Research Models

Parkinson's Disease

PD models come to Alzforum

22 Mar 2017 Selected rodent models of PD (18 of them!) are now included in the database thanks to generous support from MJFF.

Progress in Parkinson's disease research and therapeutic development depends on robust preclinical models, including rodent models. By organizing information related to the characterization of selected PD models, this resource conveys what is known about each one and facilitates comparison of phenotypes. This resource was created in collaboration with The Michael J. Fox Foundation (MJFF) as part of its commitment to make PD models more accessible and accelerate PD research.

The Parkinson's disease models were selected by experts at The Michael J. Fox Foundation (MJFF).

21 Models

Name Other Names Strain Name Genetic Background Gene Mutation Modification Info Modification Disease Neuropathology Behavior/Cognition Other Phenotype Availability Primary Paper Visualization
Rat Models (6)
DJ-1 knockout rat LE-Park7em1sage; HsdSage:LE-Park7em1Sage; formerly LEH-Park7tm1sage Long Evans Hooded Park7 (DJ1) Disruption of the DJ-1 gene by zinc finger nuclease (ZFN) technology. ZFNs were engineered to bind to a recognition sequence in exon 5 of PARK7 and cleave DNA. Park7 (DJ1): Knock-Out Parkinson's Disease Age-related decrease in dopaminergic neurons in the substantia nigra and locus coeruleus; approximately 50 percent reduction by 8 months. Striatal dopamine and serotonin levels elevated 2-3fold over wild-type levels. No change was found in TH-immunoreactivity in the ventral tegmental area or striatum. Enhanced olfactory detection, abnormal short-term memory, impaired coping behavior, reduced anxiety on light-dark box. Abnormalities in gait, paw positioning, strength, vocalizations, and tongue movements. In males, impaired licking, longer and more frequent ultrasonic vocalizations, and accelerated decrease in average call intensity with age. No increase in mortality up to 8 months of age. Available through Inotiv. Cryopreserved. (Previously available through Horizon Discovery (formerly Sage Labs), Cat #TGRL4830) Dave et al., 2014 Yes
Line 10681, hBAC G2019S-LRRK2, LRRK2 G2019S rat NTac:SD-Tg(LRRK2*G2019S)571CJLi Sprague-Dawley LRRK2 LRRK2 G2019S BAC construct carrying the human gene Lrrk2 with the G2019S mutation. LRRK2: Transgenic Parkinson's Disease No overt neurodegeneration out to 12 months of age. Elongated dopaminergic neurons. Elevated oxidative and nitrosative stress. No evidence of gliosis. No α-synuclein inclusions until challenged with exogenous α-synuclein. No change in dopamine levels. Mild abnormalities in motor behavior. Slightly more postural instability at 8 months of age (but not at 4 and 12 months). Slightly more rearing events at 12 months, but not at younger ages. Bone marrow myeloid progenitor numbers were decreased, but suppressive myeloid cells were increased at 6 to 11 months of age. Available through Taconic, Cat# 10681, Cryopreserved. Research services with this model are available from QPS Austria. West et al., 2014 Yes
Lrrk2 knockout rat LEH-Lrrk2tm1sage, HsdSage: LE-Lrrktm1sage, formerly known as TGRL4620 Long Evans Hooded Lrrk2 Zinc finger nuclease (ZFN) technology was used to generate a deletion of 10 base pairs in exon 30 of the rat Lrrk2 gene. This resulted in a frameshift and a premature stop codon in the same exon. Lrrk2: Knock-Out Parkinson's Disease Not observed. Protection against dopaminergic cell loss under conditions involving LPS or α-synuclein overexpression in the substantia nigra. No changes in basal or evoked release of dopamine. One assessment of Rotarod performance revealed no impairment at 12 months of age compared with wild-type rats. Abnormalities in peripheral organs, including the kidney, liver, and lung. Lifespan and organ function are not adversely affected by Lrrk2 knockout. Available through Inotiv, Live and cryopreserved. Baptista et al., 2013, Ness et al., 2013 Yes
Park2 KO, Parkin knockout rat HsdSage:LE-Park2em1Sage; formerly LEH-Park2TM1sage Long Evans Hooded Park2 The rat Park2 gene was disrupted using zinc finger nuclease (ZFN) technology, in which targeted ZFN RNA was injected into fertilized eggs. The ZFNs were engineered to bind to a recognition site sequence in exon 4 of Park2 and cleave the DNA. When the resulting double strand break was repaired by non-homologous end joining, a deletion of five base pairs was created in the founder rat. This deletion led to a frame shift and the creation of a premature stop codon. Park2: Knock-Out Parkinson's Disease No significant changes in dopaminergic neurons in the substantia nigra nor striatal dopamine levels, but alterations in dopaminergic signaling were detected at an early age, as were disruptions in mitochondrial protein expression in striatum. No increase in α-synuclein. Evoked release of striatal glycine greater at 12 months versus wild-type rats. No behavioral deficits detected at 4, 6, and 8 months of age. However, at 2 months, male KO rats made fewer small stereotypic movements, such as scratching and grooming, than wild-type controls. At 2 months, male KO rats had a greater preference for methamphetamine than wild-type rats. Available through Inotiv. Cryopreserved as heterozygous embryos. (Previously available through Envigo). Dave et al., 2014 Yes
Pink1 knockout rat, Park6 KO rat HsdSage:LE-Pink1em1Sage; formerly LEH-Pink1tm1Sage-/- Long Evans Hooded Pink1 The rat Pink1 gene was disrupted using zinc finger nuclease (ZFN) technology. The ZFNs were engineered to bind to a recognition site in exon 4 of Pink1 and cleave the DNA. When the resulting double strand break was repaired, a deletion of 26 base pairs was created. This deletion lead to a frameshift and the creation of a premature stop codon. Pink1: Knock-Out Parkinson's Disease Age-related decrease in dopaminergic neurons in the substantia nigra; greater than 50 percent reduction at eight months. Alterations in striatal dopamine and serotonin levels. Progressive increase in α-synuclein aggregates and reduced brain volume across numerous regions. Increased ventricular volume. Alterations in cortical and striatal mitochondria. Abnormalities in gait, coordination, and strength. As early as 5 weeks of age, KOs had increased foot slips on the tapered balance beam, at 7 weeks they showed hind limb fatigue, which progressed to hind limb dragging, and by 2 months they exhibited alterations in oromotor (lingual) behaviors. Heavier than wild-type rats at 4, 6, and 8 months of age. No increase in mortality. Early (3 months of age) gene expression changes in whole blood, indicative of inflammation-related changes. Gastrointestinal dysfunction appears within 6 months of age. Available through Inotiv. Live. (Previously available through Envigo (formerly Horizon Discovery and Sage Labs), Cat# TGRL4690)  Dave et al., 2014 Yes
α-synuclein (E46K) rat, founder line 70, Human α-synuclein E46K rat, E46K aSyn Rat, Alpha-synuclein E46K Rat (BAC Tg) NTac:SD-Tg(SNCA*E46K)70CJLi Sprague-Dawley SNCA SNCA E46K A bacterial artificial chromosome (BAC) was used to introduce human α-synuclein with the E46K mutation. SNCA: Transgenic Parkinson's Disease No overt neuronal loss. Accumulation of mutant α-synuclein in the brain, in the form of diffuse staining and intracellular aggregates. Aggregates were largely restricted to dopaminergic neurons of the substantia nigra and ventral tegmental area. Elevated nitrotyrosine in dopaminergic neurons. No overt behavioral changes until challenged with low-dose rotenone, upon which the rats exhibit bradykinesia, postural instability, and rigidity. Viable and fertile. Available through Taconic Cat #10679; Cryopreserved. Cannon et al., 2013 Yes
Mouse Models (15)
