Skip to main content
Top
Published in: Current Neurology and Neuroscience Reports 10/2018

Open Access 01-10-2018 | Dementia (K S Marder, Section Editor)

The Genetics of Dementia with Lewy Bodies: Current Understanding and Future Directions

Authors: Tatiana Orme, Rita Guerreiro, Jose Bras

Published in: Current Neurology and Neuroscience Reports | Issue 10/2018

Login to get access

Abstract

Purpose of Review

Dementia with Lewy bodies (DLB) is a neurodegenerative disease that can be clinically and pathologically similar to Parkinson’s disease (PD) and Alzheimer’s disease (AD). Current understanding of DLB genetics is insufficient and has been limited by sample size and difficulty in diagnosis. The first genome-wide association study (GWAS) in DLB was performed in 2017; a time at which the post-GWAS era has been reached in many diseases.

Recent Findings

DLB shares risk loci with AD, in the APOE E4 allele, and with PD, in variation at GBA and SNCA. Interestingly, the GWAS suggested that DLB may also have genetic risk factors that are distinct from those in AD and PD.

Summary

Although off to a slow start, recent studies have reinvigorated the field of DLB genetics and these results enable us to start to have a more complete understanding of the genetic architecture of this disease.
Literature
1.
go back to reference •• McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor J-P, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB Consortium. Neurology. 2017;89:88–100. The diagnostic criteria for DLB were updated in this manuscript to rely more heavily on biomarkers to aid diagnosis. REM sleep behaviour disorder was made a core feature. PubMedPubMedCentralCrossRef •• McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor J-P, Weintraub D, et al. Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB Consortium. Neurology. 2017;89:88–100. The diagnostic criteria for DLB were updated in this manuscript to rely more heavily on biomarkers to aid diagnosis. REM sleep behaviour disorder was made a core feature. PubMedPubMedCentralCrossRef
2.
go back to reference McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65:1863–72.PubMedCrossRef McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology. 2005;65:1863–72.PubMedCrossRef
3.
go back to reference Singleton AB, Wharton A, O’Brien KK, Walker MP, McKeith IG, Ballard CG, et al. Clinical and neuropathological correlates of apolipoprotein E genotype in dementia with Lewy bodies. Dement Geriatr Cogn Disord. 2002;14:167–75.PubMedCrossRef Singleton AB, Wharton A, O’Brien KK, Walker MP, McKeith IG, Ballard CG, et al. Clinical and neuropathological correlates of apolipoprotein E genotype in dementia with Lewy bodies. Dement Geriatr Cogn Disord. 2002;14:167–75.PubMedCrossRef
4.
go back to reference Price A, Farooq R, Yuan J-M, Menon VB, Cardinal RN, O’Brien JT. Mortality in dementia with Lewy bodies compared with Alzheimer’s dementia: a retrospective naturalistic cohort study. BMJ Open. 2017;7:e017504.PubMedPubMedCentralCrossRef Price A, Farooq R, Yuan J-M, Menon VB, Cardinal RN, O’Brien JT. Mortality in dementia with Lewy bodies compared with Alzheimer’s dementia: a retrospective naturalistic cohort study. BMJ Open. 2017;7:e017504.PubMedPubMedCentralCrossRef
6.
go back to reference Ohara K, Takauchi S, Kokai M, Morimura Y, Nakajima T, Morita Y. Familial dementia with Lewy bodies (DLB). Clin Neuropathol. 1999;18:232–9.PubMed Ohara K, Takauchi S, Kokai M, Morimura Y, Nakajima T, Morita Y. Familial dementia with Lewy bodies (DLB). Clin Neuropathol. 1999;18:232–9.PubMed
7.
go back to reference Galvin JE, Lee SL, Perry A, Havlioglu N, McKeel DW Jr, Morris JC. Familial dementia with Lewy bodies: clinicopathologic analysis of two kindreds. Neurology. 2002;59:1079–82.PubMedCrossRef Galvin JE, Lee SL, Perry A, Havlioglu N, McKeel DW Jr, Morris JC. Familial dementia with Lewy bodies: clinicopathologic analysis of two kindreds. Neurology. 2002;59:1079–82.PubMedCrossRef
8.
9.
go back to reference Clarimón J, Molina-Porcel L, Gómez-Isla T, Blesa R, Guardia-Laguarta C, González-Neira A, et al. Early-onset familial Lewy body dementia with extensive tauopathy: a clinical, genetic, and neuropathological study. J Neuropathol Exp Neurol. 2009;68:73–82.PubMedCrossRef Clarimón J, Molina-Porcel L, Gómez-Isla T, Blesa R, Guardia-Laguarta C, González-Neira A, et al. Early-onset familial Lewy body dementia with extensive tauopathy: a clinical, genetic, and neuropathological study. J Neuropathol Exp Neurol. 2009;68:73–82.PubMedCrossRef
10.
go back to reference Bonner LT, Tsuang DW, Cherrier MM, Eugenio CJ, Du Jennifer Q, Steinbart EJ, et al. Familial dementia with Lewy bodies with an atypical clinical presentation. J Geriatr Psychiatry Neurol. 2003;16:59–64.PubMedPubMedCentralCrossRef Bonner LT, Tsuang DW, Cherrier MM, Eugenio CJ, Du Jennifer Q, Steinbart EJ, et al. Familial dementia with Lewy bodies with an atypical clinical presentation. J Geriatr Psychiatry Neurol. 2003;16:59–64.PubMedPubMedCentralCrossRef
11.
go back to reference Bogaerts V, Engelborghs S, Kumar-Singh S, Goossens D, Pickut B, van der Zee J, et al. A novel locus for dementia with Lewy bodies: a clinically and genetically heterogeneous disorder. Brain. 2007;130:2277–91.PubMedCrossRef Bogaerts V, Engelborghs S, Kumar-Singh S, Goossens D, Pickut B, van der Zee J, et al. A novel locus for dementia with Lewy bodies: a clinically and genetically heterogeneous disorder. Brain. 2007;130:2277–91.PubMedCrossRef
12.
go back to reference Denson MA, Wszolek ZK, Pfeiffer RF, Wszolek EK, Paschall TM, McComb RD. Familial parkinsonism, dementia, and Lewy body disease: study of family G. Ann Neurol. 1997;42:638–43.PubMedCrossRef Denson MA, Wszolek ZK, Pfeiffer RF, Wszolek EK, Paschall TM, McComb RD. Familial parkinsonism, dementia, and Lewy body disease: study of family G. Ann Neurol. 1997;42:638–43.PubMedCrossRef
13.
go back to reference Ishikawa A, Takahashi H, Tanaka H, Hayashi T, Tsuji S. Clinical features of familial diffuse Lewy body disease. Eur Neurol. 1997;38(Suppl 1):34–8.PubMedCrossRef Ishikawa A, Takahashi H, Tanaka H, Hayashi T, Tsuji S. Clinical features of familial diffuse Lewy body disease. Eur Neurol. 1997;38(Suppl 1):34–8.PubMedCrossRef
14.
