Region-specific deficits in dopamine, but not norepinephrine, signaling in a novel A30P α-synuclein BAC transgenic mouse

Tonya N Taylor, Dawid Potgieter, Sabina Anwar, Steven L Senior, Stephanie Janezic, Sarah Threlfell, Brent Ryan, Laura Parkkinen, Thierry Deltheil, Milena Cioroch, Achilleas Livieratos, Peter L Oliver, Katie A Jennings, Kay E Davies, Olaf Ansorge, David M Bannerman, Stephanie J Cragg, Richard Wade-Martins, Tonya N Taylor, Dawid Potgieter, Sabina Anwar, Steven L Senior, Stephanie Janezic, Sarah Threlfell, Brent Ryan, Laura Parkkinen, Thierry Deltheil, Milena Cioroch, Achilleas Livieratos, Peter L Oliver, Katie A Jennings, Kay E Davies, Olaf Ansorge, David M Bannerman, Stephanie J Cragg, Richard Wade-Martins

Abstract

Parkinson's disease (PD) is a neurodegenerative disorder classically characterized by the death of dopamine (DA) neurons in the substantia nigra pars compacta and by intracellular Lewy bodies composed largely of α-synuclein. Approximately 5-10% of PD patients have a familial form of Parkinsonism, including mutations in α-synuclein. To better understand the cell-type specific role of α-synuclein on DA neurotransmission, and the effects of the disease-associated A30P mutation, we generated and studied a novel transgenic model of PD. We expressed the A30P mutant form of human α-synuclein in a spatially-relevant manner from the 111kb SNCA genomic DNA locus on a bacterial artificial chromosome (BAC) insert on a mouse null (Snca-/-) background. The BAC transgenic mice expressed α-synuclein in tyrosine hydroxylase-positive neurons and expression of either A30P α-synuclein or wildtype α-synuclein restored the sensitivity of DA neurons to MPTP in resistant Snca-/- animals. A30P α-synuclein mice showed no Lewy body-like aggregation, and did not lose catecholamine neurons in substantia nigra or locus coeruleus. However, using cyclic voltammetry at carbon-fiber microelectrodes we identified a deficit in evoked DA release in the caudate putamen, but not in the nucleus accumbens, of SNCA-A30P Snca-/- mice but no changes to release of another catecholamine, norepinephrine (NE), in the NE-rich ventral bed nucleus of stria terminalis. SNCA-A30P Snca-/- mice had no overt behavioral impairments but exhibited a mild increase in wheel-running. In summary, this refined PD mouse model shows that A30P α-synuclein preferentially perturbs the dopaminergic system in the dorsal striatum, reflecting the region-specific change seen in PD.

Keywords: Behavior; Dopamine; Norepinephrine; Parkinson's disease; Voltammetry; α-Synuclein.

© 2013.

Figures

Fig. 1
Fig. 1
Expression of human wild-type or A30P mutant α-synuclein in SNCA-BAC transgenic mice. A, Schematic representation of the human SNCA BAC transgene constructs carrying either wild-type or A30P mutant SNCA. B, Confirmation of the SNCA BAC transgene integrity by PCR in both SNCA lines. Human-specific PCR primers spanning each of the SNCA coding exons were used (exons 1–6). C, Location of transgene integration in SNCA-BAC lines. Fluorescent in situ hybridization (FISH) confirmed a single integration site for both transgenic lines using BAC probes in combination with chromosome painting. The SNCA-A30P transgene integration was observed near the telomere on one copy of chromosome 15; the SNCA-WT (hα-syn) transgene integration occurred near the telomere on one copy of chromosome 2. D, Expression of α-synuclein was quantified by western blot of whole brain homogenates from 3 month-old SNCA BAC mice. α-synuclein expression was quantified relative to actin levels and normalized to expression in mice expressing wildtype human α-synuclein (hα-syn). The difference in expression between A30P and hα-syn lines is not significant. n = 3, **p < 0.01. E, Immunohistochemical analysis in coronal brain sections of 3-month old animals revealed α-synuclein transgene expression in the cortex, SNpc, and VTA in hα-syn and SNCA-A30P mice recapitulating a spatial pattern of expression almost identical to that of endogenous α-synuclein protein in wild-type C57Bl6 mice. α-synuclein expression was not seen in Snca −/− KO animals.
