Elsevier

Neurobiology of Disease

Volume 37, Issue 2, February 2010, Pages 284-293
Neurobiology of Disease

A transgenic mouse model of spinocerebellar ataxia type 3 resembling late disease onset and gender-specific instability of CAG repeats

https://doi.org/10.1016/j.nbd.2009.08.002Get rights and content

Abstract

Spinocerebellar ataxia type 3 (SCA3), or Machado–Joseph disease (MJD), is caused by the expansion of a polyglutamine repeat in the ataxin-3 protein. We generated a mouse model of SCA3 expressing ataxin-3 with 148 CAG repeats under the control of the huntingtin promoter, resulting in ubiquitous expression throughout the whole brain. The model resembles many features of the disease in humans, including a late onset of symptoms and CAG repeat instability in transmission to offspring. We observed a biphasic progression of the disease, with hyperactivity during the first months and decline of motor coordination after about 1 year of age; however, intranuclear aggregates were not visible at this age. Few and small intranuclear aggregates appeared first at the age of 18 months, further supporting the claim that neuronal dysfunction precedes the formation of intranuclear aggregates.

Introduction

Spinocerebellar ataxia type 3 (SCA3), or Machado–Joseph disease (MJD), is an autosomal-dominantly inherited neurodegenerative disorder caused by the expansion of a CAG repeat in the MJD1 gene (Kawaguchi et al., 1994). While, in unaffected individuals, the number of CAG repeats is usually less than 45 (Padiath et al., 2005), it increases up to 86 repeats in SCA3 patients (Riess et al., 2008). The expanded CAG repeat results in an expanded polyglutamine stretch in the encoded ataxin-3 protein. SCA3 therefore belongs to the group of polyglutamine diseases, which includes other types of spinocerebellar ataxias as well as SBMA, DRPLA, and Huntington's disease (reviewed in Gatchel and Zoghbi, 2005). The protein harboring the expanded polyglutamine tract has a strong tendency for aggregation (Scherzinger et al., 1999), which usually manifests in the nucleus of neuronal cells in SCA3. Interestingly, while ataxin-3 is ubiquitously expressed throughout the whole brain, the formation of intranuclear inclusion bodies appears in specific brain regions (Paulson, 1997b, Schmidt, 1998). There is an ongoing discussion whether these intranuclear inclusions are toxic or perhaps even protective for neuronal cells (Michalik and Van Broeckhoven, 2003, Sisodia, 1998).

Clinically, SCA3 presents with a highly heterogenous phenotype, leading to differentiation into clinical subtypes (reviewed in Riess et al., 2008). In addition, the disease is characterized by a late onset, usually appearing in the late third decade of life (Dürr, 1996, Schöls, 1997), and by slow progression. In contrast to the disease course in human patients, most of the previously generated mouse models of SCA3 are characterized by an early onset and rapid progression (Bichelmeier, 2007, Cemal, 2002, Chou, 2008, Goti, 2004, Ikeda, 1996) with rather distinct regional manifestation. Thus, not all the pathogenic aspects of the disease in humans are reproduced by these models.

Here, we present a mouse model for SCA3 with late onset of symptoms that resembles major genetic and pathogenetic characteristics in humans and demonstrates that motor symptoms precede the occurrence of intranuclear inclusion bodies. For this new transgenic mouse model, we utilized the well-characterized rat huntingtin promoter (Holzmann, 1998, Holzmann, 2001) to express a full-length ataxin-3 construct containing 148 CAG repeats ubiquitously throughout the brain.

In transgenic mice, the instability of the CAG repeat expansion was obvious, and clearly correlated with the sex of the transmitting parent. We additionally observed a biphasic course of the disease and detected hyperactivity in HDPromMJD148 mice long before the onset of motor deficits, but the formation of intranuclear inclusion bodies was no prerequisite for the onset of symptoms.

The novel mouse model of SCA3 presented in this study is a valuable addition to previous published mouse models, since its slowly progressing phenotype allows us to study aspects of the disease that cannot be analyzed in other models of this disease.

Section snippets

Transgenic mice

To generate a novel transgenic mouse model for SCA3, a 764 bp XbaI restriction fragment of the rat huntingtin promoter corresponding to nucleotide positions − 777 to − 14 was cloned to the 5′ end of the full-length ataxin-3 cDNA (isoform ataxin-3c) (Goto, 1997, Schmitt, 1997) in the pBluescript SK vector. The transcriptional activity of this promoter fragment has been demonstrated before (Holzmann et al., 1998). The SV40 early mRNA polyadenylation signal was amplified from the vector pEGFP-C

The huntingtin promoter led to transgene expression throughout the whole brain

To control the expression of a full-length construct of ataxin-3 containing 148 CAG repeats, a 764-bp fragment of the well-characterized rat huntingtin promoter was used (Fig. 1A). This promoter fragment contains numerous conserved putative transcription factor binding sites, which have led to robust expression in a variety of cell lines derived from different rodents (Holzmann et al., 1998). A comparable fragment of this promoter has been successfully used before to generate a transgenic rat

Discussion

Spinocerebellar ataxia type 3 is a neurodegenerative disease with late onset, slow progression and a heterogeneous clinical phenotype (Riess et al., 2008). As previously generated mouse models of SCA3 do not reproduce all aspects of the disease in humans (Bichelmeier, 2007, Cemal, 2002, Chou, 2008, Goti, 2004, Ikeda, 1996) we generated a novel mouse model for SCA3 to reflect more closely the late manifesting and slowly progressing phenotype. To control the expression of a full-length ataxin-3

Acknowledgments

The technical help of Gabi Frommer-Kästle in electron microscopy is greatly appreciated. This study was supported by the German Research Foundation (DFG) to OR, and by a grant from the European Union (6th Framework Programme, EUROSCA).

