Adult neurogenesis and neurite outgrowth are impaired in LRRK2 G2019S mice

https://doi.org/10.1016/j.nbd.2010.12.008Get rights and content

Abstract

The generation and maturation of adult neural stem/progenitor cells are impaired in many neurodegenerative diseases, among them is Parkinson's disease (PD). In mammals, including humans, adult neurogenesis is a lifelong feature of cellular brain plasticity in the hippocampal dentate gyrus (DG) and in the subventricular zone (SVZ)/olfactory bulb system. Hyposmia, depression, and anxiety are early non-motor symptoms in PD. There are parallels between brain regions associated with non-motor symptoms in PD and neurogenic regions. In autosomal dominant PD, mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are frequent. LRRK2 homologs in non-vertebrate systems play an important role in chemotaxis, cell polarity, and neurite arborization.

We investigated adult neurogenesis and the neurite development of new neurons in the DG and SVZ/olfactory bulb system in bacterial artificial chromosome (BAC) human Lrrk2 G2019S transgenic mice. We report that mutant human Lrrk2 is highly expressed in the hippocampus in the DG and the SVZ of adult Lrrk2 G2019S mice. Proliferation of newly generated cells is significantly decreased and survival of newly generated neurons in the DG and olfactory bulb is also severely impaired. In addition, after stereotactic injection of a GFP retrovirus, newly generated neurons in the DG of Lrrk2 G2019S mice exhibited reduced dendritic arborization and fewer spines. This loss in mature, developed spines might point towards a decrease in synaptic connectivity. Interestingly, physical activity partially reverses the decrease in neuroblasts observed in Lrrk2 G2010S mice. These data further support a role for Lrrk2 in neuronal morphogenesis and provide new insights into the role of Lrrk2 in adult neurogenesis.

Research Highlights

►Decrease in olfactory and hippocampal adult neurogenesis in LRRK2 G2019S transgenic mice for proliferation and survival of new neurons. ►Impairment in neurite development of the newly generated neurons; ►Impaiment of spine maturation and ►Reversibility of reduced hippocampal neurogenesis by physical activity.

Introduction

Mutations in leucine-rich repeat kinase 2 (LRRK2) predispose carriers to Parkinson's disease (PD); (Zimprich et al., 2004a, Mata et al., 2006). Lrrk2 G2019S is found in more than 30% of patients of certain ethnicities and has the highest genotype- and population-attributable risk (Hulihan et al., 2008, Healy et al., 2008).

LRRK2 is comprised of 51 exons that encode 2527 amino acids and has a molecular mass of 286 kDa. Lrrk2 is a large, multi-domain protein kinase and consists of an ankyrin repeat region, leucine-rich repeats, a Roc GTPase domain, and a C terminal of Roc (COR) domain. In addition, a serine/threonine protein kinase domain with a high degree of homology with MAPKKK (mitogen-activated protein kinase kinase kinase, where the G2019S mutation is located) and a WD40 domain are present (Mata et al., 2006, Zimprich et al., 2004b). While Lrrk2 transgenic models are yet to recapitulate human PD neuropathology in aspects such as increased cell death of dopaminergic neurons or Lewy bodies, subtle phenotypes including impaired regulation of neurite growth and arborization have been reported. In non-vertebrate systems a role of LRRK2 homologs for chemotaxis, polarization, and regulation of axonal–dendritic polarity of synaptic vesicle proteins was described (van Egmond et al., 2008, Sakaguchi-Nakashima et al., 2007). In primary neuronal cultures, in utero transfection or AAV2 viral injection, mutant Lrrk2 slows axonal outgrowth and reduces neurite length and branching, whereas silencing of LRRK2 leads to the opposite effects (Li et al., 2009, MacLeod et al., 2006, Plowey et al., 2008).