Fig. 2
Fig. 2
Immunohistochemical analysis of α-synuclein expression in the SNpc in SNCA-BAC transgenic mice. Immunofluorescence shows human α-synuclein protein expression in 3 month-old A30P and hα-syn mice that accurately recapitulates the pattern of endogenous α-synuclein expression in TH-positive neurons in the SNpc, seen as yellow cells in the merged frame and clearly visible in the high-magnification insets. Age-matched Snca −/− KO animals display no α-synuclein expression.
Fig. 3
Fig. 3
Expression of wild-type or A30P α-synuclein restores susceptibility of DA neurons in Snca −/− mice to MPTP in 6-month old mice. A, TH immunoreactivity in striatum and SNpc in hα-syn, A30P, and Snca −/− KO mice 7 days following saline or acute dosing of MPTP free base. The A30P animals are similarly susceptible to MPTP lesion as hα-syn mice, as evidenced by a reduction of TH staining in the striatum and SNpc. Representative sections are shown. B, Quantitative unbiased stereological cell counts show a 30% reduction in TH + neurons in the SNpc of hα-syn and A30P animals. The Snca −/− KO animals were resistant to MPTP treatment. Results represent mean ± SEM for 3 animals per genotype and treatment *p < 0.05. C, D The level of striatal TH was diminished in hα-syn and A30P animals after exposure to MPTP as measured by western blot analysis of TH levels in striatal tissue, while TH expression remained unchanged in Snca −/− KO animals. TH band intensity was controlled for β-actin expression and normalized, within-genotype, to saline-treated controls (n = 3, *p < 0.05).
Fig. 4
Fig. 4
Electrically evoked dopamine transients and regulation of [DA]o by firing frequency in 3-month old hα-syn, SNCA A30P and Snca −/− KO measured by FCV. A, Mean profiles of [DA]oversus time (mean ± SEM) after a single pulse (0.2 μs; arrow) in the dorsal striatum (CPu). In CPu, there is no difference in peak [DA]o transiently evoked by single pulses in hα-syn mice compared to age-matched KO littermates (p = 0.2265, n = 60 (5 animals per genotype)). B, No significant differences seen in mean peak [DA]o evoked by a single pulse (1p) in hα-syn transgenic mice compared to KO littermates in the NAc. (p = .8680, n = 18 (5 animals per genotype)). C, Mean profiles of [DA]oversus time (mean ± SEM) after a single pulse (0.2 μs; arrow) in the dorsal striatum (CPu). In CPu, peak [DA]o transiently evoked by single pulses is slightly less in A30P than age-matched KO littermates (*p < 0.05, KO, n = 96; A30P, n = 92 (9 animals per genotype)). D, No significant differences seen in mean peak [DA]o evoked by a single pulse (1p) in A30P transgenic mice compared to KO littermates in the NAc. E, Cumulative histogram of peak [DA]o of evoked DA transients in the CPu in KO vs A30P mice for individual sampling sites. F, Comparison of the rates of decay of concentration-matched DA transients suggests that DA uptake rates are not significantly different between the two genotypes (n = 5, p > 0.05). G, No differences were observed in DA content (pmol per μg of protein) in the CPu or NAc dissected from striatal slices. Data shown represent mean ± SEM from Snca KO and SNCA A30P mouse samples (n = 8–10, p > 0.05). H, Regulation of [DA]o by firing frequency in the CPu. Mean peak [DA]o during 5 pulse trains (1–100 Hz) in the CPu varied with frequency in both genotypes in control conditions, with a slightly greater frequency sensitivity observed in A30P transgenic mice at 25 Hz. Data were normalized within genotype to the 1p value (n = 3, *p < 0.05).