go back to reference Hardy J, Crook R, Prihar G, Roberts G, Raghavan R, Perry R. Senile dementia of the Lewy body type has an apolipoprotein E epsilon 4 allele frequency intermediate between controls and Alzheimer’s disease. Neurosci Lett. 1994;182:1–2.PubMedCrossRef Hardy J, Crook R, Prihar G, Roberts G, Raghavan R, Perry R. Senile dementia of the Lewy body type has an apolipoprotein E epsilon 4 allele frequency intermediate between controls and Alzheimer’s disease. Neurosci Lett. 1994;182:1–2.PubMedCrossRef
15.
go back to reference Nalls MA, Duran R, Lopez G, Kurzawa-Akanbi M, McKeith IG, Chinnery PF, et al. A multicenter study of glucocerebrosidase mutations in dementia with Lewy bodies. JAMA Neurol. 2013;70:727–35.PubMedCrossRef Nalls MA, Duran R, Lopez G, Kurzawa-Akanbi M, McKeith IG, Chinnery PF, et al. A multicenter study of glucocerebrosidase mutations in dementia with Lewy bodies. JAMA Neurol. 2013;70:727–35.PubMedCrossRef
16.
go back to reference •• Guerreiro R, Ross OA, Kun-Rodrigues C, Hernandez DG, Orme T, Eicher JD, et al. Investigating the genetic architecture of dementia with Lewy bodies: a two-stage genome-wide association study. Lancet Neurol. 2018;17:64–74. This paper describes the first genome-wide association study in DLB and provides evidence for novel loci involved in disease. PubMedCrossRef •• Guerreiro R, Ross OA, Kun-Rodrigues C, Hernandez DG, Orme T, Eicher JD, et al. Investigating the genetic architecture of dementia with Lewy bodies: a two-stage genome-wide association study. Lancet Neurol. 2018;17:64–74. This paper describes the first genome-wide association study in DLB and provides evidence for novel loci involved in disease. PubMedCrossRef
17.
go back to reference Geiger JT, Arthur KC, Dawson TM, Rosenthal LS, Pantelyat A, Albert M, et al. C9orf72 Hexanucleotide repeat analysis in cases with pathologically confirmed dementia with Lewy bodies. Neurodegener Dis. 2016;16:370–2.PubMedPubMedCentralCrossRef Geiger JT, Arthur KC, Dawson TM, Rosenthal LS, Pantelyat A, Albert M, et al. C9orf72 Hexanucleotide repeat analysis in cases with pathologically confirmed dementia with Lewy bodies. Neurodegener Dis. 2016;16:370–2.PubMedPubMedCentralCrossRef
18.
go back to reference Snowden JS, Rollinson S, Lafon C, Harris J, Thompson J, Richardson AM, et al. Psychosis, C9ORF72 and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2012;83:1031–2.PubMedCrossRef Snowden JS, Rollinson S, Lafon C, Harris J, Thompson J, Richardson AM, et al. Psychosis, C9ORF72 and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2012;83:1031–2.PubMedCrossRef
19.
go back to reference Kun-Rodrigues C, Ross OA, Orme T, Shepherd C, Parkkinen L, Darwent L, et al. Analysis of C9orf72 repeat expansions in a large international cohort of dementia with Lewy bodies. Neurobiol Aging. 2017;49:214.e13–5.CrossRef Kun-Rodrigues C, Ross OA, Orme T, Shepherd C, Parkkinen L, Darwent L, et al. Analysis of C9orf72 repeat expansions in a large international cohort of dementia with Lewy bodies. Neurobiol Aging. 2017;49:214.e13–5.CrossRef
20.
go back to reference Blauwendraat C, Nalls MA, Federoff M, Pletnikova O, Ding J, Letson C, et al. ADORA1 mutations are not a common cause of Parkinson’s disease and dementia with Lewy bodies. Mov Disord. 2017;32:298–9.PubMedCrossRef Blauwendraat C, Nalls MA, Federoff M, Pletnikova O, Ding J, Letson C, et al. ADORA1 mutations are not a common cause of Parkinson’s disease and dementia with Lewy bodies. Mov Disord. 2017;32:298–9.PubMedCrossRef
21.
go back to reference Lorenzo-Betancor O, Ogaki K, Soto-Ortolaza AI, Labbe C, Walton RL, Strongosky AJ, et al. DNAJC13 p.Asn855Ser mutation screening in Parkinson’s disease and pathologically confirmed Lewy body disease patients. Eur J Neurol. 2015;22:1323–5.PubMedPubMedCentralCrossRef Lorenzo-Betancor O, Ogaki K, Soto-Ortolaza AI, Labbe C, Walton RL, Strongosky AJ, et al. DNAJC13 p.Asn855Ser mutation screening in Parkinson’s disease and pathologically confirmed Lewy body disease patients. Eur J Neurol. 2015;22:1323–5.PubMedPubMedCentralCrossRef
22.
go back to reference Walton RL, Soto-Ortolaza AI, Murray ME, Lorenzo-Betancor O, Ogaki K, Heckman MG, et al. TREM2 p.R47H substitution is not associated with dementia with Lewy bodies. Neurol Genet. 2016;2:e85.PubMedPubMedCentralCrossRef Walton RL, Soto-Ortolaza AI, Murray ME, Lorenzo-Betancor O, Ogaki K, Heckman MG, et al. TREM2 p.R47H substitution is not associated with dementia with Lewy bodies. Neurol Genet. 2016;2:e85.PubMedPubMedCentralCrossRef
23.
go back to reference Hodges K, Brewer SS, Labbé C, Soto-Ortolaza AI, Walton RL, Strongosky AJ, et al. RAB39B gene mutations are not a common cause of Parkinson’s disease or dementia with Lewy bodies. Neurobiol Aging. 2016;45:107–8.PubMedPubMedCentralCrossRef Hodges K, Brewer SS, Labbé C, Soto-Ortolaza AI, Walton RL, Strongosky AJ, et al. RAB39B gene mutations are not a common cause of Parkinson’s disease or dementia with Lewy bodies. Neurobiol Aging. 2016;45:107–8.PubMedPubMedCentralCrossRef
24.
go back to reference Labbé C, Heckman MG, Lorenzo-Betancor O, Soto-Ortolaza AI, Walton RL, Murray ME, et al. MAPT haplotype H1G is associated with increased risk of dementia with Lewy bodies. Alzheimers Dement. 2016;12:1297–304.PubMedPubMedCentralCrossRef Labbé C, Heckman MG, Lorenzo-Betancor O, Soto-Ortolaza AI, Walton RL, Murray ME, et al. MAPT haplotype H1G is associated with increased risk of dementia with Lewy bodies. Alzheimers Dement. 2016;12:1297–304.PubMedPubMedCentralCrossRef
25.
go back to reference Labbé C, Ogaki K, Lorenzo-Betancor O, Soto-Ortolaza AI, Walton RL, Rayaprolu S, et al. Role for the microtubule-associated protein tau variant p.A152T in risk of α-synucleinopathies. Neurology. 2015;85:1680–6.PubMedPubMedCentralCrossRef Labbé C, Ogaki K, Lorenzo-Betancor O, Soto-Ortolaza AI, Walton RL, Rayaprolu S, et al. Role for the microtubule-associated protein tau variant p.A152T in risk of α-synucleinopathies. Neurology. 2015;85:1680–6.PubMedPubMedCentralCrossRef
26.
go back to reference Heckman MG, Soto-Ortolaza AI, Contreras MYS, Murray ME, Pedraza O, Diehl NN, et al. LRRK2 variation and dementia with Lewy bodies. Parkinsonism Relat Disord. 2016;31:98–103.PubMedPubMedCentralCrossRef Heckman MG, Soto-Ortolaza AI, Contreras MYS, Murray ME, Pedraza O, Diehl NN, et al. LRRK2 variation and dementia with Lewy bodies. Parkinsonism Relat Disord. 2016;31:98–103.PubMedPubMedCentralCrossRef
27.
