Elsevier

Neurobiology of Aging

Volume 29, Issue 6, June 2008, Pages 913-925
Neurobiology of Aging

Mutant α-synuclein exacerbates age-related decrease of neurogenesis

https://doi.org/10.1016/j.neurobiolaging.2006.12.016Get rights and content

Abstract

In Parkinson disease, wild-type α-synuclein accumulates during aging, whereas α-synuclein mutations lead to an early onset and accelerated course of the disease. The generation of new neurons is decreased in regions of neurogenesis in adult mice overexpressing wild-type human α-synuclein. We examined the subventricular zone/olfactory bulb neurogenesis in aged mice expressing either wild-type human or A53T mutant α-synuclein. Aging wild-type and mutant α-synuclein-expressing animals generated significantly fewer new neurons than their non-transgenic littermates. This decreased neurogenesis was caused by a reduction in cell proliferation within the subventricular zone of mutant α-synuclein mice. In contrast, no difference was detected in mice overexpressing the wild-type allele. Also, more TUNEL-positive profiles were detected in the subventricular zone, following mutant α-synuclein expression and in the olfactory bulb, following wild-type and mutant α-synuclein expression. The impaired neurogenesis in the olfactory bulb of different transgenic α-synuclein mice during aging highlights the need to further explore the interplay between olfactory dysfunction and neurogenesis in Parkinson disease.

Introduction

In Parkinson disease (PD), as well as in other synucleinopathies, the accumulation of misfolded α-synuclein within neuronal cell bodies, axons and synapses has been proposed to be one of the crucial pathogenic hallmarks (Hashimoto and Masliah, 1999, Takeda et al., 1998). α-Synuclein is a 140-amino acid synaptic molecule that was originally identified as the non-Aβ component precursor protein of Alzheimer disease amyloid plaques (Iwai et al., 1996). A physiological role of α-synuclein has been proposed in developmental plasticity and synaptic remodeling (Chandra et al., 2004, George et al., 1995, Hsu et al., 1998). Point mutations in α-synuclein (A53T, A30P, E46K) have been identified in rare Mendelian forms of familial PD designated as PARK1 (Kruger et al., 1998, Polymeropoulos et al., 1997, Zarranz et al., 2004).

These α-synuclein mutations accelerate its aggregation and oligomerization (Conway et al., 1998, Narhi et al., 1999). As a result, in patients affected by the α-synuclein A53T mutation, the age of onset is much earlier than for patients presenting with sporadic PD (Kruger et al., 1998, Polymeropoulos et al., 1997, Zarranz et al., 2004). In PD, accumulation of α-synuclein aggregates has not only been observed in the midbrain and brain stem nuclei, but also in the olfactory bulb at a very early stage of the disease (Braak et al., 2003). Accumulation of α-synuclein in the olfactory bulb is particularly of clinical relevance, because olfactory dysfunction is one of the early symptoms in PD (Berendse et al., 2001, Sobel et al., 2001). Moreover, other neurodegenerative diseases such as Alzheimer disease or dementia with Lewy bodies result in olfactory dysfunction too (for review see Hawkes, 2006). The pathology in the olfactory bulb correlates strongly with the disease progression in dementia (Jellinger and Attems, 2005, Ohm and Braak, 1987, Tsuboi et al., 2003).

The olfactory bulb is one of the regions, where new neurons are constantly added to the olfactory circuitry throughout lifetime. The proliferating neural stem cells (NSCs) residing in the basal forebrain subventricular zone (SVZ) give constantly rise to neuronal precursors and therefore constitute the keystone of the olfactory bulb neurogenesis (Reynolds and Weiss, 1992). Newly generated neuronal precursors leave the SVZ and migrate along the rostral migratory stream (RMS) towards the olfactory bulb (Lois and Alvarez-Buylla, 1994). Upon arrival, newly generated cells mature and integrate as interneurons into the granule cell and the glomerular layer of the olfactory bulb (Betarbet et al., 1996, Winner et al., 2002). Furthermore, our previous work showed that continuous overproduction and turnover of newly generated neurons is an important regulatory feature of the adult olfactory bulb neurogenesis (Winner et al., 2002). The functional integration of the newly generated interneurons into the olfactory neuronal circuitry (Carlen et al., 2002, Carleton et al., 2003) strongly suggests a relevance of adult neurogenesis in olfaction (Magavi et al., 2005).