Fig. 5
Fig. 5
Electrically evoked noradrenergic transients and regulation of norepinephrine (NE) signals by firing frequency in 3-month old SNCA-A30P and Snca −/− KO mice measured by FCV. A, Cyclic voltammograms from a calibration using 2 μM NE (black), and 30 pulses at 50 Hz in Snca KO (red) and A30P (blue) slices. B, Maximal NE release as a function of current amplitude. Each data point was obtained with a 30-pulse, 50 Hz stimulation (n = 6 C57Bl/6 animals). C, Maximal release as a function of stimulation frequency. Each data point was obtained with a 30-pulse stimulation, and a current amplitude of 0.65 mA (n = 5 C57Bl/6 animals). D, Maximal release as a function of pulse number. Each data point was obtained with a stimulation frequency of 50 Hz, and current amplitude of 0.65 mA (n = 6 C57/Bl6 animals). E, Mean [NE]oversus time evoked by 30 pulses at 50 Hz in vBNST of C57/Bl6 mice in control conditions, in the presence of the α2 adrenergic antagonist, idazoxan (1 μM). Idazoxan significantly increased peak [NE]o in the vBNST (n = 3 animals, **p < 0.01). F, Mean [NE]oversus time evoked by 30 pulses at 50 Hz in vBNST of C57Bl/6 mice in control conditions and in the presence of 300 nM desipramine. Desipramine significantly increased peak [NE]o in the vBNST (n = 3 animals, *p < 0.05). G, Mean [DA]oversus time evoked by 30 pulses at 50 Hz in the CPu of C57/Bl6 mice in control conditions and in the presence of α2 adrenergic antagonist, idazoxan (1 μM). Unlike in the vBNST, idazoxan produces no change in peak [DA]o in the CPu (n = 3 animals). H, Mean profiles of [NE]oversus time (mean ± SEM) after a 30 pulse, 50 Hz stimulation (arrow) in the ventral bed nucleus of the stria terminalis (BNST). In vBNST, a very slight 8% difference was observed in peak [NE]o transiently evoked by 30 pulses at 50 Hz in A30P compared to age-matched KO littermates which was not significant (p > 0.05; KO, n = 79 sites; A30P, n = 82 sites (8 animals per genotype)). I, Mean peak [NE]o ± SEM vs. frequency during 30 pulse trains (10–100 Hz) in the vBNST varied with frequency in both genotypes in control conditions. No differences in frequency sensitivity were seen in A30P transgenic mice compared to KO animals (p > 0.05; 8 animals per genotype). J, Cumulative histogram of peak [NE]o of evoked NE transients in the vBNST in KO vs A30P mice for individual sampling sites. K, Immunohistochemical analysis in coronal mouse sections confirmed α-synuclein transgene expression in the LC in SNCA-A30P mice; α-synuclein expression was not seen in Snca −/− KO animals.
Fig. 6
Fig. 6
Immunohistochemical and western blot analysis of striata in aged (18-month old) hα-syn, A30P, and Snca −/− KO mice. A, TH immunoreactivity was unchanged at 18 months of age. Western blot analysis of striatal lysates from hα-syn, A30P, and Snca −/− KO mice show also no differences in TH expression with age. B, DAT immunoreactivity was unchanged at 18 months of age. Similarly, western blot analysis of striatal lysates from hα-syn, A30P, and Snca −/− KO mice does not demonstrate a reduction in DAT expression with age. Analysis was performed on 3 animals per genotype at each age. Representative sections are shown. Columns represent percentage change from the 6-month old hα-syn control. Results represent the mean ± SEM for three animals per genotype at each age. For western blot analysis, 10 μg protein was loaded per lane.
Fig. 7
Fig. 7
Stereological counts of SNpc and LC in aged hα-syn, A30P, and Snca −/− KO mice. A, B No differences were seen in TH-positive cells (A) or hematoxylin-positive (B) in the SNpc at 18 months of age in A30P animals compared with hα-syn and KO animals. C, D, Similarly, no differences were observed TH cell number (C) or hematoxylin-positive cell numbers (D) in the LC at 18 months of age between the three genotypes. Results represent the mean ± SEM for 5 animals per genotype at each age, p > 0.05.