go back to reference Geiger JT, Ding J, Crain B, Pletnikova O, Letson C, Dawson TM, et al. Next-generation sequencing reveals substantial genetic contribution to dementia with Lewy bodies. Neurobiol Dis. 2016;94:55–62.PubMedPubMedCentralCrossRef Geiger JT, Ding J, Crain B, Pletnikova O, Letson C, Dawson TM, et al. Next-generation sequencing reveals substantial genetic contribution to dementia with Lewy bodies. Neurobiol Dis. 2016;94:55–62.PubMedPubMedCentralCrossRef
28.
go back to reference Bras J, Guerreiro R, Darwent L, Parkkinen L, Ansorge O, Escott-Price V, et al. Genetic analysis implicates APOE, SNCA and suggests lysosomal dysfunction in the etiology of dementia with Lewy bodies. Hum Mol Genet Oxford Univ Press. 2014;23:6139–46.CrossRef Bras J, Guerreiro R, Darwent L, Parkkinen L, Ansorge O, Escott-Price V, et al. Genetic analysis implicates APOE, SNCA and suggests lysosomal dysfunction in the etiology of dementia with Lewy bodies. Hum Mol Genet Oxford Univ Press. 2014;23:6139–46.CrossRef
29.
go back to reference Meeus B, Verstraeten A, Crosiers D, Engelborghs S, Van den Broeck M, Mattheijssens M, et al. DLB and PDD: a role for mutations in dementia and Parkinson disease genes? Neurobiol Aging. 2012;33:629.e5–629.e18.CrossRef Meeus B, Verstraeten A, Crosiers D, Engelborghs S, Van den Broeck M, Mattheijssens M, et al. DLB and PDD: a role for mutations in dementia and Parkinson disease genes? Neurobiol Aging. 2012;33:629.e5–629.e18.CrossRef
30.
go back to reference Keogh MJ, Kurzawa-Akanbi M, Griffin H, Douroudis K, Ayers KL, Hussein RI, et al. Exome sequencing in dementia with Lewy bodies. Transl Psychiatry. 2016;6:e728.PubMedPubMedCentralCrossRef Keogh MJ, Kurzawa-Akanbi M, Griffin H, Douroudis K, Ayers KL, Hussein RI, et al. Exome sequencing in dementia with Lewy bodies. Transl Psychiatry. 2016;6:e728.PubMedPubMedCentralCrossRef
31.
go back to reference Guella I, Evans DM, Szu-Tu C, Nosova E, Bortnick SF, Group SCS, et al. α-synuclein genetic variability: a biomarker for dementia in Parkinson disease. Ann Neurol. Wiley Online Library. 2016;79:991–9. Guella I, Evans DM, Szu-Tu C, Nosova E, Bortnick SF, Group SCS, et al. α-synuclein genetic variability: a biomarker for dementia in Parkinson disease. Ann Neurol. Wiley Online Library. 2016;79:991–9.
32.
go back to reference Keogh MJ, Wei W, Wilson I, Coxhead J, Ryan S, Rollinson S, et al. Genetic compendium of 1511 human brains available through the UK Medical Research Council Brain Banks Network Resource. Genome Res. 2017;27:165–73.PubMedPubMedCentralCrossRef Keogh MJ, Wei W, Wilson I, Coxhead J, Ryan S, Rollinson S, et al. Genetic compendium of 1511 human brains available through the UK Medical Research Council Brain Banks Network Resource. Genome Res. 2017;27:165–73.PubMedPubMedCentralCrossRef
33.
go back to reference Kosaka K (2017) Dementia with Lewy bodies: clinical and biological aspects. Springer Kosaka K (2017) Dementia with Lewy bodies: clinical and biological aspects. Springer
34.
go back to reference NCI-NHGRI Working Group on Replication in Association Studies, Chanock SJ, Manolio T, Boehnke M, Boerwinkle E, Hunter DJ, et al. Replicating genotype-phenotype associations. Nature. 2007;447:655–60.CrossRef NCI-NHGRI Working Group on Replication in Association Studies, Chanock SJ, Manolio T, Boehnke M, Boerwinkle E, Hunter DJ, et al. Replicating genotype-phenotype associations. Nature. 2007;447:655–60.CrossRef
35.
go back to reference Moskvina V, Harold D, Russo G, Vedernikov A, Sharma M, Saad M, et al. Analysis of genome-wide association studies of Alzheimer disease and of Parkinson disease to determine if these 2 diseases share a common genetic risk. JAMA Neurol. 2013;70:1268–76.PubMedPubMedCentral Moskvina V, Harold D, Russo G, Vedernikov A, Sharma M, Saad M, et al. Analysis of genome-wide association studies of Alzheimer disease and of Parkinson disease to determine if these 2 diseases share a common genetic risk. JAMA Neurol. 2013;70:1268–76.PubMedPubMedCentral
36.
go back to reference Guerreiro R, Escott-Price V, Darwent L, Parkkinen L, Ansorge O, Hernandez DG, et al. Genome-wide analysis of genetic correlation in dementia with Lewy bodies, Parkinson’s and Alzheimer’s diseases. Neurobiol Aging. 2016;38:214.e7–214.e10.CrossRef Guerreiro R, Escott-Price V, Darwent L, Parkkinen L, Ansorge O, Hernandez DG, et al. Genome-wide analysis of genetic correlation in dementia with Lewy bodies, Parkinson’s and Alzheimer’s diseases. Neurobiol Aging. 2016;38:214.e7–214.e10.CrossRef
37.
go back to reference Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Ampuero I, et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol. 2004;55:164–73.PubMedCrossRef Zarranz JJ, Alegre J, Gómez-Esteban JC, Lezcano E, Ros R, Ampuero I, et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann Neurol. 2004;55:164–73.PubMedCrossRef
38.
go back to reference Morfis L, Cordato DJ. Dementia with Lewy bodies in an elderly Greek male due to α-synuclein gene mutation. J Clin Neurosci. 2006;13:942–4.PubMedCrossRef Morfis L, Cordato DJ. Dementia with Lewy bodies in an elderly Greek male due to α-synuclein gene mutation. J Clin Neurosci. 2006;13:942–4.PubMedCrossRef
39.
go back to reference Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. Alpha-synuclein locus triplication causes Parkinson’s disease. Science. 2003;302:841.PubMedCrossRef Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. Alpha-synuclein locus triplication causes Parkinson’s disease. Science. 2003;302:841.PubMedCrossRef
40.
go back to reference Obi T, Nishioka K, Ross OA, Terada T, Yamazaki K, Sugiura A, et al. Clinicopathologic study of a SNCA gene duplication patient with Parkinson disease and dementia. Neurology. 2008;70:238–41.PubMedCrossRef Obi T, Nishioka K, Ross OA, Terada T, Yamazaki K, Sugiura A, et al. Clinicopathologic study of a SNCA gene duplication patient with Parkinson disease and dementia. Neurology. 2008;70:238–41.PubMedCrossRef
41.
go back to reference Nishioka K, Hayashi S, Farrer MJ, Singleton AB, Yoshino H, Imai H, et al. Clinical heterogeneity of alpha-synuclein gene duplication in Parkinson’s disease. Ann Neurol. 2006;59:298–309.PubMedCrossRef Nishioka K, Hayashi S, Farrer MJ, Singleton AB, Yoshino H, Imai H, et al. Clinical heterogeneity of alpha-synuclein gene duplication in Parkinson’s disease. Ann Neurol. 2006;59:298–309.PubMedCrossRef
42.
go back to reference Chartier-Harlin M-C, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet. 2004;364:1167–9.PubMedCrossRef Chartier-Harlin M-C, Kachergus J, Roumier C, Mouroux V, Douay X, Lincoln S, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet. 2004;364:1167–9.PubMedCrossRef