C57BL/6-Lrrk2tm4.1Arte C57BL/6NTac LRRK2 LRRK2: Knock-In Parkinson's Disease Levels of phospho-substrates of LRRK2 (e.g., Rab10) are increased in the brain. Endocytosis and axonal transport defects in neurons. Cholinergic innervation density is lower in the prelimbic/infralimbic cortical areas and dorsomedial striatum, but not in the dorsal lateral geniculate nucleus in 2-6-month-old males. Microglial immunostaining is similar in the striatum and midbrain at 8 weeks Attention deficits, slower information processing speeds, and impaired goal-directed learning are evident in 2-6-month-old mice—deficits rescued by systemic administration of the acetylcholinesterase inhibitor donepezil. Cognitive flexibility and novel objective recognition similar to controls. Sleep behavior is perturbed at 8-10 months of age. Functional and structural synaptic alterations and impaired synaptic plasticity observed during development (P21) in the dorsal striatum and NAc, some of which may be transient. Impaired synaptic plasticity (LTP) in striatum present early (P21) and persists into adulthood. Available through Taconic, Cat#13940, Live. Research services with this model are available from QPS Austria. Matikainen-Ankney et al., 2016 Yes
BAC Lrrk2-G2019S, FLAG-Lrrk2-G2019S, BAC-Lrrk2-G2019S, LRRK2 G2019S BAC Tg Mouse (Yue) B6.Cg-Tg(Lrrk2*G2019S)2Yue/J A BAC construct was injected into B6C3 F1 oocytes. Founder line 2 was established and maintained by breeding to C57BL/6J inbred mice. LRRK2 LRRK2 G2019S A bacterial artificial chromosome (BAC) containing the entire mouse Lrrk2 gene was modified to include the G2019S mutation. The BAC (~240 kb) contained the murine Lrrk2 promoter region (~35 kb) and a FLAG-tag downstream of the start codon. LRRK2: Transgenic Parkinson's Disease Brain appears normal. No neuronal or cell death at 12 months. Impaired neurite motility and synaptic vesicle endocytosis in cultured neurons. No increase in α-synuclein or ubiquitin levels or aggregation; however, cultured neurons developed more inclusions when exposed to exogenous α-synuclein fibrils. Decreased striatal dopamine content, decreased evoked release. Apparently normal behavior. No change in activity level or motor coordination at 12 months. Motor deficits appear at 18 months. Mutant Lrrk2 protein purified from mouse brain had increased kinase activity. Age-dependent electrophysiological changes in the hippocampus including increased basal synaptic efficiency though a postsynaptic mechanism and decreased LTD. Synaptic vesicle endocytosis impaired in cultured ventral midbrain neurons. Tg mice spend less time in the REM sleep phase. Available through The Jackson Lab, Stock# 012467, cryopreserved. Li et al., 2010 Yes
G2019S-LRRK2 (line 340), G2019S-LRRK2 transgenic, LRRK2 G2019S Tg Mouse (Dawson/Moore) B6;C3-Tg(PDGFB-LRRK2*G2019S)340Djmo/J Transgene introduced into C57BL/6J x C3H/HeJ embryos. Founder mice were bred with C57BL/6J mice. LRRK2 LRRK2 G2019S Transgenic mice overexpress full-length mutant human LRRK2 with the G2019S mutation. Transgene expression is driven by a hybrid CMVe-PDGFβ promoter. LRRK2: Transgenic Parkinson's Disease Age-dependent dopaminergic neuron degeneration in the substantia nigra, though reports are mixed. No reduction in striatal dopaminergic terminals or dopamine levels. Some reports of α-synuclein accumulation. Abnormal mitochondria in striatal neurons and microglia; accumulation of autophagic vacuoles. Evidence for activated striatal microglia and increased levels of CD68 and TNF-α in whole brain. Deterioration of Rotarod performance in 14- to 18-month-old mice. Muscle weakness observed on the hanging wire test by 8 months of age. No change in pre-pulse inhibition of the acoustic startle reflex. Anxiety/depression-like symptoms at 10-12 months. No differences in body weight or survival. Reduced serotonin levels in the hippocampus at 10-12 months. Age-dependent increase in 5-HT1a receptors in the hippocampus, amygdala, and dorsal raphe nucleus. Available through The Jackson Laboratory, Stock# 016575, Cryopreserved. Ramonet et al., 2011 Yes
LRRK2 R1441C knock-in, R1441C KI, RC KI, Lrrk2 R1441C-KI B6.Cg-Lrrk2tm1.1Shn/J The mutant colony was established in B6/129 F1 mice, but backcrossed to C57BL/6J wild-type mice for at least 12 generations. LRRK2 LRRK2: Knock-In Parkinson's Disease No loss of dopaminergic (TH+) neurons in the substantia nigra pars compacta at 12 and 22 months. No loss of TH-immunoreactive neurons in the locus coeruleus. Basal levels of striatal dopamine, DOPAC, and HVA were comparable between KI and wild-type mice at 3, 12, and 23 months. However, evoked dopamine release in the striatum was reduced in adult heterozygous KI mice. Acoustic startle reflex equal to wild-type mice at 12 months of age. Motor learning impaired upon antagonism of dopamine receptors (D1 and D2). Intracellular protein transport impaired. PKA activity is elevated in the striatum. Ciliation in striatal cholinergic neurons is decreased and primary cilia formation is perturbed in the somatosensory cortex. Available through The Jackson Laboratory, Stock# 009346, Cryopreserved or frozen embryo. Tong et al., 2009 Yes
R26-LRRK2; R26-LRRK2+/+, R26-LRRK2+/+/DAT-Cre mice, ROSA26-LRRK2(R1441C), R1441C LRRK2 Tg Mouse (Moore) STOCK Gt(ROSA)26Sortm1(LRRK2*R1441C)Djmo/J R26-LRRK2 mice were generated from 129/SvJ ES cells microinjected into C57Bl/6J mouse blastocysts. Chimeras were bred with C57Bl/6J mice and maintained on a mixed genetic background (129/SvJ and C57Bl/6J). BAC-DAT-iCRE mice were maintained on a C57Bl/6J background. LRRK2 LRRK2 R1441C Full-length human LRRK2 with the R1441C mutation was targeted to the endogenous ROSA26 locus by homologous recombination. Cre-mediated excision of the transcriptional termination sequence allows for transgene expression. The Cre line expressed Cre-recombinase driven by the dopamine transporter (DAT) promoter derived from a BAC construct containing the entire mouse DAT (slc6) gene. LRRK2: Transgenic Parkinson's Disease No neurodegeneration in the brain. No proteinaceous inclusions of α-synuclein, ubiquitin, or tau. No reactive gliosis. No change in dopamine levels. Subtle morphological abnormalities in neuronal nuclei, including altered nuclear envelope. No overt behavioral differences. Activity levels and Rotarod performance are normal into advanced age. Fertile. Normal survival. No overt olfactory deficits. Available through The Jackson Laboratory, Stock# 022793, Cryopreserved. Tsika et al., 2014 Yes
WT-OX, LRRK2 WT BAC, WT LRRK2 Mouse (BAC Tg), WT LRRK2 BAC Tg Mouse (Li) FVB/N-Tg(LRRK2)1Cjli/J BAC injected into fertilized FVB zygotes. Founder mice bred with FVB/N inbred mice for many generations, and then with FVB/NJ inbred mice. LRRK2 A 188 kb human bacterial artificial chromosome (BAC) containing the entire human LRRK2 gene, with 29 kb upstream and 42 kb downstream. LRRK2: Transgenic Parkinson's Disease Overtly normal brain structure. Intact, but shorter, neurites. Motor hyperactivity at 12 months of age. Available through The Jackson Laboratory, Stock# 009610, Cryopreserved. Li et al., 2009 Yes
PARK2-Q311X, Parkin-Q311X(A), Parkin-Q311X (line A), Parkin Q311X BAC Tg Mouse (Yang) FVB/NJ-Tg(Slc6a3-PARK2*Q311X)AXwy/J The BAC was microinjected into fertilized FVB/NJ zygotes, then bred to FVB/NJ inbred mice. PRKN (parkin) Parkin Q311X Bacterial artificial chromosome (BAC)-mediated expression of mutant parkin in dopaminergic neurons. The promoter and regulatory regions of Slc6a3 (dopamine transporter) drive expression of parkin with the truncation mutation Q311X. The protein has an N-terminal Flag-tag. Two copies of the transgene integrated in tandem. PRKN (parkin): Transgenic Parkinson's Disease Degeneration of dopaminergic neurons in the substantia nigra and nerve terminals in the striatum. Reduced dopamine in the striatum. Accumulation of proteinase-K resistant α-synuclein and oxidative protein damage. Late-onset hypoactivity (about 16 months of age), other modest changes in motor behavior and coordination in tests that included traversing a beam or removing adhesive. Available through The Jackson Laboratory, Stock# 009090, Live. Lu et al., 2009 Yes
