Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jun 9.
Published in final edited form as: Handb Clin Neurol. 2023;193:3–16. doi: 10.1016/B978-0-323-85555-6.00002-3

Role of rodent models in advancing precision medicine for Parkinson’s disease

EMILY SIMONS 1, SHEILA M FLEMING 1,*
PMCID: PMC10251252  NIHMSID: NIHMS1900574  PMID: 36803818

Abstract

With a current lack of disease-modifying treatments, an initiative toward implementing a precision medicine approach for treating Parkinson’s disease (PD) has emerged. However, challenges remain in how to define and apply precision medicine in PD. To accomplish the goal of optimally targeted and timed treatment for each patient, preclinical research in a diverse population of rodent models will continue to be an essential part of the translational path to identify novel biomarkers for patient diagnosis and subgrouping, understand PD disease mechanisms, identify new therapeutic targets, and screen therapeutics prior to clinical testing. This review highlights the most common rodent models of PD and discusses how these models can contribute to defining and implementing precision medicine for the treatment of PD.

INTRODUCTION

As the second most common neurodegenerative disease, Parkinson’s disease (PD) is estimated to impact more than 12 million people worldwide by 2040 (GBD 2016 Parkinson’s Disease Collaborators, 2018; Dorsey et al., 2018). PD is clinically characterized by motor symptoms including resting tremor, bradykinesia, rigidity, and postural instability. The hallmark pathology associated with symptoms includes dysfunction and degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the development of Lewy bodies and Lewy neurites, cytoplasmic inclusions containing aggregated forms of the presynaptic protein α-synuclein (aSyn; Spillantini et al., 1997; Cheng et al., 2010). Currently, available therapeutics aim to relieve motor symptoms through dopamine replacement or deep brain stimulation; however, no disease-modifying treatments are available to prevent, slow, or stop the progression of degeneration.

Following continued failures in clinical trials and a growing appreciation for the heterogeneity in PD pathology, the notion of precision medicine has emerged as an innovative new approach for treating PD. Precision medicine is a term that has evolved over time and refers to choosing treatments that are optimally timed and targeted to the specific disease characteristics of the individual patient (Espay et al., 2017). While the concept of precision medicine is relatively agreed upon, the challenge remains in how this broad definition can be applied in the clinic. Due to the heterogeneous nature of PD, clinicians and researchers recommend stratifying PD patients into subgroups based on shared disease characteristics. However, there is a clear lack of consensus regarding which factors should be used to define these subgroups and how those factors will be measured in patients. In this review, a role for rodent models in the pursuit for a precision medicine approach in PD is discussed.

CHALLENGES TO SUBGROUPING IN PD

Several studies have attempted to subgroup PD patients based on clinical representation of motor and nonmotor symptoms (Lewis et al., 2005; Selikhova et al., 2009; Brennan et al., 2017; Fereshtehnejad et al., 2017; De Pablo-Fernández et al., 2019). Indeed, these classifications have merit in their ability to distinguish patients with motor only symptoms versus those with motor and cognitive symptoms, and to predict the rate of progression in some subgroups of patients (Lewis et al., 2005; Selikhova et al., 2009; Brennan et al., 2017; Fereshtehnejad et al., 2017; De Pablo-Fernández et al., 2019). These classifications have been useful for choosing the optimal symptom-targeted treatment regimen and beneficial in understanding diverse clinical phenotypes; however, symptomatic parameters do not necessarily reflect the underlying mechanisms of the disease and are not always represented by pathological biomarkers (Selikhova et al., 2009; De Pablo-Fernández et al., 2019). Additionally, symptom presentation typically occurs several years after the initiation of pathology and substantial irreversible damage has already occurred (Ma et al., 1997; Kilzheimer et al., 2019). Thus, biomarkers for earlier detection and patient subgrouping are crucial as disease-modifying therapies are developed and tested in clinical trials.

More recently, researchers and clinicians have been subgrouping PD patients based on genetic variations which are linked to an increased risk of PD. These studies have provided useful insight into the clinical phenotypes and rate of disease progression associated with specific genetic mutations. Furthermore, pathological features of genetic forms of PD, such as mitochondrial dysfunction or accumulation of aSyn, have been observed in patients with sporadic PD suggesting that they may provide valuable information regarding mechanistic pathways of degeneration in both familial and idiopathic PD. However, stratification of PD patients based on genetics alone, while a useful starting point, is not directly applicable to the majority of PD cases as only 1% to 2% of PD cases are caused by genetic mutations and 5% to 10% are strongly associated with risk genes (Pankratz & Foroud, 2007; Bandres-Ciga et al., 2020). Therefore, defining subgroups based on a combination of factors including both genetics and mechanistic similarities such as dysfunctional organelles (i.e., mitochondria or lysosomes), accumulation of pathological aSyn, and/or inflammatory markers may be a promising approach for stratification of idiopathic cases of PD. For mechanistic stratification to become possible, it will require a better understanding of the cellular and molecular features underlying PD to define subgroup parameters, determination of appropriate biomarkers to identify patients belonging to each subgroup, and identification of the optimal treatments to target specific mechanisms of pathology. Studies in animal models will be an important component to achieving this goal.

RATIONALE FOR MODELING PD IN RODENTS

Establishing stratification of PD patients in clinical studies is challenging in that the trials can be costly and lengthy in duration; it is difficult to obtain enough volunteers to subdivide patients while maintaining statistical power; environmental factors and lifestyle habits are difficult to control and may be confounding factors. For these reasons, mechanistic studies in animal models of PD will continue to be an essential component to better understand the heterogeneous etiologies of PD, identify subgroup biomarkers, and, ultimately, to advance the use of precision medicine in PD patients. In fine with the heterogeneity of PD in humans, a diverse array of animal models representing genetic mutations, environmental toxins, and common pathological features seen in human PD patients are available to investigate mechanisms and biomarkers of disease and to screen novel therapies prior to clinical testing. Highlighted below are a selection of the most common rodent models of PD and their value in helping to define and advance a precision medicine approach for PD.

Rodents have been used for decades to model disease and test potential therapies. They are part of the translational spectrum that includes different in vitro cell systems and patient-derived induced pluripotent stem cells (IPSCs), three dimensional in vitro systems such as stem cell-derived organoids, and drosophila and zebrafish for gene x environment studies and screening (De Souza, 2018; Valadez-Barba et al., 2020). In PD, rodent models were instrumental in the discovery of dopamine, its association with movement dysfunction in PD, and for L-DOPA administration as symptomatic treatment for PD (Carlsson, 1993; Lees et al., 2015). Rodents are good models for studying PD as they are a mammalian species with strong overlap in anatomy, physiology, and genetics to humans. Rodents also have a basic repertoire of behaviors that are similar to humans (e.g., voluntary movement, fear response, and cognitive functions that include attention, strategy switching, and different forms of memory). Their breeding, short gestation period, litter size, and physical size are amenable to genetic manipulation and studying mechanisms underlying cellular function and dysfunction. However, since there are no reported cases of PD developing spontaneously in animals, only humans are the perfect model of the disease. Some of the major limitations of using rodents to model PD include the relatively short lifespan (~2 years), limited higher order behaviors, and limited fine motor skills. In addition, while there is good anatomical overlap between rodents and humans they are obviously not the same. In the rodent brain, the caudate and putamen are not separated by the internal capsule as they are in nonhuman and human primate brains and dopamine neurons in the substantia nigra pars compacta lack melanin in rodents but not in nonhuman and human primates. Furthermore, rodents are quadrupedal animals, using all four limbs to bear weight during locomotion rather than bipedal as is the case with humans. While rodents provide a strong platform for understanding the basis of dysfunction in PD, the direct translation of results in rodents to humans needs to take into consideration the limits of the animal system.

GENETIC MODELS OF PARKINSON’S DISEASE

Numerous transgenic and knockout models have been developed to reflect genetic variations known to cause or increase the risk of developing PD (Table 1.1.). As many transgenic models of PD are generated using the same human gene mutations seen in PD patients, they can provide valuable information on the sequence of events leading to cellular and behavioral dysfunction. However, many genetic models of PD do not develop extensive and progressive degeneration of nigrostriatal dopaminergic neurons. This is likely due in part to the short lifespan of rodents and the slow progression of pathology seen in PD. While this is certainly a limitation of many genetic models, it may also present an opportunity to investigate dysfunction earlier in the disorder to facilitate identification of biomarkers of disease onset and novel therapeutic targets prior to irreversible neuron loss.