43.
go back to reference Ikeuchi T, Kakita A, Shiga A, Kasuga K, Kaneko H, Tan C-F, et al. Patients homozygous and heterozygous for SNCA duplication in a family with parkinsonism and dementia. Arch Neurol. 2008;65:514–9.PubMedCrossRef Ikeuchi T, Kakita A, Shiga A, Kasuga K, Kaneko H, Tan C-F, et al. Patients homozygous and heterozygous for SNCA duplication in a family with parkinsonism and dementia. Arch Neurol. 2008;65:514–9.PubMedCrossRef
44.
go back to reference Ibáñez P, Bonnet A-M, Débarges B, Lohmann E, Tison F, Pollak P, et al. Causal relation between alpha-synuclein gene duplication and familial Parkinson’s disease. Lancet. 2004;364:1169–71.PubMedCrossRef Ibáñez P, Bonnet A-M, Débarges B, Lohmann E, Tison F, Pollak P, et al. Causal relation between alpha-synuclein gene duplication and familial Parkinson’s disease. Lancet. 2004;364:1169–71.PubMedCrossRef
45.
go back to reference Rosborough K, Patel N, Kalia LV. α-Synuclein and Parkinsonism: updates and future perspectives. Curr Neurol Neurosci Rep. 2017;17:31.PubMedCrossRef Rosborough K, Patel N, Kalia LV. α-Synuclein and Parkinsonism: updates and future perspectives. Curr Neurol Neurosci Rep. 2017;17:31.PubMedCrossRef
46.
go back to reference Markopoulou K, Dickson DW, McComb RD, Wszolek ZK, Katechalidou L, Avery L, et al. Clinical, neuropathological and genotypic variability in SNCA A53T familial Parkinson’s disease. Variability in familial Parkinson's disease. Acta Neuropathol. 2008;116:25–35.PubMedPubMedCentralCrossRef Markopoulou K, Dickson DW, McComb RD, Wszolek ZK, Katechalidou L, Avery L, et al. Clinical, neuropathological and genotypic variability in SNCA A53T familial Parkinson’s disease. Variability in familial Parkinson's disease. Acta Neuropathol. 2008;116:25–35.PubMedPubMedCentralCrossRef
47.
go back to reference Pasanen P, Myllykangas L, Siitonen M, Raunio A, Kaakkola S, Lyytinen J, et al. Novel α-synuclein mutation A53E associated with atypical multiple system atrophy and Parkinson’s disease-type pathology. Neurobiol Aging. 2014;35:2180.e1–5.CrossRef Pasanen P, Myllykangas L, Siitonen M, Raunio A, Kaakkola S, Lyytinen J, et al. Novel α-synuclein mutation A53E associated with atypical multiple system atrophy and Parkinson’s disease-type pathology. Neurobiol Aging. 2014;35:2180.e1–5.CrossRef
48.
go back to reference Kiely AP, Asi YT, Kara E, Limousin P, Ling H, Lewis P, et al. α-Synucleinopathy associated with G51D SNCA mutation: a link between Parkinson’s disease and multiple system atrophy? Acta Neuropathol. 2013;125:753–69.PubMedPubMedCentralCrossRef Kiely AP, Asi YT, Kara E, Limousin P, Ling H, Lewis P, et al. α-Synucleinopathy associated with G51D SNCA mutation: a link between Parkinson’s disease and multiple system atrophy? Acta Neuropathol. 2013;125:753–69.PubMedPubMedCentralCrossRef
49.
go back to reference Bougea A, Koros C, Stamelou M, Simitsi A, Papagiannakis N, Antonelou R, et al. Frontotemporal dementia as the presenting phenotype of p.A53T mutation carriers in the alpha-synuclein gene. Parkinsonism Relat Disord. 2017;35:82–7CrossRef Bougea A, Koros C, Stamelou M, Simitsi A, Papagiannakis N, Antonelou R, et al. Frontotemporal dementia as the presenting phenotype of p.A53T mutation carriers in the alpha-synuclein gene. Parkinsonism Relat Disord. 2017;35:82–7CrossRef
50.
go back to reference Miller DW, Hague SM, Clarimon J, Baptista M, Gwinn-Hardy K, Cookson MR, et al. Alpha-synuclein in blood and brain from familial Parkinson disease with SNCA locus triplication. Neurology. 2004;62:1835–8.PubMedCrossRef Miller DW, Hague SM, Clarimon J, Baptista M, Gwinn-Hardy K, Cookson MR, et al. Alpha-synuclein in blood and brain from familial Parkinson disease with SNCA locus triplication. Neurology. 2004;62:1835–8.PubMedCrossRef
51.
go back to reference Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat Genet. 2014;46:989–93.PubMedPubMedCentralCrossRef Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat Genet. 2014;46:989–93.PubMedPubMedCentralCrossRef
52.
go back to reference Funahashi Y, Yoshino Y, Yamazaki K, Mori Y, Mori T, Ozaki Y, et al. DNA methylation changes at SNCA intron 1 in patients with dementia with Lewy bodies. Psychiatry Clin Neurosci. 2017;71:28–35.PubMedCrossRef Funahashi Y, Yoshino Y, Yamazaki K, Mori Y, Mori T, Ozaki Y, et al. DNA methylation changes at SNCA intron 1 in patients with dementia with Lewy bodies. Psychiatry Clin Neurosci. 2017;71:28–35.PubMedCrossRef
53.
go back to reference Beyer K, Lao JI, Carrato C, Mate JL, López D, Ferrer I, et al. Differential expression of alpha-synuclein isoforms in dementia with Lewy bodies. Neuropathol Appl Neurobiol. 2004;30:601–7.PubMedCrossRef Beyer K, Lao JI, Carrato C, Mate JL, López D, Ferrer I, et al. Differential expression of alpha-synuclein isoforms in dementia with Lewy bodies. Neuropathol Appl Neurobiol. 2004;30:601–7.PubMedCrossRef
54.
go back to reference Lim X, Yeo JM, Green A, Pal S. The diagnostic utility of cerebrospinal fluid alpha-synuclein analysis in dementia with Lewy bodies—a systematic review and meta-analysis. Parkinsonism Relat Disord. 2013;19:851–8.PubMedCrossRef Lim X, Yeo JM, Green A, Pal S. The diagnostic utility of cerebrospinal fluid alpha-synuclein analysis in dementia with Lewy bodies—a systematic review and meta-analysis. Parkinsonism Relat Disord. 2013;19:851–8.PubMedCrossRef
55.
go back to reference Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioannidis JP, Krüger R, et al. Collaborative analysis of alpha-synuclein gene promoter variability and Parkinson disease. JAMA. 2006;296:661–70.PubMedCrossRef Maraganore DM, de Andrade M, Elbaz A, Farrer MJ, Ioannidis JP, Krüger R, et al. Collaborative analysis of alpha-synuclein gene promoter variability and Parkinson disease. JAMA. 2006;296:661–70.PubMedCrossRef
56.
go back to reference Cronin KD, Ge D, Manninger P, Linnertz C, Rossoshek A, Orrison BM, et al. Expansion of the Parkinson disease-associated SNCA-Rep1 allele upregulates human alpha-synuclein in transgenic mouse brain. Hum Mol Genet. 2009;18:3274–85.PubMedPubMedCentralCrossRef Cronin KD, Ge D, Manninger P, Linnertz C, Rossoshek A, Orrison BM, et al. Expansion of the Parkinson disease-associated SNCA-Rep1 allele upregulates human alpha-synuclein in transgenic mouse brain. Hum Mol Genet. 2009;18:3274–85.PubMedPubMedCentralCrossRef
57.
