Parkinson's Disease Animal Models
Meera Saxena (meera at luminomics dot com)
Luminomics, Augusta, GA, United States
DOI
//dx.doi.org/10.13070/mm.en.3.2843
Date
last modified : 2023-05-16; original version : 2013-08-28
Cite as
MATER METHODS 2013;3:2843
Abstract

A comprehensive review of animal models for Parkinson's disease.

Parkinson’s Disease (PD) is the second most common neurodegenerative disorder after Alzheimer’s Disease, affecting 0.3% of the population in industrialized countries – 1% of the population over 60 years old [4, 5], and its growth surpasses that of Alzheimer's disease [6]. PD is caused by the dysfunction and/or degeneration of dopaminergic neurons (DN), resulting in difficulty with both motor and non-motor function. PD is a progressive disorder, typically considered idiopathic, with some known genetic and toxicological factors, including common herbicides/fungicides such as paraquat and maneb [7-9], and trichloroethylene (TCE) [10]. Current drug therapies act to suppress PD symptoms, however, significant cellular damage may have already occurred once motor deficits are evident and no neuroprotectant therapies are currently available [11, 12]. In addition, many of these drugs (such as Pramipexole, Ropinrole, Rotigotine and Apomorphine) operate by increasing dopamine signaling in the body and may have lessening effectiveness over time. Not all clinical manifestations of PD are thought to be due to DN degeneration [13]. This may explain recent data demonstrating why simply targeting a single neurotransmitter may eventually lose its therapeutic efficacy [14].

Sym Protein Top three suppliers
APOEapolipoprotein EAbcam ab1906 (14), Novus Biologicals NB110-60531 (7), Santa Cruz Biotechnology sc-13521 (4)
COMTcatechol-O-methyltransferaseAbcam ab126618 (1)
DRD2dopamine receptor D2Santa Cruz Biotechnology sc-5303 (8)
FBXO7F-box protein 7Santa Cruz Biotechnology sc-271763 (5)
GBAglucosylceramidase betaAbcam ab55080 (4), Santa Cruz Biotechnology sc-365745 (3), New England Biolabs e8023s (3)
GIGYF2GRB10 interacting GYF protein 2Santa Cruz Biotechnology sc-393918 (1)
LRRK2leucine rich repeat kinase 2Abcam ab133474 (26), Neuromab 75-253 (17), BioLegend 808201 (1)
MAOBmonoamine oxidase BATCC hb-242 (1)
MAPTmicrotubule associated protein tauInvitrogen MN1020 (734), Abcam ab80579 (30), Cell Signaling Technology 9632 (17)
MIFmacrophage migration inhibitory factor / parthanatos-associated apoptosis-inducing factor nucleaseAbcam ab175189 (6), Santa Cruz Biotechnology sc-271631 (2)
PARK7Parkinsonism associated deglycaseAbcam ab76008 (5), Santa Cruz Biotechnology sc-55572 (2), Cell Signaling Technology 5933 (2)
PINK1PTEN induced putative kinase 1Cell Signaling Technology 6946 (20), OriGene TA324354 (1), Santa Cruz Biotechnology sc-518052 (1)
PRKNparkin RBR E3 ubiquitin protein ligaseSanta Cruz Biotechnology sc-32282 (46), Cell Signaling Technology 4211 (28), Abcam ab77924 (14)
SCDstearoyl-CoA desaturaseCell Signaling Technology 2794 (16), Abcam ab19862 (8), Santa Cruz Biotechnology sc-58420 (1)
SLC6A3solute carrier family 6 member 3Novus Biologicals NBP2-22164 (12), Abcam ab5990 (4), Santa Cruz Biotechnology sc-32258 (2)
SNCAsynuclein alphaBD Biosciences 610786 (44), Abcam ab51253 (33), Invitrogen 32-8100 (19)
UBCubiquitin CInvitrogen 13-1600 (46), Cell Signaling Technology 5621 (32), Enzo Life Sciences BML-PW0600-0025 (7)
UCHL1ubiquitin C-terminal hydrolase L1Cell Signaling Technology 13179 (8), Abcam ab8189 (7), Invitrogen 480012 (7)
VPS35VPS35, retromer complex componentSanta Cruz Biotechnology sc-374372 (5), Abcam ab57632 (5), Abnova H00055737-M02 (2)
Table 1. Proteins involved in Parkinson's disease and top cited antibodies against them among the over 60,000 formal publications in Validated Antibody Database. The most cited monoclonal antibody from each supplier is listed.

The initial PD models were pharmacologically induced. Although these models did not encompass the neurodegenerative aspects of the disease, their use led to a major breakthrough in 1957 when L-DOPA administrations were noted to lessen PD-like movements in reserpine-treated mice [15]. Despite the lack of PD neuropathology, experiments using reserpine-treated mice as PD models have proven to be predictive of clinical success; all drugs currently on the market were successful in alleviating symptoms in these mice [16]. While this serves to underscore how a predictive model can be useful for screening therapies aimed at alleviating disease-associated symptoms, these models may be less useful for determining curative therapies as the disease mechanism may be dissimilar.

