You are on page 1of 8

Experimental Neurology 298 (2017) 172–179

Contents lists available at ScienceDirect

Experimental Neurology
journal homepage: www.elsevier.com/locate/yexnr

Review Article

Experimental animal models of Parkinson's disease: A transition from MARK


assessing symptomatology to α-synuclein targeted disease modification
Wai Kin D. Koa,⁎, Erwan Bezarda,b,c
a
Motac Neuroscience Ltd, Manchester, United Kingdom
b
Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
c
CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France

A R T I C L E I N F O A B S T R A C T

Keywords: With the understanding that α-synuclein plays a major role in the pathogenesis of Parkinson's disease (PD), novel
α-Synuclein animal models have been developed for conducting preclinical research in screening novel disease modifying
Parkinson's disease therapies. Advancements in research techniques in α-synuclein targeted disease modification have utilised
Animal models methods such as viral mediated expression of human α-synuclein, as well as the inoculation of pathogenic α-
Lewy Body
synuclein species from Lewy Bodies of PD patients, for accurately modelling progressive self-propagating neu-
rodegeneration. In applying these cutting-edge research tools with sophisticated trial designs in preclinical drug
trials, a useful platform has emerged for developing candidate agents with disease modifying actions, promising
a greater chance of success for clinical translation. In this article, we describe the transition of well-established
animal models of PD symptomatology to newly developed models of PD pathogenesis, with specific focus on
methods of viral-mediated and inoculation of pathogenic α-synuclein, that aim to aid scientific translation of
neuroprotective strategies.

1. Parkinson's disease pathogenesis Kruger et al., 1998; Lesage et al., 2013; Polymeropoulos et al., 1997;
Puschmann et al., 2009; Zarranz et al., 2004) and also the multi-
Parkinson's disease (PD) is a progressive neurodegenerative disorder plication and triplication of the gene locus for α-synuclein (SNCA)
affecting approximately 1% of the population over the age of 55 years, (Chartier-Harlin et al., 2004; Singleton et al., 2003).
with the highest prevalence in ages of 85 years and over (De Rijk et al., In the last two decades, the α-synuclein protein has been proposed
1997). PD patients display a clinical syndrome of motor symptoms (in as the main component in mediating the progressive neurodegeneration
particular bradykinesia, postural deficits and resting tremor) (Marsden, in PD (Dehay and Fernagut, 2016; Recasens and Dehay, 2014). The
1994), which may be preceded by a range of non-motor symptoms pathological structure of α-synuclein is a misfolded conformation of an
(including cognitive disturbance, sleep disturbance, anosmia and con- oligomeric or fibrillary nature that is of a specific phosphorylated form
stipation) (Pont-Sunyer et al., 2015). These clinical manifestations of (phosphor-Ser129), as seen in PD patient LB formations (Fujiwara et al.,
PD appear due to the extensive loss of dopaminergic neurons of the 2002; Spillantini et al., 1997). Several studies have greatly impacted the
nigrostriatal pathway (Ehringer and Hornykiewicz, 1960), as well as field of PD pathogenesis. Braak et al. first hypothesised the progression
dysfunction of neurons in dopaminergic, serotonergic, adrenergic and of PD in distinct stages; beginning at the peripheral autonomic nervous
cholinergic neurotransmitter systems (Jellinger, 1991). system, a region where α-synuclein pathology was identified (Braak
A major pathological hallmark of PD is the widespread expression of et al., 2006; Gelpi et al., 2014), in the anterior olfactory nucleus and
intraneuronal proteinaceous inclusions, known as Lewy Bodies (LBs) dorsal motor nucleus of the glossopharyngeal and vagal nerves, gra-
(Spillantini et al., 1998), found in the perikarya of neurons within the dually reaching the midbrain and cerebral cortex (Braak et al., 2003).
central and peripheral nervous systems. LBs are mainly composed of α- Pathological transmission was later suggested between neurons through
synuclein, a 14 kDa endogenous protein of 140 amino acid length. post-mortem analysis in PD patients, where LB formations were found
Autosomal dominant forms of PD are linked to missense mutations of in embryonic mesencephalic neuron grafts in the striatum that were
the α-synuclein, specifically p.A53T, p.A30P, p.E64K, p.H50Q, p.G51D implanted over a decade prior, supporting a host-to-graft transmission
and p.A53E (Appel-Cresswell et al., 2013; Athanassiadou et al., 1999; process (Kordower et al., 2008; Li et al., 2010). With the transmission of


Corresponding author at: Motac Neuroscience Ltd, PO Box 363, Manchester M15 6WE, United Kingdom.
E-mail address: d.ko@motac.com (W.K.D. Ko).

http://dx.doi.org/10.1016/j.expneurol.2017.07.020
Received 17 June 2017; Accepted 28 July 2017
Available online 29 July 2017
0014-4886/ © 2017 Elsevier Inc. All rights reserved.
W.K.D. Ko, E. Bezard Experimental Neurology 298 (2017) 172–179

α-synuclein-positive intracytoplasmic inclusions, the term ‘prionoid’ clinical scale criteria can be utilised to specifically assess movement
has been assigned, describing the pathogenic nature of intracellular range, posture, tremor, and a calculated score for overall disability (Ko
mechanisms, including release into extracellular space and uptake into et al., 2014). These translational tools are and have been instrumental
healthy neurons. Although mainly focused within dopaminergic neu- for evaluating the efficacy of symptomatic therapies (Bastide et al.,
rons of the SN, this self-propagating process with the pathogenic α- 2015; Johnston and Fox, 2015; Fox and Brotchie, 2010), which are
synuclein acting as a template for misfolding (Brundin et al., 2008; effective in providing drug efficacy in proof-of-concept tests (Ko et al.,
Dehay et al., 2016a), can lead to a spread of neurodegeneration across 2014; Rylander et al., 2010). Furthermore, in recent years, sophisti-
interconnected brain regions causing disruption of intracellular pro- cated technology in high definition video recording of limb trajectory
cesses (i.e. neurotransmission, mitochondrial structure and activity, has been adapted to nhps for assessment of detailed kinematics in swing
dynamics and mitophagy, vesicular transport and protein degradation). and gait cycles during freely moving tasks (Capogrosso et al., 2016; Yin
The pathogenic α-synuclein seeding activity is supported by detection et al., 2014). Such objective assessments of motor activity complement
of monomeric, oligomeric and Ser129-phosphorylated α-synuclein subjective behavioural ratings for detecting more subtle motor deficits,
forms in human plasma and cerebrospinal fluid (Borghi et al., 2000; El- allowing for consistent endpoint assessments between MPTP-treated
Agnaf et al., 2003; Foulds et al., 2012; Mollenhauer et al., 2011; Tokuda nhps and PD patients, in respective preclinical and clinical trials.
et al., 2010). Moreover, further supportive evidence comes from the use The MPTP-treated nhp model of PD offers further face and pre-
of synthetic recombinant α-synuclein preformed fibrils (PFFs) inducing dictive validity (i.e. phenomenological similarities between behaviours
conformation changes of endogenous soluble α-synuclein to patholo- and measurable treatment effects as seen in the human condition, re-
gical species, subsequently leading to cellular dysfunction (Hansen spectively) with clear reversal of experimental parkinsonism following
et al., 2011; Luk et al., 2009; Volpicelli-Daley et al., 2011). L-3,4-dihydroxyphenylalanine (L-DOPA) treatment. The maximum L-
In recent years, the understanding of PD pathogenesis has been DOPA dose is individually titrated based on motor symptom severity
greatly enhanced with the development of advanced research tools, and is commonly given during the stabilisation period of model re-
some taking the form of novel animal models (Angot et al., 2010; Dehay plication to aid animal feeding. Daily L-DOPA treatment (in the range of
et al., 2016b; Olanow and Brundin, 2013). In this review, we provide an 9–20 mg/kg) over 3–4 months can lead to the development of stable
overview of the commonly used symptomatic animal models of PD and and reproducible dyskinesia, with chorea and dystonia individually
newly developed models of PD pathogenesis, of which the latter are scored based on severity (Fox et al., 2012). After L-DOPA priming in
expected to aid the translation of potential therapies with focus on MPTP-treated nhps, PD disability cannot be completely reversed
disease-modifying mechanisms, paving the way for the identification of without the induction of dyskinesia, similar to that seen in PD patients.
novel agents for treating neurological disorders. This has been utilised to provide clinically relevant data such as ‘good
on-time’ (Ko et al., 2016; Pinna et al., 2016) allowing treatment stra-
2. Modelling clinical symptoms of PD tegies for dyskinesia to be thoroughly evaluated in dyskinetic MPTP-
treated nhps. Investigations utilising this model also provide for ad-
To date, neurotoxin based animal models have been well utilised in dressing specific intracellular mechanisms using novel biotechnological
translational research for the development of symptomatic therapies in approaches, such as gene therapy (e.g. modulation of regulators of G-
PD. The most commonly used neurotoxins include 6-hydroxydopamine protein signalling proteins (Gold et al., 2007)) or enzymes for dopamine
(6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), synthesis (Jarraya et al., 2009), for testing potential therapies in
which are often administered in rodents and non-human primates modifying the expression of dyskinesia.
(nhps), respectively, for inducing a parkinsonian state by causing severe The use of MPTP-treated nhps is also a valuable translational re-
loss of dopaminergic neurons. search model for studying the non-motor symptoms of PD. For example,
In rats, the unilateral injection of 6-OHDA into the medial forebrain chronic low dose (CLD) MPTP administration can be used to induce
bundle causes rapid and extensive loss (> 90%) of dopaminergic cognitive deficits found in executive and attentional tasks, which are
neurons that is seen within 14 days (Grealish et al., 2008). The re- further worsened following L-DOPA treatment, as seen in PD patients
producibility and simplicity in quantifying asymmetric motor deficits, (Gotham et al., 1988; Kulisevsky et al., 1996; Schneider et al., 2013). In
such as rotation and forelimb bias, following administration of dopa- modelling both cognitive deficits and subtle motor dysfunction in
minergic agents are major advantages for the use of 6-OHDA-lesioned MPTP-treated nhps, the translational value allows for tests of single or
rats in translational research that have allowed for efficient screening of combined drug treatment strategies that produce the most desired
anti-parkinsonian compounds (Cannon and Greenamyre, 2010; Lane clinical outcome among the myriad of disease symptoms (Ko et al.,
et al., 2006; Schwarting and Huston, 1996). In addition, the injection of 2016). Sleep disturbances seen in PD patients such as rapid eye
6-OHDA into the striatum permits for slower and less severe loss of movement sleep behaviour disorder (Postuma et al., 2009) can also be
dopaminergic nigrostriatal neurons (> 60–70%), modelling more pro- evaluated in MPTP-treated monkeys using continuous electro-
gressive neuronal degeneration (Kirik et al., 1998; Sauer and Oertel, encephalography, which has been used to assess the effects of different
1994). pharmacological treatments on sleep disturbances (Barraud et al., 2009;
The gold standard model of PD motor symptoms is produced by Hyacinthe et al., 2014). While MPTP administration causes severe loss
administration of MPTP into old world non-human primate (nhp) spe- of dopaminergic neurons in the SNc, there is also a broad neurotoxic
cies (Macaca mulatta and Macaca fascicularis). With a closer resem- effect with loss of extra-nigral dopaminergic, serotoninergic and adre-
blance to humans in physiology and brain anatomy, the PD motor nergic neurons (Forno et al., 1986; Mitchell et al., 1985; Perez-Otano
features in monkeys that are induced following a regimen of MPTP et al., 1991). Although this resembles some of the major neuropatho-
injections (typically daily doses of 0.2–2 mg/kg for 2–3 weeks) gives an logical changes seen in PD patients, this animal model remains limited
accurate model for advanced PD clinical motor signs (Bezard et al., for studying disease pathogenesis due to lack of true LB pathology,
2001; Bezard et al., 2003; Meissner et al., 2003). The MPTP-treated nhp albeit certain proteinaceous inclusions have been previously reported in
model is most commonly replicated after systemic administration of MPTP-treated baboons (Kowall et al., 2000).
MPTP, which readily crosses the blood brain barrier, inducing a par- While neurotoxin based animal models serve for the development of
kinsonian syndrome of prolonged stillness, bradykinesia, postural def- symptomatic therapies, notably seen from the early success of deep
icit and an overall reduction in movement (Crossman et al., 1985; brain stimulation in PD (Benazzouz et al., 1993), these symptomatic
Delong et al., 1985). After stabilisation of these behaviours where do- animal models remain limited for studying disease-modifying therapies
paminergic degeneration reaches > 90% in the substantia nigra pars due to the inability to accurately replicate the nature of PD pathogen-
compacta (SNc) (Bezard et al., 1997), subjective rating scales based on esis i.e. progressive self-propagating neurodegeneration with

