Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 May 1.
Published in final edited form as: Mov Disord. 2018 Feb 8;33(5):717–729. doi: 10.1002/mds.27296

New Developments in Genetic Rat Models of Parkinson’s Disease

Rose B Creed 1, Matthew S Goldberg 1,*
PMCID: PMC5992003  NIHMSID: NIHMS931185  PMID: 29418019

Abstract

Preclinical research on Parkinson’s disease (PD) has relied heavily on mouse and rat animal models. Initially, PD animal models were generated primarily by chemical neurotoxins that induce acute loss of dopaminergic neurons in the substantia nigra. Upon the discovery of genetic mutations causally linked to PD, mice were used more than rats to generate laboratory animals bearing PD-linked mutations because mutagenesis was more difficult in rats. Recent advances in technology for mammalian genome engineering and optimization of viral expression vectors have increased the use of genetic rat models of PD. Emerging research tools include “knockout” rats with disruption of genes in which mutations have been causally linked to PD, including LRRK2, α-synuclein, Parkin, PINK1 and DJ-1. Rats have also been increasingly used for transgenic and viral-mediated overexpression of genes relevant to PD, particularly α-synuclein. It may not be realistic to obtain a single animal model that completely reproduces every feature of a human disease as complex as PD. Nevertheless, compared to mice with the same mutations, many genetic rat animal models of PD better reproduce key aspects of PD including progressive loss of dopaminergic neurons in the substantia nigra, locomotor behavior deficits, and age-dependent formation of abnormal α-synuclein protein aggregates. Here we briefly review new developments in genetic rat models of PD that may have greater potential for identifying underlying mechanisms, for discovering novel therapeutic targets, and for developing greatly needed treatments to slow or halt disease progression.

Keywords: Parkinson’s disease, synuclein, LRRK2, Parkin PINK1, DJ-1, transgenic, knockout, viral, neurotoxin, rat, model

Introduction

Animal models of Parkinson’s disease (PD) are important tools to better understand PD etiology and to develop and test more effective treatments. There are many advantages of rodent PD models compared to other animal models commonly used in basic science research. The brains of mice and rats have relatively conserved neuroanatomy compared to human brains, including the function and connectivity of nuclei that are most affected in PD, such as the substantia nigra pars compacta, locus coeruleus, dorsal raphe, and ventral tegmental area. Involvement of other organs and systems including the enteric system, the olfactory system as well as the innate and adaptive immune systems, can easily be studied in mouse and rat models of PD. This is particularly important for studies of the role of inflammation in PD.1, 2 There is currently no single PD animal model that perfectly recapitulates all the central features of PD. After many years of substantial efforts to develop animal models of PD, there are now good rat models that reproduce individual or multiple aspects of PD, such as loss of dopaminergic neurons in the substantia nigra, reduced dopamine levels in the striatum, L-DOPA induced dyskinesia, formation of protein aggregates that resemble Lewy bodies or Lewy neurites, locomotor behavior deficits, age-dependent onset and age-dependent progression in severity of PD-related symptoms or pathology.

Prior to the identification of genes with mutations causally linked to PD, animal models were generated primarily using neurotoxins, such as 6-hydroxydopmine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) that can cause acute neurodegeneration and death of dopaminergic neurons in the substantia nigra upon intracranial 6-OHDA or intraperitoneal MPTP injection.3, 4 Chronic administration of L-DOPA to 6-OHDA-lesioned rats can create useful animal models to study L-DOPA induced dyskinesia.5, 6 There continues to be significant preclinical research using PD animal models generated with 6-OHDA and MPTP, along with other neurotoxins such as rotenone.7 One of the main drawbacks of neurotoxin models of PD is that the onset of neurodegeneration and behavioral deficits is induced and relatively acute rather than spontaneous and gradually progressing in severity, as occurs in PD. Furthermore, the mechanisms of neurodegeneration in neurotoxin PD models may not be physiologically relevant to the mechanisms of neurodegeneration in PD, except for cases of parkinsonism caused by inadvertent exposure to neurotoxins like MPTP.8 With the exception of rats treated chronically with rotenone, most neurotoxin animal models do not develop α-synuclein immunoreactive intraneuronal protein inclusions resembling pathognomonic Lewy bodies.7 Since the identification of human mutations linked to familial forms of PD, transgenic animal models have been generated and characterized with the goal of obtaining PD animal models that more closely model human disease physiology and progression.9, 10

Homozygous or compound heterozygous loss-of-function mutations in three genes have been causally linked to recessively inherited PD: PINK1, Parkin and DJ-1.1113 Mutations in these genes can be modeled by targeted disruption to generate “knockout” (KO) mice and rats. Gain-of-function single point mutations in LRRK2 and α-synuclein have been linked to dominantly inherited PD.1417 Also, duplication or triplication of the entire SNCA gene on chromosome 4, coding for normal α-synuclein, causes PD with more SNCA gene copies causing earlier onset, more rapid progression and prominent end-stage dementia.1820 This has important implications for generating animal models of PD because elevated expression of normal α-synuclein is sufficient to cause PD in rare families with SNCA gene multiplication.21 This enhances the genetic construct validity of transgenic α-synuclein overexpression models. Because many genetic animal models share construct validity with PD patients bearing the same or similar PD-linked mutations, genetic animal models could reveal pathogenic mechanisms and potentially provide better predictive models to expedite discovery and development of more effective therapies.

Additional methodologies for generating PD models include viral mediated expression of transgenes in various brain regions upon stereotaxic intracranial injection and, more recently, intracranial injection of monomer and fibril forms of α-synuclein, which is the major protein component of Lewy bodies and Lewy neurites.22 All of these methods continue to be widely used because each PD animal model has advantages and disadvantages, depending upon the outcome measures and the specific aspects of PD being studied (e.g. neuroprotection, regeneration, dyskinesia, progression, etc.). The recent advance in transgenic technology has enabled greater use of rats for genetic PD animal models. We focus this review on genetic rat models of PD, including rats injected with proteins or virally expressed genes with mutations linked to PD. Mouse and neurotoxin animal models of PD remain important research tools described in more detail by many comprehensive and focused reviews.10, 2328

Genetic Rat Models of PD

The identification of human mutations causally linked to inherited forms of PD prompted the generation of genetic PD animal models with the hope of reproducing the spontaneous adult-onset and progressive neurodegeneration characteristic of PD. Most of the genetic PD animal models were initially generated in mice because this was the most feasible system. Surprisingly, genetic mouse models of PD largely fail to develop greater than 50% loss of nigral dopaminergic neurons except when genes are overexpressed or disrupted exclusively in dopaminergic neurons.29, 30 In contrast to mice, some genetic rat models of PD better reproduce the significant (greater than 50%) nigral cell loss that presumably occurs in humans before the onset of PD motor symptoms.31 The reasons for the different phenotypes of mice and rats bearing similar gene mutations are not clear. In addition to many behavioral, physiological and biochemical differences between mice and rats, there are substantial differences in gene sequence and expression. For example, rats have significantly lower expression of LRRK2 in the substantia nigra compared to mice.32

At the level of individual protein domains (Figure 1), PD-linked mutations appear in highly conserved domains, consistent with the evolutionarily conserved sequences having functional importance. One noteworthy difference between human and rodent proteins is the sequence of α-synuclein at amino acid position 53 (circled in Figure 1). The mutation of alanine 53 to threonine (A53T) in humans is causally linked to familial PD.14 This presents a conundrum of how best to model the human α-synuclein A53T mutation in rodents because rats and mice already have a threonine at position 53. In fact, position 53 is threonine in α-synuclein from fish, birds, reptiles and all other mammals except primates.33 The human A53T mutations that appears “naturally” in most other species does not appear to be detrimental, even in species that live as long or longer than humans.33 Perhaps humans are uniquely susceptible to A53T due to changes at other amino acid positions in α-synuclein, due to differences in other genes, or due to physiological differences.

Figure 1.

Figure 1

Human and rat protein primary sequence comparisons for PD-linked genes. For each gene, the amino acid residues at the beginning and end of each domain are shown below the human sequence. PD-linked missense and nonsense mutations are shown on top of the human protein sequence. Functionally important amino acids are indicated at the bottom of the human protein sequence. The percent sequence identity between rat and human for each domain is indicated in the rat primary sequence. (A) α-Synuclein domains are annotated as follows: N-terminal domain (N-term); Non-Amyloid component domain (NAC); C-terminal domain (C-term). Phosphorylation of serine 129, shown below, is a marker of pathological forms of α-synuclein. (B) LRRK2 domains are as follows: Armadillo-like repeat (ARM); Ankyrin repeat (ANK); Leucine rich repeat (LRR); Roc (ras of complex) GTPase domain (GTPase); C-terminal of Roc (COR); Kinase domain (Kinase); WD40 domain (WD40). Known sites of LRRK2 phosphorylation are shown below. (C) PINK1 domains are annotated as follows: Mitochondrial Targeting Sequence (MTS); transmembrane domain (TMD); N-terminal regulatory region (NT); Kinase domain (Kinase); C-terminal regulatory sequence (C-term). Known sites of PINK1 phosphorylation are shown below. (D) Parkin domains are as follows: Ubiquitin Like domain (Ubl); really interesting new gene (RING0, RING1, RING2, respectively); In between RING domain (IBR); repressor domain (REP). Parkin is activated by PINK1 phosphorylation of serine 65, shown below the ubiquitin-like domain, and the identical serine 65 in ubiquitin. Cysteine 431 is required for Parkin mitochondrial translocation and Parkin E3 ligase activity. (E) DJ-1 domains are as follows: Core domains (Core); dimerization domain (Dimerization). Cysteine 106, shown below, is highly sensitive to oxidation and required for mitochondrial translocation.