In humans and mice, Lrrk2 is highly expressed in the hippocampus and subventricular zone (SVZ) and colocalizes with the migrating neuroblast marker PSA-NCAM (Melrose et al., 2007). The hippocampal dentate gyrus (DG) and SVZ/olfactory bulb system are the physiological areas of neurogenesis in the adult brain (Altman and Das, 1965, Curtis et al., 2007, Eriksson et al., 1998). In PD and transgenic SNCA mouse models of parkinsonism, the proliferation of dividing cells is impaired in the DG and SVZ/olfactory bulb system (Crews et al., 2008, Hoglinger et al., 2004, Winner et al., 2004). PD affects multiple neurotransmitter systems and includes a variety of non-motor symptoms (Langston, 2006). Frequently observed early symptoms in PD include hyposmia, anxiety, anhedonia, and depression (Tolosa and Poewe, 2009). These symptoms are linked to neuropathological alterations in the olfactory and limbic system, including the olfactory bulb and the hippocampus (Rodriguez-Oroz et al., 2009).

In this study, we take advantage of the hippocampal and SVZ transgene expression in G2019S Lrrk2 mice to study the impact of mutated Lrrk2 on newly generated neurons in the adult brain. With the benefit of inherent Lrrk2 expression, we examine the impact of aberrant human Lrrk2 on adult neurogenesis. We determine neurite outgrowth, and spine numbers in newly generated neurons in the hippocampal DG in vivo and test whether physical activity impacts this system.

Section snippets

Animals

All animal procedures were approved by the Mayo Clinic Institutional Animal Care and Use Committee (IACUC) and were in accordance with the National Institute of Health Guide for the Care and Use of Laboratory Animals (NIH Publications No. 80-23) revised 1996. All mice were kept in a normal light/dark cycle (12 h light/12 h dark) and had free access to food and water.

Generation of BAC transgenic mice

A Bacterial Artificial Chromosome (BAC) (RP-11 568G5) containing the entire human wild-type LRRK2 gene and regulatory sequences was

Human G2019S Lrrk2 is highly expressed in regions of adult neurogenesis

Regional expression of transgenic LRRK2 mRNA was assessed with a human-specific Taqman probe and revealed that the highest transgene expression in adult G2019S mice occurred in the hippocampal formation (Fig. 1A). Finer anatomical mapping using in situ hybridization with a LRRK2 human-specific probe confirmed that high expression was found in the Cornu Ammonis fields and, importantly, in the DG (Fig. 1B). Consistent with mRNA results, immunoblotting with a Lrrk2 specific antibody, PA0362,

Discussion

We have used a mouse model that expresses a high level of human Lrrk2 in regions of adult neurogenesis and endogenous Lrrk2 expression. We reported a significant decrease in SVZ/olfactory bulb and hippocampal adult neurogenesis in Lrrk2 G2019S transgenic mice, both at the level of proliferation and survival of newly generated cells. Moreover, impaired adult neurogenesis was accompanied by structural alterations of the newly generated cells in the DG, with decreases in neurite length and in

Acknowledgments

The authors would like to thank Ralf Burgmayer and Chris Tse for excellent technical support, J. Ecke, Munich for the artwork, and M.L. Gage for editorial comments. Funding support was provided by grants from the Bavarian State Ministry of Sciences, Research and the Arts, ForNeuroCell grant (JW), the Federal Ministry of Education and Research 01GN0979, the Mayo Clinic, the Lookout Fund, NIH Grants NIA AG17216 and NINDS NS40256, The Pacific Alzheimer's Research Foundation, Michael J Fox

References (65)

  • A. Persson et al.

    Expression of ezrin radixin moesin proteins in the adult subventricular zone and the rostral migratory stream

    Neuroscience

    (2010)
  • A.A. Pieper et al.

    Discovery of a proneurogenic, neuroprotective chemical

    Cell

    (2010)
  • M.C. Rodriguez-Oroz et al.

    Initial clinical manifestations of Parkinson's disease: features and pathophysiological mechanisms

    Lancet Neurol.

    (2009)
  • A. Sakaguchi-Nakashima et al.

    LRK-1, a C. elegans PARK8-related kinase, regulates axonal–dendritic polarity of SV proteins

    Curr. Biol.