Pink1-, Pink1-deficient mouse, PARK6 mouse, Pink1-/-, PINK1 G309D (PINK1-) KI Mouse (Auburger) B6;129-Pink1tm1Aub/J The vector was introduced into a 129/SvEV-derived embryonic stem cell line. Resulting chimeric mice were bred and maintained on a pure 129 background. PINK1 Pink1 G309D A targeting vector was used to introduce the G309D mutation into exon 4 at the orthologous mouse locus. A floxed neomycin cassette in the inverse orientation was inserted into intron 5 via homologous recombination. PINK1: Knock-In Parkinson's Disease No neuronal loss. No Lewy bodies or α-synuclein aggregates, but alpha-synuclein expression change in brainstem/midbrain. Low dopamine levels. Mitochondrial dysfunction (e.g., reduced ATP, reduced respiratory activity). Increase in factors involved in toll-like receptor signaling in the cerebellum, and increased astrocytic and microglial markers in the corticospinal tract and striatum. Reduced spontaneous locomotor activity in open-field test. No difference in strength or coordination. Electrophysiological abnormalities, including altered glutamergic activity in midbrain dopaminergic neurons and early hypersynchrony in motor cortex. Medium spiny neurons in striatal slice exhibit giant GABAergic currents. Hyperactive subthalamic nucleus neurons, indicated by spontaneous bursts instead of single spikes. Subtle alterations in gene expression in cerebellum, midbrain, and striatum. Available through The Jackson Laboratory, Stock# 013050, Cryopreserved. Gispert et al., 2009 Yes
Pink1 KO, Pink1 knockout mouse, Pink1-, Pink1-/-, PINK1 KO Mouse (Shen) B6.129S4-Pink1tm1Shn/J Congenic C57BL/6J. The construct was introduced into 129S4/SvJae-derived J1 embryonic stem cells, which were injected into C57BL/6 blastocysts. The resulting chimeric animals were crossed to generate homozygotes and then backcrossed to C57BL/6J for >7 generations. PINK1 A targeting vector containing a PGK-Neo cassette was used to disrupt exons 4 through 7 of the endogenous PINK1 gene. This creates a nonsense mutation at the beginning of exon 8; truncated RNA is degraded. PINK1: Knock-Out Parkinson's Disease Overtly normal brain structure. Normal numbers of dopaminergic neurons and tyrosine hydroxlase levels in substantia nigra at 8-9 months of age. Alterations in the dendrites of midbrain dopaminergic neurons and cultured cortical neurons. Altered shape, density, and movement of dendritic mitochondria in cultured primary neurons from embryonic mice. Reduced spontaneous locomotor activity and skill at 3-6 months. Modest vocalization deficits at 4-6 months. Heavier than wildtype mice at 5 months. Plasticity abnormalities: reduced LTP and absent LTD in response to high frequency stimulation; reversed by dopamine agonists. Abnormal rise in serum cytokines in response to exhaustive exercise which acutely stresses mitochondria. Available through The Jackson Laboratory, Stock# 017946; Live. Kitada et al., 2007 Yes
hm2α-SYN-39, HM2, hm2α-SYN, human doubly mutated α-synuclein, A30P/A53T aSyn Mouse (Tg), A30P/A53T aSyn Tg Mouse (Richfield), alpha-synuclein A30P/A53T Mouse (Tg), B6J(239) C57BL/6J-Tg(Th-SNCA*A30P*A53T)39Eric/J Transgene injected into C57/BL6 oocytes, then bred to C57/BL6J. SNCA SNCA A30P, SNCA A53T This model expresses a transgene encoding human α-synuclein with two mutations (A30P and A53T) driven by 9kb rat tyrosine hydroxylase (TH) promoter. SNCA: Transgenic Parkinson's Disease Progressively loss of dopaminergic neurons in the substantia nigra pars compacta, observed by 8.5 months. No α-synuclein inclusions. Morphological abnormalities in the dopaminergic system, including axonal and dendritic abnormalities, reduced dopamine concentration in the striatum. More active as young adults, then hypoactive compared to non-Tg. Also reduced motor coordination in old age as measured by the time to right from an inverted wire screen. No weight differences compared to non-Tg. Available through The Jackson Laboratory, Stock# 008239, Live. Richfield et al., 2002 Yes
G2-3(A53T), PrPsynA53T, A53TαS Tg mice (line G2-3), MoPrP-Huα-Syn(A53T), Hualpha-Syn(A53T), A53T aSyn Tg Mouse (Lee), alpha-synuclein A53T Mouse (Tg) B6.Cg-2310039L15RikTg(Prnp-SNCA*A53T)23Mkle/J. Formerly: B6.Cg-Tg(Prnp-SNCA*A53T)23Mkle/J Established as C3H/HeJ x C57BL6/J hybrids, then maintained by backcrossing to C57Bl6/J. SNCA SNCA A53T Transgene comprising human α-synuclein with the A53T mutation was inserted downstream of the mouse prion protein (PrP) promoter. SNCA: Transgenic Parkinson's Disease No overt neuronal loss. Alterations in dopaminergic-associated proteins in the striatum, substantia nigra, and nucleus accumbens. Region-specific neuronal accumulation of fibrillar α-synuclein, ubiquitin, and neurofilament-H, and accompanying astrocytosis. Early hyperactivity followed by severe motor impairment, manifesting as wobbling, posturing, decreased spontaneous locomotor behavior, inability to navigate the Rotarod, and ultimately paralysis and death. At 11–12 months, spatial memory impaired as assessed by the Barnes circular maze. Premature death, defects in hippocampal synaptic function. Available through The Jackson Laboratory, Stock# 006823, Live. Lee et al., 2002 Yes
PAC-Tg(SNCA-A53T) +/+; Snca-/-, dbl-PAC-Tg(SNCAA53T);Snca-/-, A53T aSyn Tg Mouse (Nussbaum), Alpha-synuclein A53T Mouse (Tg) on SNCA KO FVB;129S6-Sncatm1Nbm Tg(SNCA*A53T)1Nbm Tg(SNCA*A53T)2Nbm/J The PAC transgene was injected into FVB/N oocytes and founder mice bred to FVB/N. The knockout mice were made in a 129S6/SvEvTac background. SNCA SNCA A53T In this double-transgenic model, one parental line expresses mutant human α-synuclein (A53T mutation) via a 146 kb P1 artificial chromosome (PAC) containing full-length human SNCA gene and 34 kb upstream sequence. The other line is an Snca knockout line generated by replacing exons 4 and 5 with a neomycin resistance cassette. SNCA: Transgenic; SNCA: Knock-Out Parkinson's Disease No loss of dopaminergic neurons in the substantia nigra by 18 months of age. Rare dystrophic synapses in the hippocampus at advanced age, but no Lewy body-like pathology or α-synuclein aggregation in the brain. No change in striatal dopamine concentration. Impaired performance on the Rotarod and reduced spontaneous locomotor activity in open-field test. Gastrointestinal dysfunction (e.g., reduced fecal mass, reduced colonic motility, prolonged whole-gut transit time). α-synuclein–positive aggregates in enteric nervous system. No difference in body weight. No olfactory deficits. No difference in autonomic regulation of heart rate. Available through The Jackson Laboratory, Stock #010799; Live. Kuo et al., 2010, Cabin et al., 2002 Yes
Snca KO, Snca -/-, Snca KO Mouse (Nussbaum), Alpha-synuclein KO Mouse Inbred 129/SvEvTac background. SNCA The mouse Snca gene was disrupted using a targeting vector that replaced exons 4 and 5 with the neomycin resistance gene. SNCA: Knock-Out Parkinson's Disease No gross brain abnormalities. Electron microscopy revealed synaptic vesicle abnormalities in hippocampal neurons, i.e., fewer vesicles in the reserve pool. Behavior is largely normal. Normal performance on the Rotarod. Subtle differences in locomotor activity (e.g., less rearing) but normal overall distance travelled. Learning and memory appear intact. Possible anxiety-like phenotype. Microglial abnormalities. Reduced cardiolipin content in the brain. Some mitochondrial abnormalities. Unknown. Cabin et al., 2002 Yes
Sncaflox(neo), SNCAflox delta neo, Snca conditional knockout, SNCA KO Mouse (Buchman), Alpha-synuclein KO Mouse (Conditional) B6(Cg)-Sncatm1.1Vlb/J C57BL/6J SNCA A targeting vector containing a FRT site-flanked neomysin cassette and a loxP site was inserted downstream of exon 2 and another loxP site was inserted upstream of exon 2. Flp-mediated recombination removed the FRT-flanked neo cassette, leaving exon 2 floxed. When bred to Cre-expressing mice, the resulting offspring have the first coding exon deleted in Cre-expressing tissues. SNCA: Conditional Knock-out Parkinson's Disease No data. No data. Available through The Jackson Laboratory, Stock #025636, Live. Ninkina et al., 2015, Roman et al., 2017 Yes
mThy1-α-synuclein, mThy-1 tg, Tg(Thy1-SNCA)61Ema B6;DBA-Tg(Thy1-SNCA)61Ema (C57BL/6 x DBA/2)F1 (strain of origin) SNCA This transgenic mouse expresses human wild-type α-synuclein under the control of the mouse Thy1 promoter (Rockenstein et al., 2002). The transgene is inserted in the X chromosome. SNCA: Transgenic Parkinson's Disease Available through Eliezer Masliah, Robert Rissman for academic research; others please contact the Office of Innovation and Commercialization at the University of California, San Diego. The CRO QPS Austria offers research services with this model. Rockenstein et al., 2002 Yes