One of the most widely used pharmacological agents to create rodent PD models is 6-hydroxydopamine (6-OHDA) [17-19] ; it has been utilized in nonhuman primates as well but to a lesser degree [20, 21]. 6-OHDA is useful in that it provides a way to create an array of lesion sizes that are dose-dependent. This methodology allows for partial lesions mimicking early stage PD to much larger lesioned areas (greater than 90%). One major drawback of this approach is that 6-OHDA does not cross the blood-brain barrier and must be physically injected into the brain, increasing experimental variables. For example, animals with partial lesions (50–70%) demonstrated motor function deficits in some studies while in other experiments, the lesion had to be greater than 80% [22, 23]. Additionally, behavioral test outcomes tend to be highly variable and dependent on the chosen screening methodology [16]. Another problem these animals share with most other PD models is that they do not form the pathological PD hallmark, Lewy bodies. Classically, Lewy bodies are eosinophilic inclusions that contain ubiquitinated proteins such as α-synuclein [24]. Different conformational strains of α-synuclein are proposed to underscore various synucleinopathies like Parkinson’s disease, dementia with Lewy bodies, and multiple system atrophy [25]. Protein misfolding cyclic amplification (PMCA) assay for α-synuclein is being considered to discriminate them [25]. The exact role of Lewy bodies is unknown but as the aggregation of these bodies appears to be a critical step in the development of the disease, it is a major focus for PD therapeutics [26]. Other pesticide-induced models, such as rotenone, are problematic in that they require surgical exposure or have issues with systemic toxicity and resistance to treatment [27]. Nevertheless, linagliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor, were evaluated in a rotenone-induced rat model of PD [28]. Linagliptin impeded rotenone-induced motor disorders and histological changes and significantly suppressed the activity of pro-inflammatory cytokines, such as tumor necrosis factor-α and interleukin-6 [28]. Dopamine-receptor antagonists haloperidol or SCH23390 have been used to model PD [29].

Another common agent used for inducing both rodent and primate PD models is 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) [30]. Interestingly, it was discovered that MPTP led to chronic PD in humans after several drug users accidentally exposed themselves to the compound in the early eighties [31, 32]. In contrast, usage in rodents comes with certain caveats. For unknown reasons, rats appear to be insensitive to MPTP [33, 34]. In mice, only certain strains are sensitive to MPTP [35, 36]. Moreover, factors such as mouse supplier, gender, age, weight and dosing amounts/schedule can influence lesion reproducibility [30, 35-37].

Parkinson's Disease Animal Models figure 1
Figure 1. L-DOPA treatment increases dopaminergic cells in a monkey model of Parkinson’s Disease. Coronal sections of adult male cynomolgus monkey were stained for tyrosine hydroxylase (TH – brown; Nissl - blue) expression. A) Control MPTP treated monkey, B) MPTP monkey treated with vehicle, C) MPTP monkey additionally treated with L-DOPA. Note the increase in TH+ cells in panel C. From [1]

Thus far, five different primates have been utilized for PD research: African green, rhesus, cynomolgus, squirrel and marmoset monkeys [38]. MPTP causes a much more human-like PD syndrome in primates, although they also lack Lewy body development [39]. There is some species specificity as only African green monkeys appear to exhibit the typical PD rest tremor [40, 41]. As with mice, results may be variable due to the fact that researchers obtain their animals from a wide range of suppliers/countries [38]. However, despite these difference, PD primate models appear to be highly predictive of clinical trial success [42], mirroring some of the morphological changes that occur in humans. For example, L-DOPA appears to increase the number of tyrosine hydroxylase (TH) staining neurons — neurons which are important for dopamine synthesis — similarly in both PD humans and MPTP-treated primates (Fig 1) [1]. It should be noted that this synchronicity is not always the case. Factors that were demonstrated to be protective against 6-OHDA treatment in rodents, shown to alleviate neuronal loss in MPTP-treated primates and had initial Phase I clinical trial success, failed to show any efficacy in Phase II double-blind studies [43-46]. These results merely underscore that all animal models, regardless of how predictive they may appear to be, are simply best approximations for what may occur in humans. Another complicating issue is that gut microbes can metabolize L-DOPA, thus render any effects heterogeneous across Parkinson’s patients [47].

Parkinson's Disease Animal Models figure 2
Figure 2. Knockdown of PINK1 leads to ommatidia and DN degeneration in Drosophila. Representative images of external eye phenotypes of 3-day-old flies expressing (A), control (GMR-GAL4 driver alone) and (B), PINK1 knockdown (GMR-GAL4/UAS-dPINK1 RNAi). A’ and B’ are corresponding images of DN (as identified by anti-dTH staining of 10-day old flies. This data suggests that PINK1 plays an important role in DN survival. From [2]

All the aforementioned models have been utilized to isolate PD therapeutics that alleviate disease symptoms but as previously mentioned, drug discovery with mammalian models can occur very slowly. Toward that end, PD animal models more amenable to high-throughput drug screening have been designed. For example, researchers have created symptomatic models in flies (rotenone) and worms (MPTP) [48, 49]. MPTP decreases dopamine content in both larval and adult zebrafish with corresponding decreases in locomotor activity [50-53]. To date, results from high-throughput drug screens using these types of models have not been published.

All previously mentioned PD models can be considered “cellular” models of PD with face validity as they mimic the disease symptoms but are not an accurate depiction of the underlying mechanism. As such, many researchers are turning to genetic models of the disease, incorporating mutated genes that have been linked PD. Multiple genes have been linked to both the familial and sporadic forms of PD: SNCA, lrrk2, Parkin, PINK1, DJ1, ATP13A2, PLA2G6, FBXO7, UCHL1, GIGYF2, HTRA2, EIF4G, GBA, MAPT, BST1, PARK16, GAK, and HLA [54]. Thus far, less than 20% of all PD have been linked to a genetic cause. As seen with the pharmacological PD models, not all genetic PD models express the typical hallmarks, e.g., Lewy bodies [55, 56]. Mutations of a mitochondrial serine protease HTRA2 cause essential tremor commonly seen in Parkinson patients [57].