173
W.K.D. Ko, E. Bezard Experimental Neurology 298 (2017) 172–179

pathological hallmarks. In the next section, we look at the development neurotoxicity that are likely to involve processes of inflammatory re-
of recent animal models that allow for the screening of agents with action (Chung et al., 2009), associated exosome release (Danzer et al.,
disease modifying effects. 2012; Lee et al., 2005), ER-golgi trafficking (Cooper et al., 2006),
passive membrane diffusion (Kayed et al., 2004) and proteasome and
3. Modelling PD pathogenesis lysosome dysfunction (Venda et al., 2010).
Viral mediated overexpression of α-synuclein has also been tested in
With the advancements in the understanding of PD pathogenesis, mice species with the potential in combining the capacity of genetic
specifically the identification of misfolded α-synuclein as a pathogenic modification with progressive disease development. However, the lack
agent, novel animal models of synucleinopathies have been developed of consensus between methods has so far shown different levels of
as a useful platform for evaluating neurodegeneration, as well as for success for inducing neurodegeneration. For example, a number of
specifically investigating α-synuclein seeding and cell-to-cell transmis- studies have reported limited dopaminergic neuronal degeneration
sion (Sato et al., 2015). In regards to the latter, viral vectors have also from 7 to 12 weeks post SN injection (Lauwers et al., 2003; Theodore
been previously utilised across mice, rats and nhps for studying host-to- et al., 2008; Yasuda et al., 2009), whereas other groups, utilising AAV2/
graft transmission i.e. transmission models (Kordower et al., 2011; 7 with either wildtype and mutant A53T-α-synuclein have demon-
Dehay et al., 2016b; Dehay and Fernagut, 2016). In a series of delicate strated significant loss (82%) of nigral TH neurons at 8 weeks following
experiments, the unilateral injection of AAV2/6-huαsyn into the SN injection (Oliveras-Salva et al., 2013). In a recent study by Ip et al.
caused an approximate 23% transmission of human α-synuclein to (2017), the effects of AAV1/2-induced overexpression of A53T-α-sy-
implanted embryonic ventral mesencephalic neurons in striatum of the nuclein were well characterised in C57BL/6 mice (Ip et al., 2017). It
same hemisphere (Angot et al., 2012). was reported that at 10 weeks post unilateral injection into the SN, A-
The use of viral vectors has also been commonly used for inducing AV1/2-A53T- α-synuclein mice showed 33% reduction in TH neurons,
neurodegeneration in mammalian species and is recognised to produce with 29% loss of dopamine active transporter binding. In addition,
a more acute form of PD compared to transgenic animal models, with HPLC analysis revealed reduction in levels of dopamine (38%) and
the avoidance of compensatory mechanisms from constitutive over- DOPAC (33%), respectively (Ip et al., 2017), with histological analysis
expression, which is the case for transgenic PD models. In the first series showing pathological hallmarks of LBs and Lewy-neurites (LNs) in the
of demonstrations (Lo Bianco et al., 2002; Kirik et al., 2002), the re- nigrostriatal system. Authors also characterised the motor deficits in
combinant lenti-virus was used for overexpression of human α-synu- AAV1/2-A53T-α-synuclein mice compared to empty vector controls,
clein (wild-type, A30P mutant and A53T mutant) in the SN of rats, which were seen to have forelimb bias measured using the cylinder test
which induced modest loss of neurons expressing tyrosine hydroxylase at 5 and 9 weeks during the live-phase of the study. Together, the
(TH) (35%, 33% and 24%, respectively) (Lo Bianco et al., 2002). In construct validity in the proposed mouse model of PD pathogenesis was
another key study, Kirik et al. (2002) was first to utilise the re- recognised with degeneration associated with α-synuclein accumula-
combinant adeno-associated virus (rAAV) to overexpress wild-type and tion and histological/morphological features, but this model currently
human mutated A53T in rat nigrostriatal dopaminergic neurons. Fol- lacks predictive validation i.e. the reversal of symptomatic features
lowing unilateral injection into rats of rAAV-α-syn in the SN, patholo- with pharmacological treatments (Ip et al., 2017).
gical features of PD were observed which included α-synuclein inclu- Tests with recombinant AAV for overexpression of α-synuclein in
sions and dystrophic neurites. In addition, the overexpression of α- the SN of higher order species was originally carried out in marmosets,
synuclein caused significant loss of nigral DA neurons (30–80%) and successfully demonstrating PD pathogenesis (Eslamboli et al., 2007;
striatal dopamine (~ 40%), as well as increased dopamine turnover Kirik et al., 2003). The use of AAV2 for increased expression of wild-
(~ 160%) over 6 months (Kirik et al., 2002). However in this study, type or A53T α-synuclein was found to produce 30–60% dopaminergic
animals did not show obvious asymmetric motor deficits, possibly due neuron loss over 4 months (Kirik et al., 2003). Eslamboli et al. (2007)
to a non-substantial loss of striatal dopamine that has been recognised further demonstrated a more reliable model of PD pathogenesis in
to be at a threshold level of > 50–60% (Kirik et al., 1998; Lee et al., marmosets with use of AAV2/5-A53T α-synuclein, compared to wild-
1996). Later studies utilising a chimeric vector, specifically AAV1/2 type α-synuclein (Eslamboli et al., 2007). Recently, Koprich et al.
carrying mutated A53T-α-synuclein cDNA, for increased neuronal (2016) extended observations to macaque species, where AAV1/2-
tropism and expression, demonstrated progressive neurodegeneration driven overexpression of human A53T α-synuclein was used to model
in rats over 6 weeks reaching > 50% dopaminergic cell loss (Koprich PD pathogenesis, in a step towards developing of a drug screening
et al., 2010; Koprich et al., 2011). platform for disease modifying therapies (Koprich et al., 2016). With a
The recent studies using viral mediated overexpression of α-synu- high level of transfection (86%) in nigral dopaminergic neurons after 4
clein in rats has denoted high translational value in modelling motor separate site injections in the SN, authors demonstrated that a high titre
dysfunction as well as in PD pathology. Decressac et al. (2012a, 2012b) (20 μl of 5.1 × 1012 gp/ml) or high volume (28 μl of 1.7 × 1012 gp/ml)
recently assessed behavioural deficits and neuropathological changes of hA53T-α-synuclein caused 50% and 49% SN dopaminergic cell loss,
caused by unilateral AAV6-α-syn injections under an optimised vector respectively, as well as 72% and 60% reduction of dopamine levels in
construct (transgene driven by synapsin-1 promoter and amplified the putamen, respectively, compared to controls over 4 month ob-
WPRE sequence) in rats, which were compared to behavioural and servations (Koprich et al., 2016). Although further experiments are
pathological deficits induced by 6-OHDA-lesions (Decressac et al., needed for evaluating the PD behavioural phenotype in this model and
2012a). While loss of dopaminergic neurons in SNc of AAV6-α-syn in- for demonstrating reproducibility, these initial studies may provide a
jected rats reached approximately 70%, similar to that of two-site in- pathway towards validating a new model of PD pathogenesis for pre-
trastriatal 6-OHDA injections, the model replicated disease hallmarks of clinical drug development. Together, the studies utilising viral vectors
cytoplasmic α-synuclein inclusions with significant asymmetric motor across mammalian species provides for novel models of PD pathogen-
deficits being established over 8 weeks (Decressac et al., 2012a; esis with construct validity, replicating clinical hallmarks, specifically
Decressac et al., 2012b). Recent experiments have further characterised α-synuclein related neuropathology and progressive neurodegenera-
the motor deficiencies in AAV9-α-syn injected rats by using high speed tion.
motion caption of whole-body kinematics, where animals demonstrated Uncovering the mechanisms involved in PD pathogenesis has also
slow shuffling gait, reduced control of limb trajectory and medio-lateral utilised other research methods based on host-to-graft transmission
instability (Bourdenx et al., 2015b). The AAV-α-syn injected rat model (Angot et al., 2012; Desplats et al., 2009) which includes injection of 1.
offers major advantages in the screening of novel neuroprotective Brain samples between α-synuclein transgenic and wild-type animals
agents against motor deficits, as well as processes of α-synuclein (Luk et al., 2012b; Mougenot et al., 2012) 2. In vitro generated PFFs of