21 Visualizations

Phenotypes Examined

  • Neuronal Loss
  • Dopamine Deficiency
  • α-synuclein Inclusions
  • Neuroinflammation
  • Mitochondrial Abnormalities
  • Motor Impairment
  • Non-Motor Impairment

When visualized, these phenotypes will distributed over a 18 month timeline demarcated at the following intervals: 3mo, 6mo, 9mo, 1yr, 15mo, 18mo+.

DJ-1 KO Rat

Observed
  1. X
    Non-Motor Impairment at 17

    Olfactory detection enhanced (16 mos). Short-term memory abnormal (4.5, 15 mos). Appetitive instrumental learning normal (4, 6, 8 mos). Coping behavior (forced-swim test) impaired (6 mos). No anxiety-like behavior (elevated plus maze; 4, 8, 17 mos), less anxiety on light-dark box (6, 8 mos). No sucrose preference at 9 mos. Sensorimotor function (adhesive removal) unaffected (4, 7, 13 mos).

  2. X
    Motor Impairment at 17

    Abnormalities in gait and strength, vocalizations, and tongue movements were observed. By 4 months, the rats exhibited abnormal paw positioning and a shorter stride. Males showed impaired licking, longer and more frequent ultrasonic vocalizations, and an accelerated decrease in average call intensity with age. Fine motor skills were also impaired in KO versus wild-type rats by 7 months of age.

  3. X
    Mitochondrial Abnormalities at 13

    At 3 months of age, the mitochondrial proteome in DJ-1 KO rats was differentially expressed compared to wild-type rats. Mitochondrial respiration was also increased in KO versus wild-type rats

  4. X
    Neuronal Loss at 26

    Age-related decreases in TH-positive dopaminergic neurons were reported in the substantia nigra and locus coeruleus reaching approximately 50 percent by 8 months of age. No change was found in TH-immunoreactivity in the ventral tegmental area or striatum.

Absent
  • Dopamine Deficiency at

    Striatal dopamine level was increased 2-3 fold in KO rats compared to wild-type levels at 8 months of age. Dopaminergic innervation of the dorsal striatum was intact in DJ-1 KO rats at 4 and 6 months of age compared to wild-type rats. Basal levels of dopamine metabolites and evoked levels of dopamine in the striatum were not different between KO and wild-type rats.

  • α-synuclein Inclusions at

    Staining for α-synuclein revealed no increase in the striatum or in any other brain region assessed.

No Data
  • Neuroinflammation at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
Park7 (DJ1) Park7 (DJ1): Knock-Out Parkinson's Disease

Age-related decrease in dopaminergic neurons in the substantia nigra and locus coeruleus; approximately 50 percent reduction by 8 months. Striatal dopamine and serotonin levels elevated 2-3fold over wild-type levels. No change was found in TH-immunoreactivity in the ventral tegmental area or striatum.

Enhanced olfactory detection, abnormal short-term memory, impaired coping behavior, reduced anxiety on light-dark box. Abnormalities in gait, paw positioning, strength, vocalizations, and tongue movements. In males, impaired licking, longer and more frequent ultrasonic vocalizations, and accelerated decrease in average call intensity with age.

expand

LRRK2 R1441C KI Mouse

Observed
  1. X
    Non-Motor Impairment at 30

    Acoustic startle reflex equal to wild-type mice at 12 months of age. Intracellular protein transport impaired in primary cultured cells. PKA activity is elevated in the striatum. Ciliation in striatal cholinergic neurons is decreased at 7 months of age and primary cilia formation is perturbed in the somatosensory cortex.

Absent
  • Dopamine Deficiency at

    Basal levels of striatal dopamine, DOPAC, and HVA were comparable between KI and wild-type mice at 3, 12, and 23 months of age. However, evoked dopamine release in the striatum was reduced in adult heterozygous KI mice.

  • α-synuclein Inclusions at

    No abnormal accumulation of α-synuclein observed at 3, 12, and 22 months of age in the substantia nigra pars compacta or locus coeruleus.

  • Neuroinflammation at

    GFAP immunoreactivity was normal at 12 and 22 months of age. However, upon α-synuclein fibril injection, KI mice exhibited increased infiltration of pro-inflammatory monocytes into the brain.

  • Motor Impairment at

    Spontaneous locomotor activity (open-field test) equal to wild-type mice at 3, 12, and 24 months of age. Involuntary motor movement (Rotarod) equal to wild-type mice 3 and 12 months of age.

  • Neuronal Loss at

    No loss of dopaminergic (TH-immunoreactive) neurons in the substantia nigra pars compacta at 12 and 22 months of age. No loss of TH-immunoreactive neurons in the locus coeruleus.

No Data
  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
LRRK2 LRRK2: Knock-In Parkinson's Disease

No loss of dopaminergic (TH+) neurons in the substantia nigra pars compacta at 12 and 22 months. No loss of TH-immunoreactive neurons in the locus coeruleus. Basal levels of striatal dopamine, DOPAC, and HVA were comparable between KI and wild-type mice at 3, 12, and 23 months. However, evoked dopamine release in the striatum was reduced in adult heterozygous KI mice.

Acoustic startle reflex equal to wild-type mice at 12 months of age. Motor learning impaired upon antagonism of dopamine receptors (D1 and D2).

expand

LRRK2 G2019S KI Mouse

Observed
  1. X
    Non-Motor Impairment at 16

    Altered responses to social-defeat stress (males, 3-4 mos) which correlated with changes in striatal plasticity and intrinsic membrane excitability. Attention deficits, slower information processing, impaired goal-directed learning in 2-6-month-old male KI mice, but cognitive flexibility and novel objective recognition are intact (2-6 mos). Perturbed sleep behavior at 8-10 mos.

  2. X
    Motor Impairment at 81

    A battery of motor tests revealed no baseline deficits at 3-4, 12-13, and 18-19 months of age. However, an increased locomotor response after amphetamine challenge is observed at 18 months. Motor defects are exacerbated following a manganese stressor.

Absent
  • Dopamine Deficiency at

    Striatal dopamine levels do not differ at 2 months of age, and neither do tyrosine hydroxylase levels in the substantia nigra.

  • Neuronal Loss at

    The cytoarchitecture of the neocortex, striatum, hippocampus, and elsewhere is normal in Nissl-stained brain sections of 3-4 month-old mice, and striatal levels of tyrosine hydroxylase are similar to those of controls at P21. In another study, tyrosine hydroxylase levels are reduced in the striatum and midbrain at 2 months of age.

No Data
  • α-synuclein Inclusions at

    No data.

  • Neuroinflammation at

    No data.

  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
LRRK2 LRRK2: Knock-In Parkinson's Disease

Levels of phospho-substrates of LRRK2 (e.g., Rab10) are increased in the brain. Endocytosis and axonal transport defects in neurons. Cholinergic innervation density is lower in the prelimbic/infralimbic cortical areas and dorsomedial striatum, but not in the dorsal lateral geniculate nucleus in 2-6-month-old males. Microglial immunostaining is similar in the striatum and midbrain at 8 weeks

Attention deficits, slower information processing speeds, and impaired goal-directed learning are evident in 2-6-month-old mice—deficits rescued by systemic administration of the acetylcholinesterase inhibitor donepezil. Cognitive flexibility and novel objective recognition similar to controls. Sleep behavior is perturbed at 8-10 months of age.

expand

LRRK2 G2019S Mouse (BAC Tg)

Observed
  1. X
    Dopamine Deficiency at 52

    Age-related decline in striatal dopamine content. Levels were decreased at 12 months of age, but not significantly different from controls at 6 months of age. Also, decreased dopamine metabolite homovanillic acid (HVA).

