Elsevier

Neurobiology of Aging

Volume 24, Issue 2, March–April 2003, Pages 245-258
Neurobiology of Aging

Motor dysfunction and gliosis with preserved dopaminergic markers in human α-synuclein A30P transgenic mice

https://doi.org/10.1016/S0197-4580(02)00091-XGet rights and content

Abstract

α-Synuclein is a major component of Lewy bodies (LBs) in the substantia nigra and cortex in Parkinson’s disease (PD) and dementia with Lewy bodies (DLB), and in glial inclusions in multiple systems atrophy (MSA). Mutations in α-synuclein have been associated with autosomal dominant forms of PD. We investigated the clinical and neuropathological effects of overexpression of human α-synuclein, α-synuclein A30P, and α-synuclein A53T under the control of the hamster prion protein (PrP) promoter; 5–15× endogenous levels of protein expression were achieved with widespread neuronal, including nigral, transgene expression. High expression of α-synuclein A30P in the Tg5093 line was associated with a progressive motor disorder with rigidity, dystonia, gait impairment, and tremor. Histological analysis of this line showed aberrant expression of the protein in cell soma and progressive CNS gliosis, but no discrete Lewy body-like α-synuclein inclusions could be identified. Biochemical analysis demonstrated α-synuclein fragmentation. Despite strong expression of the transgene in the nigra, there was no specific deterioration of the nigrostriatal dopaminergic system as assessed by quantitation of nigral tyrosine hydroxylase (TH) containing neurons, striatal TH immunoreactivity, dopamine levels, or dopamine receptor number and function. Lower expressing lines had no specific behavioral or histopathological phenotype. Thus, high expression of mutant human α-synuclein resulted in a progressive motor and widespread CNS gliotic phenotype independent of dopaminergic dysfunction in the Tg5093 line.

Introduction

α-Synuclein is a highly conserved 140 amino acid presynaptic phosphoprotein implicated physiologically in synaptic plasticity and pathologically in several neurodegenerative disorders, including Parkinson’s disease (PD), dementia with Lewy bodies (DLB), multiple system atrophy (MSA), and Alzheimer’s disease (AD) [6], [15], [19], [24], [25], [27], [39], [45], [46], [49], [50]. Two distinct pathogenic mutations (A53T and A30P) in the α-synuclein gene, located on chromosome 4, have been identified in several families with autosomal dominant PD [31], [39], and α-synuclein accumulates in Lewy bodies (LBs) and Lewy neurites in both familial and sporadic PD and DLB [24], [45], [50], as well as in glial inclusions in MSA [49].

These findings suggest a pathological role for α-synuclein aggregation in these disorders [18]. The mechanisms by which α-synuclein forms neuronal and glial inclusions and becomes associated with neuronal degeneration, or by which α-synuclein mutations result in hereditary forms of PD, are unknown. Structurally, the α-synuclein protein contains seven copies of a degenerate 11 amino acid repeat which predicts four amphipathic alpha helices, structurally similar to exchangeable apolipoproteins such as apolipoprotein E [15]. The alpha helical domains may mediate reversible interactions with lipid domains including synaptosomal membranes [12], [25], [36]. α-Synuclein can form filamentous aggregates in vitro, with kinetics modified by PD associated mutations, catecholamines, and by nucleation with truncated α-synuclein [7], [8], [9], [11], [17], [44].

We tested the hypothesis that overexpression of mutant human α-synuclein in mouse brain induces clinical and neuropathological traits resembling those of human Lewy body disorders. We produced transgenic mice expressing human wild-type (wt), A53T, and A30P α-synuclein in a C57B/6jxSJL F1 hybrid background under the control of the hamster prion protein (PrP) promoter. High expression of α-synuclein A30P resulted in a progressive motor disorder associated with aberrant expression of the protein in cell soma, α-synuclein fragmentation and progressive CNS gliosis, but without specific anatomical or biochemical disruption of the nigrostriatal dopaminergic system.