go back to reference Simón-Sánchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet. 2009;41:1308–12.PubMedPubMedCentralCrossRef Simón-Sánchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat Genet. 2009;41:1308–12.PubMedPubMedCentralCrossRef
58.
go back to reference Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–40.PubMedCrossRef Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388:839–40.PubMedCrossRef
59.
go back to reference Kramer ML, Schulz-Schaeffer WJ. Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci. 2007;27:1405–10.PubMedCrossRef Kramer ML, Schulz-Schaeffer WJ. Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J Neurosci. 2007;27:1405–10.PubMedCrossRef
60.
go back to reference Colom-Cadena M, Pegueroles J, Herrmann AG, Henstridge CM, Muñoz L, Querol-Vilaseca M, et al. Synaptic phosphorylated α-synuclein in dementia with Lewy bodies. Brain. 2017;140:3204–14.PubMedCrossRef Colom-Cadena M, Pegueroles J, Herrmann AG, Henstridge CM, Muñoz L, Querol-Vilaseca M, et al. Synaptic phosphorylated α-synuclein in dementia with Lewy bodies. Brain. 2017;140:3204–14.PubMedCrossRef
61.
go back to reference • Peng C, Gathagan RJ, Covell DJ, Medellin C, Stieber A, Robinson JL, et al. Cellular milieu imparts distinct pathological α-synuclein strains in α-synucleinopathies. Nature [Internet]. 2018; Available from: https://doi.org/10.1038/s41586-018-0104-4. The authors report that pathological alpha-synuclein in glial cytoplasmic inclusions and Lewy bodies are distinct. PubMedCrossRef • Peng C, Gathagan RJ, Covell DJ, Medellin C, Stieber A, Robinson JL, et al. Cellular milieu imparts distinct pathological α-synuclein strains in α-synucleinopathies. Nature [Internet]. 2018; Available from: https://​doi.​org/​10.​1038/​s41586-018-0104-4. The authors report that pathological alpha-synuclein in glial cytoplasmic inclusions and Lewy bodies are distinct. PubMedCrossRef
62.
go back to reference Kotzbauer PT, Trojanowsk JQ, Lee VM. Lewy body pathology in Alzheimer’s disease. J Mol Neurosci. 2001;17:225–32.PubMedCrossRef Kotzbauer PT, Trojanowsk JQ, Lee VM. Lewy body pathology in Alzheimer’s disease. J Mol Neurosci. 2001;17:225–32.PubMedCrossRef
63.
go back to reference Lippa CF, Fujiwara H, Mann DM, Giasson B, Baba M, Schmidt ML, et al. Lewy bodies contain altered alpha-synuclein in brains of many familial Alzheimer’s disease patients with mutations in presenilin and amyloid precursor protein genes. Am J Pathol. 1998;153:1365–70.PubMedPubMedCentralCrossRef Lippa CF, Fujiwara H, Mann DM, Giasson B, Baba M, Schmidt ML, et al. Lewy bodies contain altered alpha-synuclein in brains of many familial Alzheimer’s disease patients with mutations in presenilin and amyloid precursor protein genes. Am J Pathol. 1998;153:1365–70.PubMedPubMedCentralCrossRef
64.
go back to reference Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci USA. 1993;90:1977–81.PubMedCrossRef Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci USA. 1993;90:1977–81.PubMedCrossRef
65.
go back to reference Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.PubMedCrossRef Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921–3.PubMedCrossRef
66.
go back to reference Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schneider JA, et al. APOE ϵ4 increases risk for dementia in pure synucleinopathies. JAMA Neurol. American Medical Association. 2013;70:223–8.CrossRef Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schneider JA, et al. APOE ϵ4 increases risk for dementia in pure synucleinopathies. JAMA Neurol. American Medical Association. 2013;70:223–8.CrossRef
67.
go back to reference Berge G, Sando SB, Rongve A, Aarsland D, White LR. Apolipoprotein E ε2 genotype delays onset of dementia with Lewy bodies in a Norwegian cohort. J Neurol Neurosurg Psychiatry. 2014;85:1227–31.PubMedPubMedCentralCrossRef Berge G, Sando SB, Rongve A, Aarsland D, White LR. Apolipoprotein E ε2 genotype delays onset of dementia with Lewy bodies in a Norwegian cohort. J Neurol Neurosurg Psychiatry. 2014;85:1227–31.PubMedPubMedCentralCrossRef
68.
go back to reference Nielsen AS, Ravid R, Kamphorst W, Jørgensen OS. Apolipoprotein E epsilon 4 in an autopsy series of various dementing disorders. J Alzheimers Dis. 2003;5:119–25.PubMedCrossRef Nielsen AS, Ravid R, Kamphorst W, Jørgensen OS. Apolipoprotein E epsilon 4 in an autopsy series of various dementing disorders. J Alzheimers Dis. 2003;5:119–25.PubMedCrossRef
69.
go back to reference Federoff M, Jimenez-Rolando B, Nalls MA, Singleton AB. A large study reveals no association between APOE and Parkinson’s disease. Neurobiol Dis. 2012;46:389–92.PubMedPubMedCentralCrossRef Federoff M, Jimenez-Rolando B, Nalls MA, Singleton AB. A large study reveals no association between APOE and Parkinson’s disease. Neurobiol Dis. 2012;46:389–92.PubMedPubMedCentralCrossRef
71.
go back to reference Carmona S, Kun-Rodrigues C, Brás J, Guerreiro R. Revisiting the genetics of APOE. Sinapse. 2017;17:27–36. Carmona S, Kun-Rodrigues C, Brás J, Guerreiro R. Revisiting the genetics of APOE. Sinapse. 2017;17:27–36.
73.
go back to reference Tulloch J, Leong L, Chen S, Keene CD, Millard SP, Shutes-David A, et al. APOE DNA methylation is altered in Lewy body dementia. Alzheimers Dement [Internet]. 2018; Available from: 10.1016/j.jalz.2018.02.005 Tulloch J, Leong L, Chen S, Keene CD, Millard SP, Shutes-David A, et al. APOE DNA methylation is altered in Lewy body dementia. Alzheimers Dement [Internet]. 2018; Available from: 10.1016/j.jalz.2018.02.005
74.
go back to reference Neudorfer O, Giladi N, Elstein D, Abrahamov A, Turezkite T, Aghai E, et al. Occurrence of Parkinson’s syndrome in type I Gaucher disease. QJM. 1996;89:691–4.PubMedCrossRef Neudorfer O, Giladi N, Elstein D, Abrahamov A, Turezkite T, Aghai E, et al. Occurrence of Parkinson’s syndrome in type I Gaucher disease. QJM. 1996;89:691–4.PubMedCrossRef
75.
go back to reference Goker-Alpan O, Schiffmann R, LaMarca ME, Nussbaum RL, McInerney-Leo A, Sidransky E. Parkinsonism among Gaucher disease carriers. J Med Genet. 2004;41:937–40.PubMedPubMedCentralCrossRef Goker-Alpan O, Schiffmann R, LaMarca ME, Nussbaum RL, McInerney-Leo A, Sidransky E. Parkinsonism among Gaucher disease carriers. J Med Genet. 2004;41:937–40.PubMedPubMedCentralCrossRef
76.
go back to reference Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER, et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. N Engl J Med. 2009;361:1651–61.PubMedPubMedCentralCrossRef Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER, et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. N Engl J Med. 2009;361:1651–61.PubMedPubMedCentralCrossRef