PINK1 KO rats

Loss-of-function mutations in the mitochondrial kinase PTEN-induced kinase 1 (PINK1) are causally linked to early onset recessive PD.11 PINK1 missense, nonsense, and frameshift mutations as well as large deletions spanning multiple exons account for about 4–9% of early onset (~24–47 years) PD in Asian populations but only 2–4% in Caucasian poulations.34 In vitro studies suggest that PINK1 functions as a sensor of mitochondria with low membrane potential and as an activator of Parkin (discussed below) to promote sequestration and degradation of defective mitochondria.35, 36 In contrast to PINK1 KO mice,37, 38 PINK1 KO rats exhibit age-dependent loss of nigral dopaminergic neurons beginning at age 6–8 months (corresponding to age 30–40 years in humans).31, 39 Locomotor behavior deficits appear as early as age 4 months and include reduced rearing frequency and distance traveled in an open field apparatus; reduced hind limb grip strength, and significantly increased foot slips and traversal time on a tapered balance beam apparatus.31, 40 Non-motor behavioral abnormalities have yet to be fully characterized, however, PINK1 KO rats have significant age-dependent vocalization deficits (reduced intensity by age 2 months, reduced bandwidth by age 4 months, and reduced peak frequency by age 8 months).40 PINK1 KO rats also exhibit mitochondrial respiration defects and proteinase K resistant α-synuclein aggregates in various brain regions.39, 40 Although the synuclein aggregates in PINK1 KO rats do not have the same appearance as Lewy bodies in PD brains, it is noteworthy that postmortem examinations of PD patients bearing PINK1 mutations have found Lewy body pathology,41, 42 which is not always the case for familial forms of PD.16, 43 The spontaneous nigral neuron loss and the spontaneous appearance of synuclein aggregates are phenotypes not found in most other genetic animal models. Therefore, this recently developed PD animal model may be particularly useful for studying the role of synuclein aggregation in PD pathogenesis and for testing methods to protect against age-dependent loss of nigral dopaminergic neurons.

DJ-1 KO rats

DJ-1 homozygous deletion and point mutations have been linked to autosomal recessive Parkinson’s disease.13 The exact function of DJ-1 remains unclear, but it may be involved in sensing or protecting against oxidative stress.44, 45 DJ-1 KO mice show mild locomotor behavioral deficits and altered nigrostriatal synaptic physiology without dopaminergic neuron loss, even though they are more susceptible to MPTP-induced neuron loss.4648 By contrast, DJ-1 KO rats show significant (~50%) age-dependent nigral dopaminergic neuron loss between 6 and 8 months of age.31 This is accompanied by locomotor impairments and altered radioligand binding to dopamine receptors in the striatum.31, 49 Mitochondria isolated from the striatum of DJ-1 KO rats showed significantly altered respiration compared to mitochondria isolated from wild-type control rats.50 These PD-related abnormalities in DJ-1 KO rats may be useful to study pathogenic mechanisms of familial and possibly idiopathic PD. Although DJ-1 KO rats are relatively new, they have already been used successfully to test neuroprotective compounds; an HDAC inhibitor was reported to alleviate locomotor behavior impairments and apoptosis in DJ-1 KO rats.51

Parkin KO rats

Deletion, truncation and missense mutations in the Parkin gene account for about half of all cases of early onset PD (before age 30) but are rarely found in PD cases with onset of symptoms after age 30.12, 52 Over 150 pathogenic mutations have been found in all exons and domains, with a clear recessive mode of inheritance and apparent 100% penetrance in homozygous or compound heterozygous individuals.34, 53 About one third of reported Parkin mutations are point mutations, while deletions or duplications of one or more exons are the most common.53 Parkin is an E3 ubiquitin ligase that is activated by PINK1 and targets dysfunctional mitochondria for degradation by autophagy, which further implicates mitochondrial dysfunction as a cause of PD.54, 55 Augmenting Parkin activity or enhancing mitochondrial quality control are potential therapeutic strategies.56 Overexpressing Parkin can prevent neurodegeneration in rat substantia nigra induced by 6-OHDA or α-synuclein overexpression.5760 Parkin KO mice have nigrostriatal mitochondrial respiration defects and increased brain markers of oxidative damage, but no loss of dopaminergic neurons, even when crossed with mice deficient for antioxidant enzymes.6164 Unlike PINK1 KO and DJ-1 KO rats, initial characterization of Parkin KO rats shows no significant behavioral deficits or age-dependent nigral dopaminergic neuron loss.31 Parkin KO rats also lack Lewy bodies or any other pathology immunoreactive to α-synuclein, consistent with the absence of Lewy bodies in most published neuropathology reports of Parkin-linked PD.65 Nevertheless, Parkin KO rats have proven useful for proteomic studies and for identifying substrates of Parkin, thereby furthering research on cellular and molecular mechanisms of PD.50, 66

α-synuclein Transgenic and KO Rats

SNCA, the gene that codes for α-synuclein, was the first gene in which point mutations were causally linked to PD.14 α-synuclein is also the major protein component of Lewy bodies and Lewy neurites, which are pathological hallmarks of PD.67 α-Synuclein is abundantly expressed in brain and localizes predominately to presynaptic terminals of neurons, where it facilitates the exocytosis of synaptic vesicles.68 Because α-synuclein mutations are dominantly inherited, and because duplication and triplication mutations of the wild-type human SNCA gene are also causally linked to PD,18, 19 many laboratories have generated mice and rats overexpressing wild-type and mutant α-synuclein to generate PD animal models.10, 26 Surprisingly, most lines of transgenic mice overexpressing wild-type or mutant α-synuclein fail to develop significant loss of dopaminergic neurons in the substantia nigra or significant synuclein-immunoreactive pathology similar to Lewy bodies, which are the hallmarks of PD. α-Synuclein knockout mice are neuropathologically normal and α-synuclein knockout rats recently developed using CRISPR/Cas9 genome targeting have yet to be characterized.69, 70

In contrast to α-synuclein transgenic mice, several lines of transgenic rats overexpressing human α-synuclein have phenotypes useful for modeling aspects of PD. Bacterial artificial chromosome (BAC) transgenic rats expressing the PD-linked E46K mutant human α-synuclein at a level 2–3 fold greater than endogenous α-synuclein develop α-synuclein protein aggregates in nigral dopamine neurons with no loss of dopamine neurons and no overt motor deficits at age 12 months, but increased sensitivity to rotenone-induced neurodegeneration even at an earlier age.71 BAC transgenic rats overexpressing wild-type human α-synuclein show significant loss of nigral TH neurons at age 18 months, reduced striatal dopamine at age 12 months, and locomotor deficits beginning at age 12 months, including decreased rearing behavior and increased time to traverse wide or narrow beams or grids.72 These rats also develop aggregates of insoluble proteinase K-resistant α-synuclein and significant non-motor behavioral abnormalities including impaired odor discrimination, and changes in novelty-seeking behavior before onset of the progressive motor deficits.72 Subsequent studies of the same line of BAC transgenic rats overexpressing wild-type human α-synuclein revealed significantly reduced serotonin levels in the hippocampus, with decreased hippocampal serotonergic fibers and unchanged raphe serotonergic neurons.73 These abnormalities occurred together with α-synuclein aggregation and anxiety-like behavior, as measured by reduced exploration and feeding. This BAC transgenic rat model could be useful for research on PD serotonergic abnormalities and anxiety symptoms, which are frequently observed but understudied aspects of PD.

Although α-synuclein transgenic rats generally do not mimic the severe loss of nigral dopaminergic neurons compared to rats with viral overexpression of α-synuclein (discussed below), advantages of transgenic rat models of PD include less phenotypic variability compared to viral overexpression and avoidance of intracranial surgeries and associated inflammatory responses. Recently, a strain of Berlin-Druckrey rats bearing a synuclein gene promoter mutation has been identified that results in accumulation of synuclein in the mesencephalic area, striatum and frontal cortex, with altered dopaminergic neuron electrophysiology.74 Together with transgenic rats, these provide valuable research tools that can be combined through breeding with other animal models relevant to PD, including transgenic rats that express green fluorescent protein (GFP) exclusively in dopaminergic neurons.75