Only two of the genes listed above (SNCA and lrrk2) have been clearly identified as autosomal dominant and therefore have been studied the most [58-61]. LRRK2 may also be involved in idiopathic PD [62]. Three clear autosomal recessive PD genes have been identified (Parkin, PINK1 and DJ1) with corresponding vertebrate and invertebrate animal models generated (for example, see Fig 2) [54].

Parkinson's Disease Animal Models figure 3
Figure 3. Lack of a functional lrrk2 causes loss of DN in zebrafish. Zebrafish expressing morphants targeting the WD40 domain of the lrrk2 gene show decreased TH and DAT (dopamine transporter) gene expression. From [3]

A great deal of research has focused on the normal role of SNCA (α-synuclein) but it is difficult to ascertain its specific role given the disparate results from the various animal models. In mice, the knock-out data does not suggest that SNCA plays a crucial role in the development or maintenance of DN [63]. In addition, neither the multiple mice nor rat SNCA models demonstrate a progressive loss of DN [64]. There are conflicting reports in Drosophila as to whether overexpression of wildtype and/or mutant SNCA causes DN loss [65-67]. In worms overexpressing exogenous SNCA mutations, a food-sensing locomotor defect was observed which the researchers correlated to a loss of DN [68]. However, it can be somewhat difficult to interpret data resulting from either fly or worm experiments as neither normally express SNCA. Currently, there are no published reports of zebrafish expressing mutant SCNA (zebrafish do not express an SCNA ortholog), however, overexpression of human wildtype SCNA does cause neurotoxicity in zebrafish [69, 70]. It does appear, however, that SNCA plays a role in regards to dopamine homeostasis as exogenous SNCA was able to rescue mutant zebrafish lacking other synuclein paralogs [69]. Although no large scale drug screening tests have been published using a mutant SNCA model, a small scale drug study was conducted using flies expressing exogenous wildtype SNCA [71]. In zebrafish, a similar model was used to determine the efficacy of a putative therapeutic that would inhibit the formation of Lewy bodies [70].

As with SNCA knockout mice, DN cells appear to develop normally in mice lacking lrrk2 [55, 72] ; these mice do demonstrate slight differences in certain behavioral tests [55]. As seen with the mice, knockout models of an lrrk2 ortholog in worms do not affect overall neuronal development/survival, however, its effects in flies appear to be a bit more complex [73-75]. In regards to PD relevant lrrk2 mutants, a common PD-linked mutant form (G2019S) is toxic in certain cell lines [76, 77]. In contrast, mice overexpressing the same mutation develop normally but have only a slight age-related decrease in overall physiological dopamine concentration, unrelated to any obvious neuronal degeneration [78]. In addition, this decrease does not translate into the altered motor function in mice less than one year old. Similar results are observed in flies and worms incorporating the same mutation [79, 80]. However, the mouse researchers found this to be consistent with the clinical data as the percentage of G2019S carriers that eventually develop PD initially is fairly low (only 28% by age 59) but increases by the age of 69 (51%). The extension of this argument would then be that dopamine levels would decrease or neuronal degeneration would increase if the mice were allowed to mature further. Overexpression of similar pathologically linked lrrk2 mutants have not been introduced into zebrafish as of yet, however, fish which have had lrrk2 function blocked did show significant DN death and locomotor defects (Fig 5 [3] ). Lrrk2 mutant model has been used to evaluate the effect of LRRK2 inhibitor DNL201 on LRRK2 functions [81]. The effects of erucic acid, a peroxisome proliferator-activated receptors (PPARs) ligand, were evaluated in a PD zebrafish model [82]. Erucic acid treatment repaired the changes in the expression of proteins linked to cytoskeletal organisation, transport and localisation and improved locomotor functions and oxidant-damage in central nervous system and intestines [82].

Ongoing Developments

Viral transduction of rodent brain with α-synuclein using a recombinant adeno-associated virus (rAAV) has been used extensively to model PD [83]. rAAV expressing either wild-type or mutant α-synuclein is able to transduce the dopaminergic cells of the substantia nigra. This model has been used a number of studies (see Table 1 of [83] for a comprehensive list). This model is attractive as it enables the overexpression of α-synuclein in the cells that are predominantly affected in the human disease. Furthermore, α-synuclein can be expressed readily in various wild-type, transgenic and knockout animals thus enabling an exploration of the effects of genetic background on PD pathology. The viral transduction of adult animal brain enables researchers to eliminate developmental effects that may complicate the analysis of transgenic and knockout models of PD. Different studies using the rAAV model have produced different phenotypic outcomes but common features are a progressive loss of striatal neurons and significant neuroinflammation (see [83] for a detailed discussion of the different phenotypes observed in various studies). By contrast, the rAAV model generates aggregates that do not fully recapitulate the structural and morphological features of the Lewy bodies seen in human PD [83, 84]. An important technical consideration of this model is that it requires injection of rAAV into the appropriate region of the brain. Whilst technically challenging, it does permit the targeting of specific, disease-relevant, brain regions.

Some groups have also used lentiviral vectors, rather than AAV, to drive expression of α-synuclein. Injection of recombinant lentivirus into the substantia nigra of mice resulted in the loss of dopaminergic neurons and motor dysfunction [85]. For a recent review of the use of viral vectors in PD research see [86].

