113 are worth examining both in cell culture pharmaceutical to surgical, only alleviate motor
and in vivo. Furthermore, researchers revealed the symptoms;116 to date, no disease-modifying treatment has
presence of α-synuclein within tunnelling nanotubes been able to stop or greatly delay neurodegeneration.
that were interconnecting co-cultured glioblastoma Although early treatment with rasagiline—a monoamine
cells,114 warranting further investigation into whether oxidase type B inhibitor—might slow progression of
tunnelling nanotubes can convey α-synuclein from one Parkinson’s disease slightly, further clinical investigations
neuron to another. are needed to assess the efficacy of this drug.117
As discussed above, the dual-hit theory suggests that Accumulating evidence supporting prion-like
a pathogenic agent with prion-like properties penetrates properties of α-synuclein indicates that cell-to-cell
the body by the nasal or gastric route, or both, and then transfer of this protein might be worth targeting.
leads to propagation of Lewy bodies and Lewy neurites Intercellular transfer of misfolded proteins followed by
from the gut to the CNS.61,62 Clearly, misfolded permissive templating could be a common pathogenetic
α-synuclein is a relevant candidate. Pathogenic forms mechanism in several cerebral proteinopathies,3,4,6,8

www.thelancet.com/neurology Vol 9 November 2010 1135


Personal View

making this approach to new treatments even more 10 Prusiner SB. Novel proteinaceous infectious particles cause scrapie.
wide-reaching. Putative novel therapies that inhibit Science 1982; 216: 136–44.
11 Prusiner SB. Shattuck lecture: neurodegenerative diseases and
transfer of disease-related proteins should ideally be prions. N Engl J Med 2001; 344: 1516–26.
initiated even before the individual has met current 12 Appleby BS, Appleby KK, Crain BJ, Onyike CU, Wallin MT,
diagnostic criteria for Parkinson’s disease—ie, at the Rabins PV. Characteristics of established and proposed sporadic
Creutzfeldt-Jakob disease variants. Arch Neurol 2009; 66: 208–15.
very beginning of progression of Lewy pathology. These
13 Diener TO, McKinley MP, Prusiner SB. Viroids and prions.
limitations will require development of biomarkers for Proc Natl Acad Sci USA 1982; 79: 5220–24.
early Parkinson’s disease—eg, brain imaging or 14 Hsiao K, Baker HF, Crow TJ, et al. Linkage of a prion protein
biological fluid biomarkers currently under development missense variant to Gerstmann-Straussler syndrome. Nature 1989;
338: 342–45.
for differential diagnosis118 and identification of a set of 15 Medori R, Tritschler HJ, LeBlanc A, et al. Fatal familial insomnia, a
non-motor symptoms (eg, olfactory or gastrointestinal prion disease with a mutation at codon 178 of the prion protein
dysfunction) with high predictive value for Parkinson’s gene. N Engl J Med 1992; 326: 444–49.
disease. The theory of prion-like spread of misfolded 16 Jackson WS, Borkowski AW, Faas H, et al. Spontaneous generation
of prion infectivity in fatal familial insomnia knockin mice. Neuron
α-synuclein suggests the process will take place in three 2009; 63: 438–50.
steps: release, uptake, and seeding. Inhibition of 17 Steele AD, Zhou Z, Jackson WS, et al. Context dependent
α-synuclein spread could interfere with one or another neuroprotective properties of prion protein (PrP). Prion 2009;
3: 240–49.
of these processes (figure 4B). Exocytosis and endocytosis 18 Si K, Choi YB, White-Grindley E, Majumdar A, Kandel ER.
inhibitors might be interesting candidates to investigate, Aplysia CPEB can form prion-like multimers in sensory neurons
because both of these cellular mechanisms are proposed that contribute to long-term facilitation. Cell 2010; 140: 421–35.
19 Prusiner SB, Groth D, Serban A, et al. Ablation of the prion protein
to participate in α-synuclein intercellular transfer,98,99,102,103 (PrP) gene in mice prevents scrapie and facilitates production of
but, of course, other treatments specific to exosomes, anti-PrP antibodies. Proc Natl Acad Sci USA 1993; 90: 10608–12.
tunnelling nanotubes, or receptors should be tested. In 20 Sailer A, Bueler H, Fischer M, Aguzzi A, Weissmann C. No
propagation of prions in mice devoid of PrP. Cell 1994; 77: 967–68.
conclusion, research into the mechanisms of α-synuclein
21 Weissmann C, Bueler H, Fischer M, Sailer A, Aguzzi A, Aguet M.