Fig. 8
Fig. 8
Absence of synuclein pathology in 23-month old A30P transgenic animals. A diffuse cytosolic α-synuclein immunostaining was seen in neuronal cells of the anterior olfactory nucleus and granular cell layer of the olfactory bulb with antibody LB509 (A), but not by Syn-1 (C), or pSyn #64 against the phosphorylated α-syn (E). Magnification in A-F × 40, inserts show higher-power magnified image × 200.
Fig. 9
Fig. 9
A30P mice display a very mild behavioral phenotype. A, A30P mice have similar total locomotor activity at all ages when introduced into a novel environment as compared to age-matched hα-syn and Snca −/− KO controls. Results represent the mean number of ambulations ± SEM for 10–13 mice per genotype. B, No deficits in forepaw stride length were apparent at 3 months or 10–13 months of age in A30P mice, compared to age-matched hα-syn and Snca −/− KO controls. Results represent average stride length (cm) ± SEM for 8–10 animals per genotype. C, No differences were seen in motor learning or performance on the rotarod at 18 months of age. Results represent latency to fall (sec) ± SEM for 4–10 animals per genotype. D, At 6 months of age, the hα-syn mice spent a greater percentage of their time in the open arms of the elevated plus maze as compared to age-matched A30P and Snca KO animals over the 5 min test period. All genotypes display an increased anxiety-like phenotype with age. Results represent the mean percentage of time spent in the open arms ± SEM for 10–13 mice per genotype. **p < 0.01 E, All genotypes had similar immobility times in the tail suspension test, which remains unchanged with age. Results represent mean time (s) ± SEM for 10–13 mice per genotype. F, A30P mice display increased wheel running during hours 16–17 of their light–dark cycle compared to hα-syn and Snca −/− KO mice at 12 months of age. Results represent total activity counts ± SEM for 5 animals per genotype *p < 0.05, **p < 0.01. G, A30P mice display normal circadian activity levels compared to hα-syn and KO animals at 12 months of age. Results represent total ambulations ± SEM for 8 animals per genotype. H, A30P mice do not display differences in stool frequency compared to age-matched hα-syn and Snca −/− KO controls. Results represent average stool frequency ± SEM for 5–11 animals per genotype. I, A30P mice do not display deficits in spontaneous alternation as measured by the T-maze at 3, 10–13, or 15–18 months of age when compared to age-matched hα-syn or Snca KO mice. Results represent the mean alternation ± SEM for 10–13 animals per genotype at each age.

References

    1. Abeliovich A. Mice lacking alpha-synuclein display functional deficits in the nigrostriatal dopamine system. Neuron. 2000;25:239–252.
    1. Alegre-Abarrategui J. LRRK2 regulates autophagic activity and localizes to specific membrane microdomains in a novel human genomic reporter cellular model. Hum. Mol. Genet. 2009;18:4022–4034.
    1. Al-Wandi A. Absence of alpha-synuclein affects dopamine metabolism and synaptic markers in the striatum of aging mice. Neurobiol. Aging. 2010;31:796–804.
    1. Anwar S. Functional alterations to the nigrostriatal system in mice lacking all three members of the synuclein family. J. Neurosci. 2011;31:7264–7274.
    1. Braak H. Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson's disease (preclinical and clinical stages) J. Neurol. 2002;249(Suppl. 3):III/1-5.
    1. Braak H. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol. Aging. 2003;24:197–211.
    1. Cabin D.E. Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking alpha-synuclein. J. Neurosci. 2002;22:8797–8807.
    1. Cabin D.E. Exacerbated synucleinopathy in mice expressing A53T SNCA on a Snca null background. Neurobiol. Aging. 2005;26:25–35.
    1. Caudle W.M. Polychlorinated biphenyl-induced reduction of dopamine transporter expression as a precursor to Parkinson's disease-associated dopamine toxicity. Toxicol. Sci. 2006;92:490–499.
    1. Chesselet M.F. In vivo alpha-synuclein overexpression in rodents: a useful model of Parkinson's disease? Exp. Neurol. 2008;209:22–27.