LRRK2 Transgenic and KO Rats

Dominantly inherited point mutations in Leucine Rich Repeat Kinase 2 (LRRK2) are the most common cause of familial PD.16, 17 LRRK2 is a large ~286 kDa protein, with multiple domains including enzymatically functional kinase and GTPase domains. It is highly unusual for a single polypeptide to contain both kinase and GTPase activities, which suggests these activities are crucially linked in the regulation or function of LRRK2.7678 In vitro studies show that PD-linked LRRK2 mutations increase LRRK2 kinase activity, which is required for LRRK2-mediated cell toxicity.7981 LRRK2 KO rats have no obvious phenotype but are surprisingly resistant to nigral dopaminergic neuron loss induced by adeno-associated viral (AAV) expression of wild-type human α-synuclein.82 LRRK2 KO rats are also resistant to recruitment and activation of myeloid cells and nigral cell loss in response to intracranial injection of lipopolysaccharide (LPS) to elicit neuroinflammation.82 These studies highlight the utility of LRRK2 KO rats for PD research, even though these rats do not have PD-related phenotypes.83, 84 Several lines of transgenic rats expressing human wild-type LRRK2 or LRRK2 with the PD-linked point mutant G2019S, R1441C or R1441G have been characterized.8589 Similar to mice, no LRRK2 transgenic rats show significant loss of nigral dopaminergic neurons. Nevertheless, Walker et al. reported that 6-month old rats overexpressing human G2019S LRRK2 had decreased ability to remain on top of a rotating rod (rotarod test of motor coordination) compared to non-transgenic rats.88 No abnormalities were found in any of the other motor behavior tests including beam walking, rearing and forelimb use in a glass cylinder, and forepaw use while being dragged backwards at a constant speed. Sloan et al. observed age-dependent motor deficits on the rotarod test in BAC transgenic rats expressing human G2019S or R1441C mutant LRRK2, but not in rats expressing wild-type human LRRK2.86 Intraperitoneal injection of L-DOPA/benserazide significantly increased the time before falling from the rotarod for the transgenic rats but had no effect on the non-transgenic controls. This suggests that these LRRK2 BAC transgenic rats have L-DOPA responsive nigrostriatal dysfunction without detectable cell death or protein aggregation. Consistent with this, fast-scan cyclic voltammetry showed impaired dopamine release evoked by a single pulse in the dorsal striatum in acute brain slices of BAC transgenic rats compared to non-transgenic controls. Furthermore, mutant but not wild-type LRRK2 BAC transgenic rats showed impaired cognitive function/spatial short-term memory, as measured by reduced performance on the alternating T-maze test. Gastrointestinal function was normal according to stool frequency and composition.86 Inducible transgenic rats with expression of human G2019S LRRK2 starting at age 5 months (early adulthood) did not result in dopaminergic neuron loss but caused impaired dopamine reuptake, measured by microdialysis.89 This model could potentially be useful for investigating the effects of elevated extracellular dopamine.

Viral Vector Animal Models of PD

More robust PD animal models of nigral dopaminergic neuron loss have been generated by injecting rats intracranially with viral vectors to overexpress α-synuclein in the substantia nigra. Recombinant adeno-associated viral (AAV) vectors and HIV-1-derived lentiviral vectors have been engineered to express transgenes, such as α-synuclein, driven by strong transcriptional promoters like the phosphoglycerate kinase (PGK) promoter or the chicken β-actin (CBA) promoter with enhancer elements from the cytomegalovirus (CMV) promoter.9092. Unlike most other viral vector systems, these can efficiently deliver genes to mature neurons and different viral coat protein serotypes can be used to selectively target different cell types, such as glia versus neurons.9395 Unlike transgenic mice, rats with viral overexpression of α-synuclein have significant nigral dopaminergic neuron loss, denervation of the striatum and abundant inclusions immunoreactive for α-synuclein.9092

Numerous recent studies have used recombinant AAV with the genomic elements of AAV serotype 2 packaged with capsid proteins from AAV serotypes 1, 2, 5, 6, 7, 8 or 9 to enhance expression of α-synuclein in the substantia nigra of wild-type rats and many other rat models of PD.58, 91, 96119 AAV vectors are typically injected unilaterally into the substantia nigra, which allows the uninjected hemisphere to be used as an internal control for histological analyses and allows locomotor asymmetry behavior to be studied as a measure of nigrostriatal function or degeneration. High purity and high titer (1 × 1010 – 1 × 1014 viral genome copies per ml) viral preparations are generally required to cause greater than 50% loss of nigral dopaminergic neurons, which is typically measured ~8 weeks after injection. In some studies ~50% nigral cell loss occurs within 1 week, however, greater nigral cell loss is generally observed after 4 weeks post-injection consistent with progressive neurodegeneration.105

Methods for viral expression of LRRK2 are limited due to the large size of full-length LRRK2 cDNA (~7.6 kilobases) that is difficult to package within viral capsids, along with the other required sequences. Second-generation recombinant human serotype 5 adenoviruses have been successfully used to express in rats full-length human wild-type and G2019S mutant LRRK2 driven by the neuron-specific synapsin-1 promoter.120 Recombinant adenoviral vectors were injected unilaterally at six locations in the striatum of adult female Wistar rats and brains were harvested and analyzed by immunohistochemistry 10, 21, and 42 days after injection. Viral titers were matched to 2.3 × 109 viral particles/microliter and 2 microliters were injected at each site. Compared to the uninjected side, injection of G2019S LRRK2 but not wild-type LRRK2 or GFP control viral vectors caused a nearly 10% reduction in nigral dopaminergic neurons by 10 days and a nearly 20% reduction at 21 and 42 days post-injection.120 This suggests that the PD-linked G2019S mutation is significantly more toxic and causes progressive loss of nigral neurons, presumably due to increased kinase activity. To further test the dependence on kinase activity, the same adenoviral system was used to inject at the same stereotaxic coordinates at similar titers (2.1 × 109 viral particles/microliter) human wild-type, G2019S and G2019S/D1994N kinase inactive control LRRK2. Surprisingly, no significant loss of nigral dopaminergic neurons was observed at 42 days post-injection, even for rats injected with the G2019S LRRK2 expressing adenovirus.121 Consistent with the absence of nigral cell loss, there was no change in motor behavior. The difference in outcome compared to the previous study was attributed to inefficient or inconsistent retrograde transport of adenoviral vectors from axon terminals in the striatum to neuronal cell bodies in the substantia nigra.121 Nevertheless, rats injected with G2019S but not wild-type or kinase inactive LRRK2 developed ubiquitin-immunoreactive inclusions in the striatum consistent with kinase-dependent pathology induced by LRRK2, even when LRRK2 fails to be transported from the striatum to the substantia nigra.121

One of the advantages of viral vector rat models of PD is the relative ease of overexpressing multiple genes or different variants of the same gene, compared to generating different lines of transgenic rats. Unlike genetic rat models of PD, viral vectors can be injected unilaterally so that the contralateral hemisphere can be used as an uninjected or vehicle injected control within the same animal for neuropathological or biochemical analyses. Moreover, different species and different brain regions can be targeted with the same recombinant viral vector, making comparisons facile relative to transgenic animal models. Another major advantage of this model is that robust behavioral, biochemical, neurochemical and neuropathological outcomes can be observed in as little as little as 3–4 weeks after viral injection. This is advantageous to minimize the time and expense of screening and testing new PD therapeutic strategies. However, genetic PD animal models generally have more consistent spatial and temporal transgene expression. The purity, titer and delivery of viral vectors can drastically affect the percentage of cells that are transduced.97 The level of viral transgene expression can diminish over time, depending on the promoter or other factors, which could confound longitudinal study designs or studies comparing interventions at different time points. Generating viral vector animal models is usually more labor intensive compared to breeding genetic PD animal models because each animal must undergo stereotaxic surgery to inject the virus into one or more brain regions.

α-Synuclein Pre-formed Fibril Rat PD Models

Direct injection of pre-formed fibrils (not monomer) of purified recombinant α-synuclein into the striatum or the substantia nigra of rats and mice has recently been shown to generate excellent PD animal models of synuclein pathology and, in some cases, loss of nigral dopaminergic neurons, depending on the length of time between injection and analysis.122125 Paumier et al. found that unilateral injection of α-synuclein pre-formed fibrils into rat striatum causes bilateral α-synuclein-immunoreactive pathology in many brain regions that innervate the striatum (including cortex, substantia nigra and amygdala) 30 and 60 days post-injection and bilateral loss of ~20–30% of nigral dopamine neurons 180 days post-injection.124 The bilateral nigral cell loss observed in unilaterally injected rats differs significantly from mice injected the same way, which only showed ipsilateral nigral degeneration.122 Although nigral cell loss was bilateral, the fibril-injected rats had reduced striatal dopamine only in the ipsilateral hemisphere. This was insufficient to cause apparent motor deficits but caused significant impairment of ultrasonic vocalizations.124 Injection of α-synuclein pre-formed fibrils into rat substantia nigra causes remarkably similar α-synuclein-immunoreactive pathology and similar ~50% nigral cell loss compared to injection of fibrils into rat striatum, as measured 6 months post-injection.125

Unique aspects of this model include the conversion of endogenous α-synuclein into visible brain inclusions or aggregates and the time-dependent spreading of the α-synuclein pathology throughout the brain. Importantly, the brain inclusions are immunoreactive for α-synuclein phosphorylated at serine 129, which is a relatively selective marker of synuclein pathology in human postmortem brains and animal models.126 The pre-formed fibrils themselves do not appear to form visible inclusions but they presumably catalyze the conversion of endogenous monomeric α-synuclein into visible inclusions as previously shown in vitro.127 This animal model may be particularly useful for determining the relationship between α-synuclein fibrilization and PD-related neurodegeneration, which has long been the subject of intense research.128 Both this model, and the model generated by intracranial injection of AAV-α-synuclein could be used to test therapies targeting α-synuclein, such as testing anti-α-synuclein antibodies to deplete normal or pathogenic forms of α-synuclein,129 testing compounds that can mitigate the effects of neuroinflammation caused by α-synuclein aggregates,130 or testing compounds that can prevent α-synuclein aggregation.131

Conclusions

A major impediment to discovering and developing more effective treatments for PD has been the lack of preclinical animal models that reproduce the age-dependent onset and progression of PD symptoms and neuropathology. Recent advances in genetics, transgenic technology and basic neuroscience research have yielded substantial improvements in PD animal models. New rat models of PD, such as those described here, provide unprecedented opportunities for advancement of preclinical research on specific aspects of PD including α-synuclein neuropathology, progressive nigral cell loss, motor deficits, and non-motor symptoms. Many of the recently developed rat PD models appear to have greater face validity by reproducing more robust and progressive nigral cell loss and α-synuclein pathology compared to mice with the same PD-linked mutations or injections.105 There is hope that this signifies greater predictive validity for determining the extent to which treatments that prevent or ameliorate neurodegeneration in these animal models will be clinically effective.