Another PD model involves the injection into the rodent brain of pre-formed α-syn fibrils [87-89]. For example, researchers from Ionis Pharmaceuticals and Biogen tested the effectiveness of antisense oligonucleotides against SNCA in the reduction of SNCA in rodent preformed fibril models [88]. The fibril injection model leads to the formation of α-syn aggregates that resemble the Lewy bodies and Lewy neurites observed in PD patients [83, 90]. H Park et al showed that pathologic α-synuclein induced neurodegeneration through parthanatos-associated apoptosis-inducing factor nuclease (PAAN) activity [89]. Genetic depletion of PAAN prevented the degeneration of dopaminergic neurons and behavioral disorders [89]. PAANIB-1, a PAAN nuclease inhibitor, suppressed neurodegeneration induced by α-synuclein in vivo [89]. As with the rAAV model, the α-syn fibril model requires material to be injected into the brains of the experimental animals (with the same advantages and disadvantages as discussed above for the rAAV model).

There has been much interest in the mechanisms of propagation of PD within the brain. It appears that the mechanism of α-synucleinopathies such as PD may have much in common with prion-based diseases [91-93]. Models have been developed to investigate this propagation within the brain. In essence, rodents overexpressing human α-synuclein (either transgenically or via AAV transduction) receive a graft of neurons which are subsequently analysed for the presence of human α-synuclein [93]. E Martin-Lopez et al observed pathological changes and functional disorders in the olfactory system in transgenic α-syn-Tg mice expressing the human A30P mutant α-synuclein [94]. The transgenic animals developed severe α-syn pathology in the projection neurons of the olfactory pathway. Furthermore. neurogenesis in α-syn-Tg mice was reduced, and proteomic analysis showed synaptic endocytosis and exocytosis defects in the olfactory bulb [94]. In another version of this model, the brains of rats overexpressing human Htt via AAV transduction received a graft of embryonic dopaminergic neurones; endocytic uptake of human α-synuclein by the grafted neurones was observed which subsequently played a role in seeding the aggregation of endogenous rat α-synuclein [95]. This recapitulates the occurrence of α-synuclein aggregates in neurones transplanted into the brains of PD patients [96, 97]. Kam TI et al reported that pathologic α-syn could activate poly(adenosine 5'-diphosphate-ribose) (PAR) polymerase-1 (PARP-1), and PAR, in turn, could accelerate the formation of pathologic α-syn [98].

A recent study hypothesized that the tyrosine nonreceptor kinase-2 (TNK2) gene and systemic RNA interference protein-3 (SID-3), which is the sole TNK2 ortholog in C. elegans, are involved in dopaminergic and epigenetic signaling regulating neuronal homeostasis [99]. CRISPR-edited nematodes, endogenously expressing SID-3 analogous to TNK2 PD-associated single-nucleotide polymorphisms, showed dopaminergic neurodegeneration and SID-3 dysfunction [99].

Researchers often use mutliple models to address the underlying questions. For example, Fanning S et al applied yeast genetics, rat cortical neurons, worms, iPSC-derived neurons, neurons from PD patients and a mouse model to study the involvement of stearoyl CoA desaturase in synuclein neurotoxicity [100].

Animal Mechanism Pro Con Research impact Expense
Rodent Pharmacologically induced
6-OHDA and MPTP treated animals most popular models.
Some transgenics/ knockouts of PD genes available
Exhibit behavioral and physiological symptoms associated with disease
Amenable to behavioral tests
Wide variety of pharmacologically induced models
Symptoms can be methodology specific, not all transgenics display expected behavioral/ pathological symptoms
Some species are not susceptible to drug treatment
Various factors (e.g., strain, supplier, gender) documented to affect experimental outcomes
Led to major breakthrough – LDOPA treatment for PD symptoms +++
Primate Pharmacologically induced, in particular MPTP Anatomy similar to human and useful for testing drug delivery
Amenable to behavioral tests
Highly predictive of human treatment outcome
Very expensive and highly regulated
Not amenable to large scale drug screening
Some species specificity in regards to symptoms
Useful for examining preclinical efficacy of various therapeutics aimed at alleviating PD symptoms ++++
Zebrafish Knockdown and knockouts of PD genes
Transgenic animals expressing PD mutations,
Pharmacologically induced models available.
Only vertebrate model amenable to large scale forward drug/genetic and behavioral screening Early stage of development as a PD animal model
Do not express all PD orthologs.
Unknown predictive efficacy in regards to clinical trials
Potential to isolate compounds that stimulate DN regrowth ++
Fly Transgenic animals expressing PD mutations
Pharmacologically induced models available.
Amenable to large scale forward drug/genetic and behavioral screening Do not express all PD orthologs.
Unknown predictive efficacy in regards to clinical trials
Useful for examining the combined effects of environmental/genetic factors +
Worm Transgenic animals expressing PD mutations
Pharmacologically induced models available.
Amenable to large scale forward drug/genetic and behavioral screening Do not express all PD orthologs
Unknown predictive efficacy in regards to clinical trials
Determining modifiers of PD proteins, thus identifying new therapeutic targets +
Table 2. Parkinson’s disease animal models.
Declarations

This article is derived from an earlier article "Animal Models for Huntington's and Parkinson's Diseases", and first appeared as a separate article in November 2019.