Table 1.1.

Pathological mechanisms in genetic models of Parkinson’s disease

Rodent model Gene Primary PD disease
mechanism(s)
Secondary disease
mechanism(s)
Dopaminergic
neuron loss
Motor
deficits
Non-motor deficits
A53T-aSyn SNCA Mutated aSyn-mediated pathology Mitochondrial dysfunction
Inflammation
No Yes Cognitive
Sensorimotor
Thy1-aSyn “Line 61” SNCA Human WT aSyn-mediated pathology Mitochondrial dysfunction
Inflammation
No Yes Cognitive
Sensorimotor
Olfactory
Disrupted sleep patterns
GBA KO GBA Lysosomal dysfunction None No No None
GBA L444P heterozygous mutation GBA Lysosomal dysfunction Disrupted mitophagy & Autophagy
aSyn pathology
No Yes
LRRK2 KO LRRK2 None observed Abnormal peripheral organ function (kidney, liver, lung, & spleen) No No None observed
LRRK2 G2019S mutation LRRK2 Vesicle trafficking, autophagy Mitochondrial dysfunction
Neuroinflammation
aSyn pathology
Yes- in some mouse models Yes Anxiety & depressive-like behavior
PINK1 KO PINK1 Mitochondrial dysfunction aSyn pathology Yes Yes Swallowing
Vocalizations
Olfactory

Furthermore, disruptions in biochemical pathways triggered by PD-associated genetic mutations are the same pathways implicated in many cases of sporadic PD, allowing for reasonable translation to sporadic forms of PD. As PD is considered to be triggered by a combination of genetic susceptibility and environmental factors, genetic models can also be combined with neurotoxin exposure to assess the vulnerability of genetic mutations to specific environmental factors. Currently, with the advancement of gene editing techniques, genetic models will continue to be invaluable for understanding PD pathogenesis, mechanistic stratification of patients, identifying novel therapeutic targets, and screening new therapies.

A53T aSyn mice

The first familial form of PD was identified as having a missense mutation with an amino acid substitution of alanine-to-threonine at codon 53 in the SNCA gene that encodes for aSyn (Polymeropoulos et al., 1997). Mice overexpressing the human A53T mutation were one of the earliest transgenic models generated and are still commonly used to study aSyn-related pathological mechanisms in PD (Giasson et al., 2002; Lee et al., 2002). The frequently used strains overexpress the human A53T mutation via the prion promoter and results in aSyn pathology in the midbrain, cerebellum, brainstem, and spinal cord in association with astrogliosis, microgliosis, and mitochondrial dysfunction (Giasson et al., 2002; Lee et al., 2002; Ordonez et al., 2018; Hu et al., 2019; Li et al., 2019). Inclusions of aSyn develop in this model and resemble filamentous aSyn inclusions seen in human PD (Giasson et al., 2002; Lee et al., 2002). However, A53T mice develop degeneration within the spinal cord and show spinal cord-related motor decline (Giasson et al., 2002; Lee et al., 2002). Furthermore, they do not develop dopaminergic neuron loss. Most studies using this model focus on aSyn-related dysfunction prior to spinal cord degeneration. Indeed, substantial molecular changes such as alterations in long-term depression and synaptic transmission and behavioral deficits such as impairments in rotarod performance and Y-maze performance develop prior to severe motor decline (Paumier et al., 2013). Although this is a commonly used model for mechanistic and target validation studies, it is important to recognize that A53T mutations in PD are very rare and findings may not be representative of what occurs in sporadic cases of PD.

Thy1-aSyn mice (a.k.a. Line 61)

In addition to missense mutations in aSyn, triplication and duplication of the aSyn locus have also been identified in familial PD (Singleton et al., 2003; Chartier-Harlin et al., 2004). These cases indicate that elevated levels of aSyn can lead to the development of PD. Multiple lines of mice that overexpress human wild-type aSyn have been generated to study the pathological effects of elevated aSyn (Rockenstein et al., 2002). Mice that overexpress wild-type human aSyn under the Thy 1 promoter are one of the most well characterized aSyn transgenic lines that continues to be utilized today (Rockenstein et al., 2002). They have a broad distribution of aSyn accumulation and age-related reduction in striatal dopamine (Rockenstein et al., 2002; Lam et al., 2011). Importantly, phosphorylated (Ser 129) and proteinase K-resistant aSyn is observed in this model (Chesselet et al., 2012; Lam et al., 2011). These aSyn aggregates are present in several brain regions including the striatum, substantia nigra, locus coeruleus, cortex, and olfactory bulb (Chesselet et al., 2012). These mice display PD-like motor symptoms as well as early non-motor symptoms often found in human PD such as cognitive impairments, olfactory deficits, sleep dysregulation, gastrointestinal dysfunction, and autonomic cardiovascular dysfunction (Fleming et al., 2004, 2008, 2013; Wang et al., 2008, 2012; Magen et al., 2012; McDowell et al., 2014). Mechanistically, early and prolonged inflammation including activated microglia, reactive astrocytes, and upregulation of the pro-inflammatory cytokine TNF-α have been reported (Watson et al., 2012). Mitochondrial alterations are also prevalent in this model with mitochondrial respiration defects shown in the ventral midbrain and striatum, and specifically inhibition of complex 1 activity and increased oxidative stress in the ventral midbrain (Subramaniam et al., 2014). While this model displays several PD-associated biochemical and behavioral features, Thy1-aSyn transgenic mice do not develop dopaminergic neuron degeneration. Despite this limitation, the myriad of PD-associated pathological features in this model make it valuable for the investigation of early biomarkers and therapeutic targets prior to the irreversible loss of dopamine neurons. Additionally, this model has been shown to display an increased vulnerability to neurotoxins such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and paraquat, which can exacerbate pathology resulting in mitochondrial dysfunction and neuronal axon degeneration (Song et al., 2004; Fernagut et al., 2007). As PD is often considered to result from a combination of genetic vulnerability and environmental exposures, this model can be combined with neurotoxins to induce specific pathological features seen in individual patients. While this model is still commonly utilized, elevated levels of aSyn are not consistently reported in sporadic PD (Duffy et al., 2018a,b).

GBA1

Gaucher’s disease is the most common lysosomal storage disorder and is associated with mutations in GBA1, the gene encoding the lysosomal enzyme glucocerebrosidase (GCase). Mutations in GBA1 are associated with an increased risk of developing Lewy body disorders including PD (Goker-Alpan et al., 2004; Satake et al., 2009). Furthermore, GBA1 variants are more common in PD patients than in age-matched controls (Sidransky et al., 2009). Additionally, patients with sporadic PD without GBA1 mutations also demonstrate reduced GCase in the brain, supporting the idea of shared mechanisms of pathology between familial and sporadic PD and suggesting that sporadic PD patients may also benefit from therapeutics developed and screened in gba genetic mouse models (Murphy et al., 2014; Chiasserini et al., 2015).

Multiple mutations in gba have been modeled in mice including L444P, D409H, and V394L in addition to knocking out gba (Migdalska-Richards et al., 2017; Yun et al., 2018; Li et al., 2019). Some models can develop aSyn accumulation, for example, at 24 months, heterozygous gba null mice show aSyn aggregation in the hippocampus and striatum (Xu et al., 2011; Migdalska-Richards et al., 2017). However, in several studies the Gaucher models are crossbred with aSyn transgenic mice or aSyn is overexpressed in specific brain regions using adeno-associated viral vectors to determine the effect of impaired GCase function on aSyn toxicity (Fishbein et al., 2014; Migdalska-Richards et al., 2017; Kim et al., 2018; Henderson et al., 2020). Indeed, combining the L444P heterozygous GBA mutation or heterozygous null mutation with A53T-aSyn mice or aSyn overexpressing mice via adeno-associated vims injection exacerbated aSyn pathology and enhanced dopaminergic neuronal degeneration (Fishbein et al., 2014; Migdalska-Richards et al., 2017). Potential therapies targeting GCase function have also been tested in PD models (Richter et al., 2014; Burbulla et al., 2021).