174
W.K.D. Ko, E. Bezard Experimental Neurology 298 (2017) 172–179

α-synuclein (Hansen et al., 2011; Luk et al., 2012a) 3. Brain homo- neurite length and branching. Using high content time-lapse imaging,
genates from patients with synucleinopathy (Bernis et al., 2015; Jones cell culture experiments have so far demonstrated the dose-dependent
et al., 2015) and 4. α-synuclein containing LB extracts from post- effect of α-synuclein-induced toxicity on neurite outgrowth of cortical
mortem PD patient brain tissue (Recasens et al., 2014). neurons over a 7 day period (Cavaliere et al., 2015). These in vitro
Studies in rodents have shown host-to-graft transmission of α-sy- methods in addition to the LB-seeding animal models in mice and nhps
nuclein between neural cells. An example of this is the use of transgenic provide for a powerful platform for studying disease modifying agents.
mice overexpressing α-synuclein (line 61 and line F28) that lead to α- For example, the ability to monitor gradual neuronal degeneration in-
synuclein immunoreactivity in grafted cells from 4 to 6 weeks post- duced by purified LB patient samples as described by Recasens et al.
transplantation (Desplats et al., 2009; Hansen et al., 2011). In vivo (2014) provides for screening of neuroprotective compounds that can
studies have also utilised the injection of brain extracts from α-synu- halt or slow the disease process. Further ex vivo analysis of post-
clein transgenic mice into healthy transgenic animals to demonstrate mortem brain tissue of animal models can be used to identify whether
pathogenic transmission (Mougenot et al., 2012). In a study by Luk potential disease modifying agents are effective against major disease
et al., brain homogenates from old transgenic mice (M83) were injected processes such as α-synuclein aggregation and cell-to-cell transmission.
into cortex and striatum of young asymptomatic transgenic mice, which
caused the spread of α-synuclein pathology in central nervous system 4. Animal model considerations
(CNS) over 90 days (Luk et al., 2012b). Although this demonstrates that
the pathogenic protein is within the brain homogenate sample for in- The implementation of novel methods in modelling PD pathogenesis
itiating disease pathogenesis, the seed pathogen remains to be eluci- through technological breakthroughs provides a potentially effective
dated within the vast content encompassed in the inoculated material. avenue for the rapid development of disease-modifying agents for
Pathological and behavioural abnormalities that resemble PD, such as clinical use. These newly developed research approaches, gives high
loss of dopaminergic neurons in the SNc and motor deficits, has also translational value in modelling progressive neurodegeneration in the
been demonstrated in wild-type mice from a striatal injection of re- closest species related to humans, mimicking effects on behaviour and
combinant α-synuclein PFFs (at 5 μg), a process dependent on host pathophysiology.
expression of endogenous α-synuclein, which supports a seeding me- With the use of the novel research tools, screening for candidate
chanism induced by pathogenic species (Luk et al., 2012a). However, agents against neurodegeneration can be carried out relatively rapidly,
while providing useful insights into the disease developmental process, where it remains important to refine and strictly orient experimental
it remains noted that large amounts of PFFs (within the microgram measures and designs for clinical translation (Camus et al., 2015). As
range) are required to initiate pathology, raising questions of the direct successful translation of research to the clinical domain has suffered
association with disease pathogenesis and calling for further studies in from poor experimental designs and methodological approaches in
identifying the pathogenic subspecies of oligomers and fibrils. animal studies (Hackam, 2007; Martic-Kehl et al., 2012; Van Der Worp
In the past few years, much progress has been made in modelling PD et al., 2010), which has led to exaggerated findings (Button et al., 2013;
pathogenesis in nhps using human α-synuclein species derived from LBs Hackam, 2007; Ioannidis, 2008; Van Der Worp and Macleod, 2011), the
of post-mortem PD patients, where it was hypothesised that patient- use of animal models in PD can benefit by strictly adhering to rando-
derived aggregated α-synuclein holds the pathogenic candidate. In the misation, treatment/assessment blinding and sample size calculated
study performed by Recasens et al. (2014), nigral LB samples from 3 PD designs (Bebarta et al., 2003; Button et al., 2013; Perel et al., 2007; Van
patient brain samples, purified by sucrose gradient fractionation and Der Worp et al., 2005), to avoid poor data reproduction and ineffective
filter retardation, were injected into the i. motor striatum (100 μl) at translational progress (Hackam, 2007; Hackam and Redelmeier, 2006;
two rostrocaudal levels or ii. substantia nigra (10 μl) in rhesus macaque Sena et al., 2007; Van Der Worp et al., 2010). Along these lines, tech-
monkeys (Recasens et al., 2014). The PD-derived LB-extracts caused nical approaches in the range of animal models can value from stan-
progressive nigrostriatal neurodegeneration, where it was found that dardising certain research methods. In the new models for example,
exogenous human α-synuclein was internalised by neurons initiating consistency in experimental factors, such as rodent strain, serotype,
conversion of host α-synuclein, which spread across anatomically in- promotor, vector (lentivirus vs. adenovirus), aggregates of wild-type or
terconnected brain areas. During the live phase of the study, dopami- forms of mutated/phosphorylated of α-synuclein and titre for sample
nergic neuron loss (~ 34%) was shown at 9 months post striatal LB preparations, are likely to enhance experimental data reproducibility.
injection by positron emission scans using radioligand marker 11C- Indeed, differences in research techniques have so far shown incon-
DTBZ. Post-termination analysis conducted at 14 months post injection, sistencies in neuropathology in rodent models, where dopaminergic
showed that nhps injected with LBs into the striatum or SNc, had sig- neuron loss has been reported to range from extensive (~ 70%)
nificant dopamine cell loss in SNc (~ 40% and ~15%, respectively). (Decressac et al., 2012a; Decressac et al., 2012b) to moderate (~ 30%)
With more extensive loss at the striatal dopaminergic axon terminals (Mcfarland et al., 2009; Recchia et al., 2008).
than SNc cell bodies, authors alluded to a putative mechanism of LB Moreover, experiments using the most relevant animal model are to
extract induced retrograde nigral neuronal degeneration or possible be ideally designed with similar or exact endpoints/outcomes to those
initiation of pathological cascades that induced distal neurodegenera- used in patients. These include the use of biomarkers for measuring
tion (Recasens et al., 2014). Moreover, the detailed cell analysis showed neurodegeneration (Delatour et al., 2006; Jack et al., 2009) i.e. in vivo
most pathological (Ser129 phosphorylated/proteinase K resistant) α- SPECT imaging measurement of [99mTc]-TRODAT-1 binding (Fernagut
synuclein diffusely accumulated in the cytosol and in the presynaptic et al., 2010; Meissner et al., 2003; Prunier et al., 2003). As such, proof
terminals. In mice, it is found that the potent reactivity species of of concept animal trials, especially in the range of new animal models
purified LB samples causing neurodegeneration is within a picogram of PD may mitigate the risk of taking a preclinical compound to clinical
dose range (Recasens et al., 2014), in comparison to a microgram dose trials.
range that is needed for recombinant PFFs to induce cell loss, sug-
gesting a higher pathogenic potential of infectivity of the former. To 5. Developing animal models of disease pathogenesis
support these in vivo investigations, an in vitro research platform has
been developed specifically for compound assessment against α-synu- With the new methods for developing drug candidates, the tech-
clein cell toxicity and cell transmission (neuron/glia ↔ glia/neuron). nological approaches employed also provide for greater ways for un-
Using 96-well plate high-throughput screening in primary cortical covering neurological disease pathogenesis. The nature of disease de-
neuronal cultures, drug efficacy against α-synuclein toxicity can be velopment in new PD pathogenesis models e.g. > 9 months in LB-
monitored through blockade of changes to cell morphology, such as injected nhps or > 4 months in AAV1/2-huA53T injected nhps,