Section snippets

Transgenic mice

cDNAs encoding human α-synuclein containing the A30P mutation, A53T mutation or wt sequence were expressed in C57B/6jxSJL F3 hybrid mice using a hamster PrP cosmid vector, in which the PrP open reading frame (ORF) was replaced with the human α-synuclein ORF. This vector has been previously shown to drive transgene product expression throughout neurons in the brain, including the substantia nigra and other vulnerable neuronal populations in Lewy body disorders [22], [43]. Subsequent generations

Transgenic mice generation

Five transgenic mouse lines were established expressing wt (lines Tg5228 and Tg5250), mutant A30P (lines Tg5093 and Tg5102) and mutant A53T (line Tg5343) α-synuclein. The lower expressing lines (Tg5228, Tg5250, Tg5102, and Tg5343) heterozygotes (Tg+/−), and Tg5102 homozygotes (Tg+/+) had 5- to 12-fold transgene protein expression relative to endogenous α-synuclein and developed normally without detectable motor or pathological deficits. The highest expressing line (Tg5093) had 15-fold

Discussion

The principal goal of this study was to determine the phenotypic and pathological effects of overexpression of mutant and wt human α-synuclein in mice, with particular emphasis on motor function and the anatomical and biochemical integrity of the nigrostriatal dopaminergic system. The transgene promoter and mouse background strain were selected to achieve high levels of transgene expression in the central nervous system. The hamster PrP cosmid vector produced high levels of brain α-synuclein

References (51)

  • P.H Jensen et al.

    Alpha-synuclein binds to Tau and stimulates the protein kinase A catalyzed tau phosphorylation of serine residues 262 and 356

    J. Biol. Chem.

    (1999)
  • P.J Kahle et al.

    Selective insolubility of alpha-synuclein in human Lewy body diseases is recapitulated in a transgenic mouse model

    Am. J. Pathol.

    (2001)
  • L Maroteaux et al.

    The rat brain synucleins; family of proteins transiently associated with neuronal membrane

    Brain Res. Mol. Brain Res.

    (1991)
  • Y Matsuoka et al.

    Lack of nigral pathology in transgenic mice expressing human alpha-synuclein driven by the tyrosine hydroxylase promoter

    Neurobiol. Dis.

    (2001)
  • P.J McLean et al.

    Membrane association and protein conformation of alpha-synuclein in intact neurons. Effect of Parkinson’s disease-linked mutations

    J. Biol. Chem.

    (2000)
  • R Morris

    Developments of a water maze procedure for studying spatial learning in the rat

    J. Neurosci. Methods

    (1984)
  • E.K Richfield et al.

    Properties of D2 dopamine receptor autoradiography: high percentage of high-affinity agonist sites and increased nucleotide sensitivity in tissue sections

    Brain Res.

    (1986)
  • E Rockenstein et al.

    Altered expression of the synuclein family mRNA in Lewy body and Alzheimer’s disease

    Brain Res.

    (2001)
  • K Wakabayashi et al.

    NACP, a presynaptic protein, immunoreactivity in Lewy bodies in Parkinson’s disease

    Neurosci. Lett.

    (1997)
  • M Baba et al.

    Aggregation of alpha-synuclein in Lewy bodies of sporadic Parkinson’s disease and dementia with Lewy bodies

    Am. J. Pathol.

    (1998)
  • Cavalieri B. Geometria degli indivisibili. Torino, Unione Tipografico, Editrice,...
  • J.-H.J Cha et al.

    Altered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human huntington disease gene

    Proc. Natl. Acad. Sci. U.S.A.

    (1998)
  • K.A Conway et al.

    Accelerated in vitro fibril formation by a mutant alpha-synuclein linked to early-onset Parkinson disease

    Nat. Med.

    (1998)
  • K.A Conway et al.

    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)
  • K.A Conway et al.

    Kinetic stabilization of the alpha-synuclein protofibril by a dopamine alpha-synuclein adduct

    Science

    (2001)
  • Cited by (0)

    1

    These authors contributed equally to this manuscript.

    2

    Present address: Neurology Department, Clinica Universitaria de Navarra, Avda. Pio XII 36, 31008 Pamplona, Navarra, Spain.

    View full text