LRRK2

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene represent the most common genetic cause of PD accounting for roughly 4% to 5% of familial cases and 1% to 3% of sporadic cases (Gilks et al., 2005; Khan et al., 2005; Lesage et al., 2007; Paisán-Ruíz et al., 2008). Importantly, LRRK2-associated PD is phenotypically and pathologically indistinguishable from sporadic PD suggesting that information gained from LRRK2 models of PD may have strong translational relevance to several other subgroups of sporadic PD patients (Paisán-Ruíz et al., 2004; Zimprich et al., 2004). Given its prominence in both familial and sporadic PD, mutant LRRK2 animal models have been important in elucidating the mechanisms underlying pathology and identifying novel potential therapeutics.

The LRRK2 gene encodes for an intracellular enzymatic protein involved in a diverse range of cellular processes, including vesicle trafficking, autophagy, and protein translation (Shin et al., 2008; Piccoli et al., 2011; Nikonova et al., 2012; Pellegrini et al., 2018; Sheehan & Yue, 2019). Mutations in LRRK2 can affect kinase and GTPase activity leading to elevated kinase activity (Healy et al., 2008; Islam & Moore, 2017; Steger et al., 2016). Mice and rats share a LRRK2 homolog with 86% to 88% conserved sequence identity to the human LRRK2 gene, including all residues impacted by pathogenic mutations identified in human PD (West et al., 2014). Multiple LRRK2 knockout, knockin, and overexpression rodent models have been developed to study the function of LRRK2 in health and disease. In general, the LRRK2 rodent models on their own show modest to no effects on the nigrostriatal system and aSyn pathology (Lin et al., 2009; Tong et al., 2010, 2012; Ramonet et al., 2011; Zhou et al., 2011; Chen et al., 2012; Hinkle et al., 2012; Baptista et al., 2013; Daher et al., 2014; West et al., 2014; Sloan et al., 2016; Weng et al., 2016; Xiong et al., 2018). However, when combined with aSyn overexpression or intracranial injection of aSyn preformed fibrils, the LRRK2 G2019S mutation can cause an increase in aSyn burden and enhanced cell loss in the substantia nigra (Lin et al., 2009; Volpicelli-Daley et al., 2016; Henderson et al., 2019; Bieri et al., 2019). Inhibition of LRRK2 activity with small-molecule inhibitors or anti-sense oligonucleotides has been shown to be beneficial in animal models (Lee et al., 2010; Daher et al., 2015; Zhao et al., 2017). LRRK2 is being pursued in biomarker development and LRRK2 inhibitors are currently in clinical trials (Kelly and West, 2020).

PINK1

PINK1 mutations are found in 4% to 9% of PD patients in the Asian population and 2% to 4% of Caucasian PD patients, making them the second most common cause of autosomal recessive PD (Schulte & Gasser, 2011). PINK1 is a mitochondrial serine/threonine protein kinase involved in mitochondrial control and dynamics. Studies show PINK1 acts as a sensor which promotes Parkin activation for the degradation of defective mitochondria. Indeed, PINK1 knockout rodents demonstrate mitochondrial dysfunction and oxidative stress similar to that seen in human PD (Gautier et al., 2008; Stauch et al., 2016; Villeneuve et al., 2016). PINK1 knock-out mice and rats are robust models for investigating early biomarkers in PINK1 related human PD cases as they display similar prodromal symptoms to humans such as olfactory, swallowing, vocalization, and gait disturbances (Dave et al., 2014; Grant et al., 2015; Cullen et al., 2018; Kelm-Nelson & Gammie, 2020). Although inconsistent, there are reports of nigrostriatal dopaminergic cell loss in the rat PINK1 knockout model (Dave et al., 2014; De Haas et al., 2019). Hallmark aSyn pathology has also been detected in the substantia nigra, nucleus ambiguous, and locus coeruleus (Grant et al., 2015). Importantly, a recent large-scale gene sequencing study matched the gene databases of PINK1 null rat brainstem tissue with human idiopathic PD and PARK6/2 tissue, demonstrating valid translational relevance of this model with human PD (Kelm-Nelson & Gammie, 2020). This model has been particularly important for studying cranial sensorimotor impairments related to PD such as vocalization and swallowing (Grant et al., 2015; Hoffmeister et al., 2021). These are significant PD symptoms that can contribute to reduced quality of life and mortality and are understudied in the disease. Exciting new studies show a relationship between brainstem norepinephrine and impairments in vocalization and anxiety in this model (Hoffmeister et al., 2021).

INDUCIBLE α-SYNUCLEIN OVEREXPRESSING MODELS

Alpha-synuclein accumulation is a hallmark feature of human pathology in both familial and sporadic cases of PD. It is clear that aSyn accumulation is an important factor in PD and, thus, aSyn models currently dominate the field (Table 1.2). In addition to transgenic models of aSyn, other models of aSyn expression include recombinant adeno-associated viral vector (AAV) mediated aSyn and the aSyn preformed fibril (PFF) models (Kirik et al., 2002; Luk et al., 2012). AAV-aSyn models have provided important information regarding the biochemical properties of aSyn aggregates, and how aSyn overexpression contributes to nigrostriatal degeneration while the aSyn PFF model is a powerful tool to study aSyn seeding.

Table 1.2.

Inducible models of aSyn pathology

α-Syn overexpression model α-Syn pathology Dopaminergic neuron loss
Viral vector-mediated aSyn overexpression Substantia nigra Yes
Preformed Fibril Injection (striatum) Striatum
Frontal cortex
Olfactory bulbs
Amygdala
Yes
Preformed Fibril Injection (gut) Medulla
Locus Coeruleus
Striatum
Prefrontal cortex
Amygdala
Hippocampus
Olfactory bulbs
One study showed DA neuron loss (Kim et al., 2019

Viral Vector-Induced Alpha-Synuclein Overexpression

Viral vector mediated overexpression of wild-type or mutant aSyn is a useful tool for inducing PD-like aSyn pathology. Mutated or wild-type aSyn genes are overexpressed via a recombinant adeno-associated viral (AAV) or lentiviral vector allowing for targeted delivery into neuronal cells of specific tissues such as dopaminergic neurons in the substantia nigra. One of the advantages of targeted viral vector-mediated aSyn overexpression is the reproducibility of dopaminergic cell loss in the substantia nigra which is generally lacking in the transgenic models. In rodents, targeted AAV-aSyn overexpression results in progressive loss of dopaminergic neurons in the substantia nigra and motor deficits (Kirik et al., 2002). Pathology initiates rapidly in the AAV-aSyn model with significantly reduced dopamine reuptake as early as 10 days postinjection, followed by a slow and progressive pattern of degeneration with synaptic and axonal deficits occurring prior to dopaminergic cell death (Butler et al., 2015; Kordower et al., 2013; Lundblad et al., 2012). AAV-aSyn models also develop a robust neuroinflammatory response that includes changes in early microglial activation, upregulation of pro-inflammatory cytokines, and infiltration of lymphocytes prior to dopaminergic neuronal loss (Chung et al., 2009; Cao et al., 2010; Daher et al., 2014; Reish & Standaert, 2015; Harms et al., 2018). Furthermore, coinjections of viral-induced aSyn overexpression and GBA or Parkin genes, or AAV-aSyn overexpression in transgenic models of PD allow for investigation of the interaction between aSyn and PD-associated genes in the progression of pathology (Lo Bianco et al., 2004; Rocha et al., 2015). Indeed, AAV-aSyn overexpression in mice expressing the G2019S-LRRK2 mutation showed enhanced inflammation and dopaminergic neuron cell loss which was ameliorated with administration of a LRRK2 inhibitor (Daher et al., 2014). AAV-aSyn models alone or in combination with other genetic or environmental factors are robust models of aSyn pathology in PD and may help elucidate aSyn mediated degeneration to identify aSyn-related targets for earlier diagnosis and therapeutic intervention. However, like the transgenic aSyn overexpression models, increased levels of aSyn is not consistently reported in sporadic PD (Duffy et al., 2018a,b).