Each PD animal model has advantages and disadvantages. Table 1 lists the major neuropathological and behavioral features of published rat PD models and their potential pre-clinical utility. Table 2 lists the specific mutations in genetic rat models of PD related to human PD-linked mutations. The primary advantage of genetic animal models of PD is the knowledge that PD-linked mutations are the root cause of all the neuropathological or behavioral phenotypes that emerge, even if they do not perfectly match humans in every way. This is a significant advantage because the pathogenic mechanisms of the more prevalent idiopathic PD remain uncertain but could overlap with familial PD. Mechanistic insight gained from genetic PD animal models could expedite the development of more effective treatments for idiopathic PD. The limited predictive validity of current PD animal models for screening and development of therapeutically effective drugs can be ascribed to insufficient understanding of the cellular and molecular abnormalities that must be targeted and specifically manipulated in order to prevent or slow PD progression. A shift in reliance away from models that phenocopy PD, even if they may not be etiologically related, toward models that share common pathogenic (e.g. genetic) mechanisms, even if they may not be perfect phenocopies, could have a profound impact on the progress of both basic and translational PD research.

Table 1.

Table of Rat Models of PD.

Rat model Nigral neuron loss Alpha synuclein pathology* Motor features Non-motor features Pre- clinical relevance References
Neurotoxin models
6-OHDA Yes (Acute) None reported Yes Cognitive impairments; DI disorder Neurodegeneration Reviewed in 24
Rotenone Yes Yes Yes (age and dose dependent) GI disorder Neurodegeneration; alpha synuclein clearance Reviewed in 24
Genetic Models
PINK1 KO rats Yes (progressive ~50% neuron loss) Yes (endogenous) Present- 4 mo. Impairments in vocalization and approach behavior Targeting mitochondrial dysfunction; Therapies that clear or reduce alpha synuclein inclusions 31, 39, 40, 49, 50
DJ-1 KO rats Yes (progressive ~50% neuron loss) None reported Present- 4 mo. None reported Targeting mitochondrial dysfunction 31, 50, 51
Parkin KO rats None observed None reported None observed None reported Targeting mitochondrial dysfunction 31, 50
LRKK2-G2019S None observed None reported Rotarod defect- 3 mo. Impaired spatial learning and memory LRRK2 Biomarkers; LRRK2 kinase inhibitors 86
LRKK2-R1441C None observed None reported Rotarod defect- 18–21 mo. Impaired spatial learning and memory LRRK2 Biomarkers; LRRK2 kinase inhibitors 86
LRKK2 KO rat None observed None reported None observed None reported LRRK2 and alpha synuclein pathological relationship 83, 84
Synuclein E46K None observed Yes (12 months) None observed None reported Therapies that clear or reduce alpha synuclein inclusions 71
Other models
AAV-α-synuclein Yes Yes (All AAV-overexpressing models) cylinder test deficits and forelimb use deficits None reported Therapies that clear or reduce alpha synuclein inclusions Reviewed in 22
Lentiviral-α– synuclein Yes Yes None reported None reported Therapies that clear or reduce alpha synuclein inclusions 90
α–Synuclein pre-formed fibrils Yes (progressive 2–6 months post injection) Yes (4 weeks post injection) None observed Impaired ultrasonic vocalizations Therapies that clear or reduce alpha synuclein inclusions 22, 124, 125
*

Synuclein-immunoreactive aggregates but no Lewy bodies have been reported in rat models of PD.

Table 2.

Human gene mutations linked to PD and modeled in rats

Gene Human Mutations Mode of Inheritance Rat Model Mutations References
Alpha synuclein (PRK1) A53T, A53E, A30P, H50Q, E46K, G51D Dominant Human E46K expressed via BAC transgenic 14, 15, 71
LRRK2 (PRK8) N1437H, R1441C/G/H, Y1699C, G2019S, I2020T Dominant Human G2019S expressed via BAC transgenic; Human R1441C expressed via BAC transgenic 16, 17
Parkin (PRK2) Deletions, Insertions, Frameshifts, Missense, and Nonsense mutations in every exon; Deletions and duplications of one or more exons are most common mutations. Recessive 5 bp deletion in exon 4 12, 31
PINK1 (PRK6) Missense and Nonsense mutations Q21X, G440E, Q456X, Q129fsX157, W437X, A271D, G309D, Exon deletions or duplications Recessive 26 bp deletion in exon 4 11, 31
DJ-1 (PRK7) L166P, D149A, R98Q, Exon deletions or splice-site alterations. Recessive 9 bp deletion and 1 bp insertion in exon 5 13, 31

Currently, α-synuclein has potentially the greatest mechanistic overlap between familial and idiopathic PD. There have been substantial recent advances in PD animal models generated by intracranial injection of pre-formed fibrils of α-synuclein.22 Because these models have exceptional spread of neuropathology immunoreactive to the pathological marker serine-129-phosphorylated α-synuclein, they could be particularly useful for studying the pathogenic role of abnormal forms of α-synuclein, for studying disease spread or progression, and for studying non-motor symptoms of disease that may be related to peripheral α-synuclein pathology. Both the model generated by injection of AAV α-synuclein and the model generated by the injection of α-synuclein pre-formed fibrils could be used to test anti-α-synuclein therapies, such as clearing, depleting, or preventing α-synuclein aggregation. The pre-formed fibril model may also be useful for developing PD biomarkers and imaging methods capable of specifically detecting abnormal α-synuclein. Disadvantages of this model include the requirement for surgically injecting the material into the brain, which is labor intensive, adds technical variability, and perturbs the system by eliciting some neuroinflammation and repair, all of which can increase variability of outcomes.

Future steps to optimize genetic rat models of PD include reducing variability caused by different “strains” of α-synuclein having different propensities to induce the aggregation of α-synuclein and other proteins, such as Tau.123, 132134 There is also a need to reduce variability caused by the effect of endogenous rodent α-synuclein on the aggregation of human wild-type or mutant α-synuclein.135 It may be necessary to “humanize” genetic animal models of PD by replacing the endogenous α-synuclein with wild-type human α-synuclein in order to obtain consistent, reproducible and robust PD models. It will also be important to determine the extent to which genetic animal models depend on background strain, because this is known to be a major source of variability in neurotoxin and other PD animal models, including intracranial injection of α-synuclein pre-formed fibrils and viral expression vectors.22 Viral gene expression can be targeted to specific subsets of cells using cell-type specific promoters and viral capsid proteins with different tropism and spreading properties.94, 136 The increasing number cell-type specific Cre transgenic rat lines, including lines for expression in selective subsets of dopaminergic neurons, will enable increased spatiotemporal gene manipulations in genetic rat models of PD.137 Much as optogenetics has revolutionized the ability to study the selective effect of specific circuits, pathways, and nuclei on neurological functions, more spatially and temporally focused manipulations in genetic rat models of PD will enhance the ability to study the role of specific proteins, pathways, and cell types in PD initiation, progression and manifestation in motor as well as non-motor symptoms.

Acknowledgments

We gratefully acknowledge funding support by grants from the Michael J. Fox Foundation for Parkinson’s Research (MJFF) and the National Institute of Neurological Disorders and Stroke under NIH award numbers R01NS082565 and R21NS082888. The content of this publication is solely the responsibility of the authors and does not necessarily represent the official views of the NIH or MJFF. These funding agencies had no role in the writing of the manuscript.

Footnotes

Authors’ Roles

1. Research project: A. Conception, B. Organization, C. Execution; 2. Statistical Analysis: A. Design, B. Execution, C. Review and Critique; 3. Manuscript: A. Writing of the first draft, B. Review and Critique.

R.B.C: 3A, 3B

M.S.G: 3A, 3B

Financial Disclosures

Relevant conflicts of interest/financial disclosures:

R.B.C: Nothing to report.

M.S.G: Grants from the Michael J. Fox Foundation for Parkinson’s Research and the National Institute of Neurological Disorders and Stroke under NIH award numbers R01NS082565 and R21NS082888.