77.
go back to reference Goker-Alpan O, Giasson BI, Eblan MJ, Nguyen J, Hurtig HI, Lee VM-Y, et al. Glucocerebrosidase mutations are an important risk factor for Lewy body disorders. Neurology. 2006;67:908–10.PubMedCrossRef Goker-Alpan O, Giasson BI, Eblan MJ, Nguyen J, Hurtig HI, Lee VM-Y, et al. Glucocerebrosidase mutations are an important risk factor for Lewy body disorders. Neurology. 2006;67:908–10.PubMedCrossRef
78.
go back to reference Mata IF, Samii A, Schneer SH, Roberts JW, Griffith A, Leis BC, et al. Glucocerebrosidase gene mutations: a risk factor for Lewy body disorders. Arch Neurol Am Med Assoc. 2008;65:379–82. Mata IF, Samii A, Schneer SH, Roberts JW, Griffith A, Leis BC, et al. Glucocerebrosidase gene mutations: a risk factor for Lewy body disorders. Arch Neurol Am Med Assoc. 2008;65:379–82.
79.
go back to reference Clark LN, Kartsaklis LA, Wolf Gilbert R, Dorado B, Ross BM, Kisselev S, et al. Association of glucocerebrosidase mutations with dementia with lewy bodies. Arch Neurol. 2009;66:578–83.PubMedPubMedCentralCrossRef Clark LN, Kartsaklis LA, Wolf Gilbert R, Dorado B, Ross BM, Kisselev S, et al. Association of glucocerebrosidase mutations with dementia with lewy bodies. Arch Neurol. 2009;66:578–83.PubMedPubMedCentralCrossRef
80.
go back to reference Shiner T, Mirelman A, Gana Weisz M, Bar-Shira A, Ash E, Cialic R, et al. High frequency of GBA gene mutations in dementia with Lewy bodies among Ashkenazi Jews. JAMA Neurol. 2016;73:1448–53.PubMedCrossRef Shiner T, Mirelman A, Gana Weisz M, Bar-Shira A, Ash E, Cialic R, et al. High frequency of GBA gene mutations in dementia with Lewy bodies among Ashkenazi Jews. JAMA Neurol. 2016;73:1448–53.PubMedCrossRef
81.
go back to reference Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schneider JA, et al. GBA mutations increase risk for Lewy body disease with and without Alzheimer disease pathology. Neurology. 2012;79:1944–50.PubMedPubMedCentralCrossRef Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schneider JA, et al. GBA mutations increase risk for Lewy body disease with and without Alzheimer disease pathology. Neurology. 2012;79:1944–50.PubMedPubMedCentralCrossRef
82.
go back to reference Gámez-Valero A, Prada-Dacasa P, Santos C, Adame-Castillo C, Campdelacreu J, Reñé R, et al. GBA mutations are associated with earlier onset and male sex in dementia with Lewy bodies. Mov Disord. 2016;31:1066–70.PubMedCrossRef Gámez-Valero A, Prada-Dacasa P, Santos C, Adame-Castillo C, Campdelacreu J, Reñé R, et al. GBA mutations are associated with earlier onset and male sex in dementia with Lewy bodies. Mov Disord. 2016;31:1066–70.PubMedCrossRef
83.
go back to reference Nishioka K, Ross OA, Vilariño-Güell C, Cobb SA, Kachergus JM, Mann DMA, et al. Glucocerebrosidase mutations in diffuse Lewy body disease. Parkinsonism Relat Disord. 2011;17:55–7.PubMedPubMedCentralCrossRef Nishioka K, Ross OA, Vilariño-Güell C, Cobb SA, Kachergus JM, Mann DMA, et al. Glucocerebrosidase mutations in diffuse Lewy body disease. Parkinsonism Relat Disord. 2011;17:55–7.PubMedPubMedCentralCrossRef
84.
go back to reference Creese B, Bell E, Johar I, Francis P, Ballard C, Aarsland D. Glucocerebrosidase mutations and neuropsychiatric phenotypes in Parkinson’s disease and Lewy body dementias: review and meta-analyses. Am J Med Genet B Neuropsychiatr Genet. 2018;177:232–41.PubMedCrossRef Creese B, Bell E, Johar I, Francis P, Ballard C, Aarsland D. Glucocerebrosidase mutations and neuropsychiatric phenotypes in Parkinson’s disease and Lewy body dementias: review and meta-analyses. Am J Med Genet B Neuropsychiatr Genet. 2018;177:232–41.PubMedCrossRef
85.
go back to reference Malini E, Grossi S, Deganuto M, Rosano C, Parini R, Dominisini S, et al. Functional analysis of 11 novel GBA alleles. Eur J Hum Genet. 2014;22:511–6.PubMedCrossRef Malini E, Grossi S, Deganuto M, Rosano C, Parini R, Dominisini S, et al. Functional analysis of 11 novel GBA alleles. Eur J Hum Genet. 2014;22:511–6.PubMedCrossRef
86.
go back to reference Berge-Seidl V, Pihlstrøm L, Maple-Grødem J, Forsgren L, Linder J, Larsen JP, et al. The GBA variant E326K is associated with Parkinson’s disease and explains a genome-wide association signal. Neurosci Lett. 2017;658:48–52.PubMedCrossRef Berge-Seidl V, Pihlstrøm L, Maple-Grødem J, Forsgren L, Linder J, Larsen JP, et al. The GBA variant E326K is associated with Parkinson’s disease and explains a genome-wide association signal. Neurosci Lett. 2017;658:48–52.PubMedCrossRef
87.
go back to reference Gegg ME, Burke D, Heales SJR, Cooper JM, Hardy J, Wood NW, et al. Glucocerebrosidase deficiency in substantia nigra of parkinson disease brains. Ann Neurol. 2012;72:455–63.PubMedPubMedCentralCrossRef Gegg ME, Burke D, Heales SJR, Cooper JM, Hardy J, Wood NW, et al. Glucocerebrosidase deficiency in substantia nigra of parkinson disease brains. Ann Neurol. 2012;72:455–63.PubMedPubMedCentralCrossRef
88.
go back to reference Parnetti L, Paciotti S, Eusebi P, Dardis A, Zampieri S, Chiasserini D, et al. Cerebrospinal fluid β-glucocerebrosidase activity is reduced in parkinson’s disease patients. Mov Disord. 2017;32:1423–31.PubMedCrossRef Parnetti L, Paciotti S, Eusebi P, Dardis A, Zampieri S, Chiasserini D, et al. Cerebrospinal fluid β-glucocerebrosidase activity is reduced in parkinson’s disease patients. Mov Disord. 2017;32:1423–31.PubMedCrossRef
90.
go back to reference Parnetti L, Balducci C, Pierguidi L, De Carlo C, Peducci M, D’Amore C, et al. Cerebrospinal fluid β-glucocerebrosidase activity is reduced in dementia with Lewy Bodies. Neurobiol Dis. 2009;34:484–6.PubMedCrossRef Parnetti L, Balducci C, Pierguidi L, De Carlo C, Peducci M, D’Amore C, et al. Cerebrospinal fluid β-glucocerebrosidase activity is reduced in dementia with Lewy Bodies. Neurobiol Dis. 2009;34:484–6.PubMedCrossRef
91.
go back to reference Guyant-Marechal I, Berger E, Laquerrière A, Rovelet-Lecrux A, Viennet G, Frebourg T, et al. Intrafamilial diversity of phenotype associated with app duplication. Neurology. 2008;71:1925–6.PubMedCrossRef Guyant-Marechal I, Berger E, Laquerrière A, Rovelet-Lecrux A, Viennet G, Frebourg T, et al. Intrafamilial diversity of phenotype associated with app duplication. Neurology. 2008;71:1925–6.PubMedCrossRef
92.