Alpha-Synuclein Preformed Fibril

The development of synthetic aSyn preformed fibrils (PFF) has been a significant advancement in modeling PD. Exogenous aSyn fibrils injected into the striatum act as seeds which trigger phosphorylation and accumulation of endogenous aSyn into neurotoxic oligomers and fibrils (Luk et al., 2012; Paumier et al., 2015; Duffy et al., 2018a,b; Patterson et al., 2019). Injection of PFFs closely mimic several pathological characteristics of human PD including aSyn accumulation, mitochondrial impairment, lysosomal dysfunction, synaptic alterations, progressive degeneration of dopaminergic neurons, and seeding of aSyn pathology through cell-to-cell transmission into regions of the brain directly connected to the site of injection be it striatum or substantia nigra (Volpicelli-Daley et al., 2011; Luk et al., 2012; Luk & Lee, 2014; Paumier et al., 2015; Duffy et al., 2018a,b; Patterson et al., 2019). This model is now widely used for target validation, mechanistic studies, and testing novel therapeutics.

Lewy body pathology is not confined to the central nervous system in PD and has been observed in peripheral tissues including the enteric nervous system (Wakabayashi et al., 1988, 1990; Braak et al., 2006; Beach et al., 2010, 2016; Barrenschee et al., 2017). Gastrointestinal (GI) dysfunction and constipation are established nonmotor symptoms in PD, often occurring prior to the onset of motor symptoms (Chen et al., 2015; Gao et al., 2011; Park et al., 2015; Sung et al., 2014). In addition, GI dysfunction is also reported in animal models overexpressing aSyn (Wang et al., 2008, 2012; Kuo et al., 2010; Hallett et al., 2012; Farrell et al., 2014). These observations combined with pathological staging studies by Braak et al. (2003a,b) led to the hypothesis that the gut may be a site of PD initiation and progress via the vagus to the central nervous system. While this hypothesis has received some pushback from studies demonstrating conflicting patterns of Lewy pathology (Jellinger, 2019), this progression of PD pathology from gut to the central nervous system may help to define subpopulations of PD patients and help to advance biomarker development and precision medicine approaches. To date, several groups have tested this hypothesis in rodents by injecting aSyn PFFs into the gut and tracking aSyn progressive pathology (Manfredsson et al., 2018; Uemura et al., 2018, 2020; Kim et al., 2019; Challis et al., 2020). Injection of aSyn PFFs in the gut have met with mixed results including development of pathological aSyn expression in enteric neurons, GI dysfunction, intestinal inflammation, and in some cases spread of aSyn pathology to CNS structures in hindbrain and one report showing aSyn pathology in the nigrostriatal system (Manfredsson et al., 2018; Uemura et al., 2018; Kim et al., 2019; Challis et al., 2020). Discrepancies in reported outcomes may be due to differences in aSyn PFF concentrations, site of injection, and timing. While it will be important to define the precise parameters for aSyn PFF injection in the gut, this model would be vital for identifying early peripheral biomarkers for a subgroup of PD patients prior to significant damage in the central nervous system and onset of motor symptoms. Furthermore, animal models of gut-brain aSyn propagation will be essential to develop novel therapeutics to prevent the spread of pathology from the enteric nervous system to central nervous system structures.

NEUROTOXIN MODELS OF PARKINSON’S DISEASE

Animal models used to study the pathological mechanisms underlying sensorimotor function in PD often involve the administration of toxins that selectively destroy nigrostriatal dopaminergic neurons (Table 1.3). These models include rodents treated with 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), both have and continue to contribute to our understanding of basal ganglia circuitry and voluntary movement (Ungerstedt, 1968; Burns et al., 1983; Carlsson, 1993). More current toxin models include paraquat and rotenone-treated rodents and these are particularly useful for studying gene–environment interactions in PD (Brooks et al., 1999; Betarbet et al., 2000: Fernagut et al., 2007). Overall, the toxin models tend to have good face validity for sensorimotor signs in that they display impairments in movement initiation, weight shifting, and postural stability that are reversible with levodopa similar to patients with PD (Tillerson et al., 2002; Alam & Schmidt, 2004; Fleming et al., 2005).

Table 1.3.

Neurotoxic models of Parkinson’s disease

Toxin Toxic mechanism(s) Dopaminergic neuron loss aSyn pathology
6-OHDA Production of ROS
Mitochondrial dysfunction
Neuroinflammation
Yes No
MPTP Mitochondrial complex I inhibitor
Oxidative stress
Neuroinflammation
Yes aSyn nitration
Paraquat Oxidative stress Yes aSyn upregulation
Rotenone Mitochondrial complex I inhibitor Yes aSyn aggregates

CONCLUSION

Parkinson’s disease is a multisystem heterogeneous neurodegenerative disorder with a wide range of genetic and environmental risk factors leading to diverse clinical and pathological phenotypes. In order to move toward precision medicine in PD, rodent models will continue to be an important component of the translational pathway (Table 1.4). Choosing the appropriate model or combination of models to represent potential subgroups of PD patients will help in elucidating the underlying mechanism of disease, identifying biomarkers to diagnose and stratify PD patients, and screening new therapeutics prior to clinical trials. Furthermore, as new therapeutics are developed, preclinical trials in animal models will continue to be a critical step in understanding the pharmacokinetics, ability to cross the blood–brain barrier, target specificity, potential toxicity, and drug interactions of each therapeutic prior to testing in humans.

Table 1.4.

The advantages of animal models in defining and implementing precision medicine for Parkinson’s disease

Model Biomarker development Elucidation of disease mechanisms &
identification of therapeutic targets
Preclinical drug screening
Transgenic/Knockout Display PD-associated prodromal symptoms and pathology Directly related to genetic mutations in human PD patients
Disease mechanisms overlap with sporadic PD
Can be combined with aSyn overexpression or neurotoxic models to study gene-aSyn or gene-toxin interactions
Can represent genetic-linked subgroups of PD patients to predict safety and efficacy of therapeutics
aSyn Overexpression Demonstrate early stages of PD pathology
Display PD-associated prodromal symptoms
Representative of a major pathological mechanism in human PD
Display PD-associated disease pathology and symptoms
Can be combined with transgenic or neurotoxic models to study interaction of aSyn with genetic or environmental factors
Can predict impact of therapeutics on preventing spread of aSyn pathology
Can demonstrate drug interaction with aSyn and related pathology
aSyn Preformed Fibril Site of injection can mimic earliest region of aSyn pathology identified in human PD Can study seeding and mechanisms of spreading pathology

Display disease pathology seen in familial and sporadic human PD
Can predict impact of therapeutics on preventing spread of aSyn pathology
Neurotoxin Mimic symptoms of human PD
Can be used to identify early vulnerabilities or triggers for neurotoxin induced PD
Disease mechanisms overlap with human PD
Some are directly related to environmental exposures impacting human PD
Can predict response of neurotoxin induced PD to drugs
Can predict symptomatic response to drugs