  2. X
    Non-Motor Impairment at 26

    Tg mice spend less time in the REM sleep phase at 12 and 18 months of age. Age-dependent increase in plasma corticosterone (present starting at 6-8 months of age). Nuclear envelope integrity is perturbed in dopaminergic neurons at 12 months.

  3. X
    Neuroinflammation at 8

    Application of α-synuclein fibrils leads to exacerbated responses (more inclusions and greater infiltration of pro-inflammatory monocytes).

  4. X
    Motor Impairment at 78

    Behavior in hemizygous mice was comparable to littermate controls in terms of activity levels (open-field test) and coordination (beam-walk test) at 6 and 12 months, , but not at 18 months of age, when Tg mice develop motor deficits (Rotarod).

  5. X
    Mitochondrial Abnormalities at 0

    Primary cultured cells from Tg mice exhibit mitochondrial fragmentation and membrane depolarization.

Absent
  • α-synuclein Inclusions at

    No evidence of α-synuclein inclusions up to 18 months of age. However, there is mixed evidence on whether cultured mutant hippocampal neurons have increased levels of α-synuclein protein. After exposure to exogenous α-synuclein fibrils, mutant neurons developed more α-synuclein inclusions than non-Tg neurons.

  • Neuronal Loss at

    No evidence of neuronal or other cell death in any brain region, including the cortex, striatum, and hippocampus. There was no difference in the number of dopaminergic neurons in the substantia nigra compared to littermate controls at 6 or 12 months.

No Data
Genes Mutations Modification Disease Neuropathology Behavior/Cognition
LRRK2 LRRK2 G2019S LRRK2: Transgenic Parkinson's Disease

Brain appears normal. No neuronal or cell death at 12 months. Impaired neurite motility and synaptic vesicle endocytosis in cultured neurons. No increase in α-synuclein or ubiquitin levels or aggregation; however, cultured neurons developed more inclusions when exposed to exogenous α-synuclein fibrils. Decreased striatal dopamine content, decreased evoked release.

Apparently normal behavior. No change in activity level or motor coordination at 12 months. Motor deficits appear at 18 months.

expand

LRRK2 G2019S Mouse (Tg)

Observed
  1. X
    Non-Motor Impairment at 45

    Anxiety/depression-like symptoms were observed at 10-12 months of age.

  2. X
    α-synuclein Inclusions at 52

    Around 2 years of age, mice did not exhibit abnormalities in α-synuclein in the ventral midbrain, striatum, or cerebral cortex. However, one study found α-synuclein accumulation in whole brain lysates of 12- to 19-month-old transgenic mice.

  3. X
    Neuroinflammation at 63

    Around 2 years of age, mice did not have GFAP abnormalities in the ventral midbrain, striatum, or cerebral cortex. However, activated microglia were reported in the striatum at 14 months, and CD68 and TNF-α levels were increased in whole brains at 4-6 months. Others have not observed differences in Iba-1 staining (microglial marker) at 6, 12, or 18 months in the striatum or substantia nigra.

  4. X
    Motor Impairment at 35

    Rotarod performance deteriorated in 14- to 18-month-old mice, but minor deficits are already observed as early as 8 months of age. Muscle weakness observed on the hanging wire test by 8 months of age. No change in pre-pulse inhibition of the acoustic startle reflex.

  5. X
    Mitochondrial Abnormalities at 63

    Increased numbers and condensation of mitochondria in striatal microglia were reported at 14 months. Abnormally high levels of condensed mitochondria were also observed in cortical and striatal neurons at 17-18 months.

  6. X
    Neuronal Loss at 83

    By 19-21 months, mice lose 18 percent of TH-positive dopaminergic neurons in the substantia nigra pars compacta and 14 percent of dopaminergic dendrites in the substantia nigra pars reticulata. At 1-2 months neuronal numbers were normal. Some authors do not see differences in TH staining up to 2 years of age. No abnormal neuronal loss is observed in the ventral tegmental area or cerebellum.

Absent
  • Dopamine Deficiency at

    At 14-15 months of age, hemizygous mice had normal levels of striatal dopamine, DOPAC, and HVA. However, in the olfactory bulb, levels of HVA and DOPAC were lower, but dopamine was unchanged.

No Data
Genes Mutations Modification Disease Neuropathology Behavior/Cognition
LRRK2 LRRK2 G2019S LRRK2: Transgenic Parkinson's Disease

Age-dependent dopaminergic neuron degeneration in the substantia nigra, though reports are mixed. No reduction in striatal dopaminergic terminals or dopamine levels. Some reports of α-synuclein accumulation. Abnormal mitochondria in striatal neurons and microglia; accumulation of autophagic vacuoles. Evidence for activated striatal microglia and increased levels of CD68 and TNF-α in whole brain.

Deterioration of Rotarod performance in 14- to 18-month-old mice. Muscle weakness observed on the hanging wire test by 8 months of age. No change in pre-pulse inhibition of the acoustic startle reflex. Anxiety/depression-like symptoms at 10-12 months.

expand

LRRK2 G2019S Rat (BAC Tg)

Observed
  1. X
    Non-Motor Impairment at 26

    Bone marrow myeloid progenitor numbers were decreased, but suppressive myeloid cells were increased at 6 to 11 months of age

  2. X
    Motor Impairment at 35

    Mild abnormalities in motor behavior. Slightly more postural instability at 8 months of age (but not at 4 and 12 months). Slightly more rearing events at 12 months, but not at younger ages.

Absent
  • Dopamine Deficiency at

    No change in striatal dopamine levels. No change in 3,4-dihydroxyphenylacetic acid (DOPAC) levels. No change in the rate of dopamine turnover. At 12 months of age Tg rats exhibited higher levels of striatal homovanillic acid (HVA).

  • α-synuclein Inclusions at

    Under basal conditions no α-synuclein inclusions were observed. 

  • Neuroinflammation at

    No increase in Iba-1 positive microglia or GFAP-positive astrocytes in the substantia nigra at 12 months of age. However, iNOS expression was elevated in nigral dopaminergic neurons.

  • Neuronal Loss at

    No overt loss of dopaminergic neurons in the substantia nigra out to 12 months of age.

No Data
  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
LRRK2 LRRK2 G2019S LRRK2: Transgenic Parkinson's Disease

No overt neurodegeneration out to 12 months of age. Elongated dopaminergic neurons. Elevated oxidative and nitrosative stress. No evidence of gliosis. No α-synuclein inclusions until challenged with exogenous α-synuclein. No change in dopamine levels.

Mild abnormalities in motor behavior. Slightly more postural instability at 8 months of age (but not at 4 and 12 months). Slightly more rearing events at 12 months, but not at younger ages.

expand

Lrrk2 KO Rat

Observed
  1. X
    Non-Motor Impairment at 5

    Abnormalities occur in peripheral organs, most notably the kidney, but also the liver, lung, and spleen. Changes are progressive, although they do not appear to shorten lifespan. The earliest reported alterations occur in the kidneys at 1 month of age.

Absent
  • Dopamine Deficiency at

    Basal levels of dopamine metabolites (3,4-dihydroxyphenylacetic and homovanillic acid) do not differ between Lrrk2 KO and wild-type rats at 4, 8, and 12 months of age. Evoked release of dopamine also does not differ between KO and wild-type rats.

  • Neuroinflammation at

    When challenged with LPS or α-synuclein overexpression, Lrrk2 KO rats show lower levels of pro-inflammatory CD68-positive myeloid cells in the substantia nigra than wild-type rats.

  • Motor Impairment at

    Assessment of Rotarod performance revealed no impairment at 12 months of age compared with wild-type rats.

  • Neuronal Loss at

    Under basal conditions, the number of TH-positive cells in the substantia nigra is comparable between Lrrk2 KO and wild-type rats. When challenged with LPS or α-synuclein overexpression, Lrrk2 KO rats develop significantly less neurodegeneration in the substantia nigra than wild-type rats.