go back to reference Ishikawa A, Piao Y-S, Miyashita A, Kuwano R, Onodera O, Ohtake H, et al. A mutant PSEN1 causes dementia with Lewy bodies and variant Alzheimer’s disease. Ann Neurol. 2005;57:429–34.PubMedCrossRef Ishikawa A, Piao Y-S, Miyashita A, Kuwano R, Onodera O, Ohtake H, et al. A mutant PSEN1 causes dementia with Lewy bodies and variant Alzheimer’s disease. Ann Neurol. 2005;57:429–34.PubMedCrossRef
93.
go back to reference Piscopo P, Marcon G, Piras MR, Crestini A, Campeggi LM, Deiana E, et al. A novel PSEN2 mutation associated with a peculiar phenotype. Neurology. 2008;70:1549–54.PubMedCrossRef Piscopo P, Marcon G, Piras MR, Crestini A, Campeggi LM, Deiana E, et al. A novel PSEN2 mutation associated with a peculiar phenotype. Neurology. 2008;70:1549–54.PubMedCrossRef
94.
go back to reference Leverenz JB, Fishel MA, Peskind ER, Montine TJ, Nochlin D, Steinbart E, et al. Lewy body pathology in familial Alzheimer disease: evidence for disease- and mutation-specific pathologic phenotype. Arch Neurol. 2006;63:370–6.PubMedPubMedCentralCrossRef Leverenz JB, Fishel MA, Peskind ER, Montine TJ, Nochlin D, Steinbart E, et al. Lewy body pathology in familial Alzheimer disease: evidence for disease- and mutation-specific pathologic phenotype. Arch Neurol. 2006;63:370–6.PubMedPubMedCentralCrossRef
95.
go back to reference Rosenberg CK, Pericak-Vance MA, Saunders AM, Gilbert JR, Gaskell PC, Hulette CM. Lewy body and Alzheimer pathology in a family with the amyloid-beta precursor protein APP717 gene mutation. Acta Neuropathol. 2000;100:145–52.PubMedCrossRef Rosenberg CK, Pericak-Vance MA, Saunders AM, Gilbert JR, Gaskell PC, Hulette CM. Lewy body and Alzheimer pathology in a family with the amyloid-beta precursor protein APP717 gene mutation. Acta Neuropathol. 2000;100:145–52.PubMedCrossRef
96.
go back to reference Hepp DH, Vergoossen DLE. Huisman E, Lemstra AW, Netherlands Brain Bank, Berendse HW, et al. Distribution and load of amyloid-β pathology in Parkinson disease and dementia with Lewy bodies. J Neuropathol Exp Neurol. 2016;75:936–45.PubMedCrossRef Hepp DH, Vergoossen DLE. Huisman E, Lemstra AW, Netherlands Brain Bank, Berendse HW, et al. Distribution and load of amyloid-β pathology in Parkinson disease and dementia with Lewy bodies. J Neuropathol Exp Neurol. 2016;75:936–45.PubMedCrossRef
97.
go back to reference Masliah E, Rockenstein E, Veinbergs I, Sagara Y, Mallory M, Hashimoto M, et al. β-Amyloid peptides enhance α-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer’s disease and Parkinson's disease. Proc Natl Acad Sci U S A. Nat Acad Sci. 2001;98:12245–50.CrossRef Masliah E, Rockenstein E, Veinbergs I, Sagara Y, Mallory M, Hashimoto M, et al. β-Amyloid peptides enhance α-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer’s disease and Parkinson's disease. Proc Natl Acad Sci U S A. Nat Acad Sci. 2001;98:12245–50.CrossRef
98.
go back to reference Vergouw LJM, van Steenoven I, van de Berg WDJ, Teunissen CE, van Swieten JC, Bonifati V, et al. An update on the genetics of dementia with Lewy bodies. Parkinsonism Relat Disord. 2017;43:1–8.PubMedCrossRef Vergouw LJM, van Steenoven I, van de Berg WDJ, Teunissen CE, van Swieten JC, Bonifati V, et al. An update on the genetics of dementia with Lewy bodies. Parkinsonism Relat Disord. 2017;43:1–8.PubMedCrossRef
99.
go back to reference Meeus B, Nuytemans K, Crosiers D, Engelborghs S, Peeters K, Mattheijssens M, et al. Comprehensive genetic and mutation analysis of familial dementia with Lewy bodies linked to 2q35-q36. J Alzheimers Dis. 2010;20:197–205.PubMedCrossRef Meeus B, Nuytemans K, Crosiers D, Engelborghs S, Peeters K, Mattheijssens M, et al. Comprehensive genetic and mutation analysis of familial dementia with Lewy bodies linked to 2q35-q36. J Alzheimers Dis. 2010;20:197–205.PubMedCrossRef
100.
go back to reference Kara E, Kiely AP, Proukakis C, Giffin N, Love S, Hehir J, et al. A 6.4 Mb duplication of the α-synuclein locus causing frontotemporal dementia and Parkinsonism: phenotype-genotype correlations. JAMA Neurol. 2014;71:1162–71.PubMedPubMedCentralCrossRef Kara E, Kiely AP, Proukakis C, Giffin N, Love S, Hehir J, et al. A 6.4 Mb duplication of the α-synuclein locus causing frontotemporal dementia and Parkinsonism: phenotype-genotype correlations. JAMA Neurol. 2014;71:1162–71.PubMedPubMedCentralCrossRef
101.
go back to reference Colom-Cadena M, Gelpi E, Martí MJ, Charif S, Dols-Icardo O, Blesa R, et al. MAPT H1 haplotype is associated with enhanced α-synuclein deposition in dementia with Lewy bodies. Neurobiol Aging. 2013;34:936–42.PubMedCrossRef Colom-Cadena M, Gelpi E, Martí MJ, Charif S, Dols-Icardo O, Blesa R, et al. MAPT H1 haplotype is associated with enhanced α-synuclein deposition in dementia with Lewy bodies. Neurobiol Aging. 2013;34:936–42.PubMedCrossRef
102.
go back to reference Coppola G, Chinnathambi S, Lee JJ, Dombroski BA, Baker MC, Soto-Ortolaza AI, et al. Evidence for a role of the rare p.A152T variant in MAPT in increasing the risk for FTD-spectrum and Alzheimer’s diseases. Hum Mol Genet. 2012;21:3500–12.PubMedPubMedCentralCrossRef Coppola G, Chinnathambi S, Lee JJ, Dombroski BA, Baker MC, Soto-Ortolaza AI, et al. Evidence for a role of the rare p.A152T variant in MAPT in increasing the risk for FTD-spectrum and Alzheimer’s diseases. Hum Mol Genet. 2012;21:3500–12.PubMedPubMedCentralCrossRef
103.
go back to reference Edwards TL, Scott WK, Almonte C, Burt A, Powell EH, Beecham GW, et al. Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease. Ann Hum Genet. 2010;74:97–109.PubMedPubMedCentralCrossRef Edwards TL, Scott WK, Almonte C, Burt A, Powell EH, Beecham GW, et al. Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease. Ann Hum Genet. 2010;74:97–109.PubMedPubMedCentralCrossRef
104.
go back to reference Robinson A, Davidson Y, Snowden JS, Mann DMA. C9ORF72 in dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2014;85:1435–6.PubMedCrossRef Robinson A, Davidson Y, Snowden JS, Mann DMA. C9ORF72 in dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2014;85:1435–6.PubMedCrossRef
105.
go back to reference Yeh T-H, Lai S-C, Weng Y-H, Kuo H-C, Wu-Chou Y-H, Huang C-L, et al. Screening for C9orf72 repeat expansions in parkinsonian syndromes. Neurobiol Aging. 2013;34:1311.e3–1311.e34.CrossRef Yeh T-H, Lai S-C, Weng Y-H, Kuo H-C, Wu-Chou Y-H, Huang C-L, et al. Screening for C9orf72 repeat expansions in parkinsonian syndromes. Neurobiol Aging. 2013;34:1311.e3–1311.e34.CrossRef
106.