References
  1. Dicaudo C, Riverol M, Mundiñano I, Ordóñez C, Hernandez M, Marcilla I, et al. Chronic levodopa administration followed by a washout period increased number and induced phenotypic changes in striatal dopaminergic cells in MPTP-monkeys. PLoS ONE. 2012;7:e50842 pubmed publisher
  2. Wang D, Qian L, Xiong H, Liu J, Neckameyer W, Oldham S, et al. Antioxidants protect PINK1-dependent dopaminergic neurons in Drosophila. Proc Natl Acad Sci U S A. 2006;103:13520-5 pubmed
  3. Sheng D, Qu D, Kwok K, Ng S, Lim A, Aw S, et al. Deletion of the WD40 domain of LRRK2 in Zebrafish causes Parkinsonism-like loss of neurons and locomotive defect. PLoS Genet. 2010;6:e1000914 pubmed publisher
  4. Lew M. Overview of Parkinson's disease. Pharmacotherapy. 2007;27:155S-160S pubmed
  5. Samii A, Nutt J, Ransom B. Parkinson's disease. Lancet. 2004;363:1783-93 pubmed
  6. Dorsey E, Bloem B. The Parkinson Pandemic-A Call to Action. JAMA Neurol. 2018;75:9-10 pubmed publisher
  7. Hatcher J, Pennell K, Miller G. Parkinson's disease and pesticides: a toxicological perspective. Trends Pharmacol Sci. 2008;29:322-9 pubmed publisher
  8. Migliore L, Coppede F. Environmental-induced oxidative stress in neurodegenerative disorders and aging. Mutat Res. 2009;674:73-84 pubmed publisher
  9. Migliore L, Coppede F. Genetics, environmental factors and the emerging role of epigenetics in neurodegenerative diseases. Mutat Res. 2009;667:82-97 pubmed publisher
  10. Goldman S, Weaver F, Stroupe K, Cao L, Gonzalez B, Colletta K, et al. Risk of Parkinson Disease Among Service Members at Marine Corps Base Camp Lejeune. JAMA Neurol. 2023;: pubmed publisher
  11. Jankovic J, Poewe W. Therapies in Parkinson's disease. Curr Opin Neurol. 2012;25:433-47 pubmed publisher
  12. Poewe W, Mahlknecht P, Jankovic J. Emerging therapies for Parkinson's disease. Curr Opin Neurol. 2012;25:448-59 pubmed publisher
  13. Langston J. The Parkinson's complex: parkinsonism is just the tip of the iceberg. Ann Neurol. 2006;59:591-6 pubmed
  14. Tritsch N, Ding J, Sabatini B. Dopaminergic neurons inhibit striatal output through non-canonical release of GABA. Nature. 2012;490:262-6 pubmed publisher
  15. Carlsson A, Lindqvist M, Magnusson T. 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature. 1957;180:1200 pubmed
  16. Duty S, Jenner P. Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease. Br J Pharmacol. 2011;164:1357-91 pubmed publisher
  17. Gon xe7 alves F, Matheus F, Silva H, Real J, Rial D, Rodrigues R, et al. Increased ATP Release and Higher Impact of Adenosine A2A Receptors on Corticostriatal Plasticity in a Rat Model of Presymptomatic Parkinson's Disease. Mol Neurobiol. 2023;60:1659-1674 pubmed publisher
  18. Spix T, Nanivadekar S, Toong N, Kaplow I, Isett B, Goksen Y, et al. Population-specific neuromodulation prolongs therapeutic benefits of deep brain stimulation. Science. 2021;374:201-206 pubmed publisher
  19. Qian H, Kang X, Hu J, Zhang D, Liang Z, Meng F, et al. Reversing a model of Parkinson's disease with in situ converted nigral neurons. Nature. 2020;582:550-556 pubmed publisher
  20. Ma K, Huang W, Chen C, Lin S, Wey S, Ting G, et al. Dual SPECT of dopamine system using [99mTc]TRODAT-1 and [123I]IBZM in normal and 6-OHDA-lesioned formosan rock monkeys. Nucl Med Biol. 2002;29:561-7 pubmed
  21. Roeling T, Docter G, Voorn P, Melchers B, Wolters E, Groenewegen H. 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;9:155-64 pubmed
  22. Barneoud P, Descombris E, Aubin N, Abrous D. Evaluation of simple and complex sensorimotor behaviours in rats with a partial lesion of the dopaminergic nigrostriatal system. Eur J Neurosci. 2000;12:322-36 pubmed
  23. Kirik D, Rosenblad C, Bjorklund A. Characterization of behavioral and neurodegenerative changes following partial lesions of the nigrostriatal dopamine system induced by intrastriatal 6-hydroxydopamine in the rat. Exp Neurol. 1998;152:259-77 pubmed
  24. Braak E, Sandmann Keil D, Rüb U, Gai W, de Vos R, Steur E, et al. alpha-synuclein immunopositive Parkinson's disease-related inclusion bodies in lower brain stem nuclei. Acta Neuropathol. 2001;101:195-201 pubmed
  25. Fellner L, Jellinger K, Wenning G, Haybaeck J. Commentary: Discriminating α-synuclein strains in parkinson's disease and multiple system atrophy. Front Neurosci. 2020;14:802 pubmed publisher
  26. Rohn T. Targeting alpha-synuclein for the treatment of Parkinson's disease. CNS Neurol Disord Drug Targets. 2012;11:174-9 pubmed
  27. Betarbet R, Sherer T, Mackenzie G, Garcia Osuna M, Panov A, Greenamyre J. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci. 2000;3:1301-6 pubmed
