Elsevier

Neurobiology of Aging

Volume 33, Issue 3, March 2012, Pages 629.e5-629.e18
Neurobiology of Aging

Genetic reports abstract
DLB and PDD: a role for mutations in dementia and Parkinson disease genes?

https://doi.org/10.1016/j.neurobiolaging.2011.10.014Get rights and content

Abstract

Based on the substantial overlap in clinical and pathological characteristics of dementia with Lewy bodies (DLB) and Parkinson disease with dementia (PDD) with Alzheimer disease (AD) and Parkinson disease (PD) we hypothesized that these disorders might share underlying genetic factors. The contribution of both sequence and copy number variants (CNVs) in known AD and PD genes to the genetic etiology of DLB and PDD however is currently unclear. Therefore, we performed a gene-based mutation analysis of all major AD and PD genes in 99 DLB and 75 PDD patients, including familial and sporadic forms, from Flanders, Belgium. Also, copy number variants in APP, SNCA, and PARK2 were determined. In the AD genes we detected proven pathogenic missense mutations in PSEN1 and PSEN2, and 2 novel missense variants in PSEN2 and MAPT. In the PD genes we identified 1 SNCA duplication, the LRRK2 R1441C founder mutation and 4 novel heterozygous missense variants with unknown pathogenicity. Our results suggest a contribution of established AD and PD genes to the genetic etiology of DLB and PDD though to a limited extent. They do support the hypothesis of a genetic overlap between members of the Lewy body disease spectrum, but additional genes still have to exist.

Introduction

Dementia with Lewy bodies (DLB) and Parkinson disease with dementia (PDD) are recognized as diverse manifestations of a distinctive disease process leading to the abnormal processing and aggregation of the synaptic protein α-synuclein (Baba et al., 1998). The core clinical features of DLB and PDD are very similar and both diseases are primarily distinguished by the different temporal manifestation of dementia (McKeith et al., 1996). By definition, patients developing dementia prior to parkinsonism or during the first year of disease are diagnosed with DLB. In PDD patients the onset of motor symptoms precedes dementia by at least 1 year. DLB is the second most frequent neurodegenerative dementia with an estimated prevalence up to 30% (Zaccai et al., 2005) of all dementias, which is confirmed by neuropathological examination (Parkkinen et al., 2001). In contrast, the prevalence of PDD is stated to be only 3.6% of all dementia cases based on studies focusing on dementia (Aarsland et al., 2005). However, up to 30% of all Parkinson disease (PD) patients develop dementia during the course of their disease (Aarsland et al., 2003), increasing the actual prevalence of PDD. The concept of DLB and PDD being separate conditions is relatively new and to date there are no markers that unequivocally differentiate both diseases.

Over the last 15 years, DLB and PDD research has mainly focused on clinical and pathological issues related to these spectrum disorders in order to establish appropriate diagnostic criteria for improved patient detection in routine patient care and clinical interventions. Amyloid beta (Aβ) deposition appears to be more frequent and widespread in DLB and is considered a potential pathological marker for differentiating DLB from PDD (Jellinger and Attems, 2006). In line with this finding, the use of Pittsburgh Compound B (PIB)-positron emission tomography (PET) neuroimaging for screening the cortical Aβ burden was shown to be a promising diagnostic tool (Gomperts et al., 2008, Maetzler et al., 2009). In addition, 1 study reported significantly higher levels of oxidized Aβ40 in cerebrospinal fluid (CSF) of DLB patients compared with PDD patients (Bibl et al., 2006).

Molecular genetic etiology studies of both diseases may also prove to be valuable in the identification of disease markers because a considerable part of our current understanding on the pathomechanisms of overlapping neurodegenerative brain diseases (NBD) originates from genetic findings. Although DLB and PDD are mainly envisaged to be sporadic diseases, families in which a mixed phenotype of dementia and parkinsonism is inherited in a Mendelian manner are reported as well (Bogaerts et al., 2007, Bonner et al., 2003, Brett et al., 2002, Denson et al., 1997, Galvin et al., 2002, Golbe et al., 1990, Ohara et al., 1999, Tsuang et al., 2002, Wakabayashi et al., 1998, Waters and Miller, 1994). A recent study even observed a statistical significant aggregation of DLB and its core features within families (Nervi et al., 2011). Genetic research in a limited number of these families already supported a potential role for genes that are known to be implicated in classic forms of either Alzheimer disease (AD; APP [Guyant-Marechal et al., 2008], PSEN1 [Ishikawa et al., 2005], PSEN2 [Piscopo et al., 2008], PGRN [Benussi et al., 2009], PRNP [Koide et al., 2002]) or PD (SNCA [Morfis and Cordato, 2006, Nishioka et al., 2010, Singleton et al., 2003, Zarranz et al., 2004], SNCB [Nishioka et al., 2010, Ohtake et al., 2004], LRRK2 [Haubenberger et al., 2007, Ross et al., 2006], GBA [Clark et al., 2009, Farrer et al., 2009, Goker-Alpan et al., 2006, Mata et al., 2008]) in the development of DLB and PDD. Despite these strong hints for a genetic component in the etiology of both diseases and their relatively high prevalence, comprehensive mutation analyses of all major dementia and PD related genes in extended, well-phenotyped clinical and/or pathologically confirmed DLB and PDD populations are still lagging behind. The primary aim of this study was to systematically determine whether mutations in dementia and PD genes play a role in the development of DLB and PDD in Flanders, Belgium.