    (2007)
  • J.L. Tillerson et al.

    Exercise induces behavioral recovery and attenuates neurochemical deficits in rodent models of Parkinson's disease

    Neuroscience

    (2003)
  • W.N. van Egmond et al.

    Intramolecular activation mechanism of the Dictyostelium LRRK2 homolog Roco protein GbpC

    J. Biol. Chem.

    (2008)
  • B. Winner et al.

    Striatal deafferentation increases dopaminergic neurogenesis in the adult olfactory bulb

    Exp. Neurol.

    (2006)
  • B. Winner et al.

    Mutant alpha-synuclein exacerbates age-related decrease of neurogenesis

    Neurobiol. Aging

    (2008)
  • A. Zimprich et al.

    Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology

    Neuron

    (2004)
  • A. Zimprich et al.

    The PARK8 locus in autosomal dominant parkinsonism: confirmation of linkage and further delineation of the disease-containing interval

    Am. J. Hum. Genet.

    (2004)
  • J. Altman et al.

    Post-natal origin of microneurones in the rat brain

    Nature

    (1965)
  • M. Babyak et al.

    Exercise treatment for major depression: maintenance of therapeutic benefit at 10 months

    Psychosom. Med.

    (2000)
  • E. Bruel-Jungerman et al.

    Inhibition of PI3K-Akt signaling blocks exercise-mediated enhancement of adult neurogenesis and synaptic plasticity in the dentate gyrus

    PLoS ONE

    (2009)
  • C.D. Clelland et al.

    Cell therapy in Huntington disease

    Neurosurg. Focus

    (2008)
  • S. Couillard-Despres et al.

    Doublecortin expression levels in adult brain reflect neurogenesis

    Eur. J. Neurosci.

    (2005)
  • L. Crews et al.

    Alpha-synuclein alters Notch-1 expression and neurogenesis in mouse embryonic stem cells and in the hippocampus of transgenic mice

    J. Neurosci.

    (2008)
  • M.A. Curtis et al.

    Human neuroblasts migrate to the olfactory bulb via a lateral ventricular extension

    Science

    (2007)
  • P.S. Eriksson et al.

    Neurogenesis in the adult human hippocampus

    Nat. Med.

    (1998)
  • K. Fabel et al.

    VEGF is necessary for exercise-induced adult hippocampal neurogenesis

    Eur. J. Neurosci.

    (2003)
  • C.J. Gloeckner et al.

    The Parkinson disease-associated protein kinase LRRK2 exhibits MAPKKK activity and phosphorylates MKK3/6 and MKK4/7, in vitro

    J. Neurochem.

    (2009)
  • H.J. Gundersen et al.

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

    APMIS

    (1988)
  • G.U. Hoglinger et al.

    Dopamine depletion impairs precursor cell proliferation in Parkinson disease

    Nat. Neurosci.

    (2004)
  • Cited by (156)

    • Mitochondrial function and dynamics in neural stem cells and neurogenesis: Implications for neurodegenerative diseases

      2022, Ageing Research Reviews
      Citation Excerpt :

      LRRK2 is a kinase that belongs to ezrin, radixin, and moesin (ERM) family of proteins, implicated in neurite growth regulation and consequently neuronal morphogenesis (Parisiadou et al., 2009). Several studies have demonstrated that a mutation in LRRK2 leading to a gain of function of the protein leads to a decrease in neurite length/outgrowth, while deletion of LRRK2 is accompanied by an increase of neurite length and arborization (Dächsel et al., 2010; Heo et al., 2010; Kawakami et al., 2012; Maekawa et al., 2012; Parisiadou et al., 2009; Winner et al., 2011). In LRRK2-deficient mouse ESCs, the differential gene expression profiling shows a faster silencing of pluripotency-associated genes during retinoic acid-induced differentiation, suggesting that LRRK2 promotes neuronal differentiation (Schulz et al., 2011).

    View all citing articles on Scopus
    1

    Contributed equally.

    View full text