No Data
  • α-synuclein Inclusions at

    No data.

  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
Lrrk2 Lrrk2: Knock-Out Parkinson's Disease

Not observed. Protection against dopaminergic cell loss under conditions involving LPS or α-synuclein overexpression in the substantia nigra. No changes in basal or evoked release of dopamine.

One assessment of Rotarod performance revealed no impairment at 12 months of age compared with wild-type rats.

expand

LRRK2 R1441C Mouse (Tg - Conditional)

Observed
Absent
  • Dopamine Deficiency at

    HPLC analysis of striata from 10-month-old mice revealed no significant differences in the levels of dopamine or its metabolites 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA).

  • Non-Motor Impairment at

    Olfactory function, as assessed by the ability to locate buried food, was normal out to 20 months of age.

  • α-synuclein Inclusions at

    Immunohistochemical analysis of the brain at 22 months did not reveal abnormalities in α-synuclein, and no proteinaceous inclusions were seen.

  • Neuroinflammation at

    Immunohistochemical analysis of the brain at 22 months found GFAP and Iba1 immunoreactivity comparable to control levels.

  • Motor Impairment at

    Around 20 months of age, R26-LRRK2 mice behaved normally, exhibiting no deficits in locomotor activity (open-field test), motor coordination (Rotarod), or gait (digital catwalk system).

  • Neuronal Loss at

    In the substantia nigra pars compacta, there was no difference in the number of tyrosine hydroxylase(TH)-positive neurons or the total number of Nissl-positive neurons at 12 and 22 months.

No Data
  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
LRRK2 LRRK2 R1441C LRRK2: Transgenic Parkinson's Disease

No neurodegeneration in the brain. No proteinaceous inclusions of α-synuclein, ubiquitin, or tau. No reactive gliosis. No change in dopamine levels. Subtle morphological abnormalities in neuronal nuclei, including altered nuclear envelope.

No overt behavioral differences. Activity levels and Rotarod performance are normal into advanced age.

expand

LRRK2 WT Mouse (BAC Tg)

Observed
  1. X
    Dopamine Deficiency at 52

    Striatal dopamine levels, as measured by PET imaging with [18F]FDOPA uptake, are higher in WT-OX versus non-Tg mice.

  2. X
    Motor Impairment at 52

    WT-OX mice (12 months) are hyperactive on several parameters of the open-field test. Gait analysis (Cat-Walk system) was also perturbed relative to non-Tg controls. However, the number of rears did not differ.

Absent
  • Mitochondrial Abnormalities at

    Mitochondrial morphology and levels of proteins involved in mitochondrial fission (Drp1 and Fis1) are normal at 12 months of age.

No Data
  • Non-Motor Impairment at

    No data.

  • α-synuclein Inclusions at

    No in vivo data, but α-syn colocalization with LAMP-2 is increased in cultured neurons from WT-OX mice.

  • Neuroinflammation at

    No data.

  • Neuronal Loss at

    No data on neuron numbers are available, but neurite length is reduced in primary hippocampal neurons and primary nigral tyrosine hydroxylase-positive neurons of WT-OX mice versus non-Tg mice.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
LRRK2 LRRK2: Transgenic Parkinson's Disease

Overtly normal brain structure. Intact, but shorter, neurites.

Motor hyperactivity at 12 months of age.

expand

Parkin KO Rat

Observed
  1. X
    Non-Motor Impairment at 9

    Orientation to an olfactory stimulus was normal. At 2 months of age, male KO rats had a greater preference for methamphetamine than wild-type rats based on self-administration and place preference tests.

  2. X
    Mitochondrial Abnormalities at 14

    Alterations in mitochondrial protein expression in synaptic and nonsynaptic striatal samples of 3-month-old KO rats.

  3. X
    Neuronal Loss at 35

    A small, non-significant reduction in dopaminergic neurons was observed in the substantia nigra at 8 months of age.

Absent
  • Dopamine Deficiency at

    No differences in striatal dopamine levels at 4, 6, or 8 months. Altered dopaminergic transmission factors in the striata, including MAO, β-phenylethylamine, trace amine-associated receptor 1, and postsynaptic dopamine D2 receptors in 2-month-old KO rats. Striatal dopamine metabolite levels decreased with age in KO rats, showing lower levels at 12 months than at 8 months.

  • α-synuclein Inclusions at

    There was no increase in α-synuclein protein in the striatum or any other brain region assessed.

  • Motor Impairment at

    No behavioral deficits were detected at 4, 6, and 8 months of age. Motor functioning, including performance on the Rotarod, was intact. However, at 2 months, male KO rats made fewer small stereotypic movements, such as scratching and grooming, than wild-type controls.

No Data
  • Neuroinflammation at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
Park2 Park2: Knock-Out Parkinson's Disease

No significant changes in dopaminergic neurons in the substantia nigra nor striatal dopamine levels, but alterations in dopaminergic signaling were detected at an early age, as were disruptions in mitochondrial protein expression in striatum. No increase in α-synuclein. Evoked release of striatal glycine greater at 12 months versus wild-type rats.

No behavioral deficits detected at 4, 6, and 8 months of age. However, at 2 months, male KO rats made fewer small stereotypic movements, such as scratching and grooming, than wild-type controls. At 2 months, male KO rats had a greater preference for methamphetamine than wild-type rats.

expand

Parkin Q311X Mouse (BAC Tg)

Observed
  1. X
    Dopamine Deficiency at 83

    Surviving nigral neurons had reduced tyrosine hydroxylase expression. By 19-21 months, striatal concentrations of dopamine and the dopamine metabolite, 3,4-dihydrooxyphenylacetic acid (DOPAC), were decreased compared with non-Tg littermates.

  2. X
    Motor Impairment at 70

    Behavior was fairly normal at 3 months, but motor abnormalities were measured by 16 months of age, including hypoactivity and deficits in coordination and in motor response to sensory stimuli.

  3. X
    Neuronal Loss at 70

    Progressive loss of dopaminergic neurons in the substantia nigra. About 40 percent loss by 16 months of age with a corresponding decrease in dopaminergic projections to the striatum. Neurons in the ventral tegmental area were relatively spared.

Absent
  • α-synuclein Inclusions at

    Inclusions were not observed at any age, however, the mice exhibit age-dependent accumulation of proteinase-K resistant endogenous α-synuclein in the substantia nigra.

No Data
  • Non-Motor Impairment at

    No data.

  • Neuroinflammation at

    No data.

  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
PRKN (parkin) Parkin Q311X PRKN (parkin): Transgenic Parkinson's Disease

Degeneration of dopaminergic neurons in the substantia nigra and nerve terminals in the striatum. Reduced dopamine in the striatum. Accumulation of proteinase-K resistant α-synuclein and oxidative protein damage.

Late-onset hypoactivity (about 16 months of age), other modest changes in motor behavior and coordination in tests that included traversing a beam or removing adhesive.

expand

PINK1 G309D (PINK1-/-) Mouse (KI)

Observed
  1. X
    Dopamine Deficiency at 39

    Decreased dopamine concentration in the striatum by 9 months of age.

  2. X
    Neuroinflammation at 81

    Expression of factors involved in toll-like receptor signaling increased in the cerebellum, as did astrocytic and microglial markers in the corticospinal tract and striatum at 18 months.

  3. X
    Motor Impairment at 70

    At 16 months of age Pink1-/- mice exhibited decreased spontaneous locomotor activity. Strength and coordination were intact.

  4. X
    Mitochondrial Abnormalities at 13

    By 3 months of age the mice exhibited a mitochondrial import defect. This phenotype was more severe at 6 months and import was reduced nearly 50% by 12 months of age. By 6 months, ATP production, respiration, and mitochondrial membrane potential were also reduced.

Absent
  • Non-Motor Impairment at

    Mutant mice performed similarly to wild-type mice in tests assessing the startle reflex, sweating, and anxiety.

  • α-synuclein Inclusions at

    No Lewy body-like inclusions of α-synuclein aggregates in the brainstem or substantia nigra, but the expression levels of alpha-synuclein are altered in brainstem/midbrain.

  • Neuronal Loss at

    Neuronal loss was not observed at 18 months of age (total neuronal population and TH-positive subset).

