Open access peer-reviewed chapter

Animal Models of Parkinson’s Disease

Written By

Javier Blesa, Ines Trigo‐Damas, Ana Quiroga‐Varela and Natalia Lopez‐Gonzalez del Rey

Submitted: 23 November 2015 Reviewed: 24 March 2016 Published: 24 August 2016

DOI: 10.5772/63328

From the Edited Volume

Challenges in Parkinson's Disease

Edited by Jolanta Dorszewska and Wojciech Kozubski

Chapter metrics overview

4,409 Chapter Downloads

View Full Metrics

Abstract

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta, the consequent dopamine deficit in the striatum and the accumulation of aggregated α‐synuclein (α‐syn) in specific brain regions. The underlying pathophysiology of PD remains poorly understood. Animal models are the best tools to study the pathogenesis of PD. Most studies in PD animal models have focused on the motor features associated with dopamine depletion but still the molecular basis of PD and the molecular pathways of cell death remain unknown. While cellular models have helped to identify specific events, in vivo animal models have simulated most, although not all, of the hallmarks of PD and are useful for testing new neuroprotective approaches. In this chapter, we provide a summary of the most used PD animal models, including their advantages and limitations. Classically, in vivo PD animal models can be divided into those using environmental or synthetic neurotoxins (toxin‐based models) or those utilizing the in vivo expression of PD‐related mutations (genetic models). These models include 6‐hydroxydopamine (6‐OHDA), 1‐methyl‐1,2,3,6‐tetrahydropyridine (MPTP), rotenone, and paraquat, as well as genetic models such as those related to α‐syn, PINK1, Parkin, DJ‐1, and LRRK2.

Keywords

  • MPTP
  • 6‐OHDA
  • Rotenone
  • Paraquat
  • α‐syn
  • LRRK2
  • Parkin
  • DJ1

1. Introduction

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the classical motor symptoms: resting tremor, bradykinesia, akinesia, rigidity, and postural instability. PD is characterized by the loss of ~50–70% of the dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the consequent loss of dopamine (DA) in the striatum, and the presence of intracytoplasmic inclusions called Lewy bodies (LB) that are composed mainly of α‐synuclein (α‐syn) and ubiquitin [1]. Although the complete PD pathogenesis is not well understood, thanks to the use of animal models, we have gained a better understanding of its etiology, pathology, and molecular mechanisms. Importantly, none of the current available models is able to fully recapitulate PD symptoms and pathology [2].

The use of animal models in PD (both in vitro and in vivo) has greatly augmented thanks to new strategies for producing sophisticated models, such as the temporal‐ and/or cell‐specific expression of mutated genes in vertebrates [3], human pluripotent cells coaxed into a specific type of neurons [4], and a host of different invertebrate organisms such as Drosophila [5], Medaka fish [6], or Caenorhabditis elegans [7]. Current PD experimental models can still be categorized into two main groups: toxic and genetic (or both of them combined). Over the years, a collection of strategies have been used to produce other animal models to model PD. Some of them included those based neither on neurotoxins nor on genetic mutations that are directly linked to familial PD. Some of these models lack transcription factors that are required for the survival of dopaminergic neurons, such as sonic hedgehog [8], nuclear receptor related protein‐1 (Nurr1) [9], pituitary homeobox 3 (Pitx3) [10], or engrailed 1 [11]. Even so, the reproducibility and reliability of most of these new models are still under debate.

Therefore, the neurotoxins covered in this chapter focus on models produced by 6‐hydroxydopamine (6‐OHDA) and 1‐methyl‐1,2,3,6‐tetrahydropyridine (MPTP) administration, and paraquat and rotenone which are more recent additions to the stable of toxic agents used to model PD. The recent identification of different genetic mutations related to PD (mainly SNCA (α‐syn, PARK1, and 4), PRKN (parkin RBR E3 ubiquitin protein ligase, PARK2), PINK1 (PTEN‐induced putative kinase 1, PARK6), DJ‐1 (PARK7), and LRRK2 (leucine‐rich repeat kinase 2, PARK8) has led to the development of a range of genetic models [12]. Although the expression of all these proteins in invertebrate models offers experimental advantages and can potentially address some important questions regarding the cellular processes underlying PD, in this chapter, we focus on the different expression of these proteins in mammalian models. Also, although the aforementioned genes are mutated in PD and are not overexpressed or knocked out (KO), these animal models are relevant in the way that may reveal specific molecular events that lead to the death of dopaminergic neurons.

In this chapter, we describe the classical and the most useful animal models to model PD. Readers with minimal knowledge of PD will eventually find out the different possibilities offered by each of these models, and their strengths and limitations.

Advertisement

2. Neurotoxic models

2.1. 6‐OHDA (2,4,5‐trihydroxyphenethylamine)

The classic and more often used neurotoxic in animal models of PD is 6‐OHDA [13, 14]. Most animals are sensitive to 6‐OHDA intoxication, including monkeys, cats, dogs, and rats. The rats were the more frequently used [15, 16]. Its effect was first described in the 1950s during the study of central nervous system; 6‐OHDA caused a noradrenaline depletion for several months and a selective loss of noradrenergic terminals [17, 18] and was firstly isolated by Ungersted to lesion the nigrostriatal pathway in the rat decades ago [19].

Although 6‐OHDA is structurally similar to DA (and noradrenaline), the presence of an additional hydroxyl group makes it toxic to dopaminergic neurons. Also, this compound does not cross the blood‐brain barrier, and it makes necessary the direct injection in the brain, normally in subtantia nigra pars compacta, medium forebrain bundle, or striatum [17, 20, 21]. Lesion size depends on the amount of 6‐OHDA, site of injection, and species. Typically, 6‐OHDA is administered in a unilateral manner and its results are very attractive since the intact side can be used as control. Furthermore, even if there is success rate in ventricular administration [22], the bilateral administration normally leads to severe adipsia, aphagia, and also death [23, 24]. When administered intrastriatally, the 6‐OHDA provokes a progressive and retrograde neuronal loss in SNpc and ventral tegmental area (VTA). Actually, in animals with full lesions (>90%) it is also observed the typical pattern seen in PD patients, with a greater loss in SNpc compared to VTA [21, 25]. Although 6‐OHDA interacts with α‐syn, it does not induce the formation of LB inclusions [17, 26]. The motor evaluation in these animal models is usually performed after the administration of drugs such as apomorphine which induces rotational behavior, but novel tests lacking the use of any drug have also been developed in rodents [27]. One use of this model is to ascertain whether the nigrostriatal degeneration is retrograde, i.e., tyrosine hydroxylase (TH) terminals die before the TH‐neurons in SNpc as it happens in patients [21, 28] (Figure 1).

This model is a good model on the base that it can replicate parkinsonian features as DA depletion, nigral DA cell loss, and behavior deficits. Nevertheless, it does not affect other regions in the brain as olfactory bulbs, lower brainstem areas, or locus coeruleus.

2.2. MPTP (1‐methyl‐4‐phenyl‐1,2,5,6‐tetrahydropyridine)

Even if the discovery of MPTP in 1982 due to an error in drug synthesis process could cause some mayhem in certain circles, for PD researchers it was an invaluable gift. Its toxicity was discovered after some young addicts developed idiopathic PD when they injected the compound intravenously. MPTP can be considered a gold standard for toxin‐based animal models since it mimics some of the hallmarks of PD such as damage to the nigrostriatal DA pathway with a profound loss of DA in the striatum and SNpc, oxidative stress, reactive oxygen species, energy failure, and inflammation [29, 30]. However, MPTP does not induce the formation of LB, definitive characteristic of PD [31, 32]. Some studies have attempted to demonstrate the production of LB‐like inclusions after MPTP administration, but those findings are not easy to replicate and make necessary to play with different dosing and timing schedules [33, 34].