go back to reference Ross OA, Toft M, Whittle AJ, Johnson JL, Papapetropoulos S, Mash DC, et al. Lrrk2 and Lewy body disease. Ann Neurol. 2006;59:388–93.PubMedCrossRef Ross OA, Toft M, Whittle AJ, Johnson JL, Papapetropoulos S, Mash DC, et al. Lrrk2 and Lewy body disease. Ann Neurol. 2006;59:388–93.PubMedCrossRef
107.
go back to reference Srivatsal S, Cholerton B, Leverenz JB, Wszolek ZK, Uitti RJ, Dickson DW, et al. Cognitive profile of LRRK2-related Parkinson’s disease. Mov Disord. 2015;30:728–33.PubMedPubMedCentralCrossRef Srivatsal S, Cholerton B, Leverenz JB, Wszolek ZK, Uitti RJ, Dickson DW, et al. Cognitive profile of LRRK2-related Parkinson’s disease. Mov Disord. 2015;30:728–33.PubMedPubMedCentralCrossRef
108.
go back to reference Kosaka K, Yoshimura M, Ikeda K, Budka H. Diffuse type of Lewy body disease: progressive dementia with abundant cortical Lewy bodies and senile changes of varying degree—a new disease? Clin Neuropathol. 1984;3:185–92.PubMed Kosaka K, Yoshimura M, Ikeda K, Budka H. Diffuse type of Lewy body disease: progressive dementia with abundant cortical Lewy bodies and senile changes of varying degree—a new disease? Clin Neuropathol. 1984;3:185–92.PubMed
109.
go back to reference Yoshita M, Taki J, Yamada M. A clinical role for [(123)I]MIBG myocardial scintigraphy in the distinction between dementia of the Alzheimer’s-type and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2001;71:583–8.PubMedPubMedCentralCrossRef Yoshita M, Taki J, Yamada M. A clinical role for [(123)I]MIBG myocardial scintigraphy in the distinction between dementia of the Alzheimer’s-type and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2001;71:583–8.PubMedPubMedCentralCrossRef
110.
go back to reference Slaets S, Van Acker F, Versijpt J, Hauth L, Goeman J, Martin J-J, et al. Diagnostic value of MIBG cardiac scintigraphy for differential dementia diagnosis. Int J Geriatr Psychiatry. 2015;30:864–9.PubMedCrossRef Slaets S, Van Acker F, Versijpt J, Hauth L, Goeman J, Martin J-J, et al. Diagnostic value of MIBG cardiac scintigraphy for differential dementia diagnosis. Int J Geriatr Psychiatry. 2015;30:864–9.PubMedCrossRef
111.
go back to reference Flanigan PM, Khosravi MA, Leverenz JB, Tousi B. Color vision impairment in dementia with lewy bodies: a novel and highly specific distinguishing feature from Alzheimer dementia. Alzheimers Dement Elsevier. 2017;13:P1460.CrossRef Flanigan PM, Khosravi MA, Leverenz JB, Tousi B. Color vision impairment in dementia with lewy bodies: a novel and highly specific distinguishing feature from Alzheimer dementia. Alzheimers Dement Elsevier. 2017;13:P1460.CrossRef
112.
go back to reference Brigo F, Turri G, Tinazzi M. 123I-FP-CIT SPECT in the differential diagnosis between dementia with Lewy bodies and other dementias. J Neurol Sci. 2015;359:161–71.PubMedCrossRef Brigo F, Turri G, Tinazzi M. 123I-FP-CIT SPECT in the differential diagnosis between dementia with Lewy bodies and other dementias. J Neurol Sci. 2015;359:161–71.PubMedCrossRef
113.
go back to reference Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536:285–91.PubMedPubMedCentralCrossRef Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536:285–91.PubMedPubMedCentralCrossRef
114.
go back to reference Birney E, Soranzo N. Human genomics: The end of the start for population sequencing. Nature. 2015;526:52–3.PubMedCrossRef Birney E, Soranzo N. Human genomics: The end of the start for population sequencing. Nature. 2015;526:52–3.PubMedCrossRef
115.
go back to reference Blauwendraat C, Kia DA, Pihlstrøm L, Gan-Or Z, Lesage S, Gibbs JR, et al. Insufficient evidence for pathogenicity of SNCA His50Gln (H50Q) in Parkinson’s disease. Neurobiol Aging. 2018;64:159.e5–8.CrossRef Blauwendraat C, Kia DA, Pihlstrøm L, Gan-Or Z, Lesage S, Gibbs JR, et al. Insufficient evidence for pathogenicity of SNCA His50Gln (H50Q) in Parkinson’s disease. Neurobiol Aging. 2018;64:159.e5–8.CrossRef
117.
go back to reference Logue MW, Panizzon MS, Elman JA, Gillespie NA, Hatton SN, Gustavson DE, et al. Use of an Alzheimer’s disease polygenic risk score to identify mild cognitive impairment in adults in their 50s. Mol Psychiatry [Internet]. 2018; Available from: https://doi.org/10.1038/s41380-018-0030-8 Logue MW, Panizzon MS, Elman JA, Gillespie NA, Hatton SN, Gustavson DE, et al. Use of an Alzheimer’s disease polygenic risk score to identify mild cognitive impairment in adults in their 50s. Mol Psychiatry [Internet]. 2018; Available from: https://​doi.​org/​10.​1038/​s41380-018-0030-8
118.
go back to reference Ibanez L, Dube U, Saef B, Budde J, Black K, Medvedeva A, et al. Parkinson disease polygenic risk score is associated with Parkinson disease status and age at onset but not with alpha-synuclein cerebrospinal fluid levels. BMC Neurol. 2017;17:198.PubMedPubMedCentralCrossRef Ibanez L, Dube U, Saef B, Budde J, Black K, Medvedeva A, et al. Parkinson disease polygenic risk score is associated with Parkinson disease status and age at onset but not with alpha-synuclein cerebrospinal fluid levels. BMC Neurol. 2017;17:198.PubMedPubMedCentralCrossRef
119.
go back to reference Donadio V, Incensi A, Rizzo G, Capellari S, Pantieri R, Stanzani Maserati M, et al. A new potential biomarker for dementia with Lewy bodies: Skin nerve α-synuclein deposits. Neurology. 2017;89:318–26.PubMedCrossRef Donadio V, Incensi A, Rizzo G, Capellari S, Pantieri R, Stanzani Maserati M, et al. A new potential biomarker for dementia with Lewy bodies: Skin nerve α-synuclein deposits. Neurology. 2017;89:318–26.PubMedCrossRef
120.
go back to reference Wood H. Dementia: Skin α-synuclein deposits - a new biomarker for DLB? Nat Rev Neurol. 2017;13:514.PubMedCrossRef Wood H. Dementia: Skin α-synuclein deposits - a new biomarker for DLB? Nat Rev Neurol. 2017;13:514.PubMedCrossRef
Metadata
Title
The Genetics of Dementia with Lewy Bodies: Current Understanding and Future Directions
Authors
Tatiana Orme
Rita Guerreiro
Jose Bras
Publication date
01-10-2018
Publisher
Springer US
Published in
Current Neurology and Neuroscience Reports / Issue 10/2018
Print ISSN: 1528-4042
Electronic ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-018-0874-y

Other articles of this Issue 10/2018

Current Neurology and Neuroscience Reports 10/2018 Go to the issue

Sleep (M Thorpy and M Billiard, Section Editors)

Sleep in Parkinson’s Disease with Impulse Control Disorder

Headache (R B Halker, Section Editor)

The Neuralgias

Sleep (M Thorpy and M Billiard, Section Editors)

Circadian Rhythm Disturbances in the Blind