  28. ElGamal R, Tadros M, Menze E. Linagliptin counteracts rotenone's toxicity in non-diabetic rat model of Parkinson's disease: Insights into the neuroprotective roles of DJ-1, SIRT-1/Nrf-2 and implications of HIF1-α. Eur J Pharmacol. 2023;941:175498 pubmed publisher
  29. Masini D, Kiehn O. Targeted activation of midbrain neurons restores locomotor function in mouse models of parkinsonism. Nat Commun. 2022;13:504 pubmed publisher
  30. Przedborski S, Jackson Lewis V, Naini A, 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;76:1265-74 pubmed
  31. Ballard P, Tetrud J, Langston J. Permanent human parkinsonism due to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP): seven cases. Neurology. 1985;35:949-56 pubmed
  32. Langston J, Ballard P, Tetrud J, Irwin I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science. 1983;219:979-80 pubmed
  33. Giovanni A, Sieber B, Heikkila R, Sonsalla P. Studies on species sensitivity to the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Part 1: Systemic administration. J Pharmacol Exp Ther. 1994;270:1000-7 pubmed
  34. Giovanni A, Sonsalla P, Heikkila R. Studies on species sensitivity to the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Part 2: Central administration of 1-methyl-4-phenylpyridinium. J Pharmacol Exp Ther. 1994;270:1008-14 pubmed
  35. Giovanni A, Sieber B, Heikkila R, Sonsalla P. Correlation between the neostriatal content of the 1-methyl-4-phenylpyridinium species and dopaminergic neurotoxicity following 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine administration to several strains of mice. J Pharmacol Exp Ther. 1991;257:691-7 pubmed
  36. Sonsalla P, Heikkila R. The influence of dose and dosing interval on MPTP-induced dopaminergic neurotoxicity in mice. Eur J Pharmacol. 1986;129:339-45 pubmed
  37. Miller D, Ali S, O Callaghan J, Laws S. The impact of gender and estrogen on striatal dopaminergic neurotoxicity. Ann N Y Acad Sci. 1998;844:153-65 pubmed
  38. Porras G, Li Q, Bezard E. Modeling Parkinson's disease in primates: The MPTP model. Cold Spring Harb Perspect Med. 2012;2:a009308 pubmed publisher
  39. Halliday G, Herrero M, 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;24:1519-23 pubmed publisher
  40. Guehl D, Pessiglione M, Francois C, Yelnik J, Hirsch E, Feger J, et al. Tremor-related activity of neurons in the 'motor' thalamus: changes in firing rate and pattern in the MPTP vervet model of parkinsonism. Eur J Neurosci. 2003;17:2388-400 pubmed
  41. Raz A, Vaadia E, Bergman H. Firing patterns and correlations of spontaneous discharge of pallidal neurons in the normal and the tremulous 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine vervet model of parkinsonism. J Neurosci. 2000;20:8559-71 pubmed
  42. Fox S, Lang A, Brotchie J. Translation of nondopaminergic treatments for levodopa-induced dyskinesia from MPTP-lesioned nonhuman primates to phase IIa clinical studies: keys to success and roads to failure. Mov Disord. 2006;21:1578-94 pubmed
  43. Gasmi M, Herzog C, Brandon E, Cunningham J, Ramirez G, Ketchum E, et al. Striatal delivery of neurturin by CERE-120, an AAV2 vector for the treatment of dopaminergic neuron degeneration in Parkinson's disease. Mol Ther. 2007;15:62-8 pubmed
  44. Kordower J, Herzog C, Dass B, Bakay R, Stansell J, Gasmi M, et al. Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys. Ann Neurol. 2006;60:706-15 pubmed
  45. Marks W, Bartus R, Siffert J, Davis C, Lozano A, Boulis N, et al. Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomised, controlled trial. Lancet Neurol. 2010;9:1164-1172 pubmed publisher
  46. Marks W, Ostrem J, Verhagen L, Starr P, Larson P, Bakay R, et al. Safety and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2-neurturin) to patients with idiopathic Parkinson's disease: an open-label, phase I trial. Lancet Neurol. 2008;7:400-8 pubmed publisher
  47. Maini Rekdal V, Bess E, Bisanz J, Turnbaugh P, Balskus E. Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism. Science. 2019;364: pubmed publisher
  48. Braungart E, Gerlach M, Riederer P, Baumeister R, Hoener M. Caenorhabditis elegans MPP+ model of Parkinson's disease for high-throughput drug screenings. Neurodegener Dis. 2004;1:175-83 pubmed
  49. Coulom H, Birman S. Chronic exposure to rotenone models sporadic Parkinson's disease in Drosophila melanogaster. J Neurosci. 2004;24:10993-8 pubmed
  50. Anichtchik O, Kaslin J, Peitsaro N, Scheinin M, Panula P. Neurochemical and behavioural changes in zebrafish Danio rerio after systemic administration of 6-hydroxydopamine and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. J Neurochem. 2004;88:443-53 pubmed
  51. Bretaud S, Lee S, Guo S. Sensitivity of zebrafish to environmental toxins implicated in Parkinson's disease. Neurotoxicol Teratol. 2004;26:857-64 pubmed