MPTP is not a dopaminergic toxin, but its high lipophilia makes it to cross the blood‐brain barrier after systemic administration. Once astrocytes enter the brain, they are metabolized to MPP+ by monoamine oxidase‐B (MAO‐B). MPP+ enters the dopaminergic neurons through the DA transporter (DAT), and once in the cytoplasm it binds to VMAT2 or it is stored in the vesicles in the mitochondria, where it inhibits the complex I of the mitochondrial electron transport chain leading to neuronal death by oxidative stress [3537]. Thus, in mice lacking DAT, MPTP is not toxic [38]. Since the storage vesicles have a limited capacity, MPP+ most likely pushes DA out into the intercellular space where it can be metabolized to a number of compounds some of which are toxic, such as DOPAL [39] and where it can be subjected to superoxide radical (5‐cysteinyl‐DA) and hydroxyl radical attack (6‐OHDA) (Figure 1). Principally, MPTP is used in primates and mice, and it is still unknown why it is not toxic in rats [40, 41]. And in primates, the resemblance with human PD features goes beyond the loss of dopaminergic neurons in the SNpc. In these animals, it also causes a greater loss of DA in SNpc than in VTA or retrorubral field [42, 43]. The classic way of administration is intravenous and systematic [44]. Some researchers also use an alternative route and they inject unilaterally in the internal carotid. This technique presents the same benefits as described before but it’s more difficult to perform [45]. In primates, traditionally, the animals have been treated with high doses of MPTP, and acute models were obtained. However, in the recent years, researchers have introduced new administration protocols in order to obtain more progressive models, which would mimic more exactly the pathology in PD patients. These progressive models would give a chance to study the compensatory mechanism which takes place before the onset of the symptoms [43, 46, 47]. Additionally, in primates treated with low doses of MPTP, a greater degeneration of dopaminergic nerve terminals has been observed in the putamen than in the caudate nucleus [43, 48]. Interestingly, in primates, there is a high variability in the animal’s susceptibility to MPTP and normally older animals are the most susceptible ones [49]. Also, primates treated with MPTP usually respond well to anti‐parkinsonian treatments such as L‐DOPA or apomorphine, and they also develop dyskinesias after long‐term treatment.

The MPTP model in primates can be used in order to study other features of the PD as the nonmotor symptoms, which have recently become a target for researchers since mice do not develop a level of impairment similar to the humans [50, 51]. In the electrophysiological field, this model has also contributed to many advances including deep brain stimulation, currently the major surgical method to alleviate PD symptoms in patients [52, 53]. In the present, MPTP is more often used in mice than monkeys, mainly because of economic and practical reasons. Mice allow researchers to understand better the molecular mechanisms involved in cell death, to explore the neuronal death process or other pathological effects of PD. One remarkable aspect of the research in mice is the possibility of working with genetically modified animals [54, 55]. In sum, MPTP can be considered as the standard bearer for toxin‐based PD animal models.

2.3. Rotenone

Rotenone is the most intoxicating member of the rotenoid family and is typically found in tropical plants. It is both an herbicide and insecticide having a half‐life of 3–5 days depending on light conditions and degrades quickly in soil and water [56]. The toxicity of rotenone comes from its high lipophilia, and it can easily cross the blood‐brain barrier (Figure 1). It is mainly used in rats since, so far, the studies attempting to lesion in mice or monkeys have not been successful [57, 58]. Recently, some studies have tested the toxicity of rotenone when administrated intragastric [59] or directly in the brain [60]. The administration of rotenone can be done via different routes. The most commonly used regime has typically been the systemic administration using osmotic pumps in rats, especially in Lewis rats which present a higher susceptibility to the toxic than other strains [61]. Oral administration is considered the least effective one [61, 62]. Intraperitoneal injections might induce behavioral and neurochemical deficits, and it also presents a high mortality [60]. In the case of intravenous administration, rotenone may lead to loss of nigrostriatal DA neurons and it is able to induce α‐syn aggregation and LB formation, apart from other features such as oxidative stress or gastrointestinal problems [63]. It is the last aspect that makes this model so attractive, since it seems to replicate almost all of the hallmarks of PD [64]. Similar to what happens in PD, rotenone intoxication is associated with 35% reduction in serotonin, 26% in noradrenergic, and 29% in cholinergic neurons [65].

On the contrary, there is some controversy about the use of rotenone as a model of PD since in spite of the DA oxidation there is not much evidence of depletion of DA in the nigrostriatal system [66], and there are no well‐documented cases of PD patients from rotenone intoxication. This makes the model not very advantageous compared to other toxic‐based ones, such as 6‐OHDA and MPTP.

2.4. Paraquat (N,N‐dimethyl‐4‐4‐4‐bypiridinium)

Paraquat (PQ) is an herbicide that exhibits similar structure to MPP+, and this is the reason why it was suggested that it could have a parkinsonian toxic effect. However, so far, only 95 cases of PD patients linked to PQ have been reported [67] even if being widely used in agriculture. Typically, PQ exerts its deleterious effect through oxidative stress mediated by redox cycling and generating reactive oxygen species, more exactly, superoxide radical, hydrogen peroxide, and the hydroxyl radical, which in turn would lead to the damage of lipids, proteins, RNA, and DNA [68, 69]. The evidence of PQ toxicity in the nigrostriatal DA system is somehow ambiguous. Some studies carried out in mice have been able to demonstrate that systemic administration can reduce motor activity, and there is a dose‐dependent loss of TH‐positive striatal fibers and SNc neurons [70, 71]. In contrast, other researchers claimed that there are no PQ‐induced changes after administration [72]. Interestingly, in a recent study, Rappold et al. [73] could evidence that when administered in high doses, PQ can employ the organic cationic transporter‐3 (OCT‐3) and the DAT becomes toxic to neurons in SNpc. They also suggest that PQ damages are caused by radicalized PQ and facilitated by glial cells, as it does MPP+. One of the most striking aspects of PQ with respect to PD is its ability to induce LB‐like structures in dopaminergic neurons of the SNpc [74] mimicking the PD‐like pathology. Nevertheless, how oxidative stress and cell death are linked because of PQ remains unknown, limiting the research to the study of the process of LB formation in dopaminergic neurons (Figure 1).

Additionally, PQ is not the only pesticide or agricultural chemical known to provoke damage in the dopaminergic system. Maneb (manganese ethylenebisdithiocarbamate) or ziram are other examples of compounds that when exposed to them have a greater risk of developing PD [75, 76]. In any case, results from studies using pesticides give credence to the theory that environmental pesticides can cause PD [77, 78]. However, further studies are required to determine the precise involvement of these compounds in the etiology of PD.

Figure 1.

Pathogenesis of toxin‐induced models. MPTP crosses the blood‐brain barrier and is metabolized to 1‐methyl‐4‐phenylpyridinium (MPP+) by the enzyme monoamine oxidase B (MAO‐B) in glial cells and then to the active toxic compound. MPP+ is then taken up by dopamine transporter where it impairs mitochondrial respiration by inhibiting complex I of the electron transport chain, causing oxidative stress and activation of programmed cell death molecular pathways. Both paraquat and 6‐hydroxydopamine (6‐OHDA) easily cross cell membrane through the dopamine transporter and may also exert their toxicities, in part, by targeting mitochondria with the subsequent production of ROS and quinones causing the degeneration of the nigrostriatal dopaminergic neurons. Rotenone is extremely hydrophobic and penetrates easily the cellular membrane inducing the formation of α‐synuclein aggregates and mitochondrial impairment with the subsequent production of ROS and quinones.