    1. Chiba-Falek O., Nussbaum R.L. Effect of allelic variation at the NACP-Rep1 repeat upstream of the alpha-synuclein gene (SNCA) on transcription in a cell culture luciferase reporter system. Hum. Mol. Genet. 2001;10:3101–3109.
    1. Chiba-Falek O. Functional analysis of intra-allelic variation at NACP-Rep1 in the alpha-synuclein gene. Hum. Genet. 2003;113:426–431.
    1. Chiba-Falek O. Regulation of alpha-synuclein expression by poly (ADP ribose) polymerase-1 (PARP-1) binding to the NACP-Rep1 polymorphic site upstream of the SNCA gene. Am. J. Hum. Genet. 2005;76:478–492.
    1. Conway K.A. Acceleration of oligomerization, not fibrillization, is a shared property of both alpha-synuclein mutations linked to early-onset Parkinson's disease: implications for pathogenesis and therapy. Proc. Natl. Acad. Sci. U. S. A. 2000;97:571–576.
    1. Cragg S.J. Variable dopamine release probability and short-term plasticity between functional domains of the primate striatum. J. Neurosci. 2003;23:4378–4385.
    1. Cragg S. Heterogeneity of electrically evoked dopamine release and reuptake in substantia nigra, ventral tegmental area, and striatum. J. Neurophysiol. 1997;77:863–873.
    1. Cragg S.J. Dopamine release and uptake dynamics within nonhuman primate striatum in vitro. J. Neurosci. 2000;20:8209–8217.
    1. Cryan J.F. Norepinephrine-deficient mice lack responses to antidepressant drugs, including selective serotonin reuptake inhibitors. Proc. Natl. Acad. Sci. U. S. A. 2004;101:8186–8191.
    1. Dauer W., Przedborski S. Parkinson's disease: mechanisms and models. Neuron. 2003;39:889–909.
    1. Dauer W. Resistance of alpha-synuclein null mice to the parkinsonian neurotoxin MPTP. Proc. Natl. Acad. Sci. U. S. A. 2002;99:14524–14529.
    1. Deacon R.M., Rawlins J.N. T-maze alternation in the rodent. Nat. Protoc. 2006;1:7–12.
    1. Exley R. Alpha6-containing nicotinic acetylcholine receptors dominate the nicotine control of dopamine neurotransmission in nucleus accumbens. Neuropsychopharmacology. 2008;33:2158–2166.
    1. Fernagut P.O., Chesselet M.F. Alpha-synuclein and transgenic mouse models. Neurobiol. Dis. 2004;17:123–130.
    1. Fortin D.L. Lipid rafts mediate the synaptic localization of alpha-synuclein. J. Neurosci. 2004;24:6715–6723.
    1. Freichel C. Age-dependent cognitive decline and amygdala pathology in alpha-synuclein transgenic mice. Neurobiol. Aging. 2007;28:1421–1435.
    1. Fu Y. A cytoarchitectonic and chemoarchitectonic analysis of the dopamine cell groups in the substantia nigra, ventral tegmental area, and retrorubral field in the mouse. Brain Struct. Funct. 2012;217:591–612.
    1. Giasson B.I. Neuronal alpha-synucleinopathy with severe movement disorder in mice expressing A53T human alpha-synuclein. Neuron. 2002;34:521–533.
    1. Giros B. Hyperlocomotion and indifference to cocaine and amphetamine in mice lacking the dopamine transporter. Nature. 1996;379:606–612.
    1. Goedert M. Alpha-synuclein and neurodegenerative diseases. Nat. Rev. Neurosci. 2001;2:492–501.
    1. Gomez-Isla T. Motor dysfunction and gliosis with preserved dopaminergic markers in human alpha-synuclein A30P transgenic mice. Neurobiol. Aging. 2003;24:245–258.
    1. Jakes R. Identification of two distinct synucleins from human brain. FEBS Lett. 1994;345:27–32.
    1. Jefferson A., Volpi E.V. Fluorescence in situ hybridization (FISH) for genomic investigations in rat. Methods Mol. Biol. 2010;659:409–426.