  52. Lam C, Korzh V, Strahle U. Zebrafish embryos are susceptible to the dopaminergic neurotoxin MPTP. Eur J Neurosci. 2005;21:1758-62 pubmed
  53. McKinley E, Baranowski T, Blavo D, Cato C, Doan T, Rubinstein A. Neuroprotection of MPTP-induced toxicity in zebrafish dopaminergic neurons. Brain Res Mol Brain Res. 2005;141:128-37 pubmed
  54. Schulte C, Gasser T. Genetic basis of Parkinson's disease: inheritance, penetrance, and expression. Appl Clin Genet. 2011;4:67-80 pubmed publisher
  55. Hinkle K, Yue M, Behrouz B, Dachsel J, Lincoln S, Bowles E, et al. LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors. Mol Neurodegener. 2012;7:25 pubmed publisher
  56. von Coelln R, Thomas B, Andrabi S, Lim K, Savitt J, Saffary R, et al. Inclusion body formation and neurodegeneration are parkin independent in a mouse model of alpha-synucleinopathy. J Neurosci. 2006;26:3685-96 pubmed
  57. Unal Gulsuner H, Gulsuner S, Mercan F, Onat O, Walsh T, Shahin H, et al. Mitochondrial serine protease HTRA2 p.G399S in a kindred with essential tremor and Parkinson disease. Proc Natl Acad Sci U S A. 2014;111:18285-90 pubmed publisher
  58. Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kösel S, et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson's disease. Nat Genet. 1998;18:106-8 pubmed
  59. Polymeropoulos M, Lavedan C, Leroy E, Ide S, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science. 1997;276:2045-7 pubmed
  60. Singleton A, Farrer M, Bonifati V. The genetics of Parkinson's disease: progress and therapeutic implications. Mov Disord. 2013;28:14-23 pubmed publisher
  61. Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, et al. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004;44:601-7 pubmed
  62. Alessi D, Sammler E. LRRK2 kinase in Parkinson's disease. Science. 2018;360:36-37 pubmed publisher
  63. Chandra S, Fornai F, Kwon H, Yazdani U, Atasoy D, Liu X, et al. Double-knockout mice for alpha- and beta-synucleins: effect on synaptic functions. Proc Natl Acad Sci U S A. 2004;101:14966-71 pubmed
  64. Dawson T, Ko H, Dawson V. Genetic animal models of Parkinson's disease. Neuron. 2010;66:646-61 pubmed publisher
  65. Feany M, Bender W. A Drosophila model of Parkinson's disease. Nature. 2000;404:394-8 pubmed
  66. Periquet M, Fulga T, Myllykangas L, Schlossmacher M, Feany M. Aggregated alpha-synuclein mediates dopaminergic neurotoxicity in vivo. J Neurosci. 2007;27:3338-46 pubmed
  67. Pesah Y, Burgess H, Middlebrooks B, Ronningen K, Prosser J, Tirunagaru V, et al. Whole-mount analysis reveals normal numbers of dopaminergic neurons following misexpression of alpha-Synuclein in Drosophila. Genesis. 2005;41:154-9 pubmed
  68. Kuwahara T, Koyama A, Gengyo Ando K, Masuda M, Kowa H, Tsunoda M, et al. Familial Parkinson mutant alpha-synuclein causes dopamine neuron dysfunction in transgenic Caenorhabditis elegans. J Biol Chem. 2006;281:334-40 pubmed
  69. Milanese C, Sager J, Bai Q, Farrell T, Cannon J, Greenamyre J, et al. Hypokinesia and reduced dopamine levels in zebrafish lacking ?- and ?1-synucleins. J Biol Chem. 2012;287:2971-83 pubmed publisher
  70. Prabhudesai S, Sinha S, ATTAR A, Kotagiri A, Fitzmaurice A, Lakshmanan R, et al. A novel "molecular tweezer" inhibitor of ?-synuclein neurotoxicity in vitro and in vivo. Neurotherapeutics. 2012;9:464-76 pubmed publisher
  71. Pendleton R, Parvez F, Sayed M, Hillman R. Effects of pharmacological agents upon a transgenic model of Parkinson's disease in Drosophila melanogaster. J Pharmacol Exp Ther. 2002;300:91-6 pubmed
  72. Tong Y, Yamaguchi H, Giaime E, Boyle S, Kopan R, Kelleher R, 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;107:9879-84 pubmed publisher
  73. Lee S, Kim W, Lee S, Chung J. Loss of LRRK2/PARK8 induces degeneration of dopaminergic neurons in Drosophila. Biochem Biophys Res Commun. 2007;358:534-9 pubmed
  74. Sakaguchi Nakashima A, Meir J, Jin Y, Matsumoto K, Hisamoto N. LRK-1, a C. elegans PARK8-related kinase, regulates axonal-dendritic polarity of SV proteins. Curr Biol. 2007;17:592-8 pubmed
  75. Wang D, Tang B, Zhao G, Pan Q, Xia K, Bodmer R, et al. Dispensable role of Drosophila ortholog of LRRK2 kinase activity in survival of dopaminergic neurons. Mol Neurodegener. 2008;3:3 pubmed publisher
  76. Greggio E, Jain S, Kingsbury A, Bandopadhyay R, Lewis P, Kaganovich A, et al. Kinase activity is required for the toxic effects of mutant LRRK2/dardarin. Neurobiol Dis. 2006;23:329-41 pubmed
  77. Smith W, Pei Z, Jiang H, Dawson V, Dawson T, Ross C. Kinase activity of mutant LRRK2 mediates neuronal toxicity. Nat Neurosci. 2006;9:1231-3 pubmed
  78. Li X, Patel J, Wang J, Avshalumov M, Nicholson C, Buxbaum J, et al. Enhanced striatal dopamine transmission and motor performance with LRRK2 overexpression in mice is eliminated by familial Parkinson's disease mutation G2019S. J Neurosci. 2010;30:1788-97 pubmed publisher