Section snippets

Study cohort

Clinical and genetic studies described in this report were approved by the medical ethical committee of the Hospital Network Antwerp (ZNA), the University Hospital of Antwerp (UZA), the University of Antwerp and the University Hospitals of Leuven, Belgium. Upon written informed consent from patients and healthy control individuals, blood samples were collected for DNA extraction, generation of Epstein-Barr virus (EBV)-transformed lymphoblast cell lines, and serum and plasma collection.

Results

Clinical and pathological features of DLB and PDD overlap substantially with AD and PD suggesting that the genetic etiologies may intertwine as well. Therefore, we focused our mutation analysis on genes which are screened on a routine basis in patients referred to the Diagnostic Service Facility for molecular diagnostics of AD and PD (www.molgen.ua.ac.be/DNADiagnostics/). Also, we investigated the contribution of 2 major risk genes, APOE for AD and GBA for PD, to the development of DLB or PDD.

Discussion

In literature there is a consensus that DLB and PDD show a marked overlap at a clinical and pathological level with AD (dementia and Aβ deposition) and PD (parkinsonism and Lewy body formation), which in turn suggests they share pathogenic or underlying disease mechanisms (Lippa et al., 2007). Extending this assumption, we stipulated that genetic commonalities between these brain diseases can be anticipated as well.

The rationale of this study originated from the growing perception that

Disclosure statement

The authors disclose no conflicts of interest.

Clinical and genetic studies described in this report were approved by the medical ethical committee of the Hospital Network Antwerp (ZNA), the University Hospital of Antwerp (UZA), the University of Antwerp and the University Hospitals of Leuven, Belgium. Patients and healthy control individuals provided written informed consent.

Acknowledgements

The work was made possible by the generous participation of the Flanders-Belgian control individuals, patients, and their families. We further acknowledge the contribution of the personnel of the VIB Genetic Service Facility (www.vibgeneticservicefacility.be) and the Biobank of the Institute Born-Bunge (www.bornbunge.be/Home/index_en.shtml). This research was in part supported by the Methusalem excellence program of the Flemish Government; a Centre of Excellence grant by the Special Research

References (108)

  • C.F. Lippa et al.

    DLB and PDD boundary issues: diagnosis, treatment, molecular pathology, and biomarkers

    Neurology

    (2007)
  • W. Maetzler et al.

    Cortical PIB binding in Lewy body disease is associated with Alzheimer-like characteristics

    Neurobiol. Dis

    (2009)
  • I.F. Mata et al.

    Glucocerebrosidase gene mutations: a risk factor for Lewy body disorders

    Arch. Neurol

    (2008)
  • L. Morfis et al.

    Dementia with Lewy bodies in an elderly Greek male due to alpha-synuclein gene mutation

    J. Clin. Neurosci

    (2006)
  • A. Nervi et al.

    Familial aggregation of dementia with lewy bodies

    Arch. Neurol

    (2011)
  • K. Nishioka et al.

    Association of alpha-, beta-, and gamma-Synuclein with diffuse lewy body disease

    Arch. Neurol

    (2010)
  • O.A. Ross et al.

    Lrrk2 and Lewy body disease

    Ann. Neurol

    (2006)
  • D. Aarsland et al.

    Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study

    Arch. Neurol

    (2003)
  • D. Aarsland et al.

    A systematic review of prevalence studies of dementia in Parkinson's disease

    Mov. Disord

    (2005)
  • T.B. Ahn et al.

    alpha-Synuclein gene duplication is present in sporadic Parkinson disease

    Neurology

    (2008)
  • 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)
  • M. Bibl et al.

    CSF amyloid-beta-peptides in Alzheimer's disease, dementia with Lewy bodies and Parkinson's disease dementia

    Brain

    (2006)
  • V. Bogaerts et al.