References

  • 1.Tansey MG, Goldberg MS. Neuroinflammation in Parkinson’s disease: its role in neuronal death and implications for therapeutic intervention. Neurobiol Dis. 2010;37(3):510–518. doi: 10.1016/j.nbd.2009.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Frank-Cannon TC, Tran T, Ruhn KA, et al. Parkin deficiency increases vulnerability to inflammation-related nigral degeneration. J Neurosci. 2008;28(43):10825–10834. doi: 10.1523/JNEUROSCI.3001-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ungerstedt U. 6-Hydroxy-dopamine induced degeneration of central monoamine neurons. Eur J Pharmacol. 1968;5(1):107–110. doi: 10.1016/0014-2999(68)90164-7. [DOI] [PubMed] [Google Scholar]
  • 4.Heikkila RE, Hess A, Duvoisin RC. Dopaminergic neurotoxicity of 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine in mice. Science. 1984;224(4656):1451–1453. doi: 10.1126/science.6610213. [DOI] [PubMed] [Google Scholar]
  • 5.Morin N, Jourdain VA, Di Paolo T. Modeling dyskinesia in animal models of Parkinson disease. Exp Neurol. 2014;256:105–116. doi: 10.1016/j.expneurol.2013.01.024. [DOI] [PubMed] [Google Scholar]
  • 6.Iderberg H, Francardo V, Pioli EY. Animal models of L-DOPA-induced dyskinesia: an update on the current options. Neuroscience. 2012;211:13–27. doi: 10.1016/j.neuroscience.2012.03.023. [DOI] [PubMed] [Google Scholar]
  • 7.Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci. 2000;3(12):1301–1306. doi: 10.1038/81834. [DOI] [PubMed] [Google Scholar]
  • 8.Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science. 1983;219(4587):979–980. doi: 10.1126/science.6823561. [DOI] [PubMed] [Google Scholar]
  • 9.Moore DJ, Dawson TM. Value of genetic models in understanding the cause and mechanisms of Parkinson’s disease. Curr Neurol Neurosci Rep. 2008;8(4):288–296. doi: 10.1007/s11910-008-0045-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lee Y, Dawson VL, Dawson TM. Animal models of Parkinson’s disease: vertebrate genetics. Cold Spring Harb Perspect Med. 2012;2(10) doi: 10.1101/cshperspect.a009324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Valente EM, Abou-Sleiman PM, Caputo V, et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science. 2004;304(5674):1158–1160. doi: 10.1126/science.1096284. [DOI] [PubMed] [Google Scholar]
  • 12.Kitada T, Asakawa S, Hattori N, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998;392(6676):605–608. doi: 10.1038/33416. [DOI] [PubMed] [Google Scholar]
  • 13.Bonifati V, Rizzu P, van Baren MJ, et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science. 2003;299(5604):256–259. doi: 10.1126/science.1077209. [DOI] [PubMed] [Google Scholar]
  • 14.Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276(5321):2045–2047. doi: 10.1126/science.276.5321.2045. [DOI] [PubMed] [Google Scholar]
  • 15.Kruger R, Kuhn W, Muller T, et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet. 1998;18(2):106–108. doi: 10.1038/ng0298-106. [DOI] [PubMed] [Google Scholar]
  • 16.Zimprich A, Biskup S, Leitner P, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44(4):601–607. doi: 10.1016/j.neuron.2004.11.005. [DOI] [PubMed] [Google Scholar]
  • 17.Paisan-Ruiz C, Jain S, Evans EW, et al. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron. 2004;44(4):595–600. doi: 10.1016/j.neuron.2004.10.023. [DOI] [PubMed] [Google Scholar]
  • 18.Singleton AB, Farrer M, Johnson J, et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science. 2003;302(5646):841. doi: 10.1126/science.1090278. [DOI] [PubMed] [Google Scholar]
  • 19.Chartier-Harlin MC, Kachergus J, Roumier C, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet. 2004;364(9440):1167–1169. doi: 10.1016/S0140-6736(04)17103-1. [DOI] [PubMed] [Google Scholar]
  • 20.Ross OA, Braithwaite AT, Skipper LM, et al. Genomic investigation of alpha-synuclein multiplication and parkinsonism. Ann Neurol. 2008;63(6):743–750. doi: 10.1002/ana.21380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Farrer M, Kachergus J, Forno L, et al. Comparison of kindreds with parkinsonism and alpha-synuclein genomic multiplications. Ann Neurol. 2004;55(2):174–179. doi: 10.1002/ana.10846. [DOI] [PubMed] [Google Scholar]
  • 22.Volpicelli-Daley LA, Kirik D, Stoyka LE, Standaert DG, Harms AS. How can rAAV-alpha-synuclein and the fibril alpha-synuclein models advance our understanding of Parkinson’s disease? J Neurochem. 2016;139(Suppl 1):131–155. doi: 10.1111/jnc.13627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Volta M, Melrose H. LRRK2 mouse models: dissecting the behavior, striatal neurochemistry and neurophysiology of PD pathogenesis. Biochem Soc Trans. 2017;45(1):113–122. doi: 10.1042/BST20160238. [DOI] [PubMed] [Google Scholar]
  • 24.Tieu K. A guide to neurotoxic animal models of Parkinson’s disease. Cold Spring Harb Perspect Med. 2011;1(1):a009316. doi: 10.1101/cshperspect.a009316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Taylor TN, Greene JG, Miller GW. Behavioral phenotyping of mouse models of Parkinson’s disease. Behav Brain Res. 2010;211(1):1–10. doi: 10.1016/j.bbr.2010.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Koprich JB, Kalia LV, Brotchie JM. Animal models of alpha-synucleinopathy for Parkinson disease drug development. Nat Rev Neurosci. 2017;18(9):515–529. doi: 10.1038/nrn.2017.75. [DOI] [PubMed] [Google Scholar]
  • 27.Blandini F, Armentero MT. Animal models of Parkinson’s disease. Febs j. 2012;279(7):1156–1166. doi: 10.1111/j.1742-4658.2012.08491.x. [DOI] [PubMed] [Google Scholar]
  • 28.Antony PM, Diederich NJ, Balling R. Parkinson’s disease mouse models in translational research. Mamm Genome. 2011;22(7–8):401–419. doi: 10.1007/s00335-011-9330-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lu XH, Fleming SM, Meurers B, et al. Bacterial artificial chromosome transgenic mice expressing a truncated mutant parkin exhibit age-dependent hypokinetic motor deficits, dopaminergic neuron degeneration, and accumulation of proteinase K-resistant alpha-synuclein. J Neurosci. 2009;29(7):1962–1976. doi: 10.1523/JNEUROSCI.5351-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shin JH, Ko HS, Kang H, et al. PARIS (ZNF746) repression of PGC-1alpha contributes to neurodegeneration in Parkinson’s disease. Cell. 2011;144(5):689–702. doi: 10.1016/j.cell.2011.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dave KD, De Silva S, Sheth NP, et al. Phenotypic characterization of recessive gene knockout rat models of Parkinson’s disease. Neurobiol Dis. 2014;70:190–203. doi: 10.1016/j.nbd.2014.06.009. [DOI] [PubMed] [Google Scholar]
  • 32.West AB, Cowell RM, Daher JP, et al. Differential LRRK2 expression in the cortex, striatum, and substantia nigra in transgenic and nontransgenic rodents. J Comp Neurol. 2014;522(11):2465–2480. doi: 10.1002/cne.23583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Larsen K, Hedegaard C, Bertelsen MF, Bendixen C. Threonine 53 in alpha-synuclein is conserved in long-living non-primate animals. Biochem Biophys Res Commun. 2009;387(3):602–605. doi: 10.1016/j.bbrc.2009.07.070. [DOI] [PubMed] [Google Scholar]
  • 34.Schulte C, Gasser T. Genetic basis of Parkinson’s disease: inheritance, penetrance, and expression. Appl Clin Genet. 2011;4:67–80. doi: 10.2147/TACG.S11639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Narendra DP, Jin SM, Tanaka A, et al. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 2010;8(1):e1000298. doi: 10.1371/journal.pbio.1000298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Matsuda N, Sato S, Shiba K, et al. PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol. 2010;189(2):211–221. doi: 10.1083/jcb.200910140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gispert S, Ricciardi F, Kurz A, et al. Parkinson phenotype in aged PINK1-deficient mice is accompanied by progressive mitochondrial dysfunction in absence of neurodegeneration. PLoS One. 2009;4(6):e5777. doi: 10.1371/journal.pone.0005777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kitada T, Tong Y, Gautier CA, Shen J. Absence of nigral degeneration in aged parkin/DJ-1/PINK1 triple knockout mice. J Neurochem. 2009;111(3):696–702. doi: 10.1111/j.1471-4159.2009.06350.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Villeneuve LM, Purnell PR, Boska MD, Fox HS. Early Expression of Parkinson’s Disease-Related Mitochondrial Abnormalities in PINK1 Knockout Rats. Mol Neurobiol. 2014 doi: 10.1007/s12035-014-8927-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Grant LM, Kelm-Nelson CA, Hilby BL, et al. Evidence for early and progressive ultrasonic vocalization and oromotor deficits in a PINK1 gene knockout rat model of Parkinson’s disease. J Neurosci Res. 2015;93(11):1713–1727. doi: 10.1002/jnr.23625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Samaranch L, Lorenzo-Betancor O, Arbelo JM, et al. PINK1-linked parkinsonism is associated with Lewy body pathology. Brain. 2010;133(Pt 4):1128–1142. doi: 10.1093/brain/awq051. [DOI] [PubMed] [Google Scholar]