  79. Ng C, Mok S, Koh C, Ouyang X, Fivaz M, Tan E, et al. Parkin protects against LRRK2 G2019S mutant-induced dopaminergic neurodegeneration in Drosophila. J Neurosci. 2009;29:11257-62 pubmed publisher
  80. Yao C, El Khoury R, Wang W, Byrd T, Pehek E, Thacker C, et al. LRRK2-mediated neurodegeneration and dysfunction of dopaminergic neurons in a Caenorhabditis elegans model of Parkinson's disease. Neurobiol Dis. 2010;40:73-81 pubmed publisher
  81. Jennings D, Huntwork Rodriguez S, Henry A, Sasaki J, Meisner R, Diaz D, et al. Preclinical and clinical evaluation of the LRRK2 inhibitor DNL201 for Parkinson's disease. Sci Transl Med. 2022;14:eabj2658 pubmed publisher
  82. xdc nal x, Cans x131 z D, S xfc rmen M, S xfc rmen S, Sezer Z, Beler M, et al. Identification of molecular network of gut-brain axis associated with neuroprotective effects of PPARδ-ligand erucic acid in rotenone-induced Parkinson's disease model in zebrafish. Eur J Neurosci. 2023;57:585-606 pubmed publisher
  83. Volpicelli Daley L, Kirik D, Stoyka L, Standaert D, Harms A. How can rAAV-?-synuclein and the fibril ?-synuclein models advance our understanding of Parkinson's disease?. J Neurochem. 2016;139 Suppl 1:131-155 pubmed publisher
  84. Azeredo da Silveira S, Schneider B, Cifuentes Diaz C, Sage D, Abbas Terki T, Iwatsubo T, et al. Phosphorylation does not prompt, nor prevent, the formation of alpha-synuclein toxic species in a rat model of Parkinson's disease. Hum Mol Genet. 2009;18:872-87 pubmed publisher
  85. Burré J, Sharma M, Sudhof T. Systematic mutagenesis of ?-synuclein reveals distinct sequence requirements for physiological and pathological activities. J Neurosci. 2012;32:15227-42 pubmed publisher
  86. Fischer D, Gombash S, Kemp C, Manfredsson F, Polinski N, Duffy M, et al. Viral Vector-Based Modeling of Neurodegenerative Disorders: Parkinson's Disease. Methods Mol Biol. 2016;1382:367-82 pubmed publisher
  87. Hansen C, Angot E, Bergström A, Steiner J, Pieri L, Paul G, et al. ?-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J Clin Invest. 2011;121:715-25 pubmed publisher
  88. Cole T, Zhao H, Collier T, Sandoval I, Sortwell C, Steece Collier K, et al. α-Synuclein antisense oligonucleotides as a disease-modifying therapy for Parkinson's disease. JCI Insight. 2021;6: pubmed publisher
  89. Park H, Kam T, Peng H, Chou S, Mehrabani Tabari A, Song J, et al. PAAN/MIF nuclease inhibition prevents neurodegeneration in Parkinson's disease. Cell. 2022;185:1943-1959.e21 pubmed publisher
  90. Volpicelli Daley L, Luk K, Patel T, Tanik S, Riddle D, Stieber A, et al. Exogenous ?-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. 2011;72:57-71 pubmed publisher
  91. Rey N, George S, Brundin P. Review: Spreading the word: precise animal models and validated methods are vital when evaluating prion-like behaviour of alpha-synuclein. Neuropathol Appl Neurobiol. 2016;42:51-76 pubmed publisher
  92. Brundin P, Ma J, Kordower J. How strong is the evidence that Parkinson's disease is a prion disorder?. Curr Opin Neurol. 2016;29:459-66 pubmed publisher
  93. Visanji N, Brotchie J, Kalia L, Koprich J, Tandon A, Watts J, et al. α-Synuclein-Based Animal Models of Parkinson's Disease: Challenges and Opportunities in a New Era. Trends Neurosci. 2016;39:750-762 pubmed publisher
  94. Martín López E, Vidyadhara D, Liberia T, Meller S, Harmon L, Hsu R, et al. α-Synuclein Pathology and Reduced Neurogenesis in the Olfactory System Affect Olfaction in a Mouse Model of Parkinson's Disease. J Neurosci. 2023;43:1051-1071 pubmed publisher
  95. Hassan T, Rage A, Ul Hassan Z. Response to internal medicine residency redesign. Am J Med. 2012;125:e11 pubmed publisher
  96. Kordower J, Chu Y, Hauser R, Freeman T, Olanow C. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease. Nat Med. 2008;14:504-6 pubmed publisher
  97. Li J, Englund E, Holton J, Soulet D, Hagell P, Lees A, et al. Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med. 2008;14:501-3 pubmed publisher
  98. Kam T, Mao X, Park H, Chou S, Karuppagounder S, Umanah G, et al. Poly(ADP-ribose) drives pathologic α-synuclein neurodegeneration in Parkinson's disease. Science. 2018;362: pubmed publisher
  99. Nourse J, Russell S, Moniz N, Peter K, Seyfarth L, Scott M, et al. Integrated regulation of dopaminergic and epigenetic effectors of neuroprotection in Parkinson's disease models. Proc Natl Acad Sci U S A. 2023;120:e2210712120 pubmed publisher
  100. Fanning S, Haque A, Imberdis T, Baru V, Barrasa M, Nuber S, et al. Lipidomic Analysis of α-Synuclein Neurotoxicity Identifies Stearoyl CoA Desaturase as a Target for Parkinson Treatment. Mol Cell. 2019;73:1001-1014.e8 pubmed publisher
ISSN : 2329-5139