175
W.K.D. Ko, E. Bezard Experimental Neurology 298 (2017) 172–179

presents a significant time frame for deciphering neurodegeneration entry and cell-to-cell transfer in primary hippocampal cell cultures
across brain regions that may relate to non-motor impairments during (Tran et al., 2014). It was also demonstrated that repeated systemic
the pre-motor symptomatic phase (Jellinger, 2011), simultaneously injection of Syn303 at 10 mg/kg in PFF-inoculated wild-type mice at-
giving translational value for development of such therapies. Recent tenuated SN dopaminergic degeneration and accumulation syn506-
studies have been extended to investigating peripheral transmission positive aggregates compared to control IgG-treated mice, where be-
into the CNS, which may help elucidate unknown processes in prion- havioural deficits in grip strength and motor coordination were also
like pathogenesis like site-specific initiation in anatomical regions i.e. improved in the group treated with α-synuclein monoclonal antibodies
olfactory bulb and internal plexuses. In wild-type mice, Rey et al. (Tran et al., 2014). Another novel candidate agent currently being
(2013) showed the injection of molecular species of recombinant tested is the specific oligomer modulator named Anle138b, a compound
human α-synuclein (monomeric, oligomeric and fibrillary forms) into discovered through high-throughput screening. So far, in vivo tests
the olfactory bulb, rapidly spread to interconnected brain regions in- have demonstrated that Anle138b has blood brain barrier permeability
cluding anterior olfactory bulb, frontal cortex, striatum and amygdala targeting the pathological aggregation process of pathological α-synu-
(Rey et al., 2013). In rat studies, the injection of rAAV serotype ex- clein oligomers, binding specifically to aggregated α-synuclein (Wagner
pressing human wild type α-synuclein into the left vagus nerve in rats et al., 2013). Following treatment in the LB-seeding mouse model of PD,
caused expression of human α-synuclein in the medulla oblongata and a reduction of oligomer accumulation and slower disease progression
caused a caudo-rostral spread to interconnected brain regions (Ulusoy has been reported. This compound is now being tested in LB-injected
et al., 2013). In addition to this, pathogenic species in the form of nhps and has so far demonstrated a promising pharmacokinetic profile.
human α-synuclein from SN of PD patients and distinct recombinant α- Further therapies in stages of development are vaccinations with short
synuclein forms (including monomers, oligomers and fibrils) can be peptide affitopes® for preventing α-synuclein aggregation by targeting
seen to pass into the CNS through the vagal nerve following injection the C-terminus region of α-synuclein, which has shown efficacy in
into the intestinal walls of rats (Holmqvist et al., 2014). The gastro- transgenic mouse models for decreasing α-synuclein oligomers in axons
intestinal route of entry for pathogenic species has also been previously and synapses and reduced loss of dopaminergic neurons (Mandler et al.,
investigated using the environmental toxin rotenone. Following the 2014; Masliah et al., 2011). The above mentioned studies together
Braak hypothesis, peripheral transmission was determined through in- demonstrate the active shift in development of disease modifying
tragastric administration of retonone in mice, which caused α-synuclein therapies using the range of novel animal models of PD pathogenesis, in
pathology in both the enteric nervous system and CNS (Pan-Montojo order to fulfil the major unmet clinical need in the treatment of neu-
et al., 2010). These new insights into the pathogenic basis of PD present rological disorders.
greater opportunities for developing disease modifying strategies,
whereby specific treatments may be designed and directed to key 7. Conclusions
anatomical regions for effectively preventing α-synuclein pathology
and subsequent disease progression. The recognition of the α-synuclein as the major component in the
Recent breakthroughs in techniques for site–specific gene editing pathogenesis of PD has given way to newly developed animal models
and transfection is also paving a new way forward for studying disease allowing for cutting-edge research studies in screening novel agents
progression in nhps (Yang et al., 2008). It has been demonstrated that with disease modifying actions. These novel disease models of patho-
intravenous injection of adeno-associated virus serotype 9 variant genesis include the use of two established methods that are viral vector
(scAAV9) in newly born macaques (post-natal day 1) mediates effica- induced α-synuclein expression and LB seeding, which replicate the
cious neuronal gene transduction in brain areas such as the cortex, progressive substantial loss of neurodegeneration and the self-propa-
midbrain, hippocampus and cerebellum (Dehay et al., 2012). Further gating spread of pathogenic α-synuclein. Although these disease models
efforts are being made for homogenous gene expression in the cortex require further predictive validity for disease modifying drug devel-
and deep brain structures using in utero delivery of viral vectors to opment, it remains a great leap towards developing a translational
monkey embryos, which have so far demonstrated widespread neuronal platform for achieving successful clinical outcomes.
transduction with scAAV2/2-GFP (Bourdenx et al., 2015a), offering the
potential for mutant gene expression for the study of neurological dis- References
orders. In addition, specific gene editing techniques in one cell embryos
for modification of the monkey genome has been shown with the Angot, E., Steiner, J.A., Hansen, C., Li, J.Y., Brundin, P., 2010. Are synucleinopathies
CRISPR/Cas9 system allowing for two gene modifications (Niu et al., prion-like disorders? Lancet Neurol. 9, 1128–1138.
Angot, E., Steiner, J.A., Lema Tome, C.M., Ekstrom, P., Mattsson, B., Bjorklund, A.,
2014). Transcription activator-like effector nuclease (TALEN)-mediated Brundin, P., 2012. Alpha-synuclein cell-to-cell transfer and seeding in grafted dopa-
gene modification has also been employed in macaque species for minergic neurons in vivo. PLoS One 7, e39465.
mutation of methyl-CpG binding protein 2 (MECP2) gene in modelling Appel-Cresswell, S., Vilarino-Guell, C., Encarnacion, M., Sherman, H., Yu, I., Shah, B.,
Weir, D., Thompson, C., Szu-Tu, C., Trinh, J., Aasly, J.O., Rajput, A., Rajput, A.H.,
Rett syndrome (Liu et al., 2014). Recent data has shown that the mi- Jon Stoessl, A., Farrer, M.J., 2013. Alpha-synuclein p.H50Q, a novel pathogenic
croinjection of MECP-2-targeting TALEN plasmids to nhp zygotes can mutation for Parkinson's disease. Mov. Disord. 28, 811–813.
produce monkeys that exhibit autistic-like behaviours (repeated cir- Athanassiadou, A., Voutsinas, G., Psiouri, L., Leroy, E., Polymeropoulos, M.H., Ilias, A.,
Maniatis, G.M., Papapetropoulos, T., 1999. Genetic analysis of families with
cling, increased stress response and reduced social interactions), as well
Parkinson disease that carry the Ala53Thr mutation in the gene encoding alpha-sy-
as germline transmission of transgene to F1 progeny, modelling general nuclein. Am. J. Hum. Genet. 65, 555–558.
phenotypes seen in patients (Liu et al., 2016). These studies to date Barraud, Q., Lambrecq, V., Forni, C., Mcguire, S., Hill, M., Bioulac, B., Balzamo, E.,
Bezard, E., Tison, F., Ghorayeb, I., 2009. Sleep disorders in Parkinson's disease: the
demonstrate the efficient use of genetic engineering techniques in nhps
contribution of the MPTP non-human primate model. Exp. Neurol. 219, 574–582.
for investigating disease pathogenesis and their potential use for de- Bastide, M.F., Meissner, W.G., Picconi, B., Fasano, S., Fernagut, P.O., Feyder, M.,
velopment of new therapies. Francardo, V., Alcacer, C., Ding, Y., Brambilla, R., Fisone, G., Jon Stoessl, A.,
Bourdenx, M., Engeln, M., Navailles, S., De Deurwaerdere, P., Ko, W.K., Simola, N.,
Morelli, M., Groc, L., Rodriguez, M.C., Gurevich, E.V., Quik, M., Morari, M., Mellone,
6. Preclinical tests of potential disease modifying agents M., Gardoni, F., Tronci, E., Guehl, D., Tison, F., Crossman, A.R., Kang, U.J., Steece-
Collier, K., Fox, S., Carta, M., Angela Cenci, M., Bezard, E., 2015. Pathophysiology of
Several studies to date have utilised novel PD pathogenesis animal L-dopa-induced motor and non-motor complications in Parkinson's disease. Prog.
Neurobiol. 132, 96–168.
models for testing candidate agents in preventing α-synuclein pa- Bebarta, V., Luyten, D., Heard, K., 2003. Emergency medicine animal research: does use
thology. In a series of experiments by Tran et al. (2014), α-synuclein of randomization and blinding affect the results? Acad. Emerg. Med. 10, 684–687.
monoclonal antibodies for misfolded α-synuclein (Syn211 and Syn303) Benazzouz, A., Gross, C., Feger, J., Boraud, T., Bioulac, B., 1993. Reversal of rigidity and
improvement in motor performance by subthalamic high-frequency stimulation in
were shown to reduce LBs and LNs pathology, blocking α-synuclein PFF