Advertisement

3. Genetic models

Although PD is mainly a sporadic disorder, about 10% of all PD cases are caused by genetic mutations [79]. Animal models of these mutations are important as they represent potential therapeutic targets. Having said that, the pathological and behavioral phenotypes of these genetic models are often quite different from the human condition [80]. For example, almost all of these genetic models failed to find significant loss of dopaminergic neurons, the main pathological hallmark of PD [8184]. Below, we describe different genetic models that reproduce the most known mutations observed in familial PD (Figure 2).

Figure 2.

Genetic animal models in Parkinson disease (PD). Many genetic mouse models have been developed in order to understand PD pathogenesis and identify potential therapeutic targets. Genetic models are adjusted based on genetic mutations identified in the human disease. These genes are part of signaling pathways important for neuronal dopaminergic function. These models contribute to know mechanisms on disease onset or progression of PD or to understand the case and effect of these genetic mutations.

3.1. α‐syn

SNCA (α‐syn) was the first gene linked to a dominant‐type, familial PD, called Park1 [85]. The duplication or triplication of α‐syn is sufficient to cause PD, suggesting that the level of α‐syn expression is a critical determinant of PD progression [86]. Three missense mutations of α‐syn, encoding the substitutions A30P, A53T, and E46K, have been identified in familial PD so far [87, 88]. The pathological accumulation of misfolded α‐syn plays an essential role in the pathogenesis of PD since α‐syn is the main component of LB. While LBs are found principally in nigral neurons of PD patients, they are also found in other brain regions such as locus coeruleus, nucleus basalis of Meynert, hypothalamus, cerebral cortex, and cranial nerve motor nuclei [89]. Numerous animal models have been developed trying to replicate α‐syn neurodegeneration and propagation. These include transgenic mice (KO and overexpression), grafting models, intracerebral protein injections, or virally induced expression of α‐syn. The main handicap of these models is that no significant nigrostriatal degeneration has been found in most of them, although some of these mice showed decreased striatal levels of TH or DA and behavioral impairments [80].

In general, the models of α‐syn overexpression in mice produced some behavioral alterations in both the A30P and A53T mice [9092]. Also, depending on the promoter, some models showed loss of terminals and DA in the striatum [9398] although almost of them failed to reproduce the dopaminergic cell loss characteristic of PD [2, 94, 99101]. Only the TH promoter led to dopaminergic cell loss in a few studies [102, 103]. Janezic et al. [104] generated bacterial artificial chromosome (BAC) transgenic mice (SNCA‐OVX) that express WT human α‐syn and display an age‐dependent loss of SNc DA neurons preceded by early deficits in DA release from terminals in the dorsal striatum, protein aggregation, and reduced firing of SNc DA neurons [104]. Regarding viral vectors injections, largely lentiviruses and adeno‐associated viruses (AAVs), have been used to drive exogenous α‐syn in mice, rats, and primates [105109]. In this case, viral vector‐mediated α‐syn models display α‐syn pathology and clear dopaminergic neurodegeneration. The injection of human mutant α‐syn by AAVs into the SNpc of rats induces a progressive, age‐dependent loss of DA neurons, motor impairment, α‐syn cytoplasmic inclusions, and degenerative changes in striatal axons both in rats [110, 111] and mice [109, 112]. In the last years, the suggested prion‐like behavior of α‐syn has been examined in animal models of PD. These models not only explore the pathology and spreading of α‐syn but the cell‐to‐cell transfer. Importantly, to date, numerous studies have demonstrated that α‐syn may be transmissible from cell to cell in animal models in different ways using different approaches [33, 113120].

Thus, despite the limitations of these α‐syn models, some of them could be useful to elucidate the role of a‐α‐syn in PD and the suggested prion‐like mechanism of propagation of this protein [121].

3.2. LRKK2

Mutations in LRRK2 are known to cause a late‐onset autosomal dominant form of PD [122]. The most frequent mutations are the G2019S and the R1441C [123]. Many different LRRK2 rodent models have been developed with different approaches but as it happens with α‐syn, although they show α‐syn or ubiquitin accumulation, progressive motor impairments, and slight reduction of striatal DA, they do not display functional disruption of the nigrostriatal dopaminergic neurons [82, 124128]. Similarly, overexpression of G2019S or R1441C LRRK2 leads to none or slight loss of dopaminergic neurons in the SNpc and no alteration in striatal DA levels or locomotor activity in both mice and rats [129131].

BAC transgenic mice expressing mutated LRKK2 have also been developed showing no nigrostriatal degeneration [132134]. On the contrary, a rat LRRK2 model with neuron‐specific, adenoviral mediated expression of LRRK2 G2019S in the nigrostriatal system has been produced, which develops a progressive degeneration of nigral dopaminergic neurons [135]. Additionally, using viral vector‐based models, Lee and colleagues [28] reported that the expression of G2019S LRRK2 resulted in a 50% neuronal loss in the ipsilateral SNc associated with reduced striatal dopaminergic fibers [136]. In summary, we can conclude that the transgenic LRRK2 animal models are not a useful model for studying the pathology of PD.

3.3. Pink1 and Parkin

Homozygous mutations in the Parkin and PINK1 genes were discovered in families with autosomal recessive PD [137]. In fact, parkin mutations are the most common cause of autosomal recessive PD. Likewise, mutations in PINK1 are the second most common. Despite this early onset, patients with these mutations have an indistinguishable phenotype from that of sporadic patients. Many PINK1 and parkin KO mice have been generated, and the phenotypes of these mice are very similar. PINK1 and Parkin KO mice have an age‐dependent, moderate reduction in striatal DA levels accompanied by low locomotor activity, but do not exhibit major abnormalities in the DA neurons or striatal DA levels, and they do not show LB formation either [81, 138145]. A new approach consisting in overexpression of T240R‐parkin and of human WT parkin in rats leads to progressive and dose‐dependent DA cell death [146]. Noteworthy, the Parkin‐Q311X‐DAT‐BAC mice exhibit multiple late onsets and progressive hypokinetic motor deficits, age‐dependent DA neuron degeneration in the SNc, and a significant reduction in striatal DA and dopaminergic terminals in the striatum [147]. Overall, PINK1 and Parkin models do not produce functional disruption of the nigrostriatal pathway or other PD‐related pathology, thus their usefulness is questionable.

3.4. DJ‐1

Missense DJ‐1 mutations are linked to autosomal recessive and early‐onset PD. DJ‐1 KO mice showed no loss of SNpc dopaminergic neurons but reduced striatal DA release and decreased locomotor activity [148, 149]. Recently, a new DJ‐1 KO mouse, backcrossed on a C57/BL6 background, displayed an early‐onset unilateral loss of DA neurons in the SNpc, progressing to bilateral degeneration with aging. Also, these mice exhibit age‐dependent bilateral degeneration in the locus coeruleus and mild motor behavioral deficits [150]. If confirmed, this model could provide a possible tool to study the progression of PD.

Advertisement

4. Concluding remarks

Our current understanding of PD pathology greatly benefited from the use of animal models. However, despite these accomplishments, current PD animal models still have to be improved a lot. It seems difficult that a single model can fully recapitulate the complexity of the human PD in the short term. Because there is no perfect model to date, it is very important to choose the correct animal model for each experiment. By providing an overview of the different animal models available to modeling PD, readers would find that there are a lot of options addressing a specific experimental need.

Advertisement

Acknowledgments

We acknowledge HM CINAC and many individuals and corporations that supported Parkinson Disease research at HM CINAC for financial support. The authors are supported by grants from CIBERNED and Plan Nacional from Spanish Ministry of Education.