No Data
Genes Mutations Modification Disease Neuropathology Behavior/Cognition
PINK1 Pink1 G309D PINK1: Knock-In Parkinson's Disease

No neuronal loss. No Lewy bodies or α-synuclein aggregates, but alpha-synuclein expression change in brainstem/midbrain. Low dopamine levels. Mitochondrial dysfunction (e.g., reduced ATP, reduced respiratory activity). Increase in factors involved in toll-like receptor signaling in the cerebellum, and increased astrocytic and microglial markers in the corticospinal tract and striatum.

Reduced spontaneous locomotor activity in open-field test. No difference in strength or coordination.

expand

PINK1 KO Mouse

Observed
  1. X
    Motor Impairment at 23

    Reduced spontaneous locomotor activity and skill reported at 3-6 months, as well as modest vocalization deficits at 4-6 months.

  2. X
    Mitochondrial Abnormalities at 0

    Altered shape, density, and movement of dendritic mitochondria were observed in cultured primary neurons from embryonic mice. Also, an abnormal rise in serum cytokines  in response to acute mitochondrial stress was reported in vivo.

Absent
  • Dopamine Deficiency at

    Overall striatal levels of dopamine did not significantly differ from levels in wild-type mice at two to three months or eight to nine months of age.

  • Neuronal Loss at

    No decrease in the number of dopaminergic neurons in the substantia nigra at 2-3 months or 8-9 months of age. Neuronal morphology also grossly intact.

No Data
  • Non-Motor Impairment at

    No data.

  • α-synuclein Inclusions at

    No data.

  • Neuroinflammation at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
PINK1 PINK1: Knock-Out Parkinson's Disease

Overtly normal brain structure. Normal numbers of dopaminergic neurons and tyrosine hydroxlase levels in substantia nigra at 8-9 months of age. Alterations in the dendrites of midbrain dopaminergic neurons and cultured cortical neurons. Altered shape, density, and movement of dendritic mitochondria in cultured primary neurons from embryonic mice.

Reduced spontaneous locomotor activity and skill at 3-6 months. Modest vocalization deficits at 4-6 months.

expand

Pink1 KO Rat

Observed
  1. X
    Non-Motor Impairment at 9

    Nociception alterations in male KO rats observed at 6 to 10 months of age, indicating thermal hyperalgesia. This effect was present in female KO rats at 2 months of age, but not at older ages. Abnormalities in ventilation frequency were also observed in male KO rats. Defects in ultrasonic vocalizations starting at 2 months of age in male and female KO rats.

  2. X
    α-synuclein Inclusions at 18

    Alpha-synuclein aggregates were found as early as 4 months of age and increased in number up to 12 months. Areas affected include the periaqueductal gray, substantia nigra pars compacta, locus coeruleus, nucleus ambiguous, cortex, thalamus, and striatum.

  3. X
    Motor Impairment at 5

    Abnormalities in gait, coordination, and strength. As early as 5 weeks of age, KOs had increased foot slips on the tapered balance beam, at 7 weeks they showed hind limb fatigue, which progressed to hind limb dragging, and by 2 months they exhibited alterations in oromotor behaviors. Deficits in gait may be transient.

  4. X
    Mitochondrial Abnormalities at 18

    Alterations in mitochondrial metabolites and mitochondrial protein expression were reported as early as 4 months of age in cortex and striatum. Oxygen consumption rates were elevated in striatal mitochondria isolated from 9-month-old rats, but not in non-synaptic samples from 3-month-old rats.

  5. X
    Neuronal Loss at 11

    Age-related decrease in tyrosine hydroxylase (TH)-positive dopaminergic neurons in the substantia nigra. A reduction of 25 and 50 percent at 6 months and 8 months, respectively. Deficits in TH staining in the substantia nigra have been observed as early as at 2.5 months of age. While some studies did not see any changes in TH-positive cells in the striatum, others have observed a 15% loss.

Absent
  • Dopamine Deficiency at

    One study found striatal dopamine levels were increased two- to threefold in Pink1 KO rats compared with wild-type levels at 8 months of age, whereas another reported a slight decrease at this age. In the dorsal striatum, KO rats have age-dependent differences in basal and evoked dopamine levels, but no differences were observed compared to wild-type rats.

No Data
  • Neuroinflammation at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
Pink1 Pink1: Knock-Out Parkinson's Disease

Age-related decrease in dopaminergic neurons in the substantia nigra; greater than 50 percent reduction at eight months. Alterations in striatal dopamine and serotonin levels. Progressive increase in α-synuclein aggregates and reduced brain volume across numerous regions. Increased ventricular volume. Alterations in cortical and striatal mitochondria.

Abnormalities in gait, coordination, and strength. As early as 5 weeks of age, KOs had increased foot slips on the tapered balance beam, at 7 weeks they showed hind limb fatigue, which progressed to hind limb dragging, and by 2 months they exhibited alterations in oromotor (lingual) behaviors.

expand

Thy1-αSyn “Line 61” Mouse

Observed
  1. X
    Dopamine Deficiency at 63

    Line 61 mice lose striatal dopamine progressively, starting at about 14 months of age. Between approximately 6 and 10 months, however, the neurotransmitter’s extracellular levels are transiently increased.

  2. X
    Non-Motor Impairment at 11

    Disruptions in olfaction, circadian rhythms, sleep, cognition, social behavior, and autonomic function have been reported. Several are reminiscent of PD non-motor impairments. Alterations in olfaction, circadian rhythms, and the autonomic regulation of heartrate occur as early as 3 months of age.

  3. X
    α-synuclein Inclusions at 4

    Proteinase K-resistant aggregates are seen at 1 month and increase with age, including the substantia nigra, periqueductal gray, cortex, striatum, vagus, olfactory bulb, thalamus, locus coeruleus, and cerebellum, as well as cholinergic neurons in the colon. Elevated levels of phospho-serine 129 α-synuclein are found in the substantia nigra, striatum, cortex, frontal cortex, and hippocampus.

  4. X
    Neuroinflammation at 5

    Neuroinflammation markers have been seen in the cortex, striatum, substantia nigra, and hippocampus. The time course and profile vary between brain regions and some features remain subject to debate. However, neuroinflammation appears to affect the striatum first (1 month), and then the substantia nigra (5–6 months). At older ages, particularly in the substantia nigra, inflammation attenuates.

  5. X
    Motor Impairment at 4

    Impairments in balance, coordination, muscle strength, fine motor skills, vocalizations, and stress-induced defecation arise between 1 and 3 months. Transient hyperactivity is seen between 4 and 9 months of age, followed by hypoactivity and sensorimotor deficits at about 15 months. As the phenotype becomes more severe, the mice develop difficulty eating, akinesia, and hunched posture.

  6. X
    Mitochondrial Abnormalities at 18

    The functions of mitochondrial respiratory complexes in midbrain and striatum are impaired, with the earliest reported deficit affecting complex I in the midbrain at 4 months. Elevated α-synuclein accumulation was found in mitochondria of the midbrain, striatum, and cortex.

  7. X
    Neuronal Loss at 16

    Although neuron loss occurs in the neocortex and hippocampal CA3 region as early as 3–4 months of age, there is no reduction in the PD-relevant dopaminergic neurons of the substantia nigra, even at 22 months of age.

Absent
No Data
Genes Mutations Modification Disease Neuropathology Behavior/Cognition
SNCA SNCA: Transgenic Parkinson's Disease

expand

α-synuclein A30P/A53T Mouse (Tg)

Observed
  1. X
    Dopamine Deficiency at 9

    Striatal dopamine concentrations were lower at all ages tested, including the earliest age, 2-3 months. Dopamine concentrations dropped with age, and levels of metabolites (e.g., DOPAC and HVA) were also lower in HM2 mice than non-Tg by 13-23 months of age.

  2. X
    Motor Impairment at 30

    At young age 2-3 months, HM2 mice were more active than non-Tg controls, but by middle age (7-9 months) they were less active. At advanced ages (13-23 months), they also exhibited impaired coordination as measured by the time it took to right themselves from an inverted wire screen. However, no deficiencies in Rotarod performance, grip strength, or open-field movements were detected at 6 months.

  3. X
    Neuronal Loss at 34

    Progressive loss of dopaminergic neurons was reported in the substantia nigra pars compacta (19 percent reduction at 8.5 months and 55 percent at 19 months).

Absent
  • α-synuclein Inclusions at

    Inclusions were not observed at any age. Diffuse α- synuclein protein was both cytoplasmic and nuclear.