References (69)

  • LüesseH.G.

    Evaluation of R6/2 HD transgenic mice for therapeutic studies in Huntington's disease: behavioral testing and impact of diabetes mellitus

    Behav. Brain Res.

    (2001)
  • MunozE.

    Intranuclear inclusions, neuronal loss and CAG mosaicism in two patients with Machado–Joseph disease

    J. Neurol. Sci.

    (2002)
  • PaulsonH.L.

    Intranuclear inclusions of expanded polyglutamine protein in spinocerebellar ataxia type 3

    Neuron

    (1997)
  • RübU.

    Degeneration of the external cuneate nucleus in spinocerebellar ataxia type 3 (Machado–Joseph disease)

    Brain Res.

    (2002)
  • SaudouF.

    Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions

    Cell

    (1998)
  • SchillingG.

    Distinct behavioral and neuropathological abnormalities in transgenic mouse models of HD and DRPLA

    Neurobiol. Dis.

    (2001)
  • SisodiaS.S.

    Nuclear inclusions in glutamine repeat disorders: are they pernicious, coincidental, or beneficial?

    Cell

    (1998)
  • TrottierY.

    Heterogeneous intracellular localization and expression of ataxin-3

    Neurobiol. Dis.

    (1998)
  • BichelmeierU.

    Nuclear localization of ataxin-3 is required for the manifestation of symptoms in SCA3: in vivo evidence

    J. Neurosci

    (2007)
  • BrownS.D.

    EMPReSS: standardized phenotype screens for functional annotation of the mouse genome

    Nat. Genet.

    (2005)
  • CancelG.

    Marked phenotypic heterogeneity associated with expansion of a CAG repeat sequence at the spinocerebellar ataxia 3/Machado–Joseph disease locus

    Am. J. Hum. Genet.

    (1995)
  • CemalC.K.

    YAC transgenic mice carrying pathological alleles of the MJD1 locus exhibit a mild and slowly progressive cerebellar deficit

    Hum. Mol. Genet.

    (2002)
  • ChungM.Y.

    Evidence for a mechanism predisposing to intergenerational CAG repeat instability in spinocerebellar ataxia type I

    Nat. Genet.

    (1993)
  • DürrA.

    Spinocerebellar ataxia 3 and Machado–Joseph disease: clinical, molecular, and neuropathological features

    Ann. Neurol.

    (1996)
  • GarthwaiteG. et al.

    AMPA neurotoxicity in rat cerebellar and hippocampal slices: histological evidence for three mechanisms

    Eur. J. Neurosci.

    (1991)
  • GarthwaiteG. et al.

    Mechanisms of AMPA neurotoxicity in rat brain slices

    Eur. J. Neurosci

    (1991)
  • GatchelJ.R. et al.

    Diseases of unstable repeat expansion: mechanisms and common principles

    Nat. Rev. Genet.

    (2005)
  • GotiD.

    A mutant ataxin-3 putative-cleavage fragment in brains of Machado–Joseph disease patients and transgenic mice is cytotoxic above a critical concentration

    J. Neurosci.

    (2004)
  • GouwL.G.

    Analysis of the dynamic mutation in the SCA7 gene shows marked parental effects on CAG repeat transmission

    Hum. Mol. Genet.

    (1998)
  • GreenE.C.

    EMPReSS: European mouse phenotyping resource for standardized screens

    Bioinformatics

    (2005)
  • HolzmannC.

    Isolation and characterization of the rat huntingtin promoter

    Biochem. J.

    (1998)
  • IgarashiS.

    Intergenerational instability of the CAG repeat of the gene for Machado-Joseph disease (MJD1) is affected by the genotype of the normal chromosome: implications for the molecular mechanisms of the instability of the CAG repeat

    Hum. Mol. Genet.

    (1996)
  • IkedaH.

    Expanded polyglutamine in the Machado–Joseph disease protein induces cell death in vitro and in vivo

    Nat. Genet.

    (1996)
  • KawaguchiY.

    CAG expansions in a novel gene for Machado–Joseph disease at chromosome 14q32.1

    Nat. Genet.

    (1994)
  • Cited by (48)

    • RNA toxicity in tandem nucleotide repeats mediated neurodegenerative disorders

      2020, RNA-Based Regulation in Human Health and Disease
    • Autosomal-dominant cerebellar ataxias

      2018, Handbook of Clinical Neurology
    • Interaction between the AAA ATPase p97 and its cofactor ataxin3 in health and disease: Nucleotide-induced conformational changes regulate cofactor binding

      2017, Journal of Biological Chemistry
      Citation Excerpt :

      For example, ataxin3 knock-out animals feature increased levels of polyubiquitinated proteins and increased levels of the stress response (46–48). Interestingly, a mutant form of ataxin3 containing an expanded polyglutamine tract gives rise to aggregates of misfolded protein and causes the neurodegenerative disorder spinocerebellar ataxia type-3 (SCA3, also known as Machado-Joseph disease (49–54)). Therefore, it appears that ataxin3 can both support normal protein degradation and (in its mutant form) disrupt this same process.

    • Mouse Models of SCA3 and Other Polyglutamine Repeat Ataxias

      2015, Movement Disorders: Genetics and Models: Second Edition
    View all citing articles on Scopus
    1

    These authors contributed equally to this project.

    2

    Present address: Prince of Wales Medical Research Institute, Randwick, Australia.

    View full text