References

  1. 1. Lees AJ, Hardy J, Revesz T. Parkinson's disease. Lancet. 2009 Jun 13;373(9680):2055–66.
  2. 2. Blesa J, Przedborski S. Parkinson's disease: animal models and dopaminergic cell vulnerability. Front Neuroanat. 2014 Dec 15;8:155.
  3. 3. Dawson TM, Ko HS, Dawson VL. Genetic animal models of Parkinson's disease. Neuron. 2010 Jun 10;66(5):646–61.
  4. 4. Berg J, Roch M, Altschüler J, Winter C, Schwerk A, Kurtz A, et al. Human adipose‐derived mesenchymal stem cells improve motor functions and are neuroprotective in the 6‐hydroxydopamine‐rat model for Parkinson's disease when cultured in monolayer cultures but suppress hippocampal neurogenesis and hippocampal memory Function when cultured in spheroids. Stem Cell Rev. 2015 Feb;11(1):133–49.
  5. 5. Guo M. Drosophila as a model to study mitochondrial dysfunction in Parkinson's disease. Cold Spring Harb Perspect Med. 2012 Nov 1;2(11).
  6. 6. Matsui H, Uemura N, Yamakado H, Takeda S, Takahashi R. Exploring the pathogenetic mechanisms underlying Parkinson's disease in medaka fish. J Parkinsons Dis. 2014 Jan;4(2):301–10.
  7. 7. Chege PM, McColl G. Caenorhabditis elegans: a model to investigate oxidative stress and metal dyshomeostasis in Parkinson's disease. Front Aging Neurosci. 2014 Jan;6:89.
  8. 8. Gonzalez‐Reyes LE, Verbitsky M, Blesa J, Jackson‐Lewis V, Paredes D, Tillack K, et al. Sonic hedgehog maintains cellular and neurochemical homeostasis in the adult nigrostriatal circuit. Neuron. 2012 Jul 26;75(2):306–19.
  9. 9. Jankovic J, Chen S, Le WD. The role of Nurr1 in the development of dopaminergic neurons and Parkinson's disease. Prog Neurobiol. 2005 Sep–Oct;77(1–2):128–38.
  10. 10. Li J, Dani JA, Le W. The role of transcription factor Pitx3 in dopamine neuron development and Parkinson's disease. Curr Top Med Chem. 2009 Jan;9(10):855–9.
  11. 11. Nordström U, Beauvais G, Ghosh A, Pulikkaparambil Sasidharan BC, Lundblad M, Fuchs J, Joshi RL, Lipton JW, Roholt A, Medicetty S, Feinstein TN, Steiner JA, Escobar Galvis ML, Prochiantz A, Brundin P. Progressive nigrostriatal terminal dysfunction and degeneration in the engrailed1 heterozygous mouse model of Parkinson's disease. Neurobiol Dis. 2015 Jan;73:70–82.
  12. 12. Chesselet M‐F, Richter F. Modelling of Parkinson's disease in mice. Lancet Neurol. 2011 Dec;10(12):1108–18.
  13. 13. Schwarting RK, Huston JP. Unilateral 6‐hydroxydopamine lesions of meso‐striatal dopamine neurons and their physiological sequelae. Prog Neurobiol. 1996;49(3):215–66.
  14. 14. Schwarting RK, Huston JP. The unilateral 6‐hydroxydopamine lesion model in behavioral brain research. Analysis of functional deficits, recovery and treatments. Prog Neurobiol. 1996;50(2–3):275–331.
  15. 15. Valette H, Deleuze P, Syrota A, Delforge J, Crouzel C, Fuseau C, et al. Canine myocardial beta‐adrenergic, muscarinic receptor densities after denervation: a PET study. J Nucl Med. 1995;36(1):140–6.
  16. 16. Roeling TA, Docter GJ, Voorn P, Melchers BP, Wolters EC, Groenewegen HJ. Effects of unilateral 6‐hydroxydopamine lesions on neuropeptide immunoreactivity in the basal ganglia of the common marmoset, Callithrix jacchus, a quantitative immunohistochemical analysis. J Chem Neuroanat. 1995;9(3):155–64.
  17. 17. Blandini F, Armentero M‐T, Martignoni E. The 6‐hydroxydopamine model: news from the past. Parkinsonism Relat Disord. 2008 Jan;14 Suppl 2:S124–9.
  18. 18. Porter CC, Totaro JA, Stone CA. Effect of 6‐hydroxydopamine and some other compounds on the concentration of norepinephrine in the hearts of mice. J Pharmacol Exp Ther. 1963 Jun;140:308–16.
  19. 19. Ungerstedt U. 6‐Hydroxy‐dopamine induced degeneration of central monoamine neurons. Eur J Pharmacol. 1968;5(1):107–10.
  20. 20. Perese DA, Ulman J, Viola J, Ewing SE, Bankiewicz KS. A 6‐hydroxydopamine‐induced selective parkinsonian rat model. Brain Res. 1989;494(2):285–93.
  21. 21. Przedborski S, Levivier M, Jiang H, Ferreira M, Jackson‐Lewis V, Donaldson D, et al. Dose‐dependent lesions of the dopaminergic nigrostriatal pathway induced by intrastriatal injection of 6‐hydroxydopamine. Neuroscience. 1995;67(3):631–47.
  22. 22. Rodríguez M, Barroso‐Chinea P, Abdala P, Obeso J, González‐Hernández T. Dopamine cell degeneration induced by intraventricular administration of 6‐hydroxydopamine in the rat: similarities with cell loss in Parkinson's disease. Exp Neurol. 2001 May;169(1):163–81.
  23. 23. Ungerstedt U. Adipsia and aphagia after 6‐hydroxydopamine induced degeneration of the nigro‐striatal dopamine system. Acta Physiol Scand Suppl. 1971;367:95–122.
  24. 24. Bourn WM, Chin L, Picchioni AL. Enhancement of audiogenic seizure by 6‐hydroxydopamine. J Pharm Pharmacol. 1972;24(11):913–4.
  25. 25. Sauer H, Oertel WH. Progressive degeneration of nigrostriatal dopamine neurons following intrastriatal terminal lesions with 6‐hydroxydopamine: a combined retrograde tracing and immunocytochemical study in the rat. Neuroscience. 1994;59(2):401–15.
  26. 26. Alves da Costa C, Dunys J, Brau F, Wilk S, Cappai R, Checler F. 6‐Hydroxydopamine but not 1‐methyl‐4‐phenylpyridinium abolishes alpha‐synuclein anti‐apoptotic phenotype by inhibiting its proteasomal degradation and by promoting its aggregation. J Biol Chem. 2006 Apr;281(14):9824–31.
  27. 27. Glajch KE, Fleming SM, Surmeier DJ, Osten P. Sensorimotor assessment of the unilateral 6‐hydroxydopamine mouse model of Parkinson's disease. Behav Brain Res. 2012 May 1;230(2):309–16.
  28. 28. Lee CS, Sauer H, Bjorklund A. Dopaminergic neuronal degeneration and motor impairments following axon terminal lesion by instrastriatal 6‐hydroxydopamine in the rat. Neuroscience. 1996;72(3):641–53.
  29. 29. Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in humans due to a product of meperidine‐analog synthesis. Science. 1983 Feb 25;219(4587):979–80.
  30. 30. Dauer W, Przedborski S. Parkinson's disease: mechanisms and models. Neuron. 2003 Sep 11;39(6):889–909.
  31. 31. Forno LS, DeLanney LE, Irwin I, Langston JW. Similarities and differences between MPTP‐induced parkinsonism and Parkinson's disease. Neuropathologic considerations. Adv Neurol. 1993;60:600–8.
  32. 32. Halliday G, Herrero MT, Murphy K, McCann H, Ros‐Bernal F, Barcia C, et al. No Lewy pathology in monkeys with over 10 years of severe MPTP Parkinsonism. Mov Disord. 2009 Jul 30;24(10):1519–23.