    1. Jensen P.H. Binding of alpha-synuclein to brain vesicles is abolished by familial Parkinson's disease mutation. J. Biol. Chem. 1998;273:26292–26294.
    1. Jo E. Defective membrane interactions of familial Parkinson's disease mutant A30P alpha-synuclein. J. Mol. Biol. 2002;315:799–807.
    1. Kilts C.D., Anderson C.M. The simultaneous quantification of dopamine, norepinephrine and epinephrine in micropunched rat brain nuclei by on-line trace enrichment HPLC with electrochemical detection: distribution of catecholamines in the limbic system. Neurochem. Int. 1986;9:437–445.
    1. Knab A.M. Altered dopaminergic profiles: implications for the regulation of voluntary physical activity. Behav. Brain Res. 2009;204:147–152.
    1. Knab A.M. Pharmacological manipulation of the dopaminergic system affects wheel-running activity in differentially active mice. J. Biol. Regul. Homeost. Agents. 2012;26:119–129.
    1. Kruger R. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson's disease. Nat. Genet. 1998;18:106–108.
    1. Kruger R. Familial parkinsonism with synuclein pathology: clinical and PET studies of A30P mutation carriers. Neurology. 2001;56:1355–1362.
    1. Kulnane L.S. Rapid and efficient detection of transgene homozygosity by FISH of mouse fibroblasts. Mamm. Genome. 2002;13:223–226.
    1. Kuo Y.M. Extensive enteric nervous system abnormalities in mice transgenic for artificial chromosomes containing Parkinson disease-associated alpha-synuclein gene mutations precede central nervous system changes. Hum. Mol. Genet. 2010;19:1633–1650.
    1. Langston J.W. The Parkinson's complex: parkinsonism is just the tip of the iceberg. Ann. Neurol. 2006;59:591–596.
    1. Lee M.K. Human alpha-synuclein-harboring familial Parkinson's disease-linked Ala-53 – > Thr mutation causes neurodegenerative disease with alpha-synuclein aggregation in transgenic mice. Proc. Natl. Acad. Sci. U. S. A. 2002;99:8968–8973.
    1. Li Z.S. Physiological modulation of intestinal motility by enteric dopaminergic neurons and the D2 receptor: analysis of dopamine receptor expression, location, development, and function in wild-type and knock-out mice. J. Neurosci. 2006;26:2798–2807.
    1. Matsuoka Y. Lack of nigral pathology in transgenic mice expressing human alpha-synuclein driven by the tyrosine hydroxylase promoter. Neurobiol. Dis. 2001;8:535–539.
    1. McCormack A.L. Environmental risk factors and Parkinson's disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol. Dis. 2002;10:119–127.
    1. Miles P.R. Release and uptake of catecholamines in the bed nucleus of the stria terminalis measured in the mouse brain slice. Synapse. 2002;44:188–197.
    1. Miller G.W. Immunochemical analysis of dopamine transporter protein in Parkinson's disease. Ann. Neurol. 1997;41:530–539.
    1. Miller G.W. Immunochemical analysis of vesicular monoamine transporter (VMAT2) protein in Parkinson's disease. Exp. Neurol. 1999;156:138–148.
    1. Nemani V.M. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron. 2010;65:66–79.
    1. Neumann M. Misfolded proteinase K-resistant hyperphosphorylated alpha-synuclein in aged transgenic mice with locomotor deterioration and in human alpha-synucleinopathies. J. Clin. Invest. 2002;110:1429–1439.
    1. Olanow C.W., Tatton W.G. Etiology and pathogenesis of Parkinson's disease. Annu. Rev. Neurosci. 1999;22:123–144.
    1. Oliver P.L. Disrupted circadian rhythms in a mouse model of schizophrenia. Curr. Biol. 2012;22:314–319.
    1. Outeiro T.F., Lindquist S. Yeast cells provide insight into alpha-synuclein biology and pathobiology. Science. 2003;302:1772–1775.