To further elucidate the α-synuclein-associated pathological mechanisms, animal models for synucleinopathies have been established using various promoters to drive expression of α-synuclein. The neuropathological changes observed in these models included protein accumulation in different areas of the brain according to a promoter-dependent topography (Masliah et al., 2000, Rockenstein et al., 2002). For example, PDGF-promoter driven expression of mutant A53T α-synuclein has resulted in a transgenic mouse model with a severe motor impairment (Hashimoto et al., 2003). α-Synuclein is endogenously expressed in NSCs and their progeny in the adult SVZ/olfactory bulb system as well as in the hippocampus (Li et al., 2002, Winner et al., 2004). Recently, using a transgenic mouse line that expresses high levels of wild-type human α-synuclein (WTS) we reported that increased levels of WTS affected adult neurogenesis. In particular, increased α-synuclein, obtained via transgenic expression of human α-synuclein led to the death of immature neurons in the brain of adult animals, indicating that α-synuclein has an impact on adult neurogenesis (Winner et al., 2004).

Clinical manifestations of synucleinopathies occur to a large extent in advanced age. We therefore examined the impact of α-synuclein on olfactory bulb neurogenesis during aging using the models of (i) transgenic mice overexpressing WTS and (ii) transgenic mice expressing the mutant A53T α-synuclein (MTS), a mutation accelerating the α-synuclein aggregation.

Section snippets

Animals and paradigm

For the present study, aged human wild-type α-synuclein (aged WTS) and A53T mutant α-synuclein transgenic mice (aged MTS) were compared to littermate controls (aged CTR). The aged animals were 15 month-old at the beginning of the experiment (n = 6 per group) and their genetic background was C57BL6/DBA. The synuclein transgenic mice (WTS and MTS) expressed α-synuclein under the regulatory control of the PDGF-promoter and the WTS group belongs to the human WTS high expresser line D (Masliah et al.,

Synuclein expression in regions of adult neurogenesis

The transgenic animal models used in this study express human wild-type α-synuclein (WTS) or A53T mutant allele (MTS) in the olfactory bulb and the hippocampus, as well as in other regions of the adult brain (Hashimoto et al., 2003, Masliah et al., 2000). In adult WTS, human α-synuclein is coexpressed in DCX-immunopositive neuroblasts (Couillard-Despres et al., 2005, Winner et al., 2004). This expression pattern is also observed in the SVZ/rostral migratory stream of aged WTS mice (Fig. 1A–C,

Discussion

Considering the limited success for supplying exogenously cells for the treatment of PD (Freed et al., 2001), the interest in understanding the characteristics of endogenous neural precursor raised considerably over the past years (for examples, see Baker et al., 2004, Hoglinger et al., 2004). To further explore the potential of these cells, it is important to characterize the biology of endogenous NSCs populations in models of PD. Recently, we reported that adult neurogenesis is decreased in

Acknowledgements

Beate Winner was supported by fellowships sponsored by the “Hochschul-und Wissenschaftsprogramm” (University of Regensburg) and Glaxo-Smith-Kline (Munich, Germany). Moreover, this study was supported by the Adalbert Raps Foundation (Kulmbach, Germany), the Bavarian State Ministry of Sciences, Research and the Arts, ForNeuroCell (Regensburg, Germany), and the NIH grants AG18440 and AG10435 and AG022074. The authors thank Ludwig Aigner, Department of Neurology, University of Regensburg for

References (68)

  • C.G. Specht et al.

    Subcellular localisation of recombinant alpha- and gamma-synuclein

    Mol. Cell Neurosci.

    (2005)
  • T. Takenouchi et al.

    Reduced neuritic outgrowth and cell adhesion in neuronal cells transfected with human alpha-synuclein

    Mol. Cell Neurosci.

    (2001)
  • B. Winner et al.

    Striatal deafferentation increases dopaminergic neurogenesis in the adult olfactory bulb

    Exp. Neurol.