  33. 33. Fornai F, Schlüter OM, Lenzi P, Gesi M, Ruffoli R, Ferrucci M, et al. Parkinson‐like syndrome induced by continuous MPTP infusion: convergent roles of the ubiquitin‐proteasome system and alpha‐synuclein. Proc Natl Acad Sci U S A. 2005 Mar;102(9):3413–8.
  34. 34. Kowall NW, Hantraye P, Brouillet E, Beal MF, McKee AC, Ferrante RJ. MPTP induces alpha‐synuclein aggregation in the substantia nigra of baboons. Neuroreport. 2000 Jan 17;11(1):211–3.
  35. 35. Guillot TS, Shepherd KR, Richardson JR, Wang MZ, Li Y, Emson PC, et al. Reduced vesicular storage of dopamine exacerbates methamphetamine‐induced neurodegeneration and astrogliosis. J Neurochem. 2008 Sep;106(5):2205–17.
  36. 36. Guillot TS, Miller GW. Protective actions of the vesicular monoamine transporter 2 (VMAT2) in monoaminergic neurons. Mol Neurobiol. 2009 Apr;39(2):149–70.
  37. 37. Gainetdinov RR, Fumagalli F, Wang YM, Jones SR, Levey AI, Miller GW, et al. Increased MPTP neurotoxicity in vesicular monoamine transporter 2 heterozygote knockout mice. J Neurochem. 1998;70(5):1973–8.
  38. 38. Bezard E, Gross CE, Fournier MC, Dovero S, Bloch B, Jaber M. Absence of MPTP‐induced neuronal death in mice lacking the dopamine transporter. Exp Neurol. 1999;155(2):268–73.
  39. 39. Panneton WM, Kumar VB, Gan Q, Burke WJ, Galvin JE. The neurotoxicity of DOPAL: behavioral and stereological evidence for its role in Parkinson disease pathogenesis. PLoS One. 2010;5(12):e15251.
  40. 40. Chiueh CC, Markey SP, Burns RS, Johannessen JN, Pert A, Kopin IJ. Neurochemical and behavioral effects of systemic and intranigral administration of N‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine in the rat. Eur J Pharmacol. 1984;100(2):189–94.
  41. 41. Tieu K. A guide to neurotoxic animal models of Parkinson's disease. Cold Spring Harb Perspect Med. 2011 Sep;1(1):a009316.
  42. 42. Blesa J, Pifl C, Sánchez‐González MA, Juri C, García‐Cabezas MA, Adánez R, et al. The nigrostriatal system in the presymptomatic and symptomatic stages in the MPTP monkey model: a PET, histological and biochemical study. Neurobiol Dis. 2012 Oct;48(1):79–91.
  43. 43. Blesa J, Juri C, Collantes M, Peñuelas I, Prieto E, Iglesias E, et al. Progression of dopaminergic depletion in a model of MPTP‐induced Parkinsonism in non‐human primates. An (18)F‐DOPA and (11)C‐DTBZ PET study. Neurobiol Dis. 2010 Jun;38(3):456–63.
  44. 44. Przedborski S, Jackson‐Lewis V, Naini AB, Jakowec M, Petzinger G, Miller R, et al. The parkinsonian toxin 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP): a technical review of its utility and safety. J Neurochem. 2001 Mar;76(5):1265–74.
  45. 45. Bankiewicz KS, Oldfield EH, Chiueh CC, Doppman JL, Jacobowitz DM, Kopin IJ. Hemiparkinsonism in monkeys after unilateral internal carotid artery infusion of 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP). Life Sci. 1986;39(1):7–16.
  46. 46. Hantraye P, Varastet M, Peschanski M, Riche D, Cesaro P, Willer JC, et al. Stable parkinsonian syndrome and uneven loss of striatal dopamine fibres following chronic MPTP administration in baboons. Neuroscience. 1993;53(1):169–78.
  47. 47. Bezard E, Imbert C, Deloire X, Bioulac B, Gross CE. A chronic MPTP model reproducing the slow evolution of Parkinson's disease: evolution of motor symptoms in the monkey. Brain Res. 1997 Aug 22;766(1–2):107–12.
  48. 48. Moratalla R, Quinn B, DeLanney LE, Irwin I, Langston JW, Graybiel AM. Differential vulnerability of primate caudate‐putamen and striosome‐matrix dopamine systems to the neurotoxic effects of 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine. Proc Natl Acad Sci U S A. 1992 May 1;89(9):3859–63.
  49. 49. Ovadia A, Zhang Z, Gash DM. Increased susceptibility to MPTP toxicity in middle‐aged rhesus monkeys. Neurobiol Aging. 1995;16(6):931–7.
  50. 50. Pessiglione M, Guehl D, Hirsch EC, Féger J, Tremblay L. Disruption of self‐organized actions in monkeys with progressive MPTP‐induced parkinsonism. I. Effects of task complexity. Eur J Neurosci. 2004 Jan;19(2):426–36.
  51. 51. Schneider JS. Modeling cognitive deficits associated with Parkinsonism in the chronic‐low‐dose MPTP‐treated monkey. In: Levin ED, Buccafusco JJ, editors. Animal Models of Cognitive Impairment. Boca Raton (FL): CRC Press/Taylor & Francis; 2006. Chapter 9.
  52. 52. Guridi J, Herrero MT, Luquin MR, Guillen J, Ruberg M, Laguna J, et al. Subthalamotomy in parkinsonian monkeys. Behavioural and biochemical analysis. Brain. 1996;119 (Pt 5):1717–27.
  53. 53. Bergman H, Wichmann T, DeLong MR. Reversal of experimental parkinsonism by lesions of the subthalamic nucleus. Science 1990 Sep 21;249(4975):1436–8.
  54. 54. Dauer W, Kholodilov N, Vila M, Trillat A‐CC, Goodchild R, Larsen KE, et al. Resistance of alpha ‐synuclein null mice to the parkinsonian neurotoxin MPTP. Proc Natl Acad Sci U S A. 2002 Oct 29;99(22):14524–9.
  55. 55. Vila M, Jackson‐Lewis V, Vukosavic S, Djaldetti R, Liberatore G, Offen D, et al. Bax ablation prevents dopaminergic neurodegeneration in the 1‐methyl‐ 4‐phenyl‐1,2,3,6‐tetrahydropyridine mouse model of Parkinson's disease. Proc Natl Acad Sci U S A. 2001 Feb 27;98(5):2837–42.
  56. 56. Hisata J. Lake and stream rehabilitation: rotenone use and health risks. 2002.
  57. 57. Lake and Stream Rehabilitation: Rotenone Use and Health Risks, Final Supplemental Environmental Impact Statement (January 2002).
  58. 58. Ferrante RJ, Schulz JB, Kowall NW, Beal MF. Systemic administration of rotenone produces selective damage in the striatum and globus pallidus, but not in the substantia nigra. Brain Res. 1997;753(1):157–62.
  59. 59. Pan‐Montojo F, Anichtchik O, Dening Y, Knels L, Pursche S, Jung R, et al. Progression of Parkinson's disease pathology is reproduced by intragastric administration of rotenone in mice. PLoS One. 2010 Jan;5(1):e8762.
  60. 60. Alam M, Mayerhofer A, Schmidt WJ. The neurobehavioral changes induced by bilateral rotenone lesion in medial forebrain bundle of rats are reversed by L‐DOPA. Behav Brain Res. 2004;151(1–2):117–24.
  61. 61. Betarbet R, Sherer TB, MacKenzie G, Garcia‐Osuna M, Panov A V, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci. 2000;3(12):1301–6.