References

  1. Alam M, Schmidt WJ (2004). L-DOPA reverses the hypokinetic behaviour and rigidity in rotenone-treated rats. Behav Brain Res 153: 439–446. 10.1016/j.bbr.2003.12.021. [DOI] [PubMed] [Google Scholar]
  2. Bandres-Ciga S, Diez-Fairen M, Kim JJ et al. (2020). Genetics of Parkinson’s disease: An introspection of its journey towards precision medicine. In: Neurobiology of Disease, 137: Academic Press Inc. 10.1016/j.nbd.2020.104782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Baptista MA, Dave KD, Frasier MA et al. (2013). Loss of leucine-rich repeat kinase 2 (LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs. PLoS One 8: e80705. 10.1371/journal.pone.0080705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Barrenschee M, Zorenkov D, Böttner M et al. (2017). Distinct pattern of enteric phospho-alpha-synuclein aggregates and gene expression profiles in patients with Parkinson’s disease. Cerebellum Ataxias 4: 1–14. 10.1186/s40478-016-0408-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Beach TG, Adler CH, Sue LI et al. (2010). Multi-organ distribution of phosphorylated α-synuclein histopathology in subjects with Lewy body disorders. Acta Neuropathol 119: 689–702. 10.1007/s00401-010-0664-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Beach TG, Corbillé AG, Letournel F et al. (2016). Multicenter assessment of immunohistochemical methods for pathological alpha-synuclein in sigmoid colon of autopsied Parkinson’s disease and control subjects. J Parkinsons Dis 6: 761–770. 10.3233/JPD-160888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Betarbet R, Sherer TB, MacKenzie G et al. (2000). Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci 3: 1301–1306. 10.1038/81834. [DOI] [PubMed] [Google Scholar]
  8. Bieri G, Brahic M, Bousset L et al. (2019). LRRK2 modifies α-syn pathology and spread in mouse models and human neurons. Acta Neuropathol 137: 961–980. 10.1007/s00401-019-01995-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Braak H, Rüb U, Gai WP et al. (2003a). Idiopathic Parkinson’s disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J Neural Transm 110: 517–536. 10.1007/s00702-002-0808-2. [DOI] [PubMed] [Google Scholar]
  10. Braak H, Del Tredici K, Rüb U et al. (2003b). Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging 24: 197–211. 10.1016/S0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
  11. Braak H, De Vos RAI, Bohl J et al. (2006). Gastric α-synuclein immunoreactive inclusions in Meissner’s and Auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci Lett 396: 67–72. 10.1016/j.neulet.2005.11.012. [DOI] [PubMed] [Google Scholar]
  12. Brennan L, Devlin KM, Xie SX et al. (2017). Neuropsychological subgroups in non-demented Parkinson’s disease: a latent class analysis. J Parkinsons Dis 7: 385–395. 10.3233/JPD-171081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Brooks AI, Chadwick CA, Gelbard HA et al. (1999). Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Res 823: 1–10. 10.1016/S0006-8993(98)01192-5. [DOI] [PubMed] [Google Scholar]
  14. Burbulla LF, Zheng J, Song P et al. (2021). Direct targeting of wild-type glucocerebrosidase by antipsychotic quetiapine improves pathogenic phenotypes in Parkinson’s disease models. JCI Insight 6: e148649. 10.1172/jci.insight.148649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Burns RS, Chiueh CC, Markey SP et al. (1983). A primate model of parkinsonism: selective destruction of dopaminergic neurons in the pars compacta of the substantia nigra by N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc Natl Acad Sci U S A 80: 4546–4550. 10.1073/pnas.80.14.4546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Butler B, Saha K, Rana T et al. (2015). Dopamine transporter activity is modulated by α-synuclein. J Biol Chem 290: 29542–29554. 10.1074/jbc.M115.691592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Cao S, Theodore S, Standaert DG (2010). Fcy receptors are required for NF-kB signaling, microglial activation and dopaminergic neurodegeneration in an AAV-synuclein mouse model of Parkinson’s disease. Mol Neurodegener 5: 42. 10.1186/1750-1326-5-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Carlsson A (1993). Thirty years of dopamine research. Adv Neurol 60: 1–10. [PubMed] [Google Scholar]
  19. Challis C, Hori A, Sampson TR et al. (2020). Gut-seeded α-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat Neurosci 23: 327–336. 10.1038/s41593-020-0589-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chartier-Harlin MC, Kachergus J, Roumier C et al. (2004). Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet 364: 1167–1169. 10.1016/S0140-6736(04)17103-1. [DOI] [PubMed] [Google Scholar]
  21. Chen CY, Weng YH, Chien KY et al. (2012). (G2019S) LRRK2 activates MKK4-JNK pathway and causes degeneration of SN dopaminergic neurons in a transgenic mouse model of PD. Cell Death Differ 19: 1623–1633. 10.1038/cdd.2012.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chen H, Zhao EJ, Zhang W et al. (2015). Meta-analyses on prevalence of selected Parkinson’s nonmotor symptoms before and after diagnosis. Transl Neurodegener 4: 4–11. 10.1186/2047-9158-4-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Cheng H-C, Ulane CM, Burke RE (2010). Clinical progression in Parkinson’s disease and the neurobiology of axons. Ann Neurol 67: 715–725. 10.1002/ana.21995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chesselet MF, Richter F, Zhu C et al. (2012). A progressive mouse model of Parkinson’s disease: the Thy1-aSyn (“Line 61”) mice. Neurotherapeutics 9: 297–314. 10.1007/s13311-012-0104-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Chiasserini D, Paciotti S, Eusebi P et al. (2015). Selective loss of glucocerebrosidase activity in sporadic Parkinson’s disease and dementia with Lewy bodies. Mol Neurodegener 10: 4–9. 10.1186/s13024-015-0010-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Chung CY, Koprich JB, Siddiqi H et al. (2009). Dynamic changes in presynaptic and axonal transport proteins combined with striatal neuroinflammation precede dopaminergic neuronal loss in a rat model of AAV α-synucleinopathy. J Neurosci 29: 3365–3373. 10.1523/JNEUROSCI.5427-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Cullen KP, Grant LM, Kelm-Nelson CA et al. (2018). Pink1 −/− rats show early-onset swallowing deficits and correlative brainstem pathology. Dysphagia 33: 749–758. 10.1007/s00455-018-9896-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Daher JPL, Volpicelli-Daley LA, Blackburn JP et al. (2014). Abrogation of α-synuclein-mediated dopaminergic neurodegeneration in LRRK2-deficient rats. Proc Natl Acad Sci U S A 111: 9289–9294. 10.1073/pnas.1403215111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Daher JP, Abdelmotilib HA, Hu X et al. (2015). Leucine-rich repeat kinase 2 (LRRK2) pharmacological inhibition abates alpha-synuclein gene-induced neurodegeneration. J Biol Chem 290: 19433–19444. 10.1074/jbc.M115.660001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Dave KD, De Silva S, Sheth NP et al. (2014). Phenotypic characterization of recessive gene knockout rat models of Parkinson’s disease. Neurobiol Dis 70: 190–203. 10.1016/j.nbd.2014.06.009. [DOI] [PubMed] [Google Scholar]
  31. De Haas R, Heltzel LCMW, Tax D et al. (2019). To be or not to be pink(1): contradictory findings in an animal model for Parkinson’s disease. Brain Commun 1: fcz016. 10.1093/braincomms/fcz016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. De Pablo-Fernández E, Lees AJ, Holton JL et al. (2019). Prognosis and neuropathologic correlation of clinical subtypes of Parkinson disease. JAMA Neurol 76: 470–479. 10.1001/jamaneurol.2018.4377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. De Souza N (2018). Organoids. Nat Methods 15: 23. [Google Scholar]
  34. Dorsey ER, Sherer T, Okun MS et al. (2018). The emerging evidence of the Parkinson pandemic. J Parkinsons Dis 8: S3–S8. 10.3233/JPD-181474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Duffy MF, Collier TJ, Patterson JR et al. (2018a). Quality over quantity: advantages of using alpha-synuclein preformed fibril triggered synucleinopathy to model idiopathic Parkinson’s disease. Front Neurosci 4: 621. 10.3389/fnins.2018.00621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Duffy MF, Collier TJ, Patterson JR et al. (2018b). Lewy body-like alpha-synuclein inclusions trigger reactive microgliosis prior to nigral degeneration. J Neuroinflammation 15: 129. 10.1186/s12974-018-1171-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Espay AJ, Brundin P, Lang AE (2017). Precision medicine for disease modification in Parkinson disease. Nat Rev Neurol 13: 119–126. 10.1038/nrneurol.2016.196. [DOI] [PubMed] [Google Scholar]