cell-to-cell transfer in combination with other ongoing PrP-deficient mice are resistant to scrapie. Ann N Y Acad Sci 1994;
strategies could lead to promising advances in clinical 724: 235–40.
treatment of synucleinopathies. 22 Riek R, Hornemann S, Wider G, Billeter M, Glockshuber R,
Wüthrich K. NMR structure of the mouse prion protein domain
Contributors PrP(121-321). Nature 1996; 382: 180–82.
EA and JAS wrote the manuscript and prepared the figures, and CH, 23 Wille H, Bian W, McDonald M, et al. Natural and synthetic prion
JYL, and PB edited the paper. structure from X-ray fiber diffraction. Proc Natl Acad Sci USA 2009;
106: 16990–95.
Conflicts of interest
PB has received honoraria from Pfizer, H Lundbeck A/S, Roche, and 24 Tanaka M, Collins SR, Toyama BH, Weissman JS. The physical
basis of how prion conformations determine strain phenotypes.
Orion Pharma, consultancy fees from Teva Pharmaceutical Industries
Nature 2006; 442: 585–89.
and H Lundbeck A/S, and research support from Teva. The other
25 Yam AY, Gao CM, Wang X, Wu P, Peretz D. The octarepeat region
authors have no conflicts of interest.
of the prion protein is conformationally altered in PrP(Sc).
Acknowledgments PLoS One 2010; 5: e9316.
We thank the Human Frontier Science Program, the ERA-net Neuron 26 Bergasa-Caceres F, Rabitz HA. Low entropic barrier to the
program MIPROTRAN, the Michael J Fox Foundation for Parkinson’s hydrophobic collapse of the prion protein: effects of intermediate
Research, the Swedish Brain Foundation, the Swedish Parkinson states and conformational flexibility. J Phys Chem A 2010;
Foundation, the Ragnar and Thorsten Söderbergs Foundation, 114: 6978–82.
Anna-Lisa Rosenberg Foundation, and the Swedish Research Council. 27 Colby DW, Giles K, Legname G, et al. Design and construction of
The funding sources had no role in preparation of this paper. diverse mammalian prion strains. Proc Natl Acad Sci USA 2009;
106: 20417–22.
References 28 Alais S, Simoes S, Baas D, et al. Mouse neuroblastoma cells release
1 Uversky VN. α-synuclein misfolding and neurodegenerative prion infectivity associated with exosomal vesicles. Biol Cell 2008;
diseases. Curr Protein Pept Sci 2008; 9: 507–40. 100: 603–15.
2 Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, 29 Février B, Vilette D, Archer F, et al. Cells release prions in
Goedert M. α-synuclein in Lewy bodies. Nature 1997; 388: 839–40. association with exosomes. Proc Natl Acad Sci USA 2004;
3 Aguzzi A. Cell biology: beyond the prion principle. Nature 2009; 101: 9683–88.
459: 924–25. 30 Gousset K, Schiff E, Langevin C, et al. Prions hijack tunnelling
4 Aguzzi A, Rajendran L. The transcellular spread of cytosolic nanotubes for intercellular spread. Nat Cell Biol 2009; 11: 328–36.
amyloids, prions, and prionoids. Neuron 2009; 64: 783–90. 31 Jen A, Parkyn CJ, Mootoosamy RC, et al. Neuronal low-density
5 Angot E, Brundin P. Dissecting the potential molecular mechanisms lipoprotein receptor-related protein 1 binds and endocytoses prion
underlying α-synuclein cell-to-cell transfer in Parkinson’s disease. fibrils via receptor cluster 4. J Cell Sci 2010; 123: 246–55.
Parkinsonism Relat Disord 2009; 15 (suppl 3): S143–47. 32 Veith NM, Plattner H, Stuermer CA, Schulz-Schaeffer WJ,
6 Frost B, Diamond MI. Prion-like mechanisms in neurodegenerative Bürkle A. Immunolocalisation of PrPSc in scrapie-infected N2a
diseases. Nat Rev Neurosci 2010; 11: 155–59. mouse neuroblastoma cells by light and electron microscopy.
7 Olanow CW, Prusiner SB. Is Parkinson’s disease a prion disorder? Eur J Cell Biol 2009; 88: 45–63.
Proc Natl Acad Sci USA 2009; 106: 12571–72. 33 Vella LJ, Sharples RA, Lawson VA, Masters CL, Cappai R, Hill AF.
8 Brundin P, Melki R, Kopito R. Prion-like transmission of protein Packaging of prions into exosomes is associated with a novel
aggregates in neurodegenerative diseases. Nat Rev Mol Cell Biol pathway of PrP processing. J Pathol 2007; 211: 582–90.
2010; 11: 301–07. 34 Simons M, Raposo G. Exosomes: vesicular carriers for intercellular
9 Goedert M, Clavaguera F, Tolnay M. The propagation of prion-like communication. Curr Opin Cell Biol 2009; 21: 575–81.
protein inclusions in neurodegenerative diseases. Trends Neurosci 35 Février B, Vilette D, Laude H, Raposo G. Exosomes: a bubble ride
2010; 33: 317–25. for prions? Traffic 2005; 6: 10–17.