  62. 62. Inden M, Kitamura Y, Takeuchi H, Yanagida T, Takata K, Kobayashi Y, et al. Neurodegeneration of mouse nigrostriatal dopaminergic system induced by repeated oral administration of rotenone is prevented by 4‐phenylbutyrate, a chemical chaperone. J Neurochem. 2007 Jun;101(6):1491–504.
  63. 63. Cannon JR, Tapias V, Na HM, Honick AS, Drolet RE, Greenamyre JT. A highly reproducible rotenone model of Parkinson's disease. Neurobiol Dis. 2009 May;34(2):279–90.
  64. 64. Sherer TB, Kim JH, Betarbet R, Greenamyre JT. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha‐synuclein aggregation. Exp Neurol. 2003 Jan;179(1):9–16.
  65. 65. Höglinger GU, Féger J, Prigent A, Michel PP, Parain K, Champy P, et al. Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. J Neurochem. 2003 Feb;84(3):491–502.
  66. 66. Wu Y‐N, Johnson SW. Dopamine oxidation facilitates rotenone‐dependent potentiation of N‐methyl‐D‐aspartate currents in rat substantia nigra dopamine neurons. Neuroscience. 2011 Nov;195:138–44.
  67. 67. Berry C, La Vecchia C, Nicotera P. Paraquat and Parkinson's disease. Cell Death Differ. 2010 Jul;17(7):1115–25.
  68. 68. Day BJ, Patel M, Calavetta L, Chang LY, Stamler JS. A mechanism of paraquat toxicity involving nitric oxide synthase. Proc Natl Acad Sci U S A. 1999;96(22):12760–5.
  69. 69. Przedborski S, Ischiropoulos H. Reactive oxygen and nitrogen species: weapons of neuronal destruction in models of Parkinson's disease. Antioxid Redox Signal. 2005 May–Jun;7(5–6):685–93
  70. 70. Brooks AI, Chadwick CA, Gelbard HA, Cory‐Slechta DA, Federoff HJ. Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Res. 1999;823(1–2):1–10.
  71. 71. McCormack AL, Thiruchelvam M, Manning‐Bog AB, Thiffault C, Langston JW, Cory‐Slechta DA, et al. Environmental risk factors and Parkinson's disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol Dis. 2002 Jul;10(2):119–27.
  72. 72. Thiruchelvam M, Brockel BJ, Richfield EK, Baggs RB, Cory‐Slechta DA. Potentiated and preferential effects of combined paraquat and maneb on nigrostriatal dopamine systems: environmental risk factors for Parkinson's disease? Brain Res. 2000;873(2):225–34.
  73. 73. Rappold PM, Cui M, Chesser AS, Tibbett J, Grima JC, Duan L, et al. Paraquat neurotoxicity is mediated by the dopamine transporter and organic cation transporter‐3. Proc Natl Acad Sci U S A. 2011 Dec 20;108(51):20766–71.
  74. 74. Manning‐Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Di Monte DA. The herbicide paraquat causes up‐regulation and aggregation of alpha‐synuclein in mice: paraquat and alpha‐synuclein. J Biol Chem. 2002 Jan 18;277(3):1641–4.
  75. 75. Tang CC, Poston KL, Dhawan V, Eidelberg D. Abnormalities in metabolic network activity precede the onset of motor symptoms in Parkinson's disease. J Neurosci. 2010 Jan;30(3):1049–56.
  76. 76. Tanner CM, Kamel F, Ross GW, Hoppin JA, Goldman SM, Korell M, et al. Rotenone, paraquat, and Parkinson's disease. Environ Health Perspect. 2011 Jun;119(6):866–72.
  77. 77. Butterfield PG, Valanis BG, Spencer PS, Lindeman CA, Nutt JG. Environmental antecedents of young‐onset Parkinson's disease. Neurology. 1993;43(6):1150–8.
  78. 78. Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Richardson RJ. The risk of Parkinson's disease with exposure to pesticides, farming, well water, and rural living. Neurology. 1998;50(5):1346–50.
  79. 79. Kalinderi K, Bostantjopoulou S, Fidani L. The genetic background of Parkinson's disease: current progress and future prospects. Acta Neurol Scand. 2016 Feb 12.
  80. 80. Chesselet MF, Fleming S, Mortazavi F, Meurers B. Strengths and limitations of genetic mouse models of Parkinson's disease. Park Relat Disord. 2008;14 Suppl 2:S84–7.
  81. 81. Goldberg MS, Fleming SM, Palacino JJ, Cepeda C, Lam HA, Bhatnagar A, et al. Parkin‐deficient mice exhibit nigrostriatal deficits but not loss of dopaminergic neurons. J Biol Chem. 2003 Oct 31;278(44):43628–35.
  82. 82. Hinkle KM, Yue M, Behrouz B, Dächsel JC, Lincoln SJ, Bowles EE, et al. LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co‐ordination behaviors. Mol Neurodegener. 2012 Jan;7:25.
  83. 83. Sanchez G, Varaschin RK, Büeler H, Marcogliese PC, Park DS, Trudeau L‐E. Unaltered striatal dopamine release levels in young Parkin knockout, Pink1 knockout, DJ‐1 knockout and LRRK2 R1441G transgenic mice. PLoS One. 2014 Jan;9(4):e94826.
  84. 84. Andres‐Mateos E, Perier C, Zhang L, Blanchard‐Fillion B, Greco TM, Thomas B, et al. DJ‐1 gene deletion reveals that DJ‐1 is an atypical peroxiredoxin‐like peroxidase. Proc Natl Acad Sci U S A. 2007;104(37):14807–12.
  85. 85. Goedert M, Spillantini MG, Del Tredici K, Braak H. 100 years of Lewy pathology. Nat Rev Neurol. 2013 Jan;9(1):13–24.
  86. 86. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. alpha‐Synuclein locus triplication causes Parkinson's disease. Science. 2003 Oct 31;302(5646):841.
  87. 87. Schapira AH, Jenner P. Etiology and pathogenesis of Parkinson's disease. Mov Disord. 2011 May;26(6):1049–55.
  88. 88. Vekrellis K, Xilouri M, Emmanouilidou E, Rideout HJ, Stefanis L. Pathological roles of α‐synuclein in neurological disorders. Lancet Neurol. 2011 Nov;10(11):1015–25.
  89. 89. Recasens A, Dehay B. Alpha‐synuclein spreading in Parkinson's disease. Front Neuroanat. 2014 Jan;8:159.
  90. 90. Oaks AW, Frankfurt M, Finkelstein DI, Sidhu A. Age‐dependent effects of A53T alpha‐synuclein on behavior and dopaminergic function. PLoS One. 2013 Jan;8(4):e60378.
  91. 91. Sotiriou E, Vassilatis DK, Vila M, Stefanis L. Selective noradrenergic vulnerability in α‐synuclein transgenic mice. Neurobiol Aging. 2010 Dec;31(12):2103–14.
  92. 92. Paumier KL, Sukoff Rizzo SJ, Berger Z, Chen Y, Gonzales C, Kaftan E, et al. Behavioral characterization of A53T mice reveals early and late stage deficits related to Parkinson's disease. PLoS One. 2013 Jan;8(8):e70274.
  93. 93. Masliah E, Rockenstein E, Veinbergs I, Mallory M, Hashimoto M, Takeda A, Sagara Y, Sisk A, Mucke L. Dopaminergic loss and inclusion body formation in alpha‐synuclein mice: implications for neurodegenerative disorders. Science 2000 Feb 18;287(5456):1265–9. PubMed PMID: 10678833.