    1. Paxinos G., Franklin K.B.J. Academic Press; San Diego: 2001. The Mouse Brain in Stereotaxic Coordinates.
    1. Pellow S. Validation of open:closed arm entries in an elevated plus-maze as a measure of anxiety in the rat. J. Neurosci. Methods. 1985;14:149–167.
    1. Peruzzi P.P. Physiological transgene regulation and functional complementation of a neurological disease gene deficiency in neurons. Mol. Ther. 2009;17:1517–1526.
    1. Platt N.J. Striatal dopamine transmission is subtly modified in human A53Talpha-synuclein overexpressing mice. PLoS One. 2012;7:e36397.
    1. Polymeropoulos M.H. Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science. 1997;276:2045–2047.
    1. Rathke-Hartlieb S. Sensitivity to MPTP is not increased in Parkinson's disease-associated mutant alpha-synuclein transgenic mice. J. Neurochem. 2001;77:1181–1184.
    1. Reveron M.E. L-DOPA does not cause neurotoxicity in VMAT2 heterozygote knockout mice. Neurotoxicology. 2002;23:611–619.
    1. Rice M.E., Cragg S.J. Nicotine amplifies reward-related dopamine signals in striatum. Nat. Neurosci. 2004;7:583–584.
    1. Rye D., DeLong M.R. Time to focus on the locus. Arch. Neurol. 2003;60:320.
    1. Schank J.R. Dopamine beta-hydroxylase knockout mice have alterations in dopamine signaling and are hypersensitive to cocaine. Neuropsychopharmacology. 2006;31:2221–2230.
    1. Schank J.R. Norepinephrine signaling through beta-adrenergic receptors is critical for expression of cocaine-induced anxiety. Biol. Psychiatry. 2008;63:1007–1012.
    1. Seidel K. First appraisal of brain pathology owing to A30P mutant alpha-synuclein. Ann. Neurol. 2010;67:684–689.
    1. Senior S.L. Increased striatal dopamine release and hyperdopaminergic-like behaviour in mice lacking both alpha-synuclein and gamma-synuclein. Eur. J. Neurosci. 2008;27:947–957.
    1. Specht C.G., Schoepfer R. Deletion of the alpha-synuclein locus in a subpopulation of C57BL/6 J inbred mice. BMC Neurosci. 2001;2:11.
    1. Spillantini M.G. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–840.
    1. Sulzer D. Clues to how alpha-synuclein damages neurons in Parkinson's disease. Mov. Disord. 2010;25(Suppl. 1):S27–S31.
    1. Taylor T.N. Nonmotor symptoms of Parkinson's disease revealed in an animal model with reduced monoamine storage capacity. J. Neurosci. 2009;29:8103–8113.
    1. Thomas B. Resistance to MPTP-neurotoxicity in alpha-synuclein knockout mice is complemented by human alpha-synuclein and associated with increased beta-synuclein and Akt activation. PLoS One. 2011;6:e16706.
    1. Tillerson J.L. Detection of behavioral impairments correlated to neurochemical deficits in mice treated with moderate doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Exp. Neurol. 2002;178:80–90.
    1. Touchman J.W. Human and mouse alpha-synuclein genes: comparative genomic sequence analysis and identification of a novel gene regulatory element. Genome Res. 2001;11:78–86.
    1. Ueda K. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc. Natl. Acad. Sci. U. S. A. 1993;90:11282–11286.
    1. van der Putten H. Neuropathology in mice expressing human alpha-synuclein. J. Neurosci. 2000;20:6021–6029.
    1. West M.J. Unbiased stereological estimation of the total number of neurons in thesubdivisions of the rat hippocampus using the optical fractionator. Anat. Rec. 1991;231:482–497.
    1. Xia Y. Characterization of the human alpha-synuclein gene: genomic structure, transcription start site, promoter region and polymorphisms. J. Alzheimers Dis. 2001;3:485–494.
    1. Yavich L. Locomotor activity and evoked dopamine release are reduced in mice overexpressing A30P-mutated human alpha-synuclein. Neurobiol. Dis. 2005;20:303–313.
    1. Zarranz J.J. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann. Neurol. 2004;55:164–173.

Source: PubMed

3
Prenumerera