1136 www.thelancet.com/neurology Vol 9 November 2010


Personal View

36 Davis DM, Sowinski S. Membrane nanotubes: dynamic long- 61 Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: a dual-hit
distance connections between animal cells. Nat Rev Mol Cell Biol hypothesis. Neuropathol Appl Neurobiol 2007; 33: 599–614.
2008; 9: 431–36. 62 Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: the dual
37 Mabbott NA, MacPherson GG. Prions and their lethal journey to hit theory revisited. Ann N Y Acad Sci 2009; 1170: 615–22.
the brain. Nat Rev Microbiol 2006; 4: 201–11. 63 Daniel SE, Hawkes CH. Preliminary diagnosis of Parkinson’s
38 Cohen AS, Connors LH. The pathogenesis and biochemistry of disease by olfactory bulb pathology. Lancet 1992; 340: 186.
amyloidosis. J Pathol 1987; 151: 1–10. 64 Ubeda-Bañon I, Saiz-Sanchez D, de la Rosa-Prieto C,
39 Meyer-Luehmann M, Coomaraswamy J, Bolmont T, et al. Argandoña-Palacios L, Garcia-Muñozguren S, Martinez-Marcos A.
Exogenous induction of cerebral β-amyloidogenesis is governed by alpha-Synucleinopathy in the human olfactory system in
agent and host. Science 2006; 313: 1781–84. Parkinson’s disease: involvement of calcium-binding protein- and
40 Eisele YS, Bolmont T, Heikenwalder M, et al. Induction of cerebral substance P-positive cells. Acta Neuropathol 2010; 119: 723–35.
β-amyloidosis: intracerebral versus systemic Aβ inoculation. 65 Duda JE, Shah U, Arnold SE, Lee VM, Trojanowski JQ. The
Proc Natl Acad Sci USA 2009; 106: 12926–31. expression of α-, β-, and γ-synucleins in olfactory mucosa from
41 Frost B, Jacks RL, Diamond MI. Propagation of tau misfolding from patients with and without neurodegenerative diseases.
the outside to the inside of a cell. J Biol Chem 2009; 284: 12845–52. Exp Neurol 1999; 160: 515–22.
42 Clavaguera F, Bolmont T, Crowther RA, et al. Transmission and 66 Braak H, de Vos RA, Bohl J, Del Tredici K. Gastric α-synuclein
spreading of tauopathy in transgenic mouse brain. Nat Cell Biol immunoreactive inclusions in Meissner’s and Auerbach’s plexuses
2009; 11: 909–13. in cases staged for Parkinson’s disease-related brain pathology.
43 Halliday GM, McCann H. The progression of pathology in Neurosci Lett 2006; 396: 67–72.
Parkinson’s disease. Ann N Y Acad Sci 2010; 1184: 188–95. 67 Chaudhuri KR, Naidu Y. Early Parkinson’s disease and non-motor
44 Barone P, Antonini A, Colosimo C, et al. The PRIAMO study: issues. J Neurol 2008; 255 (suppl 5): 33–38.
a multicenter assessment of nonmotor symptoms and their impact 68 Park A, Stacy M. Non-motor symptoms in Parkinson’s disease.
on quality of life in Parkinson’s disease. Mov Disord 2009; J Neurol 2009; 256 (suppl 3): 293–98.
24: 1641–49. 69 Maetzler W, Liepelt I, Berg D. Progression of Parkinson’s disease
45 Olanow CW, Perl DP, DeMartino GN, McNaught KStP. Lewy-body in the clinical phase: potential markers. Lancet Neurol 2009;
formation is an aggresome-related process: a hypothesis. 8: 1158–71.
Lancet Neurol 2004; 3: 496–503. 70 Mendez I, Vinuela A, Astradsson A, et al. Dopamine neurons
46 Braak H, Del Tredici K, Bratzke H, Hamm-Clement J, implanted into people with Parkinson’s disease survive without
Sandmann-Keil D, Rüb U. Staging of the intracerebral inclusion pathology for 14 years. Nat Med 2008; 14: 507–09.
body pathology associated with idiopathic Parkinson’s disease 71 Björklund A, Dunnett SB, Brundin P, et al. Neural transplantation
(preclinical and clinical stages). J Neurol 2002; 249 (suppl 3): III/1–5. for the treatment of Parkinson’s disease. Lancet Neurol 2003;
47 Braak H, Del Tredici K, Rüb U, de Vos RA, Jansen Steur EN, 2: 437–45.
Braak E. Staging of brain pathology related to sporadic Parkinson’s 72 Olanow CW, Kordower JH, Lang AE, Obeso JA. Dopaminergic
disease. Neurobiol Aging 2003; 24: 197–211. transplantation for Parkinson’s disease: current status and future
48 Braak H, Ghebremedhin E, Rüb U, Bratzke H, Del Tredici K. Stages prospects. Ann Neurol 2009; 66: 591–96.
in the development of Parkinson’s disease-related pathology. 73 Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy
Cell Tissue Res 2004; 318: 121–34. body-like pathology in long-term embryonic nigral transplants in
49 Stefanova N, Bücke P, Duerr S, Wenning GK. Multiple system Parkinson’s disease. Nat Med 2008; 14: 504–06.
atrophy: an update. Lancet Neurol 2009; 8: 1172–78. 74 Kordower JH, Chu Y, Hauser RA, Olanow CW, Freeman TB.
50 Uéda K, Fukushima H, Masliah E, et al. Molecular cloning of cDNA Transplanted dopaminergic neurons develop PD pathologic
encoding an unrecognized component of amyloid in Alzheimer changes: a second case report. Mov Disord 2008; 23: 2303–06.
disease. Proc Natl Acad Sci USA 1993; 90: 11282–86. 75 Li JY, Englund E, Holton JL, et al. Lewy bodies in grafted neurons in
51 Jakes R, Spillantini MG, Goedert M. Identification of two distinct subjects with Parkinson’s disease suggest host-to-graft disease
synucleins from human brain. FEBS Lett 1994; 345: 27–32. propagation. Nat Med 2008; 14: 501–03.
52 Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the 76 Chu Y, Kordower JH. Lewy body pathology in fetal grafts.
α-synuclein gene identified in families with Parkinson’s disease. Ann N Y Acad Sci 2010; 1184: 55–67.
Science 1997; 276: 2045–47. 77 Li JY, Englund E, Widner H, et al. Characterization of Lewy body
53 Krüger R, Kuhn W, Müller T, et al. Ala30Pro mutation in the gene pathology in 12- and 16-year-old intrastriatal mesencephalic grafts
encoding α-synuclein in Parkinson’s disease. Nat Genet 1998; surviving in a patient with Parkinson’s disease. Mov Disord 2010;
18: 106–08. 25: 1091–96.
54 Singleton AB, Farrer M, Johnson J, et al. α-synuclein locus 78 Brundin P, Li JY, Holton JL, Lindvall O, Revesz T. Research in
triplication causes Parkinson’s disease. Science 2003; 302: 841. motion: the enigma of Parkinson’s disease pathology spread.
55 Chartier-Harlin M-C, Kachergus J, Roumier C, et al. α-synuclein Nat Rev Neurosci 2008; 9: 741–45.
locus duplication as a cause of familial Parkinson’s disease. 79 Pan-Montojo F, Anichtchik O, Dening Y, et al. Progression of
Lancet 2004; 364: 1167–69. Parkinson’s disease pathology is reproduced by intragastric
56 Zarranz JJ, Alegre J, Gómez-Esteban JC, et al. The new mutation, administration of rotenone in mice. PLoS One 2010; 5: e8762.
E46K, of α-synuclein causes Parkinson and Lewy body dementia. 80 Elbaz A, Clavel J, Rathouz PJ, et al. Professional exposure to
Ann Neurol 2004; 55: 164–73. pesticides and Parkinson disease. Ann Neurol 2009; 66: 494–504.
57 El-Agnaf OM, Salem SA, Paleologou KE, et al. α-synuclein 81 Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Richardson RJ.
implicated in Parkinson’s disease is present in extracellular The risk of Parkinson’s disease with exposure to pesticides,
biological fluids, including human plasma. FASEB J 2003; farming, well water, and rural living. Neurology 1998; 50: 1346–50.
17: 1945–47. 82 Hatcher JM, Pennell KD, Miller GW. Parkinson’s disease and
58 El-Agnaf OM, Salem SA, Paleologou KE, et al. Detection of pesticides: a toxicological perspective. Trends Pharmacol Sci 2008;
oligomeric forms of α-synuclein protein in human plasma as 29: 322–29.
a potential biomarker for Parkinson’s disease. FASEB J 2006; 83 Lee HJ, Shin SY, Choi C, Lee YH, Lee SJ. Formation and removal of
20: 419–25. α-synuclein aggregates in cells exposed to mitochondrial inhibitors.
59 Noguchi-Shinohara M, Tokuda T, Yoshita M, et al. CSF α-synuclein J Biol Chem 2002; 277: 5411–17.
levels in dementia with Lewy bodies and Alzheimer’s disease. 84 Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV,
Brain Res 2009; 1251: 1–6. Greenamyre JT. Chronic systemic pesticide exposure reproduces
60 Chu Y, Kordower JH. Age-associated increases of α-synuclein in features of Parkinson’s disease. Nat Neurosci 2000; 3: 1301–06.
monkeys and humans are associated with nigrostriatal dopamine 85 Cannon JR, Tapias V, Na HM, Honick AS, Drolet RE,
depletion: is this the target for Parkinson’s disease? Neurobiol Dis Greenamyre JT. A highly reproducible rotenone model of
2007; 25: 134–49. Parkinson’s disease. Neurobiol Dis 2009; 34: 279–90.