  94. 94. van der Putten H, Wiederhold KH, Probst A, Barbieri S, Mistl C, Danner S, et al. Neuropathology in mice expressing human alpha‐synuclein. J Neurosci. 2000 Aug 15;20(16):6021–9.
  95. 95. Rockenstein E, Mallory M, Hashimoto M, Song D, Shults CW, Lang I, et al. Differential neuropathological alterations in transgenic mice expressing alpha‐synuclein from the platelet‐derived growth factor and Thy‐1 promoters. J Neurosci Res. 2002 Jun 1;68(5):568–78.
  96. 96. Ikeda M, Kawarabayashi T, Harigaya Y, Sasaki A, Yamada S, Matsubara E, et al. Motor impairment and aberrant production of neurochemicals in human alpha‐synuclein A30P+A53T transgenic mice with alpha‐synuclein pathology. Brain Res. 2009 Jan 23;1250:232–41.
  97. 97. Ono K, Ikemoto M, Kawarabayashi T, Ikeda M, Nishinakagawa T, Hosokawa M, et al. A chemical chaperone, sodium 4‐phenylbutyric acid, attenuates the pathogenic potency in human alpha‐synuclein A30P + A53T transgenic mice. Parkinsonism Relat Disord. 2009 Nov;15(9):649–54.
  98. 98. Lam HA, Wu N, Cely I, Kelly RL, Hean S, Richter F, et al. Elevated tonic extracellular dopamine concentration and altered dopamine modulation of synaptic activity precede dopamine loss in the striatum of mice overexpressing human α‐synuclein. J Neurosci Res. 2011 Jul;89(7):1091–102.
  99. 99. Giasson BI, Duda JE, Quinn SM, Zhang B, Trojanowski JQ, Lee VM. Neuronal alpha‐synucleinopathy with severe movement disorder in mice expressing A53T human alpha‐synuclein. Neuron. 2002;34(4):521–33.
  100. 100. Gispert S, Del Turco D, Garrett L, Chen A, Bernard DJ, Hamm‐Clement J, et al. Transgenic mice expressing mutant A53T human alpha‐synuclein show neuronal dysfunction in the absence of aggregate formation. Mol Cell Neurosci. 2003 Oct;24(2):419–29.
  101. 101. Gomez‐Isla T, Irizarry MC, Mariash A, Cheung B, Soto O, Schrump S, et al. Motor dysfunction and gliosis with preserved dopaminergic markers in human alpha‐synuclein A30P transgenic mice. Neurobiol Aging. 2003;24(2):245–58.
  102. 102. Thiruchelvam MJ, Powers JM, Cory‐Slechta DA, Richfield EK. Risk factors for dopaminergic neuron loss in human alpha‐synuclein transgenic mice. Eur J Neurosci. 2004 Feb;19(4):845–54.
  103. 103. Wakamatsu M, Ishii A, Iwata S, Sakagami J, Ukai Y, Ono M, et al. Selective loss of nigral dopamine neurons induced by overexpression of truncated human alpha‐synuclein in mice. Neurobiol Aging. 2008 Apr;29(4):574–85.
  104. 104. Janezic S, Threlfell S, Dodson PD, Dowie MJ, Taylor TN, Potgieter D, et al. Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model. Proc Natl Acad Sci U S A. 2013 Oct 15;110(42):E4016–25.
  105. 105. Kirik D, Rosenblad C, Burger C, Lundberg C, Johansen TE, Muzyczka N, et al. Parkinson‐like neurodegeneration induced by targeted overexpression of alpha‐synuclein in the nigrostriatal system. J Neurosci. 2002 Apr 1;22(7):2780–91.
  106. 106. Lo Bianco C, Ridet J‐L, 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 Aug 6;99(16):10813–8.
  107. 107. 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 Mar 20;13(5):605–12.
  108. 108. Lauwers E, Bequé D, Van Laere K, Nuyts J, Bormans G, Mortelmans L, et al. Non‐invasive imaging of neuropathology in a rat model of alpha‐synuclein overexpression. Neurobiol Aging. 2007 Feb;28(2):248–57.
  109. 109. Lauwers E, Debyser Z, Van Dorpe J, De Strooper B, Nuttin B, Baekelandt V. Neuropathology and neurodegeneration in rodent brain induced by lentiviral vector‐mediated overexpression of alpha‐synuclein. Brain Pathol. 2003 Jul;13(3):364–72.
  110. 110. Decressac M, Mattsson B, Lundblad M, Weikop P, Björklund A. Progressive neurodegenerative and behavioural changes induced by AAV‐mediated overexpression of α‐synuclein in midbrain dopamine neurons. Neurobiol Dis. 2012 Mar;45(3):939–53.
  111. 111. Kirik D, Annett LE, Burger C, Muzyczka N, Mandel RJ, Björklund A. Nigrostriatal alpha‐synucleinopathy induced by viral vector‐mediated overexpression of human alpha‐synuclein: a new primate model of Parkinson's disease. Proc Natl Acad Sci U S A. 2003 Mar 4;100(5):2884–9.
  112. 112. Oliveras‐Salvá M, Van der Perren A, Casadei N, Stroobants S, Nuber S, D’Hooge R, et al. rAAV2/7 vector‐mediated overexpression of alpha‐synuclein in mouse substantia nigra induces protein aggregation and progressive dose‐dependent neurodegeneration. Mol Neurodegener. 2013 Jan;8:44.
  113. 113. Luk KC, Lee VM‐Y. Modeling Lewy pathology propagation in Parkinson's disease. Parkinsonism Relat Disord. 2014 Jan;20 Suppl 1:S85–7.
  114. 114. Recasens A, Dehay B, Bové J, Carballo‐Carbajal I, Dovero S, Pérez‐Villalba A, et al. Lewy body extracts from Parkinson disease brains trigger α‐synuclein pathology and neurodegeneration in mice and monkeys. Ann Neurol. 2014 Mar;75(3):351–62.
  115. 115. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Trojanowski JQ, et al. Pathological α‐synuclein transmission initiates Parkinson‐like neurodegeneration in nontransgenic mice. Science. 2012 Nov 16;338(6109):949–53.
  116. 116. Luk KC, Kehm VM, Zhang B, O’Brien P, Trojanowski JQ, Lee VMY. Intracerebral inoculation of pathological α‐synuclein initiates a rapidly progressive neurodegenerative α‐synucleinopathy in mice. J Exp Med. 2012 May 7;209(5):975–86.
  117. 117. Masuda‐Suzukake M, Nonaka T, Hosokawa M, Kubo M, Shimozawa A, Akiyama H, et al. Pathological alpha‐synuclein propagates through neural networks. Acta Neuropathol Commun. 2014 Jan;2:88.
  118. 118. Sacino AN, Brooks M, Thomas MA, McKinney AB, Lee S, Regenhardt RW, et al. Intramuscular injection of α‐synuclein induces CNS α‐synuclein pathology and a rapid‐onset motor phenotype in transgenic mice. Proc Natl Acad Sci U S A. 2014 Jul 7;111(29):10732–7.
  119. 119. Grofova I. The identification of striatal and pallidal neurons projecting to substantia nigra. An experimental study by means of retrograde axonal transport of horseradish peroxidase. Brain Res. 1975;91(2):286–91.
  120. 120. Peelaerts W, Bousset L, Van der Perren A, Moskalyuk A, Pulizzi R, Giugliano M, et al. α‐Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature. 2015 Jun 18;522(7556):340–4.
  121. 121. Gubellini P, Kachidian P. Animal models of Parkinson's disease: an updated overview. Rev Neurol (Paris). 2015 Nov;171(11):750–61.