  38. Farrell KF, Krishnamachari S, Villanueva E et al. (2014). Non-motor parkinsonian pathology in aging A53T α-synuclein mice is associated with progressive synucleinopathy and altered enzymatic function. J Neurochem 128: 536–546. 10.1111/jnc.12481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Fereshtehnejad SM, Zeighami Y, Dagher A et al. (2017). Clinical criteria for subtyping Parkinson’s disease: biomarkers and longitudinal progression. Brain 140: 1959–1976. 10.1093/brain/awx118. [DOI] [PubMed] [Google Scholar]
  40. Fernagut PO, Hutson CB, Fleming SM et al. (2007). Behavioral and histopathological consequences of paraquat intoxication in mice: effects of alpha-synuclein over-expression. Synapse 61: 991–1001. 10.1002/syn.20456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Fishbein I, Kuo YM, Giasson BI et al. (2014). Augmentation of phenotype in a transgenic Parkinson mouse heterozygous for a Gaucher mutation. Brain 137: 3235–3247. 10.1093/brain/awu291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Fleming SM, Salcedo J, Fernagut PO et al. (2004). Early and progressive sensorimotor anomalies in mice overexpressing wild-type human alpha-synuclein. J Neurosci 24: 9434–9440. 10.1523/JNEUROSCI.3080-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Fleming SM, Delville Y, Schallert T (2005). An intermittent, controlled-rate, slow progressive degeneration model of Parkinson’s disease: antiparkinson effects of Sinemet and protective effects of methylphenidate. Behav Brain Res 156: 201–213. 10.1016/j.bbr.2004.05.024. [DOI] [PubMed] [Google Scholar]
  44. Fleming SM, Tetreault N, Mulligan C et al. (2008). Olfactory deficits in mice overexpressing human wildtype α-synuclein. Eur J Neurosci 28: 247–256. 10.1111/j.1460-9568.2008.06346.x.Olfactory. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Fleming SM, Jordan MC, Mulligan CK et al. (2013). Impaired baroreflex function in mice overexpressing alpha-synuclein. Front Neurol 23: 103. 10.3389/fneur.2013.00103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Gao X, Chen H, Schwarzschild MA et al. (2011). A prospective study of bowel movement frequency and risk of Parkinson’s disease. Am J Epidemiol 174: 546–551. 10.1093/aje/kwr119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Gautier CA, Kitada T, Shen J (2008). Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress. Proc Natl Acad Sci U S A 105: 11364–11369. 10.1073/pnas.0802076105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. GBD 2016 Parkinson’s Disease Collaborators (2018). Global, regional, and national burden of Parkinson’s disease, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 17: 939–953. 10.1016/S1474-4422(18)30295-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Giasson BI, Duda JE, Quinn SM et al. (2002). Neuronal alpha-synucleinopathy with severe movement disorder in mice expressing A53T human alpha-synuclein. Neuron 34: 521–533. 10.1016/s0896-6273(02)00682-7. [DOI] [PubMed] [Google Scholar]
  50. Gilks WP, Abou-Sleiman PM, Gandhi S et al. (2005). A common LRRK2 mutation in idiopathic Parkinson’s disease. Lancet 365: 415–416. 10.1016/S0140-6736(05)17830-1. [DOI] [PubMed] [Google Scholar]
  51. Goker-Alpan O, Schiffmann R, LaMarca ME et al. (2004). Parkinsonism among Gaucher disease carriers. J Med Genet 41: 937–940. 10.1136/jmg.2004.024455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Grant LM, Kelm-nelson CA, Hilby BL et al. (2015). Evidence for early and progressive ultrasonic vocalization and oromotor deficits in a PINK1 knockout rat model of Parkinson disease. J Neurosci Res 93: 1713–1727. 10.1002/jnr.23625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Hallett PJ, McLean JR, Kartunen A et al. (2012). Alpha-synuclein overexpressing transgenic mice show internal organ pathology and autonomic deficits. Neurobiol Dis 47: 258–267. 10.1016/j.nbd.2012.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Harms AS, Thome AD, Yan Z et al. (2018). Peripheral monocyte entry is required for alpha-synuclein induced inflammation and neurodegeneration in a model of Parkinson disease. Exp Neurol 300: 179–187. 10.1016/j.expneurol.2017.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Healy DG, Falchi M, O’Sullivan SS et al. (2008). Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson’s disease: a case-control study. Lancet Neurol 7: 583–590. 10.1016/S1474-4422(08)70117-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Henderson MX, Comblath EJ, Darwich A et al. (2019). Spread of α-synuclein pathology through the brain connectome is modulated by selective vulnerability and predicted by network analysis. Nat Neurosci 22: 1248–1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Henderson MX, Sedor S, McGeary I et al. (2020). Glucocerebrosidase activity modulates neuronal susceptibility to pathological alpha-synuclein insult. Neuron 105: 822–836. 10.1016/j.neuron.2019.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Hinkle KM, Yue M, Behrouz B et al. (2012). LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors. Mol Neurodegener 7: 1–18. 10.1186/1750-1326-7-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hoffmeister JD, Kelm-Nelson CA, Ciucci MR (2021). Quantification of brainstem norepinephrine relative to vocal impairment and anxiety in the Pink1−/− rat model of Parkinson disease. Behav Brain Res 24: 113514. 10.1016/j.bbr.2021.113514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Hu D, Sun X, Liao X et al. (2019). Alpha-synuclein suppresses mitochondrial protease ClpP to trigger mitochondrial oxidative damage and neurotoxicity. Acta Neuropathol 137: 939–960. 10.1007/s00401-019-01993-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Islam MS, Moore DJ (2017). Mechanisms of LRRK2-dependent neurodegeneration: role of enzymatic activity and protein aggregation. Biochem Soc Trans 45: 163–172. 10.1042/BST20160264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Jellinger KA (2019). Is Braak staging valid for all types of Parkinson’s disease? J Neural Transm 126: 423–431. 10.1007/s00702-018-1898-9. [DOI] [PubMed] [Google Scholar]
  63. Kelly K, West AB (2020). Pharmacodynamic biomarkers for emerging LRRK2 therapeutics. Front Neurosci 6: 807. 10.3389/fnins.2020.00807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kelm-Nelson CA, Gammie S (2020). Gene expression within the periaqueductal gray is linked to vocal behavior and early-onset parkinsonism in Pink1 knockout rats. BMC Genomics 21: 1–13. 10.1186/s12864-020-07037-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Khan NL, Jain S, Lynch JM et al. (2005). Mutations in the gene LRRK2 encoding dardarin (PARK8) cause familial Parkinson’s disease: Clinical, pathological, olfactory and functional imaging and genetic data. Brain 128: 2786–2796. 10.1093/brain/awh667. [DOI] [PubMed] [Google Scholar]
  66. Kilzheimer A, Hentrich T, Burkhardt S et al. (2019). The challenge and opportunity to diagnose Parkinson’s disease in midlife. Front Neurol 10. 10.3389/fneur.2019.01328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kim D, Hwang H, Choi S et al. (2018). D409H GBA1 mutation accelerates the progression of pathology in A53T α-synuclein transgenic mouse model. Acta Neuropathol Commun 6: 32. 10.1186/s40478-018-0538-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kim S, Kwon SH, Kam TI et al. (2019). Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron 103: 627–641. 10.1016/j.neuron.2019.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kirik D, Rosenblad C, Burger C et al. (2002). Parkinson-like neurodegeneration induced by targeted overexpression of α-synuclein in the nigrostriatal system. J Neurosci 22: 2780–2791. 10.1523/jneurosci.22-07-02780.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Kordower JH, Olanow CW, Dodiya HB et al. (2013). Disease duration and the integrity of the nigrostriatal system in Parkinson’s disease. Brain 136: 2419–2431. 10.1093/brain/awt192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Kuo YM, Li Z, Jiao Y et al. (2010). Extensive enteric nervous system abnormalities in mice transgenic for artificial chromosomes containing Parkinson disease-associated α-synuclein gene mutations precede central nervous system changes. Hum Mol Genet 19: 1633–1650. 10.1093/hmg/ddq038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Lam HA, Wu N, Cely I et al. (2011). Elevated tonic extracellular dopamine concentration and altered dopamine modulation of synaptic activity precede dopamine loss in the striatum of mice overexpressing human α-synuclein Hoa. J Neurosci Res 89: 1091–1102. 10.1002/jnr.22611.Elevated. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lee MK, Stirling W, Xu Y et al. (2002). Human α-synuclein-harboring familial Parkinson’s disease-linked Ala-53 → Thr mutation causes neurodegenerative disease with α-synuclein aggregation in transgenic mice. Proc Natl Acad Sci U S A 99: 8968–8973. 10.1073/pnas.132197599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Lee BD, Shin JH, VanKampen J et al. (2010). Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson’s disease. Nat Med 16: 998–1000. 10.1038/nm.2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Lees AJ, Tolosa E, Olanow CW (2015). Four pioneers of L-dopa treatment: Arvid Carlsson, Oleh Homykiewicz, George Cotzias, and Melvin Yahr. Mov Disord 30: 19–36. 10.1002/mds.26120. [DOI] [PubMed] [Google Scholar]