www.thelancet.com/neurology Vol 9 November 2010 1137


Personal View

86 Desplats P, Lee HJ, Bae EJ, et al. Inclusion formation and neuronal 102 Lee HJ, Suk JE, Bae EJ, Lee JH, Paik SR, Lee SJ.
cell death through neuron-to-neuron transmission of α-synuclein. Assembly-dependent endocytosis and clearance of extracellular
Proc Natl Acad Sci USA 2009; 106: 13010–15. α-synuclein. Int J Biochem Cell Biol 2008; 40: 1835–49.
87 Lee HJ, Suk JE, Patrick C, et al. Direct transfer of α-synuclein from 103 Sung JY, Kim J, Paik SR, Park JH, Ahn YS, Chung KC. Induction of
neuron to astroglia causes inflammatory responses in neuronal cell death by Rab5A-dependent endocytosis of α-synuclein.
synucleinopathies. J Biol Chem 2010; 285: 9262–72. J Biol Chem 2001; 276: 27441–48.
88 Miller DW, Johnson JM, Solano SM, Hollingsworth ZR, 104 Liu J, Zhang JP, Shi M, et al. Rab11a and HSP90 regulate recycling
Standaert DG, Young AB. Absence of α-synuclein mRNA of extracellular α-synuclein. J Neurosci 2009; 29: 1480–85.
expression in normal and multiple system atrophy oligodendroglia. 105 Danzer KM, Haasen D, Karow AR, et al. Different species of
J Neural Transm 2005; 112: 1613–24. α-synuclein oligomers induce calcium influx and seeding.
89 Waxman EA, Giasson BI. Molecular mechanisms of α-synuclein J Neurosci 2007; 27: 9220–32.
neurodegeneration. Biochim Biophys Acta 2009; 1792: 616–24. 106 Danzer KM, Krebs SK, Wolff M, Birk G, Hengerer B. Seeding
90 Conway KA, Harper JD, Lansbury PT. Accelerated in vitro fibril induced by α-synuclein oligomers provides evidence for spreading
formation by a mutant α-synuclein linked to early-onset Parkinson of α-synuclein pathology. J Neurochem 2009; 111: 192–203.
disease. Nat Med 1998; 4: 1318–20. 107 Luk KC, Song C, O’Brien P, et al. Exogenous α-synuclein fibrils
91 El-Agnaf OM, Jakes R, Curran MD, Wallace A. Effects of the seed the formation of Lewy body-like intracellular inclusions in
mutations Ala30 to Pro and Ala53 to Thr on the physical and cultured cells. Proc Natl Acad Sci USA 2009; 106: 20051–56.
morphological properties of α-synuclein protein implicated in 108 Waxman EA, Giasson BI. A novel, high-efficiency cellular model of
Parkinson’s disease. FEBS Lett 1998; 440: 67–70. fibrillar α-synuclein inclusions and the examination of mutations
92 Giasson BI, Uryu K, Trojanowski JQ, Lee VM. Mutant and wild type that inhibit amyloid formation. J Neurochem 2010; 113: 374–88.
human α-synucleins assemble into elongated filaments 109 Jellinger KA. Formation and development of Lewy pathology: a
with distinct morphologies in vitro. J Biol Chem 1999; 274: 7619–22. critical update. J Neurol 2009; 256 (suppl 3): 270–79.
93 Hashimoto M, Hsu LJ, Sisk A, et al. Human recombinant 110 Choi DY, Liu M, Hunter RL, et al. Striatal neuroinflammation
NACP/α-synuclein is aggregated and fibrillated in vitro: relevance promotes Parkinsonism in rats. PLoS One 2009; 4: e5482.