176
W.K.D. Ko, E. Bezard Experimental Neurology 298 (2017) 172–179

MPTP-treated monkeys. Eur. J. Neurosci. 5, 382–389. mediates brain transduction in newborn rhesus macaques. Sci Rep 2, 253.
Bernis, M.E., Babila, J.T., Breid, S., Wusten, K.A., Wullner, U., Tamguney, G., 2015. Prion- Dehay, B., Decressac, M., Bourdenx, M., Guadagnino, I., Fernagut, P.O., Tamburrino, A.,
like propagation of human brain-derived alpha-synuclein in transgenic mice ex- Bassil, F., Meissner, W.G., Bezard, E., 2016a. Targeting alpha-synuclein: therapeutic
pressing human wild-type alpha-synuclein. Acta Neuropathol. Commun. 3, 75. options. Mov. Disord. 31, 882–888.
Bezard, E., Imbert, C., Deloire, X., Bioulac, B., Gross, C.E., 1997. A chronic MPTP model Dehay, B., Vila, M., Bezard, E., Brundin, P., Kordower, J.H., 2016b. Alpha-synuclein
reproducing the slow evolution of Parkinson's disease: evolution of motor symptoms propagation: new insights from animal models. Mov. Disord. 31, 161–168.
in the monkey. Brain Res. 766, 107–112. Delatour, B., Guegan, M., Volk, A., Dhenain, M., 2006. In vivo MRI and histological
Bezard, E., Dovero, S., Prunier, C., Ravenscroft, P., Chalon, S., Guilloteau, D., Crossman, evaluation of brain atrophy in APP/PS1 transgenic mice. Neurobiol. Aging 27,
A.R., Bioulac, B., Brotchie, J.M., Gross, C.E., 2001. Relationship between the ap- 835–847.
pearance of symptoms and the level of nigrostriatal degeneration in a progressive 1- Delong, M.R., Crutcher, M.D., Georgopoulos, A.P., 1985. Primate globus pallidus and
methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson's subthalamic nucleus: functional organization. J. Neurophysiol. 53, 530–543.
disease. J. Neurosci. 21, 6853–6861. Desplats, P., Lee, H.J., Bae, E.J., Patrick, C., Rockenstein, E., Crews, L., Spencer, B.,
Bezard, E., Ferry, S., Mach, U., Stark, H., Leriche, L., Boraud, T., Gross, C., Sokoloff, P., Masliah, E., Lee, S.J., 2009. Inclusion formation and neuronal cell death through
2003. Attenuation of levodopa-induced dyskinesia by normalizing dopamine D3 re- neuron-to-neuron transmission of alpha-synuclein. Proc. Natl. Acad. Sci. U. S. A. 106,
ceptor function. Nat. Med. 9, 762–767. 13010–13015.
Borghi, R., Marchese, R., Negro, A., Marinelli, L., Forloni, G., Zaccheo, D., Abbruzzese, G., Ehringer, H., Hornykiewicz, O., 1960. Distribution of noradrenaline and dopamine (3-
Tabaton, M., 2000. Full length alpha-synuclein is present in cerebrospinal fluid from hydroxytyramine) in the human brain and their behavior in diseases of the extra-
Parkinson's disease and normal subjects. Neurosci. Lett. 287, 65–67. pyramidal system. Klin. Wochenschr. 38, 1236–1239.
Bourdenx, M., Chansel-Debordeaux, L., Dovero, S., Grouthier, V., Uranga, J., Dutheil, N., El-Agnaf, O.M., Salem, S.A., Paleologou, K.E., Cooper, L.J., Fullwood, N.J., Gibson, M.J.,
Brun, S., Espagna, A., Groc, L., Li, Q., Jimenez, C., Bezard, E., Dehay, B., 2015a. In Curran, M.D., Court, J.A., Mann, D.M., Ikeda, S., Cookson, M.R., Hardy, J., Allsop, D.,
utero delivery of rAAV mediates widespread brain transduction in rats and monkeys: 2003. Alpha-synuclein implicated in Parkinson's disease is present in extracellular
towards new models of PD. In: Society of Neuroscience Conference. biological fluids, including human plasma. FASEB J. 17, 1945–1947.
Bourdenx, M., Dovero, S., Engeln, M., Bido, S., Bastide, M.F., Dutheil, N., Vollenweider, I., Eslamboli, A., Romero-Ramos, M., Burger, C., Bjorklund, T., Muzyczka, N., Mandel, R.J.,
Baud, L., Piron, C., Grouthier, V., Boraud, T., Porras, G., Li, Q., Baekelandt, V., Baker, H., Ridley, R.M., Kirik, D., 2007. Long-term consequences of human alpha-
Scheller, D., Michel, A., Fernagut, P.O., Georges, F., Courtine, G., Bezard, E., Dehay, synuclein overexpression in the primate ventral midbrain. Brain 130, 799–815.
B., 2015b. Lack of additive role of ageing in nigrostriatal neurodegeneration triggered Fernagut, P.O., Li, Q., Dovero, S., Chan, P., Wu, T., Ravenscroft, P., Hill, M., Chen, Z.,
by alpha-synuclein overexpression. Acta Neuropathol. Commun. 3, 46. Bezard, E., 2010. Dopamine transporter binding is unaffected by L-DOPA adminis-
Braak, H., Del Tredici, K., Rub, U., De Vos, R.A., Jansen Steur, E.N., Braak, E., 2003. tration in normal and MPTP-treated monkeys. PLoS One 5, e14053.
Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol. Aging Forno, L.S., Langston, J.W., Delanney, L.E., Irwin, I., Ricaurte, G.A., 1986. Locus ceruleus
24, 197–211. lesions and eosinophilic inclusions in MPTP-treated monkeys. Ann. Neurol. 20,
Braak, H., De Vos, R.A., Bohl, J., Del Tredici, K., 2006. Gastric alpha-synuclein im- 449–455.
munoreactive inclusions in Meissner's and Auerbach's plexuses in cases staged for Foulds, P.G., Yokota, O., Thurston, A., Davidson, Y., Ahmed, Z., Holton, J., Thompson,
Parkinson's disease-related brain pathology. Neurosci. Lett. 396, 67–72. J.C., Akiyama, H., Arai, T., Hasegawa, M., Gerhard, A., Allsop, D., Mann, D.M., 2012.
Brundin, P., Li, J.Y., Holton, J.L., Lindvall, O., Revesz, T., 2008. Research in motion: the Post mortem cerebrospinal fluid alpha-synuclein levels are raised in multiple system
enigma of Parkinson's disease pathology spread. Nat. Rev. Neurosci. 9, 741–745. atrophy and distinguish this from the other alpha-synucleinopathies, Parkinson's
Button, K.S., Ioannidis, J.P., Mokrysz, C., Nosek, B.A., Flint, J., Robinson, E.S., Munafo, disease and Dementia with Lewy bodies. Neurobiol. Dis. 45, 188–195.
M.R., 2013. Power failure: why small sample size undermines the reliability of Fox, S.H., Brotchie, J.M., 2010. The MPTP-lesioned non-human primate models of
neuroscience. Nat. Rev. Neurosci. 14, 365–376. Parkinson's disease. Past, present, and future. Prog. Brain Res. 184, 133–157.
Camus, S., Ko, W.K., Pioli, E., Bezard, E., 2015. Why bother using non-human primate Fox, S.H., Johnston, T.H., Li, Q., Brotchie, J., Bezard, E., 2012. A critique of available
models of cognitive disorders in translational research? Neurobiol. Learn. Mem. 124, scales and presentation of the Non-Human Primate Dyskinesia Rating Scale. Mov.
123–129. Disord. 27, 1373–1378.
Cannon, J.R., Greenamyre, J.T., 2010. Neurotoxic in vivo models of Parkinson's disease Fujiwara, H., Hasegawa, M., Dohmae, N., Kawashima, A., Masliah, E., Goldberg, M.S.,
recent advances. Prog. Brain Res. 184, 17–33. Shen, J., Takio, K., Iwatsubo, T., 2002. Alpha-synuclein is phosphorylated in synu-
Capogrosso, M., Milekovic, T., Borton, D., Wagner, F., Moraud, E.M., Mignardot, J.B., cleinopathy lesions. Nat. Cell Biol. 4, 160–164.
Buse, N., Gandar, J., Barraud, Q., Xing, D., Rey, E., Duis, S., Jianzhong, Y., Ko, W.K., Gelpi, E., Navarro-Otano, J., Tolosa, E., Gaig, C., Compta, Y., Rey, M.J., Marti, M.J.,
Li, Q., Detemple, P., Denison, T., Micera, S., Bezard, E., Bloch, J., Courtine, G., 2016. Hernandez, I., Valldeoriola, F., Rene, R., Ribalta, T., 2014. Multiple organ involve-
A brain-spine interface alleviating gait deficits after spinal cord injury in primates. ment by alpha-synuclein pathology in Lewy body disorders. Mov. Disord. 29,
Nature 539, 284–288. 1010–1018.
Cavaliere, F., Cerf, L., Dehay, B., 2015. Lewy Body extracts from Parkinson's Disease Gold, S.J., Hoang, C.V., Potts, B.W., Porras, G., Pioli, E., Kim, K.W., Nadjar, A., Qin, C.,
Brains as models for high-throughput screens of neurotoxicity and a-synuclein Lahoste, G.J., Li, Q., Bioulac, B.H., Waugh, J.L., Gurevich, E., Neve, R.L., Bezard, E.,
spreading from cell-to-cell through endocytosis. In: Neuroscience Sf (Ed.), 45th 2007. RGS9-2 negatively modulates L-3,4-dihydroxyphenylalanine-induced dyski-
Annual Meeting of Society for Neuroscience. Society for Neuroscience, Chicago. nesia in experimental Parkinson's disease. J. Neurosci. 27, 14338–14348.
Chartier-Harlin, M.C., Kachergus, J., Roumier, C., Mouroux, V., Douay, X., Lincoln, S., Gotham, A.M., Brown, R.G., Marsden, C.D., 1988. ‘Frontal’ cognitive function in patients
Levecque, C., Larvor, L., Andrieux, J., Hulihan, M., Waucquier, N., Defebvre, L., with Parkinson's disease ‘on’ and ‘off’ levodopa. Brain 111 (Pt 2), 299–321.
Amouyel, P., Farrer, M., Destee, A., 2004. Alpha-synuclein locus duplication as a Grealish, S., Xie, L., Kelly, M., Dowd, E., 2008. Unilateral axonal or terminal injection of
cause of familial Parkinson's disease. Lancet 364, 1167–1169. 6-hydroxydopamine causes rapid-onset nigrostriatal degeneration and contralateral
Chung, C.Y., Koprich, J.B., Siddiqi, H., Isacson, O., 2009. Dynamic changes in presynaptic motor impairments in the rat. Brain Res. Bull. 77, 312–319.
and axonal transport proteins combined with striatal neuroinflammation precede Hackam, D.G., 2007. Translating animal research into clinical benefit. BMJ 334, 163–164.
dopaminergic neuronal loss in a rat model of AAV alpha-synucleinopathy. J. Hackam, D.G., Redelmeier, D.A., 2006. Translation of research evidence from animals to
Neurosci. 29, 3365–3373. humans. JAMA 296, 1731–1732.
Cooper, A.A., Gitler, A.D., Cashikar, A., Haynes, C.M., Hill, K.J., Bhullar, B., Liu, K., Xu, Hansen, C., Angot, E., Bergstrom, A.L., Steiner, J.A., Pieri, L., Paul, G., Outeiro, T.F.,
K., Strathearn, K.E., Liu, F., Cao, S., Caldwell, K.A., Caldwell, G.A., Marsischky, G., Melki, R., Kallunki, P., Fog, K., Li, J.Y., Brundin, P., 2011. Alpha-synuclein propa-
Kolodner, R.D., Labaer, J., Rochet, J.C., Bonini, N.M., Lindquist, S., 2006. Alpha- gates from mouse brain to grafted dopaminergic neurons and seeds aggregation in
synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. cultured human cells. J. Clin. Invest. 121, 715–725.
Science 313, 324–328. Holmqvist, S., Chutna, O., Bousset, L., Aldrin-Kirk, P., Li, W., Bjorklund, T., Wang, Z.Y.,
Crossman, A.R., Mitchell, I.J., Sambrook, M.A., 1985. Regional brain uptake of 2-deox- Roybon, L., Melki, R., Li, J.Y., 2014. Direct evidence of Parkinson pathology spread
yglucose in N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkin- from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 128, 805–820.
sonism in the macaque monkey. Neuropharmacology 24, 587–591. Hyacinthe, C., Barraud, Q., Tison, F., Bezard, E., Ghorayeb, I., 2014. D1 receptor agonist
Danzer, K.M., Kranich, L.R., Ruf, W.P., Cagsal-Getkin, O., Winslow, A.R., Zhu, L., improves sleep-wake parameters in experimental parkinsonism. Neurobiol. Dis. 63,
Vanderburg, C.R., Mclean, P.J., 2012. Exosomal cell-to-cell transmission of alpha 20–24.
synuclein oligomers. Mol. Neurodegener. 7, 42. Ioannidis, J.P., 2008. Why most discovered true associations are inflated. Epidemiology
De Rijk, M.C., Tzourio, C., Breteler, M.M., Dartigues, J.F., Amaducci, L., Lopez-Pousa, S., 19, 640–648.
Manubens-Bertran, J.M., Alperovitch, A., Rocca, W.A., 1997. Prevalence of parkin- Ip, C.W., Klaus, L.C., Karikari, A.A., Visanji, N.P., Brotchie, J.M., Lang, A.E., Volkmann, J.,
sonism and Parkinson's disease in Europe: the EUROPARKINSON Collaborative Koprich, J.B., 2017. AAV1/2-induced overexpression of A53T-alpha-synuclein in the
Study. European Community Concerted Action on the Epidemiology of Parkinson's substantia nigra results in degeneration of the nigrostriatal system with Lewy-like
disease. J. Neurol. Neurosurg. Psychiatry 62, 10–15. pathology and motor impairment: a new mouse model for Parkinson's disease. Acta
Decressac, M., Mattsson, B., Bjorklund, A., 2012a. Comparison of the behavioural and Neuropathol. Commun. 5 (11).
histological characteristics of the 6-OHDA and alpha-synuclein rat models of Jack Jr., C.R., Lowe, V.J., Weigand, S.D., Wiste, H.J., Senjem, M.L., Knopman, D.S.,
Parkinson's disease. Exp. Neurol. 235, 306–315. Shiung, M.M., Gunter, J.L., Boeve, B.F., Kemp, B.J., Weiner, M., Petersen, R.C., 2009.
Decressac, M., Mattsson, B., Lundblad, M., Weikop, P., Bjorklund, A., 2012b. Progressive Serial PIB and MRI in normal, mild cognitive impairment and Alzheimer's disease:
neurodegenerative and behavioural changes induced by AAV-mediated over- implications for sequence of pathological events in Alzheimer's disease. Brain 132,
expression of alpha-synuclein in midbrain dopamine neurons. Neurobiol. Dis. 45, 1355–1365.
939–953. Jarraya, B., Boulet, S., Ralph, G.S., Jan, C., Bonvento, G., Azzouz, M., Miskin, J.E., Shin,
Dehay, B., Fernagut, P.O., 2016. Alpha-synuclein-based models of Parkinson's disease. M., Delzescaux, T., Drouot, X., Herard, A.S., Day, D.M., Brouillet, E., Kingsman, S.M.,
Rev. Neurol. (Paris) 172, 371–378. Hantraye, P., Mitrophanous, K.A., Mazarakis, N.D., Palfi, S., 2009. Dopamine gene
Dehay, B., Dalkara, D., Dovero, S., Li, Q., Bezard, E., 2012. Systemic scAAV9 variant therapy for Parkinson's disease in a nonhuman primate without associated