  76. Lesage S, Janin S, Lohmann E et al. (2007). LRRK2 exon 41 mutations in sporadic Parkinson disease in Europeans. Arch Neurol 64: 425–430. 10.1001/archneur.64.3.425. [DOI] [PubMed] [Google Scholar]
  77. Lewis SJG, Foltynie T, Blackwell AD et al. (2005). Heterogeneity of Parkinson’s disease in the early clinical stages using a data driven approach. J Neurol Neurosurg Psychiatry 76: 343–348. 10.1136/jnnp.2003.033530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Li H, Ham A, Ma TC et al. (2019). Mitochondrial dysfunction and mitophagy defect triggered by heterozygous GBA mutations. Autophagy 15: 113–130. 10.1080/15548627.2018.1509818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Lin X, Parisiadou L, Gu XL et al. (2009). Leucine-rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson’s-disease-related mutant α-synuclein. Neuron 64: 807–827. 10.1016/j.neuron.2009.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Lo Bianco C, Schneider BL, Bauer M et al. (2004). Lentiviral vector delivery of parkin prevents dopaminergic degeneration in an α-synuclein rat model of Parkinson’s disease. Proc Nad Acad Sci U S A 101: 17510–17515. 10.1073/pnas.0405313101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Luk KC, Lee VM (2014). Addition of exogenous α-synuclein-pre-formed fibrils to primaiy.pdf. Nat Protoc 9: 2135–2146. 10.1038/nprot.2014.143.Addition. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Luk KC, Kehm VM, Carroll J et al. (2012). Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in non-transgenic mice. Science 338: 949–953. 10.1126/science.1227157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Lundblad M, Decressac M, Mattsson B et al. (2012). Impaired neurotransmission caused by overexpression of α-synuclein in nigral dopamine neurons. Proc Natl Acad Sci U S A 109: 3213–3219. 10.1073/pnas.1200575109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Ma SY, Röyttä M, Rinne JO et al. (1997). Correlation between neuromorphometry in the substantia nigra and clinical features in Parkinson’s disease using disector counts. J Neurol Sci 151: 83–87. 10.1016/S0022-510X(97)00100-7. [DOI] [PubMed] [Google Scholar]
  85. Magen I, Fleming SM, Zhu C et al. (2012). Cognitive deficits in a mouse model of pre-manifest Parkinson’s disease. Eur J Neurosci 35: 870–882. 10.1111/j.l460-9568.2012.08012.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Manfredsson FP, Luk KC, Benskey MJ et al. (2018). Induction of alpha-synuclein pathology in the enteric nervous system of the rat and non-human primate results in gastrointestinal dysmotility and transient CNS pathology. Neurobiol Dis 112: 106–118. 10.1016/j.nbd.2018.01.008.Induction. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. McDowell KA, Shin D, Roos KP et al. (2014). Sleep dysfunction and EEG alterations in mice overexpressing alpha-synuclein. J Parkinsons Dis 4: 531–539. 10.3233/JPD-140374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Migdalska-Richards A, Wegrzynowicz M, Rusconi R et al. (2017). The L444P Gba1 mutation enhances alpha-synuclein induced loss of nigral dopaminergic neurons in mice. Brain 140: 2706–2721. 10.1093/brain/awx221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Murphy KE, Gysbers AM, Abbott SK et al. (2014). Reduced glucocerebrosidase is associated with increased α-synuclein in sporadic Parkinson’s disease. Brain 137: 834–848. 10.1093/brain/awt367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Nikonova EV, Xiong Y, Tanis KQ et al. (2012). Transcriptional responses to loss or gain of function of the leucine-rich repeat kinase 2 (LRRK2) gene uncover biological processes modulated by LRRK2 activity. Hum Mol Genet 21: 163–174. 10.1093/hmg/ddr451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Ordonez DG, Lee MK, Feany MB (2018). α-Synuclein induces mitochondrial dysfunction through spectrin and the actin cytoskeleton. Neuron 97: 108–124. 10.1016/j.neuron.2017.11.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Paisán-Ruíz C, Jain S, Evans EW et al. (2004). Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron 44: 595–600. 10.1016/j.neuron.2004.10.023. [DOI] [PubMed] [Google Scholar]
  93. Paisán-Ruíz C, Nath P, Washecka N et al. (2008). Comprehensive analysis of LRRK2 in publicly available Parkinson’s disease cases and neurologically normal controls. Hum Mutat 29: 485–490. 10.1002/humu.20668. [DOI] [PubMed] [Google Scholar]
  94. Pankratz N, Foroud T (2007). Genetics of Parkinson disease. Genet Med 9: 801–811. 10.1097/GIM.0b013e31815bf97c. [DOI] [PubMed] [Google Scholar]
  95. Park H, Lee JY, Shin CM et al. (2015). Characterization of gastrointestinal disorders in patients with parkinsonian syndromes. Parkinsonism Relat Disord 21: 455–460. 10.1016/j.parkreldis.2015.02.005. [DOI] [PubMed] [Google Scholar]
  96. Patterson JR, Duffy MF, Kemp CJ et al. (2019). Time course and magnitude of alpha-synuclein inclusion formation and nigrostriatal degeneration in the rat model of synucleinopathy triggered by intrastriatal α-synuclein preformed fibrils. Neurobiol Dis 130: 104525. 10.1016/j.nbd.2019.104525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Paumier KL, Sukoff Rizzo SJ, Berger Z et al. (2013). Behavioral characterization of A53T mice reveals early and late stage deficits related to Parkinson’s disease. PLoS One 8. 10.1371/journal.pone.0070274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Paumier KL, Luk KC, Manfredsson FP et al. (2015). Intrastriatal injection of pre-formed mouse α-synuclein fibrils into rats triggers α-synuclein pathology and bilateral nigrostriatal degeneration. Physiol Behav 82: 185–199. 10.1016/j.nbd.2015.06.003.Intrastriatal. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Pellegrini L, Hauser DN, Li Y et al. (2018). Proteomic analysis reveals co-ordinated alterations in protein synthesis and degradation pathways in LRRK2 knockout mice. Hum Mol Genet 27: 3257–3271. 10.1093/hmg/ddy232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Piccoli G, Condliffe SB, Bauer M et al. (2011). LRRK2 controls synaptic vesicle storage and mobilization within the recycling pool. J Neurosci 31: 2225–2237. 10.1523/JNEUROSCI.3730-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Polymeropoulos MH, Lavedan C, Leroy E et al. (1997). Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 276: 2045–2047. 10.1126/science.276.5321.2045. [DOI] [PubMed] [Google Scholar]
  102. Ramonet D, Daher JPL, Lin BM et al. (2011). Dopaminergic neuronal loss, reduced neurite complexity and autophagic abnormalities in transgenic mice expressing G2019S mutant LRRK2. PLoS One 6: 13–19. 10.1371/journal.pone.0018568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Reish HEA, Standaert DG (2015). Role of α-synuclein in inducing innate and adaptive immunity in Parkinson disease. J Parkinsons Dis 5: 1–19. 10.3233/JPD-140491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Richter F, Fleming SM, Watson M et al. (2014). A GCase chaperone improves motor function in a mouse model of synucleinopathy. Neurotherapeutics 11: 840–856. 10.1007/s13311-014-0294-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Rocha EM, Smith GA, Park E et al. (2015). Glucocerebrosidase gene therapy prevents α-synucleinopathy of midbrain dopamine neurons. Neurobiol Dis 82: 495–503. 10.1016/j.nbd.2015.09.009. [DOI] [PubMed] [Google Scholar]
  106. Rockenstein E, Mallory M, Hashimoto M et al. (2002). Differential neuropathological alterations in transgenic mice expressing α-synuclein from the platelet-derived growth factor and Thy-1 promoters. J Neurosci Res 68: 568–578. 10.1002/jnr.10231. [DOI] [PubMed] [Google Scholar]
  107. Satake W, Nakabayashi Y, Mizuta I et al. (2009). Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson’s disease. Nat Genet 41: 1303–1307. 10.1038/ng.485. [DOI] [PubMed] [Google Scholar]