    (2006)
  • M. Yamada et al.

    Neurogenesis in olfactory bulb identified by retroviral labeling in normal and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated adult mice

    Neuroscience

    (2004)
  • M.A. Alim et al.

    Demonstration of a role for alpha-synuclein as a functional microtubule-associated protein

    J. Alzheimers Dis.

    (2004)
  • A. Arvidsson et al.

    Neuronal replacement from endogenous precursors in the adult brain after stroke

    Nat. Med.

    (2002)
  • S.A. Baker et al.

    Dopaminergic nigrostriatal projections regulate neural precursor proliferation in the adult mouse subventricular zone

    Eur. J. Neurosci.

    (2004)
  • H.W. Berendse et al.

    Subclinical dopaminergic dysfunction in asymptomatic Parkinson's disease patients’ relatives with a decreased sense of smell

    Ann. Neurol.

    (2001)
  • M. Biebl et al.

    Caspase inhibition decreases cell death in regions of adult neurogenesis

    Neuroreport

    (2005)
  • J. Brown et al.

    Enriched environment and physical activity stimulate hippocampal but not olfactory bulb neurogenesis

    Eur. J. Neurosci.

    (2003)
  • H.A. Cameron et al.

    Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus

    J. Comp. Neurol.

    (2001)
  • A. Carleton et al.

    Becoming a new neuron in the adult olfactory bulb

    Nat. Neurosci.

    (2003)
  • S. Chandra et al.

    Double-knockout mice for alpha- and beta-synucleins: effect on synaptic functions

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

    (2004)
  • K.A. Conway et al.

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

    Nat. Med.

    (1998)
  • S. Couillard-Despres et al.

    Doublecortin expression levels in adult brain reflect neurogenesis

    Eur. J. Neurosci.

    (2005)
  • V. Darsalia et al.

    Stroke-induced neurogenesis in aged brain

    Stroke

    (2005)
  • T.M. Dawson et al.

    Molecular pathways of neurodegeneration in Parkinson's disease

    Science

    (2003)
  • E. Enwere et al.

    Aging results in reduced epidermal growth factor receptor signaling, diminished olfactory neurogenesis, and deficits in fine olfactory discrimination

    J. Neurosci.

    (2004)
  • C.R. Freed et al.

    Transplantation of embryonic dopamine neurons for severe Parkinson's disease

    N. Engl. J. Med.

    (2001)
  • J. Goers et al.

    Nuclear localization of alpha-synuclein and its interaction with histones

    Biochemistry

    (2003)
  • H.J. Gundersen et al.

    Some new, simple and efficient stereological methods and their use in pathological research and diagnosis

    Apmis

    (1988)
  • M. Hashimoto et al.

    Alpha-synuclein in Lewy body disease and Alzheimer's disease

    Brain Pathol.

    (1999)
  • M. Hashimoto et al.

    Transgenic models of alpha-synuclein pathology: past, present, and future

    Ann. NY Acad. Sci.

    (2003)
  • C. Hawkes

    Olfaction in neurodegenerative disorder

    Adv. Otorhinolaryngol.

    (2006)
  • Cited by (104)

    • Mesenchymal stem cell-derived exosomes altered neuron cholesterol metabolism via Wnt5a-LRP1 axis and alleviated cognitive impairment in a progressive Parkinson's disease model

      2022, Neuroscience Letters
      Citation Excerpt :

      Among them, the A53T missense mutation of α-Syn is considered to be most closely related to the occurrence and development of PD, which can lead to the formation of intraneuronal inclusion bodies and the occurrence of apoptosis. A53Tα-Syn transgenic mice or A53Tα-Syn overexpressing mouse models can exhibit severe dyskinesia, mimicking the clinical symptoms and neuropathological processes of PD [4,32]. Specifically, survival of hippocampal adult newborn neurons is regulated by synaptic input and was reduced in A53T mutant α-syn mouse model.

    • Neurogenesis in the damaged mammalian brain

      2020, Patterning and Cell Type Specification in the Developing CNS and PNS: Comprehensive Developmental Neuroscience, Second Edition
    View all citing articles on Scopus
    View full text