    A novel locus for dementia with Lewy bodies: a clinically and genetically heterogeneous disorder

    Brain

    (2007)
  • L.T. Bonner et al.

    Familial dementia with Lewy bodies with an atypical clinical presentation

    J. Geriatr. Psychiatry Neurol

    (2003)
  • F.M. Brett et al.

    Familial diffuse Lewy body disease, eye movement abnormalities, and distribution of pathology

    Arch. Neurol

    (2002)
  • N. Brouwers et al.

    Genetic variability in progranulin contributes to risk for clinically diagnosed Alzheimer disease

    Neurology

    (2008)
  • N. Brouwers et al.

    Molecular genetics of Alzheimer's disease: an update

    Ann. Med

    (2008)
  • R. Calabrese et al.

    Functional annotations improve the predictive score of human disease-related mutations in proteins

    Hum. Mutat

    (2009)
  • J. Clarimón et al.

    Early-onset familial lewy body dementia with extensive tauopathy: a clinical, genetic, and neuropathological study

    J. Neuropathol. Exp. Neurol

    (2009)
  • L.N. Clark et al.

    Association of glucocerebrosidase mutations with dementia with lewy bodies

    Arch. Neurol

    (2009)
  • L.N. Clark et al.

    Mutations in the glucocerebrosidase gene are associated with early-onset Parkinson disease

    Neurology

    (2007)
  • E.H. Corder et al.

    Protective effect of apolipoprotein E type 2 allele for late onset Alzheimer disease

    Nat. Genet

    (1994)
  • E.H. Corder et al.

    Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families

    Science

    (1993)
  • D. Crosiers et al.

    Self-reported non-motor symptoms in a cohort of 139 Parkinson's disease patients

    Mov. Disord

    (2010)
    Crosiers, D., Meeus, B., Nuytemans, K., Van Broeckhoven, C., Theuns, J., Cras, P., 2010. Self-reported non-motor...
  • M.A. Denson et al.

    Familial parkinsonism, dementia, and Lewy body disease: study of family G

    Ann. Neurol

    (1997)
  • B. Dermaut et al.

    A novel presenilin 1 mutation associated with Pick's disease but not beta-amyloid plaques

    Ann. Neurol

    (2004)
  • B. Dermaut et al.

    Tau is central in the genetic Alzheimer-frontotemporal dementia spectrum

    Trends Genet

    (2005)
  • S. Engelborghs et al.

    Prospective Belgian study of neurodegenerative and vascular dementia: APOE genotype effects

    J. Neurol. Neurosurg. Psychiatry

    (2003)
  • S. Engelborghs et al.

    Dose dependent effect of APOE epsilon4 on behavioral symptoms in frontal lobe dementia

    Neurobiol. Aging

    (2006)
  • M.J. Farrer et al.

    Glucosidase-beta variations and Lewy body disorders

    Parkinsonism Relat. Disord

    (2009)
  • J. Fuchs et al.

    Phenotypic variation in a large Swedish pedigree due to SNCA duplication and triplication

    Neurology

    (2007)
  • D. Galasko et al.

    The apolipoprotein E allele epsilon 4 is overrepresented in patients with the Lewy body variant of Alzheimer's disease

    Neurology

    (1994)
  • J.E. Galvin et al.

    Familial dementia with Lewy bodies: clinicopathologic analysis of two kindreds

    Neurology

    (2002)
  • Z. Gan-Or et al.

    Differential phenotype in Parkinson's disease patients with severe versus mild GBA mutations

    Brain

    (2009)
  • Z. Gan-Or et al.

    Genotype-phenotype correlations between GBA mutations and Parkinson disease risk and onset

    Neurology

    (2008)
  • O. Goker-Alpan et al.

    Glucocerebrosidase mutations are an important risk factor for Lewy body disorders

    Neurology

    (2006)
  • L.I. Golbe et al.

    A large kindred with autosomal dominant Parkinson's disease

    Ann. Neurol

    (1990)
  • E. Gómez-Tortosa et al.

    Clinical-genetic correlations in familial Alzheimer's disease caused by presenilin 1 mutations

    J. Alzheimers Dis

    (2010)
  • S.N. Gomperts et al.

    Imaging amyloid deposition in Lewy body diseases

    Neurology

    (2008)
  • R.J. Guerreiro et al.

    Genetic screening of Alzheimer's disease genes in Iberian and African samples yields novel mutations in presenilins and APP

    Neurobiol. Aging

    (2010)
  • Cited by (0)

    View full text