No Data
  • Non-Motor Impairment at

    No data.

  • Neuroinflammation at

    No data.

  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
SNCA SNCA A30P, SNCA A53T SNCA: Transgenic Parkinson's Disease

Progressively loss of dopaminergic neurons in the substantia nigra pars compacta, observed by 8.5 months. No α-synuclein inclusions. Morphological abnormalities in the dopaminergic system, including axonal and dendritic abnormalities, reduced dopamine concentration in the striatum.

More active as young adults, then hypoactive compared to non-Tg. Also reduced motor coordination in old age as measured by the time to right from an inverted wire screen.

expand

α-synuclein A53T Mouse (Tg)

Observed
  1. X
    Dopamine Deficiency at 23

    In symptomatic mice, striatal dopamine and metabolites DOPAC and HVA are comparable to wildtype, but at 5 months, striatal tyrosine hydroxylase is reduced.  Increased D1 receptors in the substantia nigra and decreased dopamine transporters in the nucleus accumbens and striatum have been reported.

  2. X
    Non-Motor Impairment at 50

    At 11–12 months, spatial memory was impaired as assessed by the Barnes circular maze.

  3. X
    α-synuclein Inclusions at 35

    Prior to motor deficits, these mice develop accumulations of α-synuclein in select neuronal populations, including the midbrain, cerebellum, brainstem, and spinal cord. The protein aggregates do not resemble Lewy bodies, but are thioflavin-S-positive, indicating fibrillar structure.

  4. X
    Neuroinflammation at 40

    In symptomatic mice, increased GFAP immunoreactivity was observed in select brain regions, including the dorsal midbrain, deep cerebellar nuclei, brainstem, and spinal cord. Cortex, hippocampus, and substantia nigra did not have increased reactivity compared with non-Tg controls.

  5. X
    Motor Impairment at 32

    These mice develop severe motor impairment starting around 9-16 months of age. The deficits start out with mild hyperactivity at 7 months and progress to a wobbling movement, decreased activity, and ultimately paralysis and death.

  6. X
    Mitochondrial Abnormalities at 56

    At 11–14 months, mitochondria in brainstem neurons were enlarged and their co-localization with the mitochondrial fission protein Drp1 was reduced.

Absent
  • Neuronal Loss at

    Overt neuronal loss was not reported in these mice.

No Data
Genes Mutations Modification Disease Neuropathology Behavior/Cognition
SNCA SNCA A53T SNCA: Transgenic Parkinson's Disease

No overt neuronal loss. Alterations in dopaminergic-associated proteins in the striatum, substantia nigra, and nucleus accumbens. Region-specific neuronal accumulation of fibrillar α-synuclein, ubiquitin, and neurofilament-H, and accompanying astrocytosis.

Early hyperactivity followed by severe motor impairment, manifesting as wobbling, posturing, decreased spontaneous locomotor behavior, inability to navigate the Rotarod, and ultimately paralysis and death. At 11–12 months, spatial memory impaired as assessed by the Barnes circular maze.

expand

α-synuclein A53T Mouse (Tg) on SNCA KO

Observed
  1. X
    Non-Motor Impairment at 14

    By 3 months of age, the mice develop gastrointestinal dysfunction.

  2. X
    Motor Impairment at 26

    By 6 months of age, homozygous mice became hypoactive, traveling less distance. This was not attributed to changes in exploratory behavior caused by anxiety. Also at 6 months, differences in performance on the accelerating Rotarod were seen.

Absent
  • Dopamine Deficiency at

    No differences in striatal dopamine concentrations, or dopaminergic metabolites, homovanillic acid (HVA) and 3,4-dihydroxyphenylacetic acid (DOPAC) at 11 and 18 months of age.

  • α-synuclein Inclusions at

    No evidence of Lewy body-like inclusions in the brain at any age. Likewise α-synuclein aggregates were not observed in the brain, although they did occur in enteric neurons in the gut.

  • Neuronal Loss at

    No evidence of neuronal cell loss in the substantia nigra at 11 and 18 months of age, including dopaminergic neurons (TH-positive neurons) and total neurons.

No Data
  • Neuroinflammation at

    No data.

  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
SNCA SNCA A53T SNCA: Transgenic; SNCA: Knock-Out Parkinson's Disease

No loss of dopaminergic neurons in the substantia nigra by 18 months of age. Rare dystrophic synapses in the hippocampus at advanced age, but no Lewy body-like pathology or α-synuclein aggregation in the brain. No change in striatal dopamine concentration.

Impaired performance on the Rotarod and reduced spontaneous locomotor activity in open-field test.

expand

α-synuclein E46K Rat (BAC Tg)

Observed
  1. X
    α-synuclein Inclusions at 52

    By 12 months of age, intracellular aggregates were observed in dopaminergic neurons of the substantia nigra and ventral tegmental area. Aggregates noted to be fairly small compared to those observed in PD brain. In the striatum and cortex α -synuclein accumulation appeared primarily in neuronal processes.

Absent
  • Dopamine Deficiency at

    No dopamine deficiency in the striatum at 12 months of age. No serotonin deficiency in the striatum. Dopamine metabolites dihydroxyphenylacetic acid and homovanillic acid were reduced by approximately 25 percent and transmitter turnover was decreased.

  • Motor Impairment at

    No overt motor differences out to 12 months of age, unless challenged with low-dose rotenone, upon which the rats exhibit bradykinesia, postural instability, and rigidity.

  • Neuronal Loss at

    No overt loss of dopaminergic neurons out to 12 months of age.

No Data
  • Non-Motor Impairment at

    No data.

  • Neuroinflammation at

    No data.

  • Mitochondrial Abnormalities at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
SNCA SNCA E46K SNCA: Transgenic Parkinson's Disease

No overt neuronal loss. Accumulation of mutant α-synuclein in the brain, in the form of diffuse staining and intracellular aggregates. Aggregates were largely restricted to dopaminergic neurons of the substantia nigra and ventral tegmental area. Elevated nitrotyrosine in dopaminergic neurons.

No overt behavioral changes until challenged with low-dose rotenone, upon which the rats exhibit bradykinesia, postural instability, and rigidity.

expand

α-synuclein KO Mouse

Observed
  1. X
    Neuroinflammation at 16

    Microglia cultured from Snca KO brain were more reactive, ramified. They had vacuole-like structures. Snca KO microglia exhibited exacerbated response to LPS, with greater secretion of pro-inflammatory cytokines.

  2. X
    Motor Impairment at 26

    Motor function was largely intact. Normal performance on the Rotarod and in total distance travelled in the open field test. Subtle differences only (e.g., less rearing behavior than controls). They also spent less time in the center of the field, suggesting a possible anxiety-related phenotype.

  3. X
    Mitochondrial Abnormalities at 39

    Mitochondrial abnormalities include reduced levels of the mitochondrial phospholipid cardiolipin and reduced activity of electron transport chain complex I/III.

Absent
  • Non-Motor Impairment at

    The mice had normal reflexes and sensory abilities. Also, learning and memory appeared intact at 6-10 months of age, as assesed by the Morris water maze and tests of conditioned fear memory.

No Data
  • Dopamine Deficiency at

    A possible modest reduction in striatal dopamine level, but highly variable from mouse to mouse.

  • α-synuclein Inclusions at

    No data.

  • Neuronal Loss at

    No gross abnormalities in the brain.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
SNCA SNCA: Knock-Out Parkinson's Disease

No gross brain abnormalities. Electron microscopy revealed synaptic vesicle abnormalities in hippocampal neurons, i.e., fewer vesicles in the reserve pool.

Behavior is largely normal. Normal performance on the Rotarod. Subtle differences in locomotor activity (e.g., less rearing) but normal overall distance travelled. Learning and memory appear intact. Possible anxiety-like phenotype.

expand

α-synuclein KO Mouse (Conditional)

Observed
Absent
No Data
  • Dopamine Deficiency at

    No data.

  • Non-Motor Impairment at

    No data.

  • α-synuclein Inclusions at

    No data.

  • Neuroinflammation at

    No data.

  • Motor Impairment at

    No data.

  • Mitochondrial Abnormalities at

    No data.

  • Neuronal Loss at

    No data.

Genes Mutations Modification Disease Neuropathology Behavior/Cognition
SNCA SNCA: Conditional Knock-out Parkinson's Disease

No data.

No data.

expand