177
W.K.D. Ko, E. Bezard Experimental Neurology 298 (2017) 172–179

dyskinesia. Sci. Transl. Med. 1, 2ra4. monkeys. Cell Stem Cell 14, 323–328.
Jellinger, K.A., 1991. Pathology of Parkinson's disease. Changes other than the nigros- Liu, Z., Li, X., Zhang, J.T., Cai, Y.J., Cheng, T.L., Cheng, C., Wang, Y., Zhang, C.C., Nie,
triatal pathway. Mol. Chem. Neuropathol. 14, 153–197. Y.H., Chen, Z.F., Bian, W.J., Zhang, L., Xiao, J., Lu, B., Zhang, Y.F., Zhang, X.D., Sang,
Jellinger, K.A., 2011. Synuclein deposition and non-motor symptoms in Parkinson dis- X., Wu, J.J., Xu, X., Xiong, Z.Q., Zhang, F., Yu, X., Gong, N., Zhou, W.H., Sun, Q., Qiu,
ease. J. Neurol. Sci. 310, 107–111. Z., 2016. Autism-like behaviours and germline transmission in transgenic monkeys
Johnston, T.M., Fox, S.H., 2015. Symptomatic models of Parkinson's disease and L-DOPA- overexpressing MeCP2. Nature 530, 98–102.
induced dyskinesia in non-human primates. Curr. Top. Behav. Neurosci. 22, 221–235. Lo Bianco, C., Ridet, J.L., Schneider, B.L., Deglon, N., Aebischer, P., 2002. Alpha-synu-
Jones, D.R., Delenclos, M., Baine, A.T., Deture, M., Murray, M.E., Dickson, D.W., Mclean, cleinopathy and selective dopaminergic neuron loss in a rat lentiviral-based model of
P.J., 2015. Transmission of soluble and insoluble alpha-synuclein to mice. J. Parkinson's disease. Proc. Natl. Acad. Sci. U. S. A. 99, 10813–10818.
Neuropathol. Exp. Neurol. 74, 1158–1169. Luk, K.C., Song, C., O'brien, P., Stieber, A., Branch, J.R., Brunden, K.R., Trojanowski, J.Q.,
Kayed, R., Sokolov, Y., Edmonds, B., Mcintire, T.M., Milton, S.C., Hall, J.E., Glabe, C.G., Lee, V.M., 2009. Exogenous alpha-synuclein fibrils seed the formation of Lewy body-
2004. Permeabilization of lipid bilayers is a common conformation-dependent ac- like intracellular inclusions in cultured cells. Proc. Natl. Acad. Sci. U. S. A. 106,
tivity of soluble amyloid oligomers in protein misfolding diseases. J. Biol. Chem. 279, 20051–20056.
46363–46366. Luk, K.C., Kehm, V., Carroll, J., Zhang, B., O'brien, P., Trojanowski, J.Q., Lee, V.M.,
Kirik, D., Rosenblad, C., Bjorklund, A., 1998. Characterization of behavioral and neuro- 2012a. Pathological alpha-synuclein transmission initiates Parkinson-like neurode-
degenerative changes following partial lesions of the nigrostriatal dopamine system generation in nontransgenic mice. Science 338, 949–953.
induced by intrastriatal 6-hydroxydopamine in the rat. Exp. Neurol. 152, 259–277. Luk, K.C., Kehm, V.M., Zhang, B., O'brien, P., Trojanowski, J.Q., Lee, V.M., 2012b.
Kirik, D., Rosenblad, C., Burger, C., Lundberg, C., Johansen, T.E., Muzyczka, N., Mandel, Intracerebral inoculation of pathological alpha-synuclein initiates a rapidly pro-
R.J., Bjorklund, A., 2002. Parkinson-like neurodegeneration induced by targeted gressive neurodegenerative alpha-synucleinopathy in mice. J. Exp. Med. 209,
overexpression of alpha-synuclein in the nigrostriatal system. J. Neurosci. 22, 975–986.
2780–2791. Mandler, M., Valera, E., Rockenstein, E., Weninger, H., Patrick, C., Adame, A., Santic, R.,
Kirik, D., Annett, L.E., Burger, C., Muzyczka, N., Mandel, R.J., Bjorklund, A., 2003. Meindl, S., Vigl, B., Smrzka, O., Schneeberger, A., Mattner, F., Masliah, E., 2014.
Nigrostriatal alpha-synucleinopathy induced by viral vector-mediated overexpression Next-generation active immunization approach for synucleinopathies: implications
of human alpha-synuclein: a new primate model of Parkinson's disease. Proc. Natl. for Parkinson's disease clinical trials. Acta Neuropathol. 127, 861–879.
Acad. Sci. U. S. A. 100, 2884–2889. Marsden, C.D., 1994. Parkinson's disease. J. Neurol. Neurosurg. Psychiatry 57, 672–681.
Ko, W.K., Pioli, E., Li, Q., Mcguire, S., Dufour, A., Sherer, T.B., Bezard, E., Facheris, M.F., Martic-Kehl, M.I., Schibli, R., Schubiger, P.A., 2012. Can animal data predict human
2014. Combined fenobam and amantadine treatment promotes robust antidyskinetic outcome? Problems and pitfalls of translational animal research. Eur. J. Nucl. Med.
effects in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate Mol. Imaging 39, 1492–1496.
model of Parkinson's disease. Mov. Disord. 29, 772–779. Masliah, E., Rockenstein, E., Mante, M., Crews, L., Spencer, B., Adame, A., Patrick, C.,
Ko, W.K., Camus, S.M., Li, Q., Yang, J., Mcguire, S., Pioli, E.Y., Bezard, E., 2016. An Trejo, M., Ubhi, K., Rohn, T.T., Mueller-Steiner, S., Seubert, P., Barbour, R.,
evaluation of istradefylline treatment on Parkinsonian motor and cognitive deficits in Mcconlogue, L., Buttini, M., Games, D., Schenk, D., 2011. Passive immunization re-
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated macaque models. duces behavioral and neuropathological deficits in an alpha-synuclein transgenic
Neuropharmacology 110, 48–58. model of Lewy body disease. PLoS One 6, e19338.
Koprich, J.B., Johnston, T.H., Reyes, M.G., Sun, X., Brotchie, J.M., 2010. Expression of Mcfarland, N.R., Fan, Z., Xu, K., Schwarzschild, M.A., Feany, M.B., Hyman, B.T., Mclean,
human A53T alpha-synuclein in the rat substantia nigra using a novel AAV1/2 vector P.J., 2009. Alpha-synuclein S129 phosphorylation mutants do not alter nigrostriatal
produces a rapidly evolving pathology with protein aggregation, dystrophic neurite toxicity in a rat model of Parkinson disease. J. Neuropathol. Exp. Neurol. 68,
architecture and nigrostriatal degeneration with potential to model the pathology of 515–524.
Parkinson's disease. Mol. Neurodegener. 5, 43. Meissner, W., Prunier, C., Guilloteau, D., Chalon, S., Gross, C.E., Bezard, E., 2003. Time-
Koprich, J.B., Johnston, T.H., Huot, P., Reyes, M.G., Espinosa, M., Brotchie, J.M., 2011. course of nigrostriatal degeneration in a progressive MPTP-lesioned macaque model
Progressive neurodegeneration or endogenous compensation in an animal model of of Parkinson's disease. Mol. Neurobiol. 28, 209–218.
Parkinson's disease produced by decreasing doses of alpha-synuclein. PLoS One 6, Mitchell, I.J., Cross, A.J., Sambrook, M.A., Crossman, A.R., 1985. Sites of the neurotoxic
e17698. action of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in the macaque monkey in-
Koprich, J.B., Johnston, T.H., Reyes, G., Omana, V., Brotchie, J.M., 2016. Towards a non- clude the ventral tegmental area and the locus coeruleus. Neurosci. Lett. 61,