  108. Schulte C, Gasser T (2011). Genetic basis of Parkinson’s disease: Inheritance, penetrance, and expression. Appl Clin Genet 4: 67–80. 10.2147/TACG.S11639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Selikhova M, Williams DR, Kempster PA et al. (2009). A clinico-pathological study of subtypes in Parkinson’s disease. Brain 132: 2947–2957. 10.1093/brain/awp234. [DOI] [PubMed] [Google Scholar]
  110. Sheehan P, Yue Z (2019). Deregulation of autophagy and vesicle trafficking in Parkinson’s disease. Physiol Behav 697: 59–65. 10.1016/j.neulet.2018.04.013.Deregulation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Shin N, Jeong H, Kwon J et al. (2008). LRRK2 regulates synaptic vesicle endocytosis. Exp Cell Res 314: 2055–2065. 10.1016/j.yexcr.2008.02.015. [DOI] [PubMed] [Google Scholar]
  112. Sidransky E, Nalls MA, Aasly JO et al. (2009). Multicenter Analysis of Glucocerebrosidase Mutations in Parkinson’s Disease. N Engl J Med 361: 1651–1661. 10.1056/nejmoa0901281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Singleton AB, Farrer M, Johnson J et al. (2003). alpha-Synuclein locus triplication causes Parkinson’s disease. Science 302: 841. 10.1126/science.1090278. [DOI] [PubMed] [Google Scholar]
  114. Sloan M, Alegre-Abarrategui J, Potgieter D et al. (2016). LRRK2 BAC transgenic rats develop progressive, L-DOPA-responsive motor impairment, and deficits in dopamine circuit function. Hum Mol Genet 25: 951–963. 10.1093/hmg/ddv628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Song DD, Shults CW, Sisk A et al. (2004). Enhanced substantia nigra mitochondrial pathology in human α-synuclein transgenic mice after treatment with MPTP. Exp Neurol 186: 158–172. 10.1016/S0014886(03)00342-X. [DOI] [PubMed] [Google Scholar]
  116. Spillantini MG, Schmidt ML, Lee VMY et al. (1997). α-Synuclein in Lewy bodies. Nature 388: 839–840. [DOI] [PubMed] [Google Scholar]
  117. Stauch KL, Villeneuve LM, Purnell PR et al. (2016). Loss of Pink1 modulates synaptic mitochondrial bioenergetics in the rat striatum prior to motor symptoms: concomitant complex I respiratory defects and increased complex II-mediated respiration. Proteomics Clin Appl 10: 1205–1217. 10.1002/prca.201600005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Steger M, Tonelli F, Ito G et al. (2016). Phosphoproteomics reveals that Parkinson’s disease kinase LRRK2 regulates a subset of Rab GTPases. Elife 5: 1–28. 10.7554/elife.12813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Subramaniam SR, Vergnes L, Franich NR et al. (2014). Region specific mitochondrial impairment in mice with wide-spread overexpression of alpha-synuclein. Neurobiol Dis 70: 204–213. 10.1016/j.nbd.2014.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Sung H-Y, Park J-W, Kim J-S (2014). The frequency and severity of gastrointestinal symptoms in patients with early Parkinson’s disease. J Mov Disord 7: 7–12. 10.14802/jmd.14002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Tillerson JL, Caudle WM, Reverón ME et al. (2002). Detection of behavioral impairments correlated to neurochemical deficits in mice treated with moderate doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Exp Neurol 178: 80–90. 10.1006/exnr.2002.8021. [DOI] [PubMed] [Google Scholar]
  122. Tong Y, Yamaguchi H, Giaime E et al. (2010). Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of α-synuclein, and apoptotic cell death in aged mice. Proc Natl Acad Sci U S A 107: 9879–9884. 10.1073/pnas.1004676107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Tong Y, Giaime E, Yamaguchi H et al. (2012). Loss of leucine-rich repeat kinase 2 causes age-dependent bi-phasic alterations of the autophagy pathway. Mol Neurodegener 9: 2. 10.1186/1750-1326-7-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Uemura N, Yagi H, Uemura MT et al. (2018). Inoculation of α-synuclein preformed fibrils into the mouse gastrointestinal tract induces Lewy body-like aggregates in the brain-stem via the vagus nerve. Mol Neurodegener 13: 1–11. 10.1186/s13024-018-0257-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Uemura N, Yagi H, Uemura MT et al. (2020). Limited spread of pathology within the brainstem of α-synuclein BAC transgenic mice inoculated with preformed fibrils into the gastrointestinal tract. Neurosci Lett 716: 134651. 10.1016/j.neulet.2019.134651. [DOI] [PubMed] [Google Scholar]
  126. Ungerstedt U (1968). 6-Hydroxy-dopamine induced degeneration of central monoamine neurons. Eur J Pharmacol 5: 107–110 10.1016/0014-2999(68)90164-7. [DOI] [PubMed] [Google Scholar]
  127. Valadez-Barba V, Cota-Coronado A, Hernández-Pérez OR et al. (2020). iPSC for modeling neurodegenerative disorders. Regen Med 15: 332–339. 10.1016/j.reth.2020.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Villeneuve LM, Purnell PR, Boska MD et al. (2016). Early expression of Parkinson’s disease-related mitochondrial abnormalities in PINK1 knockout rats. Mol Neurobiol 53: 171–186. 10.1007/s12035-014-8927-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Volpicelli-Daley LA, Luk KC, Patel TP et al. (2011). α-Syn fibrils. Neuron 72: 57–71. 10.1016/j.neuron.2011.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Volpicelli-Daley LA, Abdelmotilib H, Liu Z et al. (2016). G2019S-LRRK2 expression augments α-synuclein sequestration into inclusions in neurons. J Neurosci 36: 7415–7427. 10.1523/JNEUROSCI.3642-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Wakabayashi K, Takahashi H, Takeda S et al. (1988). Parkinson’s disease: the presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol 76: 217–221. 10.1007/BF00687767. [DOI] [PubMed] [Google Scholar]
  132. Wakabayashi K, Takahashi H, Ohama E et al. (1990). Parkinson’s disease: an immunohistochemical study of Lewy body-containing neurons in the enteric nervous system. Acta Neuropathol 79: 581–583. 10.1007/BF00294234. [DOI] [PubMed] [Google Scholar]
  133. Wang L, Fleming SM, Chesselet M-F et al. (2008). Abnormal colonic motility in mice overexpressing human wild-type α-synuclein Lixin. Neuroreport 19: 873–876. 10.1097/WNR.0b013e3282ffda5e.Abnormal. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Wang L, Magen I, Yuan PQ et al. (2012). Mice overexpressing wild-type human alpha-synuclein display alterations in colonic myenteric ganglia and defecation. Neurogastroenterol Motil 24: e425–e436. 10.1111/j.l365-2982.2012.01974.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Watson MB, Richter F, Lee SK et al. (2012). Regionally-specific microglial activation in young mice overexpressing human wildtype alpha-synuclein. Exp Neurol 237: 318–334. 10.1016/j.expneurol.2012.06.025.Regionally-specific. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Weng YH, Chen CY, Lin KJ et al. (2016). (R1441C) LRRK2 induces the degeneration of SN dopaminergic neurons and alters the expression of genes regulating neuronal survival in a transgenic mouse model. Exp Neurol 275: 104–115. 10.1016/j.expneurol.2015.09.001. [DOI] [PubMed] [Google Scholar]
  137. West AB, Cowell RM, Daher PL (2014). Differential LRRK2 expression in the cortex, striatum, and substantia nigra in transgenic and nontransgenic rodents. J Comp Neurol 522: 2465–2480. 10.1002/cne.23583.Differential. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Xiong Y, Neifert S, Karuppagounder SS et al. (2018). Robust kinase- and age-dependent dopaminergic and norepinephrine neurodegeneration in LRRK2 G2019S transgenic mice. Proc Natl Acad Sci U S A 115: 1635–1640. 10.1073/pnas.1712648115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Xu Y, Sun Y, Ran H et al. (2011). Accumulation and distribution of α-synuclein and ubiquitin in the CNS of Gaucher disease mouse models YH. Mol Genet Metab 102: 436–447. 10.1016/j.ymgme.2010.12.014.Accumulation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Yun SP, Kim D, Kim S et al. (2018). α-Synuclein accumulation and GBA deficiency due to L444P GBA mutation contributes to MPTP-induced parkinsonism. Mol Neurodegener 13: 1–19. 10.1186/s13024-017-0233-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Zhao HT, John N, Delic V et al. (2017). LRRK2 antisense oligonucleotides ameliorate alpha-synuclein inclusion formation in a Parkinson’s disease mouse model. Mol Ther Nucleic Acids 15: 508–519. 10.1016/j.omtn.2017.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Zhou H, Huang C, Tong J et al. (2011). Temporal expression of mutant LRRK2 in adult rats impairs dopamine reuptake. Int J Biol Sci 7: 753–761. 10.7150/ijbs.7.753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Zimprich A, Biskup S, Leitner P et al. (2004). Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 44: 601–607. 10.1016/j.neuron.2004.11.005. [DOI] [PubMed] [Google Scholar]

RESOURCES