  • 42.Steele JC, Guella I, Szu-Tu C, et al. Defining neurodegeneration on Guam by targeted genomic sequencing. Ann Neurol. 2015;77(3):458–468. doi: 10.1002/ana.24346. [DOI] [PubMed] [Google Scholar]
  • 43.Farrer M, Chan P, Chen R, et al. Lewy bodies and parkinsonism in families with parkin mutations. Ann Neurol. 2001;50(3):293–300. doi: 10.1002/ana.1132. [DOI] [PubMed] [Google Scholar]
  • 44.Baulac S, Lu H, Strahle J, et al. Increased DJ-1 expression under oxidative stress and in Alzheimer’s disease brains. Mol Neurodegener. 2009;4:12. doi: 10.1186/1750-1326-4-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lev N, Ickowicz D, Melamed E, Offen D. Oxidative insults induce DJ-1 upregulation and redistribution: Implications for neuroprotection. Neurotoxicology. 2008;29(3):397–405. doi: 10.1016/j.neuro.2008.01.007. [DOI] [PubMed] [Google Scholar]
  • 46.Kim RH, Smith PD, Aleyasin H, et al. Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci U S A. 2005;102(14):5215–5220. doi: 10.1073/pnas.0501282102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chen L, Cagniard B, Mathews T, et al. Age-dependent motor deficits and dopaminergic dysfunction in DJ-1 null mice. J Biol Chem. 2005;280(22):21418–21426. doi: 10.1074/jbc.M413955200. [DOI] [PubMed] [Google Scholar]
  • 48.Goldberg MS, Pisani A, Haburcak M, et al. Nigrostriatal dopaminergic deficits and hypokinesia caused by inactivation of the familial Parkinsonism-linked gene DJ-1. Neuron. 2005;45(4):489–496. doi: 10.1016/j.neuron.2005.01.041. [DOI] [PubMed] [Google Scholar]
  • 49.Sun J, Kouranova E, Cui X, Mach RH, Xu J. Regulation of dopamine presynaptic markers and receptors in the striatum of DJ-1 and Pink1 knockout rats. Neurosci Lett. 2013;557(Pt B):123–128. doi: 10.1016/j.neulet.2013.10.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Stauch KL, Villeneuve LM, Purnell PR, Pandey S, Guda C, Fox HS. SWATH-MS proteome profiling data comparison of DJ-1, Parkin, and PINK1 knockout rat striatal mitochondria. Data Brief. 2016;9:589–593. doi: 10.1016/j.dib.2016.09.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chiu S, Terpstra KJ, Bureau Y, et al. Liposomal-formulated curcumin [Lipocurc] targeting HDAC (histone deacetylase) prevents apoptosis and improves motor deficits in Park 7 (DJ-1)-knockout rat model of Parkinson’s disease: implications for epigenetics-based nanotechnology-driven drug platform. Journal of complementary & integrative medicine. 2013:10. doi: 10.1515/jcim-2013-0020. [DOI] [PubMed] [Google Scholar]
  • 52.Lucking CB, Durr A, Bonifati V, et al. Association between early-onset Parkinson’s disease and mutations in the parkin gene. N Engl J Med. 2000;342(21):1560–1567. doi: 10.1056/NEJM200005253422103. [DOI] [PubMed] [Google Scholar]
  • 53.Klein C, Westenberger A. Genetics of Parkinson’s disease. Cold Spring Harb Perspect Med. 2012;2(1):a008888. doi: 10.1101/cshperspect.a008888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nguyen TN, Padman BS, Lazarou M. Deciphering the Molecular Signals of PINK1/Parkin Mitophagy. Trends Cell Biol. 2016;26(10):733–744. doi: 10.1016/j.tcb.2016.05.008. [DOI] [PubMed] [Google Scholar]
  • 55.Pickrell AM, Youle RJ. The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron. 2015;85(2):257–273. doi: 10.1016/j.neuron.2014.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kazlauskaite A, Muqit MM. PINK1 and Parkin - mitochondrial interplay between phosphorylation and ubiquitylation in Parkinson’s disease. FEBS J. 2015;282(2):215–223. doi: 10.1111/febs.13127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Vercammen L, Van der Perren A, Vaudano E, et al. Parkin protects against neurotoxicity in the 6-hydroxydopamine rat model for Parkinson’s disease. Mol Ther. 2006;14(5):716–723. doi: 10.1016/j.ymthe.2006.06.009. [DOI] [PubMed] [Google Scholar]
  • 58.Yamada M, Mizuno Y, Mochizuki H. Parkin gene therapy for alpha-synucleinopathy: a rat model of Parkinson’s disease. Hum Gene Ther. 2005;16(2):262–270. doi: 10.1089/hum.2005.16.262. [DOI] [PubMed] [Google Scholar]
  • 59.Lo Bianco C, Schneider BL, Bauer M, et al. Lentiviral vector delivery of parkin prevents dopaminergic degeneration in an alpha-synuclein rat model of Parkinson’s disease. Proc Natl Acad Sci U S A. 2004;101(50):17510–17515. doi: 10.1073/pnas.0405313101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Manfredsson FP, Burger C, Sullivan LF, Muzyczka N, Lewin AS, Mandel RJ. rAAV-mediated nigral human parkin over-expression partially ameliorates motor deficits via enhanced dopamine neurotransmission in a rat model of Parkinson’s disease. Exp Neurol. 2007;207(2):289–301. doi: 10.1016/j.expneurol.2007.06.019. [DOI] [PubMed] [Google Scholar]
  • 61.Goldberg MS, Fleming SM, Palacino JJ, et al. Parkin-deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons. J Biol Chem. 2003;278(44):43628–43635. doi: 10.1074/jbc.M308947200. [DOI] [PubMed] [Google Scholar]
  • 62.Hennis MR, Marvin MA, Taylor CM, 2nd, Goldberg MS. Surprising behavioral and neurochemical enhancements in mice with combined mutations linked to Parkinson’s disease. Neurobiol Dis. 2014;62:113–123. doi: 10.1016/j.nbd.2013.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hennis MR, Seamans KW, Marvin MA, Casey BH, Goldberg MS. Behavioral and neurotransmitter abnormalities in mice deficient for Parkin, DJ-1 and superoxide dismutase. PLoS One. 2013;8(12):e84894. doi: 10.1371/journal.pone.0084894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Palacino JJ, Sagi D, Goldberg MS, et al. Mitochondrial dysfunction and oxidative damage in parkin-deficient mice. J Biol Chem. 2004;279(18):18614–18622. doi: 10.1074/jbc.M401135200. [DOI] [PubMed] [Google Scholar]
  • 65.Doherty KM, Hardy J. Parkin disease and the Lewy body conundrum. Mov Disord. 2013;28(6):702–704. doi: 10.1002/mds.25486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kurup PK, Xu J, Videira RA, et al. STEP61 is a substrate of the E3 ligase parkin and is upregulated in Parkinson’s disease. Proc Natl Acad Sci U S A. 2015;112(4):1202–1207. doi: 10.1073/pnas.1417423112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839–840. doi: 10.1038/42166. [DOI] [PubMed] [Google Scholar]
  • 68.Xu J, Wu XS, Sheng J, et al. alpha-Synuclein Mutation Inhibits Endocytosis at Mammalian Central Nerve Terminals. J Neurosci. 2016;36(16):4408–4414. doi: 10.1523/JNEUROSCI.3627-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Abeliovich A, Schmitz Y, Farinas I, et al. Mice lacking alpha-synuclein display functional deficits in the nigrostriatal dopamine system. Neuron. 2000;25(1):239–252. doi: 10.1016/s0896-6273(00)80886-7. [DOI] [PubMed] [Google Scholar]
  • 70.Cabin DE, Shimazu K, Murphy D, et al. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking alpha-synuclein. J Neurosci. 2002;22(20):8797–8807. doi: 10.1523/JNEUROSCI.22-20-08797.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Cannon JR, Geghman KD, Tapias V, et al. Expression of human E46K-mutated alpha-synuclein in BAC-transgenic rats replicates early-stage Parkinson’s disease features and enhances vulnerability to mitochondrial impairment. Exp Neurol. 2013;240:44–56. doi: 10.1016/j.expneurol.2012.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nuber S, Harmuth F, Kohl Z, et al. A progressive dopaminergic phenotype associated with neurotoxic conversion of alpha-synuclein in BAC-transgenic rats. Brain. 2013;136(Pt 2):412–432. doi: 10.1093/brain/aws358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kohl Z, Ben Abdallah N, Vogelgsang J, et al. Severely impaired hippocampal neurogenesis associates with an early serotonergic deficit in a BAC alpha-synuclein transgenic rat model of Parkinson’s disease. Neurobiol Dis. 2016;85:206–217. doi: 10.1016/j.nbd.2015.10.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Guatteo E, Rizzo FR, Federici M, et al. Functional alterations of the dopaminergic and glutamatergic systems in spontaneous alpha-synuclein overexpressing rats. Exp Neurol. 2017;287(Pt 1):21–33. doi: 10.1016/j.expneurol.2016.10.009. [DOI] [PubMed] [Google Scholar]