for Lewy body disease. Brain Res 1998; 799: 301–06. 111 Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, Lee VM.
94 Narhi L, Wood SJ, Steavenson S, et al. Both familial Parkinson’s Neuroinflammation and oxidation/nitration of α-synuclein linked
disease mutations accelerate α-synuclein aggregation. J Biol Chem to dopaminergic neurodegeneration. J Neurosci 2008; 28: 7687–98.
1999; 274: 9843–46. 112 Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease:
95 Conway KA, Harper JD, Lansbury PT Jr. Fibrils formed in vitro a target for neuroprotection? Lancet Neurol 2009; 8: 382–97.
from α-synuclein and two mutant forms linked to Parkinson’s 113 Fauré J, Lachenal G, Court M, et al. Exosomes are released by
disease are typical amyloid. Biochemistry 2000; 39: 2552–63. cultured cortical neurones. Mol Cell Neurosci 2006; 31: 642–48.
96 Wood SJ, Wypych J, Steavenson S, Louis JC, Citron M, Biere AL. 114 Agnati LF, Guidolin D, Baluska F, et al. A new hypothesis of
α-synuclein fibrillogenesis is nucleation-dependent: implications pathogenesis based on the divorce between mitochondria and their
for the pathogenesis of Parkinson’s disease. J Biol Chem 1999; host cells: possible relevance for the Alzheimer’s disease.
274: 19509–12. Curr Alzheimer Res 2010; 7: 307–22.
97 Choi W, Zibaee S, Jakes R, et al. Mutation E46K increases 115 Kuo YM, Li Z, Jiao Y, et al. Extensive enteric nervous system
phospholipid binding and assembly into filaments of human abnormalities in mice transgenic for artificial chromosomes
α-synuclein. FEBS Lett 2004; 576: 363–68. containing Parkinson disease-associated α-synuclein gene
98 Lee HJ, Patel S, Lee SJ. Intravesicular localization and exocytosis of mutations precede central nervous system changes. Hum Mol Genet
α-synuclein and its aggregates. J Neurosci 2005; 25: 6016–24. 2010; 19: 1633–50.
99 Jang A, Lee HJ, Suk JE, Jung JW, Kim KP, Lee SJ. Non-classical 116 Olanow CW, Stern MB, Sethi K. The scientific and clinical basis for
exocytosis of α-synuclein is sensitive to folding states and promoted the treatment of Parkinson disease. Neurology 2009; 72: S1–136.
under stress conditions. J Neurochem 2010; 113: 1263–74. 117 Olanow CW, Rascol O, Hauser R, et al. A double-blind, delayed-start
100 Emmanouilidou E, Melachroinou K, Roumeliotis T, et al. trial of rasagiline in Parkinson’s disease. N Engl J Med 2009;
Cell-produced α-synuclein is secreted in a calcium-dependent 361: 1268–78.
manner by exosomes and impacts neuronal survival. J Neurosci 118 Eller M, Williams DR. Biological fluid biomarkers in
2010; 30: 6838–51. neurodegenerative parkinsonism. Nat Rev Neurol 2009; 5: 561–70.
101 Ahn KJ, Paik SR, Chung KC, Kim J. Amino acid sequence motifs
and mechanistic features of the membrane translocation of
α-synuclein. J Neurochem 2006; 97: 265–79.

1138 www.thelancet.com/neurology Vol 9 November 2010

You might also like