  122. 122. Healy DG, Falchi M, O’Sullivan SS, Bonifati V, Durr A, Bressman S, et al. Phenotype, genotype, and worldwide genetic penetrance of LRRK2‐associated Parkinson's disease: a case‐control study. Lancet Neurol. 2008 Jul;7(7):583–90.
  123. 123. Rudenko IN, Cookson MR. Heterogeneity of leucine-rich repeat kinase 2 mutations: genetics, mechanisms and therapeutic implications. Neurotherapeutics. 2014 Oct;11(4):738-50. doi: 10.1007/s13311-014-0284-z.
  124. 124. Andres‐Mateos E, Mejias R, Sasaki M, Li X, Lin BM, Biskup S, et al. Unexpected lack of hypersensitivity in LRRK2 knock‐out mice to MPTP (1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine). J Neurosci. 2009 Dec 16;29(50):15846–50.
  125. 125. Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher RJ, et al. Loss of leucine‐rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of alpha‐synuclein, and apoptotic cell death in aged mice. Proc Natl Acad Sci U S A. 2010 May 25;107(21):9879–84.
  126. 126. Lin X, Parisiadou L, Gu X‐L, Wang L, Shim H, Sun L, et al. Leucine‐rich repeat kinase 2 regulates the progression of neuropathology induced by Parkinson's‐disease‐related mutant alpha‐synuclein. Neuron. 2009 Dec 24;64(6):807–27.
  127. 127. Tong Y, Pisani A, Martella G, Karouani M, Yamaguchi H, Pothos EN, et al. R1441C mutation in LRRK2 impairs dopaminergic neurotransmission in mice. Proc Natl Acad Sci U S A. 2009 Aug 25;106(34):14622–7.
  128. 128. Herzig MC, Kolly C, Persohn E, Theil D, Schweizer T, Hafner T, et al. LRRK2 protein levels are determined by kinase function and are crucial for kidney and lung homeostasis in mice. Hum Mol Genet. 2011 Nov 1;20(21):4209–23.
  129. 129. Ramonet D, Daher JPL, Lin BM, Stafa K, Kim J, Banerjee R, et al. Dopaminergic neuronal loss, reduced neurite complexity and autophagic abnormalities in transgenic mice expressing G2019S mutant LRRK2. PLoS One. 2011 Jan;6(4):e18568.
  130. 130. Chen C‐Y, Weng Y‐H, Chien K‐Y, Lin K‐J, Yeh T‐H, Cheng Y‐P, et al. (G2019S) LRRK2 activates MKK4‐JNK pathway and causes degeneration of SN dopaminergic neurons in a transgenic mouse model of PD. Cell Death Differ. 2012 Oct;19(10):1623–33.
  131. 131. Shaikh KT, Yang A, Youshin E, Schmid S. Transgenic LRRK2 (R1441G) rats‐a model for Parkinson disease? PeerJ. 2015 Jan;3:e945.
  132. 132. Li Y, Liu W, Oo TF, Wang L, Tang Y, Jackson‐Lewis V, et al. Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal features of Parkinson's disease. Nat Neurosci. 2009 Jul;12(7):826–8.
  133. 133. Winner B, Melrose HL, Zhao C, Hinkle KM, Yue M, Kent C, et al. Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol Dis. 2011 Mar;41(3):706–16.
  134. 134. Tagliaferro P, Kareva T, Oo TF, Yarygina O, Kholodilov N, Burke RE. An early axonopathy in a hLRRK2(R1441G) transgenic model of Parkinson disease. Neurobiol Dis. 2015 Oct;82:359–71.
  135. 135. Dusonchet J, Kochubey O, Stafa K, Young SM, Zufferey R, Moore DJ, et al. A rat model of progressive nigral neurodegeneration induced by the Parkinson's disease‐associated G2019S mutation in LRRK2. J Neurosci. 2011 Jan 19;31(3):907–12.
  136. 136. Lee BD, Shin J‐H, VanKampen J, Petrucelli L, West AB, Ko HS, et al. Inhibitors of leucine‐rich repeat kinase‐2 protect against models of Parkinson's disease. Nat Med. 2010 Sep;16(9):998–1000.
  137. 137. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998 Apr;392(6676):605–8.
  138. 138. Gautier CA, Kitada T, Shen J. Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress. Proc Natl Acad Sci U S A. 2008 Aug 12;105(32):11364–9.
  139. 139. Gispert S, Ricciardi F, Kurz A, Azizov M, Hoepken H‐H, Becker D, et al. Parkinson phenotype in aged PINK1‐deficient mice is accompanied by progressive mitochondrial dysfunction in absence of neurodegeneration. PLoS One. 2009 Jan;4(6):e5777.
  140. 140. Martella G, Platania P, Vita D, Sciamanna G, Cuomo D, Tassone A, et al. Enhanced sensitivity to group II mGlu receptor activation at corticostriatal synapses in mice lacking the familial parkinsonism‐linked genes PINK1 or Parkin. Exp Neurol. 2009 Feb;215(2):388–96.
  141. 141. Palacino JJ, Sagi D, Goldberg MS, Krauss S, Motz C, Wacker M, et al. Mitochondrial dysfunction and oxidative damage in parkin‐deficient mice. J Biol Chem. 2004 Apr 30;279(18):18614–22.
  142. 142. Zhu X‐R, Maskri L, Herold C, Bader V, Stichel CC, Güntürkün O, et al. Non‐motor behavioural impairments in parkin‐deficient mice. Eur J Neurosci. 2007 Oct;26(7):1902–11.
  143. 143. Itier J‐M, Ibanez P, Mena MA, Abbas N, Cohen‐Salmon C, Bohme GA, et al. Parkin gene inactivation alters behaviour and dopamine neurotransmission in the mouse. Hum Mol Genet. 2003 Sep 15;12(18):2277–91.
  144. 144. Von Coelln R, Thomas B, Savitt JM, Lim KL, Sasaki M, Hess EJ, et al. Loss of locus coeruleus neurons and reduced startle in parkin null mice. Proc Natl Acad Sci U S A. 2004 Jul 20;101(29):10744–9.
  145. 145. Perez FA, Palmiter RD. Parkin‐deficient mice are not a robust model of parkinsonism. Proc Natl Acad Sci U S A. 2005 Feb 8;102(6):2174–9.
  146. 146. Van Rompuy A‐S, Lobbestael E, Van der Perren A, Van den Haute C, Baekelandt V. Long‐term overexpression of human wild‐type and T240R mutant Parkin in rat substantia nigra induces progressive dopaminergic neurodegeneration. J Neuropathol Exp Neurol. 2014 Feb;73(2):159–74.
  147. 147. Lu X‐H, Fleming SM, Meurers B, Ackerson LC, Mortazavi F, Lo V, 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 Feb 18;29(7):1962–76.
  148. 148. Goldberg MS, Pisani A, Haburcak M, Vortherms TA, Kitada T, Costa C, et al. Nigrostriatal dopaminergic deficits and hypokinesia caused by inactivation of the familial Parkinsonism‐linked gene DJ‐1. Neuron. 2005 Feb 17;45(4):489–96.
  149. 149. Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, 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 Apr 5;102(14):5215–20.
  150. 150. Rousseaux MWC, Marcogliese PC, Qu D, Hewitt SJ, Seang S, Kim RH, et al. Progressive dopaminergic cell loss with unilateral‐to‐bilateral progression in a genetic model of Parkinson disease. Proc Natl Acad Sci U S A. 2012 Sep 25;109(39):15918–23.

Written By

Javier Blesa, Ines Trigo‐Damas, Ana Quiroga‐Varela and Natalia Lopez‐Gonzalez del Rey

Submitted: 23 November 2015 Reviewed: 24 March 2016 Published: 24 August 2016