  • 75.Iacovitti L, Wei X, Cai J, et al. The hTH-GFP reporter rat model for the study of Parkinson’s disease. PLoS One. 2014;9(12):e113151. doi: 10.1371/journal.pone.0113151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Webber PJ, Smith AD, Sen S, Renfrow MB, Mobley JA, West AB. Autophosphorylation in the leucine-rich repeat kinase 2 (LRRK2) GTPase domain modifies kinase and GTP-binding activities. J Mol Biol. 2011;412(1):94–110. doi: 10.1016/j.jmb.2011.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Biosa A, Trancikova A, Civiero L, et al. GTPase activity regulates kinase activity and cellular phenotypes of Parkinson’s disease-associated LRRK2. Hum Mol Genet. 2013;22(6):1140–1156. doi: 10.1093/hmg/dds522. [DOI] [PubMed] [Google Scholar]
  • 78.Islam MS, Moore DJ. Mechanisms of LRRK2-dependent neurodegeneration: role of enzymatic activity and protein aggregation. Biochem Soc Trans. 2017;45(1):163–172. doi: 10.1042/BST20160264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Smith WW, Pei Z, Jiang H, Dawson VL, Dawson TM, Ross CA. Kinase activity of mutant LRRK2 mediates neuronal toxicity. Nat Neurosci. 2006;9(10):1231–1233. doi: 10.1038/nn1776. [DOI] [PubMed] [Google Scholar]
  • 80.West AB, Moore DJ, Choi C, et al. Parkinson’s disease-associated mutations in LRRK2 link enhanced GTP-binding and kinase activities to neuronal toxicity. Hum Mol Genet. 2007;16(2):223–232. doi: 10.1093/hmg/ddl471. [DOI] [PubMed] [Google Scholar]
  • 81.Greggio E, Jain S, Kingsbury A, et al. Kinase activity is required for the toxic effects of mutant LRRK2/dardarin. Neurobiol Dis. 2006;23(2):329–341. doi: 10.1016/j.nbd.2006.04.001. [DOI] [PubMed] [Google Scholar]
  • 82.Daher JP, Volpicelli-Daley LA, Blackburn JP, Moehle MS, West AB. Abrogation of alpha-synuclein-mediated dopaminergic neurodegeneration in LRRK2-deficient rats. Proc Natl Acad Sci U S A. 2014;111(25):9289–9294. doi: 10.1073/pnas.1403215111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Baptista MA, Dave KD, Frasier MA, et al. Loss of leucine-rich repeat kinase 2 (LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs. PLoS One. 2013;8(11):e80705. doi: 10.1371/journal.pone.0080705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ness D, Ren Z, Gardai S, et al. Leucine-rich repeat kinase 2 (LRRK2)-deficient rats exhibit renal tubule injury and perturbations in metabolic and immunological homeostasis. PLoS One. 2013;8(6):e66164. doi: 10.1371/journal.pone.0066164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lee JW, Tapias V, Di Maio R, Greenamyre JT, Cannon JR. Behavioral, neurochemical, and pathologic alterations in bacterial artificial chromosome transgenic G2019S leucine-rich repeated kinase 2 rats. Neurobiol Aging. 2015;36(1):505–518. doi: 10.1016/j.neurobiolaging.2014.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sloan M, Alegre-Abarrategui J, Potgieter D, et al. LRRK2 BAC transgenic rats develop progressive, L-DOPA-responsive motor impairment, and deficits in dopamine circuit function. Hum Mol Genet. 2016;25(5):951–963. doi: 10.1093/hmg/ddv628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Shaikh KT, Yang A, Youshin E, Schmid S. Transgenic LRRK2 (R1441G) rats-a model for Parkinson disease? PeerJ. 2015;3:e945. doi: 10.7717/peerj.945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Walker MD, Volta M, Cataldi S, et al. Behavioral deficits and striatal DA signaling in LRRK2 p. G2019S transgenic rats: a multimodal investigation including PET neuroimaging. J Parkinsons Dis. 2014;4(3):483–498. doi: 10.3233/JPD-140344. [DOI] [PubMed] [Google Scholar]
  • 89.Zhou H, Huang C, Tong J, Hong WC, Liu YJ, Xia XG. Temporal expression of mutant LRRK2 in adult rats impairs dopamine reuptake. Int J Biol Sci. 2011;7(6):753–761. doi: 10.7150/ijbs.7.753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lo Bianco C, Ridet JL, Schneider BL, Deglon N, Aebischer P. alpha -Synucleinopathy and selective dopaminergic neuron loss in a rat lentiviral-based model of Parkinson’s disease. Proc Natl Acad Sci U S A. 2002;99(16):10813–10818. doi: 10.1073/pnas.152339799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kirik D, Rosenblad C, Burger C, et al. Parkinson-like neurodegeneration induced by targeted overexpression of alpha-synuclein in the nigrostriatal system. J Neurosci. 2002;22(7):2780–2791. doi: 10.1523/JNEUROSCI.22-07-02780.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Klein RL, King MA, Hamby ME, Meyer EM. Dopaminergic cell loss induced by human A30P alpha-synuclein gene transfer to the rat substantia nigra. Hum Gene Ther. 2002;13(5):605–612. doi: 10.1089/10430340252837206. [DOI] [PubMed] [Google Scholar]
  • 93.Naldini L, Blomer U, Gallay P, et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science. 1996;272(5259):263–267. doi: 10.1126/science.272.5259.263. [DOI] [PubMed] [Google Scholar]
  • 94.Cannon JR, Sew T, Montero L, Burton EA, Greenamyre JT. Pseudotype-dependent lentiviral transduction of astrocytes or neurons in the rat substantia nigra. Exp Neurol. 2011;228(1):41–52. doi: 10.1016/j.expneurol.2010.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Howard DB, Powers K, Wang Y, Harvey BK. Tropism and toxicity of adeno-associated viral vector serotypes 1, 2, 5, 6, 7, 8, and 9 in rat neurons and glia in vitro. Virology. 2008;372(1):24–34. doi: 10.1016/j.virol.2007.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Landeck N, Buck K, Kirik D. Toxic effects of human and rodent variants of alpha-synuclein in vivo. Eur J Neurosci. 2016 doi: 10.1111/ejn.13493. [DOI] [PubMed] [Google Scholar]
  • 97.Van der Perren A, Casteels C, Van Laere K, Gijsbers R, Van den Haute C, Baekelandt V. Development of an Alpha-synuclein Based Rat Model for Parkinson’s Disease via Stereotactic Injection of a Recombinant Adeno-associated Viral Vector. J Vis Exp. 2016;(108):53670. doi: 10.3791/53670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.He Q, Koprich JB, Wang Y, et al. Treatment with Trehalose Prevents Behavioral and Neurochemical Deficits Produced in an AAV alpha-Synuclein Rat Model of Parkinson’s Disease. Mol Neurobiol. 2016;53(4):2258–2268. doi: 10.1007/s12035-015-9173-7. [DOI] [PubMed] [Google Scholar]
  • 99.Alvarsson A, Caudal D, Bjorklund A, Svenningsson P. Emotional memory impairments induced by AAV-mediated overexpression of human alpha-synuclein in dopaminergic neurons of the ventral tegmental area. Behav Brain Res. 2016;296:129–133. doi: 10.1016/j.bbr.2015.08.034. [DOI] [PubMed] [Google Scholar]
  • 100.Van der Perren A, Toelen J, Casteels C, et al. Longitudinal follow-up and characterization of a robust rat model for Parkinson’s disease based on overexpression of alpha-synuclein with adeno-associated viral vectors. Neurobiol Aging. 2015;36(3):1543–1558. doi: 10.1016/j.neurobiolaging.2014.11.015. [DOI] [PubMed] [Google Scholar]
  • 101.Van der Perren A, Macchi F, Toelen J, et al. FK506 reduces neuroinflammation and dopaminergic neurodegeneration in an alpha-synuclein-based rat model for Parkinson’s disease. Neurobiol Aging. 2015;36(3):1559–1568. doi: 10.1016/j.neurobiolaging.2015.01.014. [DOI] [PubMed] [Google Scholar]
  • 102.Daniel G, Musso A, Tsika E, et al. alpha-Synuclein-induced dopaminergic neurodegeneration in a rat model of Parkinson’s disease occurs independent of ATP13A2 (PARK9) Neurobiol Dis. 2015;73:229–243. doi: 10.1016/j.nbd.2014.10.007. [DOI] [PubMed] [Google Scholar]
  • 103.Daher JP, Abdelmotilib HA, Hu X, et al. Leucine-rich Repeat Kinase 2 (LRRK2) Pharmacological Inhibition Abates alpha-Synuclein Gene-induced Neurodegeneration. J Biol Chem. 2015;290(32):19433–19444. doi: 10.1074/jbc.M115.660001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Caudal D, Alvarsson A, Bjorklund A, Svenningsson P. Depressive-like phenotype induced by AAV-mediated overexpression of human alpha-synuclein in midbrain dopaminergic neurons. Exp Neurol. 2015;273:243–252. doi: 10.1016/j.expneurol.2015.09.002. [DOI] [PubMed] [Google Scholar]