human primate model of alpha-synucleinopathy for development of therapeutics for 195–200.
Parkinson's disease: optimization of AAV1/2 delivery parameters to drive sustained Mollenhauer, B., Locascio, J.J., Schulz-Schaeffer, W., Sixel-Doring, F., Trenkwalder, C.,
expression of alpha synuclein and dopaminergic degeneration in macaque. PLoS One Schlossmacher, M.G., 2011. Alpha-synuclein and tau concentrations in cerebrospinal
11, e0167235. fluid of patients presenting with parkinsonism: a cohort study. Lancet Neurol. 10,
Kordower, J.H., Chu, Y., Hauser, R.A., Freeman, T.B., Olanow, C.W., 2008. Lewy body- 230–240.
like pathology in long-term embryonic nigral transplants in Parkinson's disease. Nat. Mougenot, A.L., Nicot, S., Bencsik, A., Morignat, E., Verchere, J., Lakhdar, L., Legastelois,
Med. 14, 504–506. S., Baron, T., 2012. Prion-like acceleration of a synucleinopathy in a transgenic
Kordower, J.H., Dodiya, H.B., Kordower, A.M., Terpstra, B., Paumier, K., Madhavan, L., mouse model. Neurobiol. Aging 33, 2225–2228.
Sortwell, C., Steece-Collier, K., Collier, T.J., 2011. Transfer of host-derived alpha Niu, Y., Shen, B., Cui, Y., Chen, Y., Wang, J., Wang, L., Kang, Y., Zhao, X., Si, W., Li, W.,
synuclein to grafted dopaminergic neurons in rat. Neurobiol. Dis. 43, 552–557. Xiang, A.P., Zhou, J., Guo, X., Bi, Y., Si, C., Hu, B., Dong, G., Wang, H., Zhou, Z., Li,
Kowall, N.W., Hantraye, P., Brouillet, E., Beal, M.F., Mckee, A.C., Ferrante, R.J., 2000. T., Tan, T., Pu, X., Wang, F., Ji, S., Zhou, Q., Huang, X., Ji, W., Sha, J., 2014.
MPTP induces alpha-synuclein aggregation in the substantia nigra of baboons. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated gene
Neuroreport 11, 211–213. targeting in one-cell embryos. Cell 156, 836–843.
Kruger, R., Kuhn, W., Muller, T., Woitalla, D., Graeber, M., Kosel, S., Przuntek, H., Epplen, Olanow, C.W., Brundin, P., 2013. Parkinson's disease and alpha synuclein: is Parkinson's
J.T., Schols, L., Riess, O., 1998. Ala30Pro mutation in the gene encoding alpha-sy- disease a prion-like disorder? Mov. Disord. 28, 31–40.
nuclein in Parkinson's disease. Nat. Genet. 18, 106–108. Oliveras-Salva, M., Van Der Perren, A., Casadei, N., Stroobants, S., Nuber, S., D'hooge, R.,
Kulisevsky, J., Avila, A., Barbanoj, M., Antonijoan, R., Berthier, M.L., Gironell, A., 1996. Van Den Haute, C., Baekelandt, V., 2013. rAAV2/7 vector-mediated overexpression
Acute effects of levodopa on neuropsychological performance in stable and fluctu- of alpha-synuclein in mouse substantia nigra induces protein aggregation and pro-
ating Parkinson's disease patients at different levodopa plasma levels. Brain 119 (Pt gressive dose-dependent neurodegeneration. Mol. Neurodegener. 8, 44.
6), 2121–2132. Pan-Montojo, F., Anichtchik, O., Dening, Y., Knels, L., Pursche, S., Jung, R., Jackson, S.,
Lane, E.L., Cheetham, S.C., Jenner, P., 2006. Does contraversive circling in the 6-OHDA- Gille, G., Spillantini, M.G., Reichmann, H., Funk, R.H., 2010. Progression of
lesioned rat indicate an ability to induce motor complications as well as therapeutic Parkinson's disease pathology is reproduced by intragastric administration of rote-
effects in Parkinson's disease? Exp. Neurol. 197, 284–290. none in mice. PLoS One 5, e8762.
Lauwers, E., Debyser, Z., Van Dorpe, J., De Strooper, B., Nuttin, B., Baekelandt, V., 2003. Perel, P., Roberts, I., Sena, E., Wheble, P., Briscoe, C., Sandercock, P., Macleod, M.,
Neuropathology and neurodegeneration in rodent brain induced by lentiviral vector- Mignini, L.E., Jayaram, P., Khan, K.S., 2007. Comparison of treatment effects be-
mediated overexpression of alpha-synuclein. Brain Pathol. 13, 364–372. tween animal experiments and clinical trials: systematic review. BMJ 334, 197.
Lee, C.S., Sauer, H., Bjorklund, A., 1996. Dopaminergic neuronal degeneration and motor Perez-Otano, I., Herrero, M.T., Oset, C., De Ceballos, M.L., Luquin, M.R., Obeso, J.A., Del
impairments following axon terminal lesion by instrastriatal 6-hydroxydopamine in Rio, J., 1991. Extensive loss of brain dopamine and serotonin induced by chronic
the rat. Neuroscience 72, 641–653. administration of MPTP in the marmoset. Brain Res. 567, 127–132.
Lee, H.J., Patel, S., Lee, S.J., 2005. Intravesicular localization and exocytosis of alpha- Pinna, A., Ko, W.K., Costa, G., Tronci, E., Fidalgo, C., Simola, N., Li, Q., Tabrizi, M.A.,
synuclein and its aggregates. J. Neurosci. 25, 6016–6024. Bezard, E., Carta, M., Morelli, M., 2016. Antidyskinetic effect of A2A and 5HT1A/1B
Lesage, S., Anheim, M., Letournel, F., Bousset, L., Honore, A., Rozas, N., Pieri, L., receptor ligands in two animal models of Parkinson's disease. Mov. Disord. 31,
Madiona, K., Durr, A., Melki, R., Verny, C., Brice, A., 2013. G51D alpha-synuclein 501–511.
mutation causes a novel parkinsonian-pyramidal syndrome. Ann. Neurol. 73, Polymeropoulos, M.H., Lavedan, C., Leroy, E., Ide, S.E., Dehejia, A., Dutra, A., Pike, B.,
459–471. Root, H., Rubenstein, J., Boyer, R., Stenroos, E.S., Chandrasekharappa, S.,
Li, J.Y., Englund, E., Widner, H., Rehncrona, S., Bjorklund, A., Lindvall, O., Brundin, P., Athanassiadou, A., Papapetropoulos, T., Johnson, W.G., Lazzarini, A.M., Duvoisin,
2010. Characterization of Lewy body pathology in 12- and 16-year-old intrastriatal R.C., Di Iorio, G., Golbe, L.I., Nussbaum, R.L., 1997. Mutation in the alpha-synuclein
mesencephalic grafts surviving in a patient with Parkinson's disease. Mov. Disord. 25, gene identified in families with Parkinson's disease. Science 276, 2045–2047.
1091–1096. Pont-Sunyer, C., Hotter, A., Gaig, C., Seppi, K., Compta, Y., Katzenschlager, R., Mas, N.,
Liu, H., Chen, Y., Niu, Y., Zhang, K., Kang, Y., Ge, W., Liu, X., Zhao, E., Wang, C., Lin, S., Hofeneder, D., Brucke, T., Bayes, A., Wenzel, K., Infante, J., Zach, H., Pirker, W.,
Jing, B., Si, C., Lin, Q., Chen, X., Lin, H., Pu, X., Wang, Y., Qin, B., Wang, F., Wang, H., Posada, I.J., Alvarez, R., Ispierto, L., De Fabregues, O., Callen, A., Palasi, A., Aguilar,
Si, W., Zhou, J., Tan, T., Li, T., Ji, S., Xue, Z., Luo, Y., Cheng, L., Zhou, Q., Li, S., Sun, M., Marti, M.J., Valldeoriola, F., Salamero, M., Poewe, W., Tolosa, E., 2015. The onset
Y.E., Ji, W., 2014. TALEN-mediated gene mutagenesis in rhesus and cynomolgus of nonmotor symptoms in Parkinson's disease (the ONSET PD study). Mov. Disord.

178
W.K.D. Ko, E. Bezard Experimental Neurology 298 (2017) 172–179

30, 229–237. dementia with lewy bodies. Proc. Natl. Acad. Sci. U. S. A. 95, 6469–6473.
Postuma, R.B., Gagnon, J.F., Vendette, M., Montplaisir, J.Y., 2009. Markers of neurode- Theodore, S., Cao, S., Mclean, P.J., Standaert, D.G., 2008. Targeted overexpression of
generation in idiopathic rapid eye movement sleep behaviour disorder and human alpha-synuclein triggers microglial activation and an adaptive immune re-
Parkinson's disease. Brain 132, 3298–3307. sponse in a mouse model of Parkinson disease. J. Neuropathol. Exp. Neurol. 67,
Prunier, C., Bezard, E., Montharu, J., Mantzarides, M., Besnard, J.C., Baulieu, J.L., Gross, 1149–1158.
C., Guilloteau, D., Chalon, S., 2003. Presymptomatic diagnosis of experimental Tokuda, T., Qureshi, M.M., Ardah, M.T., Varghese, S., Shehab, S.A., Kasai, T., Ishigami,
Parkinsonism with 123I-PE2I SPECT. NeuroImage 19, 810–816. N., Tamaoka, A., Nakagawa, M., El-Agnaf, O.M., 2010. Detection of elevated levels of
Puschmann, A., Ross, O.A., Vilarino-Guell, C., Lincoln, S.J., Kachergus, J.M., Cobb, S.A., alpha-synuclein oligomers in CSF from patients with Parkinson disease. Neurology
Lindquist, S.G., Nielsen, J.E., Wszolek, Z.K., Farrer, M., Widner, H., Van Westen, D., 75, 1766–1772.
Hagerstrom, D., Markopoulou, K., Chase, B.A., Nilsson, K., Reimer, J., Nilsson, C., Tran, H.T., Chung, C.H., Iba, M., Zhang, B., Trojanowski, J.Q., Luk, K.C., Lee, V.M., 2014.
2009. A Swedish family with de novo alpha-synuclein A53T mutation: evidence for Alpha-synuclein immunotherapy blocks uptake and templated propagation of mis-
early cortical dysfunction. Parkinsonism Relat. Disord. 15, 627–632. folded alpha-synuclein and neurodegeneration. Cell Rep. 7, 2054–2065.
Recasens, A., Dehay, B., 2014. Alpha-synuclein spreading in Parkinson's disease. Front. Ulusoy, A., Rusconi, R., Perez-Revuelta, B.I., Musgrove, R.E., Helwig, M., Winzen-
Neuroanat. 8, 159. Reichert, B., Di Monte, D.A., 2013. Caudo-rostral brain spreading of alpha-synuclein
Recasens, A., Dehay, B., Bove, J., Carballo-Carbajal, I., Dovero, S., Perez-Villalba, A., through vagal connections. EMBO Mol. Med. 5, 1119–1127.
Fernagut, P.O., Blesa, J., Parent, A., Perier, C., Farinas, I., Obeso, J.A., Bezard, E., Van Der Worp, H.B., Macleod, M.R., 2011. Preclinical studies of human disease: time to
Vila, M., 2014. Lewy body extracts from Parkinson disease brains trigger alpha-sy- take methodological quality seriously. J. Mol. Cell. Cardiol. 51, 449–450.
nuclein pathology and neurodegeneration in mice and monkeys. Ann. Neurol. 75, Van Der Worp, H.B., De Haan, P., Morrema, E., Kalkman, C.J., 2005. Methodological
351–362. quality of animal studies on neuroprotection in focal cerebral ischaemia. J. Neurol.
Recchia, A., Rota, D., Debetto, P., Peroni, D., Guidolin, D., Negro, A., Skaper, S.D., Giusti, 252, 1108–1114.
P., 2008. Generation of a alpha-synuclein-based rat model of Parkinson's disease. Van Der Worp, H.B., Howells, D.W., Sena, E.S., Porritt, M.J., Rewell, S., O'collins, V.,
Neurobiol. Dis. 30, 8–18. Macleod, M.R., 2010. Can animal models of disease reliably inform human studies?
Rey, N.L., Petit, G.H., Bousset, L., Melki, R., Brundin, P., 2013. Transfer of human alpha- PLoS Med. 7, e1000245.
synuclein from the olfactory bulb to interconnected brain regions in mice. Acta Venda, L.L., Cragg, S.J., Buchman, V.L., Wade-Martins, R., 2010. Alpha-synuclein and
Neuropathol. 126, 555–573. dopamine at the crossroads of Parkinson's disease. Trends Neurosci. 33, 559–568.
Rylander, D., Iderberg, H., Li, Q., Dekundy, A., Zhang, J., Li, H., Baishen, R., Danysz, W., Volpicelli-Daley, L.A., Luk, K.C., Patel, T.P., Tanik, S.A., Riddle, D.M., Stieber, A.,
Bezard, E., Cenci, M.A., 2010. A mGluR5 antagonist under clinical development Meaney, D.F., Trojanowski, J.Q., Lee, V.M., 2011. Exogenous alpha-synuclein fibrils
improves L-DOPA-induced dyskinesia in parkinsonian rats and monkeys. Neurobiol. induce Lewy body pathology leading to synaptic dysfunction and neuron death.
Dis. 39, 352–361. Neuron 72, 57–71.
Sato, H., Kato, T., Arawaka, S., 2015. Potential of cellular and animal models based on a Wagner, J., Ryazanov, S., Leonov, A., Levin, J., Shi, S., Schmidt, F., Prix, C., Pan-Montojo,
prion-like propagation of alpha-synuclein for assessing antiparkinson agents. Mol. F., Bertsch, U., Mitteregger-Kretzschmar, G., Geissen, M., Eiden, M., Leidel, F.,
Neurobiol. 52, 226–235. Hirschberger, T., Deeg, A.A., Krauth, J.J., Zinth, W., Tavan, P., Pilger, J.,
Sauer, H., Oertel, W.H., 1994. Progressive degeneration of nigrostriatal dopamine neu- Zweckstetter, M., Frank, T., Bahr, M., Weishaupt, J.H., Uhr, M., Urlaub, H.,
rons following intrastriatal terminal lesions with 6-hydroxydopamine: a combined Teichmann, U., Samwer, M., Botzel, K., Groschup, M., Kretzschmar, H., Griesinger,
retrograde tracing and immunocytochemical study in the rat. Neuroscience 59, C., Giese, A., 2013. Anle138b: a novel oligomer modulator for disease-modifying
401–415. therapy of neurodegenerative diseases such as prion and Parkinson's disease. Acta
Schneider, J.S., Pioli, E.Y., Jianzhong, Y., Li, Q., Bezard, E., 2013. Levodopa improves Neuropathol. 125, 795–813.
motor deficits but can further disrupt cognition in a macaque Parkinson model. Mov. Yang, S.H., Cheng, P.H., Banta, H., Piotrowska-Nitsche, K., Yang, J.J., Cheng, E.C.,
Disord. 28, 663–667. Snyder, B., Larkin, K., Liu, J., Orkin, J., Fang, Z.H., Smith, Y., Bachevalier, J., Zola,
Schwarting, R.K., Huston, J.P., 1996. The unilateral 6-hydroxydopamine lesion model in S.M., Li, S.H., Li, X.J., Chan, A.W., 2008. Towards a transgenic model of Huntington's
behavioral brain research. Analysis of functional deficits, recovery and treatments. disease in a non-human primate. Nature 453, 921–924.
Prog. Neurobiol. 50, 275–331. Yasuda, T., Nihira, T., Ren, Y.R., Cao, X.Q., Wada, K., Setsuie, R., Kabuta, T., Wada, K.,
Sena, E., Van Der Worp, H.B., Howells, D., Macleod, M., 2007. How can we improve the Hattori, N., Mizuno, Y., Mochizuki, H., 2009. Effects of UCH-L1 on alpha-synuclein
pre-clinical development of drugs for stroke? Trends Neurosci. 30, 433–439. over-expression mouse model of Parkinson's disease. J. Neurochem. 108, 932–944.
Singleton, A.B., Farrer, M., Johnson, J., Singleton, A., Hague, S., Kachergus, J., Hulihan, Yin, M., Borton, D.A., Komar, J., Agha, N., Lu, Y., Li, H., Laurens, J., Lang, Y., Li, Q., Bull,
M., Peuralinna, T., Dutra, A., Nussbaum, R., Lincoln, S., Crawley, A., Hanson, M., C., Larson, L., Rosler, D., Bezard, E., Courtine, G., Nurmikko, A.V., 2014. Wireless
Maraganore, D., Adler, C., Cookson, M.R., Muenter, M., Baptista, M., Miller, D., neurosensor for full-spectrum electrophysiology recordings during free behavior.
Blancato, J., Hardy, J., Gwinn-Hardy, K., 2003. Alpha-synuclein locus triplication Neuron 84, 1170–1182.
causes Parkinson's disease. Science 302 (841). Zarranz, J.J., Alegre, J., Gomez-Esteban, J.C., Lezcano, E., Ros, R., Ampuero, I., Vidal, L.,
Spillantini, M.G., Schmidt, M.L., Lee, V.M., Trojanowski, J.Q., Jakes, R., Goedert, M., Hoenicka, J., Rodriguez, O., Atares, B., Llorens, V., Gomez Tortosa, E., Del Ser, T.,
1997. Alpha-synuclein in Lewy bodies. Nature 388, 839–840. Munoz, D.G., De Yebenes, J.G., 2004. The new mutation, E46K, of alpha-synuclein
Spillantini, M.G., Crowther, R.A., Jakes, R., Hasegawa, M., Goedert, M., 1998. Alpha- causes Parkinson and Lewy body dementia. Ann. Neurol. 55, 164–173.
synuclein in filamentous inclusions of Lewy bodies from Parkinson's disease and

179

You might also like