  • 105.Bourdenx M, Dovero S, Engeln M, et al. Lack of additive role of ageing in nigrostriatal neurodegeneration triggered by alpha-synuclein overexpression. Acta neuropathologica communications. 2015;3:46. doi: 10.1186/s40478-015-0222-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Moloney TC, Hyland R, O’Toole D, et al. Heat shock protein 70 reduces alpha-synuclein-induced predegenerative neuronal dystrophy in the alpha-synuclein viral gene transfer rat model of Parkinson’s disease. CNS neuroscience & therapeutics. 2014;20(1):50–58. doi: 10.1111/cns.12200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Febbraro F, Sahin G, Farran A, et al. Ser129D mutant alpha-synuclein induces earlier motor dysfunction while S129A results in distinctive pathology in a rat model of Parkinson’s disease. Neurobiol Dis. 2013;56:47–58. doi: 10.1016/j.nbd.2013.03.014. [DOI] [PubMed] [Google Scholar]
  • 108.Febbraro F, Andersen KJ, Sanchez-Guajardo V, Tentillier N, Romero-Ramos M. Chronic intranasal deferoxamine ameliorates motor defects and pathology in the alpha-synuclein rAAV Parkinson’s model. Exp Neurol. 2013;247:45–58. doi: 10.1016/j.expneurol.2013.03.017. [DOI] [PubMed] [Google Scholar]
  • 109.Lindgren HS, Lelos MJ, Dunnett SB. Do alpha-synuclein vector injections provide a better model of Parkinson’s disease than the classic 6-hydroxydopamine model? Exp Neurol. 2012;237(1):36–42. doi: 10.1016/j.expneurol.2012.05.022. [DOI] [PubMed] [Google Scholar]
  • 110.Decressac M, Mattsson B, Lundblad M, Weikop P, Bjorklund A. Progressive neurodegenerative and behavioural changes induced by AAV-mediated overexpression of alpha-synuclein in midbrain dopamine neurons. Neurobiol Dis. 2012;45(3):939–953. doi: 10.1016/j.nbd.2011.12.013. [DOI] [PubMed] [Google Scholar]
  • 111.Decressac M, Mattsson B, Bjorklund A. Comparison of the behavioural and histological characteristics of the 6-OHDA and alpha-synuclein rat models of Parkinson’s disease. Exp Neurol. 2012;235(1):306–315. doi: 10.1016/j.expneurol.2012.02.012. [DOI] [PubMed] [Google Scholar]
  • 112.Ulusoy A, Decressac M, Kirik D, Bjorklund A. Viral vector-mediated overexpression of alpha-synuclein as a progressive model of Parkinson’s disease. Prog Brain Res. 2010;184:89–111. doi: 10.1016/S0079-6123(10)84005-1. [DOI] [PubMed] [Google Scholar]
  • 113.Yamada M, Iwatsubo T, Mizuno Y, Mochizuki H. Overexpression of alpha-synuclein in rat substantia nigra results in loss of dopaminergic neurons, phosphorylation of alpha-synuclein and activation of caspase-9: resemblance to pathogenetic changes in Parkinson’s disease. J Neurochem. 2004;91(2):451–461. doi: 10.1111/j.1471-4159.2004.02728.x. [DOI] [PubMed] [Google Scholar]
  • 114.Gorbatyuk OS, Li S, Nguyen FN, et al. alpha-Synuclein expression in rat substantia nigra suppresses phospholipase D2 toxicity and nigral neurodegeneration. Mol Ther. 2010;18(10):1758–1768. doi: 10.1038/mt.2010.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gorbatyuk OS, Li S, Sullivan LF, et al. The phosphorylation state of Ser-129 in human alpha-synuclein determines neurodegeneration in a rat model of Parkinson disease. Proc Natl Acad Sci U S A. 2008;105(2):763–768. doi: 10.1073/pnas.0711053105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Taschenberger G, Garrido M, Tereshchenko Y, Bahr M, Zweckstetter M, Kugler S. Aggregation of alphaSynuclein promotes progressive in vivo neurotoxicity in adult rat dopaminergic neurons. Acta Neuropathol. 2012;123(5):671–683. doi: 10.1007/s00401-011-0926-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Gaugler MN, Genc O, Bobela W, et al. Nigrostriatal overabundance of alpha-synuclein leads to decreased vesicle density and deficits in dopamine release that correlate with reduced motor activity. Acta Neuropathol. 2012;123(5):653–669. doi: 10.1007/s00401-012-0963-y. [DOI] [PubMed] [Google Scholar]
  • 118.Gombash SE, Manfredsson FP, Kemp CJ, et al. Morphological and behavioral impact of AAV2/5-mediated overexpression of human wildtype alpha-synuclein in the rat nigrostriatal system. PLoS One. 2013;8(11):e81426. doi: 10.1371/journal.pone.0081426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Rocha EM, Smith GA, Park E, et al. Glucocerebrosidase gene therapy prevents alpha-synucleinopathy of midbrain dopamine neurons. Neurobiol Dis. 2015;82:495–503. doi: 10.1016/j.nbd.2015.09.009. [DOI] [PubMed] [Google Scholar]
  • 120.Dusonchet J, Kochubey O, Stafa K, et al. A rat model of progressive nigral neurodegeneration induced by the Parkinson’s disease-associated G2019S mutation in LRRK2. J Neurosci. 2011;31(3):907–912. doi: 10.1523/JNEUROSCI.5092-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Tsika E, Nguyen AP, Dusonchet J, Colin P, Schneider BL, Moore DJ. Adenoviral-mediated expression of G2019S LRRK2 induces striatal pathology in a kinase-dependent manner in a rat model of Parkinson’s disease. Neurobiol Dis. 2015;77:49–61. doi: 10.1016/j.nbd.2015.02.019. [DOI] [PubMed] [Google Scholar]
  • 122.Luk KC, Kehm V, Carroll J, et al. Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science. 2012;338(6109):949–953. doi: 10.1126/science.1227157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Peelaerts W, Bousset L, Van der Perren A, et al. alpha-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015;522(7556):340–344. doi: 10.1038/nature14547. [DOI] [PubMed] [Google Scholar]
  • 124.Paumier KL, Luk KC, Manfredsson FP, et al. Intrastriatal injection of pre-formed mouse alpha-synuclein fibrils into rats triggers alpha-synuclein pathology and bilateral nigrostriatal degeneration. Neurobiol Dis. 2015;82:185–199. doi: 10.1016/j.nbd.2015.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Abdelmotilib H, Maltbie T, Delic V, et al. alpha-Synuclein fibril-induced inclusion spread in rats and mice correlates with dopaminergic Neurodegeneration. Neurobiol Dis. 2017;105:84–98. doi: 10.1016/j.nbd.2017.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Fujiwara H, Hasegawa M, Dohmae N, et al. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat Cell Biol. 2002;4(2):160–164. doi: 10.1038/ncb748. [DOI] [PubMed] [Google Scholar]
  • 127.Volpicelli-Daley LA, Luk KC, Patel TP, et al. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. 2011;72(1):57–71. doi: 10.1016/j.neuron.2011.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Goldberg MS, Lansbury PT., Jr Is there a cause-and-effect relationship between alpha-synuclein fibrillization and Parkinson’s disease? Nat Cell Biol. 2000;2(7):E115–119. doi: 10.1038/35017124. [DOI] [PubMed] [Google Scholar]
  • 129.Schenk DB, Koller M, Ness DK, et al. First-in-human assessment of PRX002, an anti-alpha-synuclein monoclonal antibody, in healthy volunteers. Mov Disord. 2017;32(2):211–218. doi: 10.1002/mds.26878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Jucaite A, Svenningsson P, Rinne JO, et al. Effect of the myeloperoxidase inhibitor AZD3241 on microglia: a PET study in Parkinson’s disease. Brain. 2015;138(Pt 9):2687–2700. doi: 10.1093/brain/awv184. [DOI] [PubMed] [Google Scholar]
  • 131.Wrasidlo W, Tsigelny IF, Price DL, et al. A de novo compound targeting alpha-synuclein improves deficits in models of Parkinson’s disease. Brain. 2016 doi: 10.1093/brain/aww238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Peng C, Gathagan RJ, Lee VM. Distinct alpha-Synuclein strains and implications for heterogeneity among alpha-Synucleinopathies. Neurobiol Dis. 2017 doi: 10.1016/j.nbd.2017.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Melki R. Role of Different Alpha-Synuclein Strains in Synucleinopathies, Similarities with other Neurodegenerative Diseases. J Parkinsons Dis. 2015;5(2):217–227. doi: 10.3233/JPD-150543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Bousset L, Pieri L, Ruiz-Arlandis G, et al. Structural and functional characterization of two alpha-synuclein strains. Nat Commun. 2013;4:2575. doi: 10.1038/ncomms3575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Fares MB, Maco B, Oueslati A, et al. Induction of de novo alpha-synuclein fibrillization in a neuronal model for Parkinson’s disease. Proc Natl Acad Sci U S A. 2016;113(7):E912–921. doi: 10.1073/pnas.1512876113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Aschauer DF, Kreuz S, Rumpel S. Analysis of transduction efficiency, tropism and axonal transport of AAV serotypes 1, 2, 5, 6, 8 and 9 in the mouse brain. PLoS One. 2013;8(9):e76310. doi: 10.1371/journal.pone.0076310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Liu Z, Brown A, Fisher D, Wu Y, Warren J, Cui X. Tissue Specific Expression of Cre in Rat Tyrosine Hydroxylase and Dopamine Active Transporter-Positive Neurons. PLoS One. 2016;11(2):e0149379. doi: 10.1371/journal.pone.0149379. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES