Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jul 1.
Published in final edited form as: Lancet Neurol. 2014 Jul;13(7):686–699. doi: 10.1016/S1474-4422(14)70065-1

Frontotemporal dementia and its subtypes: a genome-wide association study

Raffaele Ferrari 1,*, Dena G Hernandez 1,*, Michael A Nalls 1,*, Jonathan D Rohrer 1,*, Adaikalavan Ramasamy 1, John B J Kwok 1, Carol Dobson-Stone 1, William S Brooks 1, Peter R Schofield 1, Glenda M Halliday 1, John R Hodges 1, Olivier Piguet 1, Lauren Bartley 1, Elizabeth Thompson 1, Eric Haan 1, Isabel Hernández 1, Agustín Ruiz 1, Mercè Boada 1, Barbara Borroni 1, Alessandro Padovani 1, Carlos Cruchaga 1, Nigel J Cairns 1, Luisa Benussi 1, Giuliano Binetti 1, Roberta Ghidoni 1, Gianluigi Forloni 1, Daniela Galimberti 1, Chiara Fenoglio 1, Maria Serpente 1, Elio Scarpini 1, Jordi Clarimón 1, Alberto Lleó 1, Rafael Blesa 1, Maria Landqvist Waldö 1, Karin Nilsson 1, Christer Nilsson 1, Ian R A Mackenzie 1, Ging-Yuek R Hsiung 1, David M A Mann 1, Jordan Grafman 1, Christopher M Morris 1, Johannes Attems 1, Timothy D Griffiths 1, Ian G McKeith 1, Alan J Thomas 1, P Pietrini 1, Edward D Huey 1, Eric M Wassermann 1, Atik Baborie 1, Evelyn Jaros 1, Michael C Tierney 1, Pau Pastor 1, Cristina Razquin 1, Sara Ortega-Cubero 1, Elena Alonso 1, Robert Perneczky 1, Janine Diehl-Schmid 1, Panagiotis Alexopoulos 1, Alexander Kurz 1, Innocenzo Rainero 1, Elisa Rubino 1, Lorenzo Pinessi 1, Ekaterina Rogaeva 1, Peter St George-Hyslop 1, Giacomina Rossi 1, Fabrizio Tagliavini 1, Giorgio Giaccone 1, James B Rowe 1, J C M Schlachetzki 1, James Uphill 1, John Collinge 1, S Mead 1, Adrian Danek 1, Vivianna M Van Deerlin 1, Murray Grossman 1, John Q Trojanowsk 1, Julie van der Zee 1, William Deschamps 1, Tim Van Langenhove 1, Marc Cruts 1, Christine Van Broeckhoven 1, Stefano F Cappa 1, Isabelle Le Ber 1, Didier Hannequin 1, Véronique Golfier 1, Martine Vercelletto 1, Alexis Brice 1, Benedetta Nacmias 1, Sandro Sorbi 1, Silvia Bagnoli 1, Irene Piaceri 1, Jørgen E Nielsen 1, Lena E Hjermind 1, Matthias Riemenschneider 1, Manuel Mayhaus 1, Bernd Ibach 1, Gilles Gasparoni 1, Sabrina Pichler 1, Wei Gu 1, Martin N Rossor 1, Nick C Fox 1, Jason D Warren 1, Maria Grazia Spillantini 1, Huw R Morris 1, Patrizia Rizzu 1, Peter Heutink 1, Julie S Snowden 1, Sara Rollinson 1, Anna Richardson 1, Alexander Gerhard 1, Amalia C Bruni 1, Raffaele Maletta 1, Francesca Frangipane 1, Chiara Cupidi 1, Livia Bernardi 1, Maria Anfossi 1, Maura Gallo 1, Maria Elena Conidi 1, Nicoletta Smirne 1, Rosa Rademakers 1, Matt Baker 1, Dennis W Dickson 1, Neill R Graff-Radford 1, Ronald C Petersen 1, David Knopman 1, Keith A Josephs 1, Bradley F Boeve 1, Joseph E Parisi 1, William W Seeley 1, Bruce L Miller 1, Anna M Karydas 1, Howard Rosen 1, John C van Swieten 1, Elise G P Dopper 1, Harro Seelaar 1, Yolande AL Pijnenburg 1, Philip Scheltens 1, Giancarlo Logroscino 1, Rosa Capozzo 1, Valeria Novelli 1, Annibale A Puca 1, M Franceschi 1, Alfredo Postiglione 1, Graziella Milan 1, Paolo Sorrentino 1, Mark Kristiansen 1, Huei-Hsin Chiang 1, Caroline Graff 1, Florence Pasquier 1, Adeline Rollin 1, Vincent Deramecourt 1, Florence Lebert 1, Dimitrios Kapogiannis 1, Luigi Ferrucci 1, Stuart Pickering-Brown 1, Andrew B Singleton 1,, John Hardy 1,, Parastoo Momeni 1,
PMCID: PMC4112126  NIHMSID: NIHMS599470  PMID: 24943344

Summary

Background

Frontotemporal dementia (FTD) is a complex disorder characterised by a broad range of clinical manifestations, differential pathological signatures, and genetic variability. Mutations in three genes—MAPT, GRN, and C9orf72—have been associated with FTD. We sought to identify novel genetic risk loci associated with the disorder.

Methods

We did a two-stage genome-wide association study on clinical FTD, analysing samples from 3526 patients with FTD and 9402 healthy controls. All participants had European ancestry. In the discovery phase (samples from 2154 patients with FTD and 4308 controls), we did separate association analyses for each FTD subtype (behavioural variant FTD, semantic dementia, progressive non-fluent aphasia, and FTD overlapping with motor neuron disease [FTD-MND]), followed by a meta-analysis of the entire dataset. We carried forward replication of the novel suggestive loci in an independent sample series (samples from 1372 patients and 5094 controls) and then did joint phase and brain expression and methylation quantitative trait loci analyses for the associated (p<5 × 10−8) and suggestive single-nucleotide polymorphisms.

Findings

We identified novel associations exceeding the genome-wide significance threshold (p<5 × 10−8) that encompassed the HLA locus at 6p21.3 in the entire cohort. We also identified a potential novel locus at 11q14, encompassing RAB38/CTSC, for the behavioural FTD subtype. Analysis of expression and methylation quantitative trait loci data suggested that these loci might affect expression and methylation incis.

Interpretation

Our findings suggest that immune system processes (link to 6p21.3) and possibly lysosomal and autophagy pathways (link to 11q14) are potentially involved in FTD. Our findings need to be replicated to better define the association of the newly identified loci with disease and possibly to shed light on the pathomechanisms contributing to FTD.

Funding

The National Institute of Neurological Disorders and Stroke and National Institute on Aging, the Wellcome/ MRC Centre on Parkinson’s disease, Alzheimer’s Research UK, and Texas Tech University Health Sciences Center.

Introduction

Frontotemporal dementia (FTD) is the second most common form of young-onset dementia after Alzheimer’s disease and comprises about 10–20% of all dementias worldwide.1 FTD occurs in roughly three to 15 per 100 000 individuals aged between 55 years and 65 year2 The disease has an insidious onset: it is familial in 30–50% of patients and affects men and women almost equally.3 The main clinical syndromes are the behavioural variant1,4 and the language variants (semantic dementia and progressive nonfluent aphasia).1,5 FTD also overlap with motor neuron disease (FTD-MND), and atypical parkinsonian disorders.3 The molecular pathology is heterogeneous and based on the type of neuronal lesions and protein inclusions: 40% or more of patients have frontotemporal lobar degeneration (FTLD) with tau pathology (FTLD-tau), about 50% have TDP-43 (TAR DNA-binding protein 43) pathology (FTLD-TDP),6 and the remaining 10% have inclusions positive for fused in sarcoma (FUS; FTLD-FUS) or ubiquitin/p62 (FTLD-UPS [ubiquitin proteasome system]).7 Mutations in three main genes are commonly associated with FTD: the microtubule-associated protein tau (MAPT),8 granulin (GRN),9,10 and C9orf72.1115 Mutations in the charged multivesicular body protein 2B (CHMP2B), the valosin-containing protein (VCP), and ubiquilin 2 (UBQLN2) genes are rare causes of disease.13,16 Findings from a previous genome-wide association study (GWAS) of neuropathologically confirmed FTLD-TDP (515 patients vs 2509) showed TMEM106B to be a disease risk factor.17

We did a larger GWAS in samples from people with clinical FTD, and we report results for the discovery, replication, and joint phase analyses, as well as for assessment of the effect on expression and methylation quantitative trait loci (QTL) exerted by associated or suggestive SNPs. We aimed to identify novel genetic risk loci associated with FTD and its subtypes.

Methods

Study population

44 international research groups (appendix) contributed samples to this two-stage (discovery phase and replication phase) GWAS of clinical FTD. Investigators at every site obtained appropriate written informed consent from patients and control individuals. Every participating group provided consent for the use of these samples for the purposes of this study. The patients included in the discovery phase were diagnosed according to the Neary criteria1 for FTD, whereas those included in replication phase were diagnosed according to the Neary criteria,1 or the revised criteria for behavioural FTD4 and the language variants of FTD5 at every collaborative site. For each patient, the diagnosis was made by a neurologist with an interest in FTD or (the minority: <5%) by pathological diagnosis. To cover the most relevant FTD clinical signatures, we included patients diagnosed with behavioural FTD, semantic dementia, progressive nonfluent aphasia, or FTD-MND.18 We reviewed all patients with a diagnosis of language impairment to exclude cases of the logopenic variant of primary progressive aphasia,5 most of which are associated with Alzheimer’s disease pathology. Samples were obtained from North America (USA and Canada), UK, France, the Netherlands, Belgium, Germany, Denmark, Sweden, Spain, and Italy and all patients were of confirmed European ancestry.

DNA was collected at the three institutions leading this project: the Department of Molecular Neuroscience at University College London (UCL), UK; the Laboratory of Neurogenetics of the National Institute on Aging at the National Institutes of Health (NIH), MD, USA; and the Laboratory of Neurogenetics at the Texas Tech University Health Sciences Center (TTUHSC); TX, USA. All samples were anonymous and stored with a patientspecific coded identification number. Each DNA sample was assessed for quality with gel electrophoresis and DNA concentrations were assessed via spectrophotometer (Nanodrop; Wilmington, DE, USA) or fluorometer (Qubit; Life Technologies, Grand Island, NY, USA). Samples from non-overlapping patients were genotyped at the Laboratory of Neurogenetics of the National Institute on Aging, NIH (40%) or at the core facility at the Institute of Child Health, UCL (60%). We obtained standardised clinical, pathological, and genetic data for each patient from all the collaborating groups (appendix). Sporadic cases along with probands from FTD families were included in the study.. We excluded carriers of mutations in MAPT and GRN. We did not exclude individuals with C9orf72 expansions because this locus was identified subsequent to sample collection. After quality control of genotyping data and detailed assessment of the clinical diagnosis, we used 2154 and 1372 samples in the discovery phase and replication phase, respectively, for association analysis (table 1). In total, after quality control, we analysed 3526 FTD samples (table 1). Further details about cases included in the study are provided in the appendix.

Table 1.

Sample characteristics

Samples collected (n) Samples included in analysis (n) Samples from women (% [n/N]) Mean age at onset




Discovery
phase
Replication
phase
Total Discovery
phase
Replication
phase
Total Discovery phase Replication phase Discovery phase Replication phase
USA 706 209 915 579 175 754 44% (257/579) 49% (85/174) 60 (23–85); 520 63 (24–93); 120

Canada 25 37 62 24 29 53 52% (12/23) 57% (8/14) 64 (43–85); 15 59 (43–75); 9

UK 494 372 866 401 284 685 43% (171/400) 40% (108/272) 60 (23–83); 372 61 (35–86); 167

Spain 100 330 430 0 309 309 NA 43% (133/309) NA 65 (32–89); 308

France 238 54 292 205 42 247 44% (91/205) 48% (20/42) 62 (39–79); 190 NA

Belgium 240 51 291 191 42 233 46% (88/191) 29% (12/42) 63 (29–90); 191 64 (43–84); 42

Netherlands 333 93 426 250 77 327 52 (129/250) 40% (31/77) 58 (29–76); 250 61 (51–69); 59

Denmark 35 0 35 7 0 7 71% (5/7) NA 57 (40–62); 7 NA

Germany 349 34 383 320 33 353 NA 50% (15/30) 61 (36–83); 243 57 (29–72); 30

Sweden 26 112 138 18 98 116 56% (10/18) 61% (60/98) 57 (38–75); 16 62 (28–78); 93

Italy 1035 563 1598 564 371 935 53% (297/561) 45% (168/371) 64 (31–83); 429 65 (31–87); 353

Australia 0 138 138 0 121 121 NA 36% (44/121) NA 59 (32–77); 112

Meta-analysis 3581 1993 5574 2559 (2154*) 1581 (1372*) 4140 (3526*) 47% (1186/2552) 44% (684/1550) 61 (23–90); 2233 62 (24–93); 1293

NA=.

*

The number of the samples that passed genotyping data quality control and were used for association analyses.

Control samples for the discovery phase were taken from studies previously done at the Laboratory of Neurogenetics of the National Institute on Aging at the NIH or at UCL. Control individuals were matched to patients on the basis of population ancestry and genotyping platform. Aggregate data for control samples were merged based on overlapping single-nucleotide polymorphisms (SNPs). The selected 7444 control samples were from the USA, UK, Italy, Germany, France, Sweden, and the Netherlands, and were used as controls in previous GWAS;19 all individuals had given consent for their samples to be used as controls. All were free of neurological illness at the time of sampling, but most had not been screened for the absence of a family history of FTD. For each patient, at least two controls were matched based on compatibility of genetic ancestry estimates by principal components analysis to accommodate the lack of precisely matched clinical controls. After quality control, we included 4308 control samples in this study. The genotyping of controls for the replication phase was done at the Laboratory of Neurogenetics of the National Institute on Aging, NIH (90%) and at the core facility at the Institute of Child Health, UCL (10%). All control samples used in the replication phase were collected from the groups participating in the study (5094 samples passed quality control) and were of European ancestry from the following countries: USA (European American), UK, Italy, France, Germany, Sweden, Spain, and the Netherlands.

Procedures

For every sample, 2 µg of DNA extracted from either blood or the brain at each collaborative site was collected (whole genome amplified DNA samples were excluded). Samples were securely stored at −20°C. Every sample was first screened for integrity and purity by means of gel electrophoresis on 1% agarose gel, and concentrations were analysed by spectrophotometric (Nanodrop) or fluorometric (Qubit) quantification. The same procedure was implemented at NIH, UCL, and TTUHSC.

Samples from patients and control individuals included in the discovery phase were genotyped using Illumina human 370K, 550K, and 660K Quad Beadchips and Omni Express chips (Illumina Inc, CA, USA). We used Illumina NeuroX custom chips for all samples included in replication phase genotyping. The NeuroX chip is a partially custom-designed chip that specifically targets the main loci associated with several different neurological disorders obtained from GWAS or whole-exome sequencing data. The NeuroX chip holds about 267K SNPs, of which 3759 were FTD-specific, being selected from SNPs that had p values of less than 1 × 10−4 during the discovery phase of the study. These SNPs were tag SNPs based on European ancestry linkage-disequilibrium patterns from the most up-to-date data for samples of European ancestry from the 1000 Genomes project.20 For all GWAS significant hits and candidate SNPs, five linkage-disequilibrium-based proxies or technical replicates were included on the array per locus, tagging associations within +/−250 kb and r2 >0·5 from the most strongly associated proximal SNP. To replicate each locus, we picked the tag SNP most significant in the discovery phase before beginning. If no linkage-disequilibrium-based proxies were available, technical replicates were included. All genotyping arrays (discovery phase and replication phase) were assayed on the Illumina Infinium platform (Illumina, San Diego, CA, USA) at the Laboratory of Neurogenetics of the National Institute on Aging, NIH and at the core facility at the Institute of Child Health, UCL. All genotypes for this project were called centrally using Illumina Genome Studio and all 3759 SNPs of interest for FTD were manually examined to ensure high-quality genotype clusters before data export.

For the purpose of assessing possible biological relevance for any associated SNPs we used quantitative trait loci (QTL) data generated by the UK Brain Expression Consortium (UKBEC) and the North American Brain Expression Consortium (NABEC) for brain tissues assayed for genome-wide expression and methylation. Details about sample collection, RNA/DNA extraction, and genotyping are provided in the appendix.

Statistical analysis

We did standard quality control for GWAS data before association analyses. Briefly, for the discovery phase, we extracted overlapping SNPs across all Illumina arrays used. This was done as a means of dealing with the low numbers of matched cases and controls per study site or chip type to facilitate the FTD subtype analyses. We maximised sample size for the subtype analyses by pooling as many possible samples while sacrificing some array content, leaving 228 189 autosomal SNPs as a basis for imputation after the quality control was completed. We excluded samples possibly mismatched for sex by assessing X chromosome heterozygosity. Samples with a call rate of greater than 95% and SNPs with a minor allele frequency greater than 1% were filtered and included in the analyses. We calculated Hardy-Weinberg equilibrium p values (exclusion at p values <1 × 10−5). We assessed non-random missingness per SNP by case-control status with exclusion at p values of less than 1 × 10−5 and non-random missingness per SNP by haplotype at p values for exclusion <1 × 10−5. We assessed the presence of relatedness by identifying and excluding first-degree relatives (through identity by descent for any pairwise with an estimate of less than 0·125) and verified European ancestry by principal components analysis compared with HapMap3 populations, with European ancestry ascertained at values for the first two eigenvectors less than six SDs from the population mean for the combined Europeans from Utah and Tuscans from Italy reference samples.21 After preliminary quality assessment, principal components analysis as implemented in EIGENSTRAT22 was used to assess matching between cases and controls based on all available cases and controls. Custom coding in R was used to match cases to controls. We treated each subtype (behavioural FTD, semantic dementia, progressive nonfluent aphasia, and FTD-MND) as a separate group in which the two most genetically similar unique controls per case were selected based on eigenvectors 1 and 2 in order to compensate for a lack of precisely matched controls at recruitment. In this respect, matched controls were unique per case and non-redundant across subtype datasets. Thus, cases and controls were matched for each subtype (behavioural FTD, semantic dementia, progressive nonfluent aphasia, and FTD-MND) based on similarity of the first two eigenvectors from principal components analysis and did not overlap across subtypes. We used logistic regression based on imputed dosages to assess the association between each SNP and any of the FTD subtypes, adjusting for eigenvectors 1 and 2 from principal components analysis as covariates. Eigenvectors were generated separately for each subtype, as in the overall sample pool, parameter estimates for the first two were associated with case status at p values of less than 0·05. We did fixed-effects meta-analyses to combine results across subtypes and quantify heterogeneity across subtypes. Genomic inflation was minimal across subtypes and in the meta analysis across subtypes (λ<1·05), therefore we did not use genomic control (see appendix for quantile-quantile plots and λ values per discovery phase analysis). SNPs were imputed to August, 2010 release of the 1000 Genomes haplotypes using default settings of minimac and were excluded if their minor allele frequency was less than 0·01 or imputation quality (Rsq) was less than 0·30 across all samples, leaving 6 026 385 SNPs for analyses.

For the replication phase, we did standard quality control as for the discovery phase with slight adjustments to account for the bias in NeuroX array content (candidate neurological or neurodegenerative disease SNPs and exonic content). Standard content variants included on the NeuroX array that were used for sample quality control were called using a publicly available cluster file based on more than 60 000 samples.23 For quality control, variants with GenTrain scores greater than 0·70 (indicative of high-quality genotype clusters) were extracted first to calculate call rates. Samples with call rates greater than 95% were excluded, as were samples whose genetically determined sex conflicted with that from the clinical data and samples exhibiting excess heterozygosity. Next, SNPs overlapping with HapMap phase 3 samples were extracted from the previous subset and pruned for linkage disequilibrium (SNPs excluded if r2 >0·50 within a 50 SNP sliding window), and SNPs with minor allele frequency less than 5%, Hardy-Weinberg equilibrium p values less than 1 × 10−5, and per SNP missingness rates greater than 5%. At this stage, we used pairwise identity-by-descent filtering to remove samples that were cryptically related and principal components analysis to identify samples to be excluded when genetic ancestry was not consistent with European descent based on comparisons with HapMap phase 3 reference populations. For replication analyses and due to an effort to maximise the restricted power of this phase compared to the discovery phase, analyses of each subtype included all control samples available, adjusting for the first five eigenvectors only from principal components analysis as covariates in the logistic regression model. No other adjustments were implemented. Additionally, we pooled the individual genotypes from different subsets in the replication phase to help increase statistical power. For details about QTL statistical analysis, see appendix.

Role of the funding source

The sponsors of the study had no role in study design, data collection, data analysis, data interpretation, or writing of the report. No pharmaceutical company or other agency paid to write this article. The corresponding author had full access to all the data in the study and had final responsibility for the decision to submit for publication.

Results

In the discovery phase, we analysed samples from 2154 patients (table 1) and 4308 controls. We first did separate association analyses for each subtype (behavioural FTD, semantic dementia, progressive nonfluent aphasia, and FTD-MND; table 2) and then undertook a meta-analysis of the entire dataset. Findings from the meta-analysis showed 29 SNPs (appendix) exceeding genome-wide significance (p value <5×10−8) at the HLA locus (6p21.3), encompassing the butyrophilinlike 2 (MHC class II associated) gene (BTNL2) and the major histocompatibility complex, class II, DR alpha (HLA-DRA), and DR beta 5 (HLA-DRB5; figure, table 3). To identify susceptibility loci for the behavioural FTD subtype we analysed 1377 patient samples (table 2) and 2754 control samples. Two non-coding SNPs at 11q14, locating to intron 1 of the gene RAB38, member RAS oncogene family (RAB38; rs302652) and encompassing RAB38 and cathepsin C (CTSC; rs74977128), passed the genome-wide significance threshold (figure, table 3). Similarly, we did analyses on the other subtypes (table 1): 308 semantic dementia versus 616 controls, 269 progressive nonfluent aphasia versus 538 controls, and 200 FTD-MND versus 400 controls. No SNP reached genome-wide significance in either subtype, probably due to the small sample size. However, several SNPs (appendix) showed suggestive associations (p values between 1 × 10−6 and 1 × 10−7; figure) and warrant further investigation in future screenings.

Table 2.

Sample characteristics, by subtype

Behavioural frontotemporal
dementia
Semantic dementia Progressive nonfluent aphasia Frontotemporal dementia/
myotrophic lateral sclerosis
FTLD-U





Discovery
phase
Replication
phase
Total Discovery
phase
Replication
phase
Total Discovery
phase
Replication
phase
Total Discovery
phase
Replication
phase
Total Discovery
phase
Replication
phase
Total
USA 315 25 340 147 12 159 81 15 96 36 21 57 0 102 102

Canada 22 5 27 1 1 2 0 5 5 1 7 8 0 11 11

UK* 207 152 359 75 53 128 69 44 113 50 16 66 0 19 19

Spain NA 194 194 NA 41 41 NA 51 51 NA 13 13 NA 10 10

France 135 30 165 3 0 3 8 3 11 59 8 67 0 1 1

Belgium 135 27 162 13 1 14 22 2 24 21 2 23 0 10 10

Netherlands 159 37 196 47 31 78 24 6 30 20 3 23 0 0 0

Denmark 2 NA 2 0 NA 0 1 NA 1 4 NA 4 0 NA 0

Germany 209 18 227 45 8 53 55 6 61 11 1 12 0 0 0

Sweden 7 53 60 2 20 22 6 10 16 3 8 11 0 7 7

Italy 443 186 629 28 22 50 69 86 155 24 16 40 0 61 61

Australia* NA 56 56 NA 26 26 NA 19 19 NA 20 20 NA 0 0

Meta-analysis 1634
(1377)
783
(690)
2417
(2061)
361
(308)
215
(190)
576
(495)
335
(269)
247
(221)
582
(486)
229
(200)
115
(94)
344
(294)
0 221
(177)
221
(177)

NA=.

*

Used the same control samples.

The number of the samples that passed genotyping data quality control and were used for association analyses.

Table 3.

Characteristics of single-nucleotide polymorphisms exceeding genome-wide significance in the discovery phase

Chromosome Base pair Candidate gene Minor
allele
Major
allele
Frequency of
minor allele
(r2 when
applicable)
Imputation
quality
Odds ratio (95% CI) Standard
error
p value
Discovery phase

Behavioural frontotemporal dementia
rs302652 11 87894831 RAB38 A T 0·259 0·9296 0·730 (0·65–0·82) 0·057 2·02×10−8
rs74977128 11 87936874 RAB38/CTSC C T 0·118 0·4182 1·815 (1·48–2·24) 0·107 3·06×10−8
All frontotemporal dementia*
  rs9268877 6 32431147 HLA-DRA/HLA-DRB5 A G 0·440 0·7783 1·331 (1·22–1·45) 0·045 1·65×10−10
  rs9268856 6 32429719 HLA-DRA/HLA-DRB5 A C 0·251 0·8563 0·752 (0·68–0·83) 0·050 1·30×10−8
  rs1980493 6 32363215 BTNL2 C T 0·147 0·9642 0·720 (0·69–0·81) 0·060 4·94×10−8

Replication phase

Behavioural frontotemporal dementia
  rs302668 (proxy) 11 87876911 RAB38 C T 0·325 (0·65) NA 0·877 (0.77–0.99) 0·064 0·041
  rs16913634 (proxy) 11 87934068 RAB38/CTSC A G 0·104 (0·54) NA 0·964 (0.79–1.17) 0·098 0·710
All frontotemporal dementia*
  rs9268877 6 32431147 HLA-DRA/HLA-DRB5 A G 0·449 NA 1·080 (0·98–1·18) 0·047 0·104
  rs9268856 6 32429719 HLA-DRA/HLA-DRB5 A C 0·253 NA 0·878 (0·79–0·97) 0·053 0·014
  rs1980493 6 32363215 BTNL2 C T 0·145 NA 0.85 (0·75–0·97) 0·068 0·020

Discovery and replication combined

Behavioural frontotemporal dementia
  rs302668 (proxy) 11 87876911 RAB38 C T 0·292 (0·65) NA 0·814 (0·71–0·92) 0·064 2·44×10−7
  rs16913634 (proxy) 11 87934068 RAB38/CTSC A G 0·111 (0·54) NA 1·248 (1·14–1·37) 0·049 8·15×10−4
All frontotemporal dementia*
  rs9268877 6 32431147 HLA-DRA/HLA-DRB5 A G 0·4445 NA 1·204 (1·11–1·30) 0·039 1·05×10−8
  rs9268856 6 32429719 HLA-DRA/HLA-DRB5 A C 0·252 NA 0·809 (0·76–0·86) 0·029 5·51×10−9
  rs1980493 6 32363215 BTNL2 C T 0·146 NA 0·775 (0·69–0·86) 0·058 1·57×10−8

Replication and joint analyses were assessed for the same single-nucleotide polymorphisms (SNPs) at 6p21.3, whereas proxy SNPs were used to assess the association at 11q14 (for which r2 values are included). The odds ratio is shown for the minor allele.

NA=.

*

Denotes only minimal cross-subtype heterogeneity, with heterogeneity p values ranging from 0·793 to 0·944 based on Cochran’s Q test.

Heterogeneity p value <0·01 in the meta-analyses of the discovery and replication phases combined.

In the replication phase, we analysed samples from 1372 patients (table 1) and 5094 controls. We assessed the associated SNPs at 6p21.3 (rs9268877, rs9268856, and rs1980493) in the whole replication cohort (table 3). Table 3 shows findings from the surrogate or proxy SNPs assessed for replication at 11q14 in 690 behavioural FTD cases: rs302668 and rs16913634. Combined analyses of discovery and replication phases showed genome-wide significant association at 6p21.3 for all SNPs (table 3). Joint p values of the SNPs at 11q14 only revealed suggestive association for rs302668 (table 3) possibly because of decreased power due to proxy-based replication (r2 of rs302652 to rs302668=0·65).

We then assessed biological relevance for the novel potential loci in human brain cortex tissues assayed for genome-wide expression and methylation. There was no eQTL in our dataset, but assessment of Zeller and colleagues’ dataset38 showed a cis-eQTL (p=5·05 × 10−32; appendix) at 11q14 for rs302652 (chr11:87894881, risk allele T) causing a decreased expression of RAB38 (Illumina ILMN_2134974 located on chr11:87846656-87846705) in monocytes. These data suggest a role in transcriptional processes in cis for this SNP. Furthermore, we identified significant cis-mQTL at 6p21.3 after multiple test correction for rs1980493 (risk allele T) that associated with changes in the methylation levels related to HLADRA in the frontal cortex (table 4).

Table 4.

Summary of association of top hits with cis-methylation levels at 6p21.3

CpG probe Single-
nucleotide
polymorphism
Chromosome Position
(base pair)
Reference
allele
Alternate
allele
Frequency
of
reference
allele
Imputation
quality
Effect
estimate of
alternate
allele (in
Z units)
Standard
error
p value FDR adjsuted
p value
Probe start
(base pair)
Probe end
(base pair)
Symbol
Frontal cortex
(CpG
methylation)
cg21415604 rs1980493 6 32363215 T C 0·8361 0·9888 −0·463 0·116 0·0000701 0·00834666 31948433 31948483 C4B
Frontal cortex
(CpG
methylation)
cg25764570 rs1980493 6 32363215 T C 0·8361 0·9888 −0·652 0·116 2·17×10−8 0·00000773 32407239 32407289 HLA-DRA
Frontal cortex
(CpG
methylation)
cg25764570 rs9268856 6 32429719 C A 0·748 0·9687 −0·484 0·1 1·16×10−6 0·000207417 32407239 32407289 HLA-DRA

Association is shown for rs1980493 and rs9268877, which indicates an involvement of methylation processes and patterns in relation to HLA-DRA.

To assess potential genetic overlap between FTD and closely related forms of neurodegenerative diseases we selected relevant SNPs for candidate loci and analysed them in our dataset. This analysis included published association studies for amyotrophic lateral sclerosis,39 progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD),40 Alzheimer’s disease,41 and FTLD-TDP.17 We also assessed whether the two loci identified through this study had also been reported previously in other studies of neurological disorders.

For the C9orf72 locus (for amyotrophic lateral sclerosis), the SNP rs3849942 (effect allele A) was associated with the FTD-MND subtype, which was consistent with our post-hoc analyses (about 23% of expansion carriers in this subtype; table 5; appendix). Association was modest in behavioural FTD (p=7·38×10−3, OR=1·155) as well as in the entire discovery cohort, but we saw no evidence for association in the semantic dementia or progressive nonfluent aphasia subtypes (table 5). These results confirm that the C9orf72 locus associates mainly with FTD-MND and to a lesser extent with behavioural FTD (appendix).

Table 5.

Comparison with single-nucleotide polymorphisms and loci associated with other disorders

Reference
allele
Alternate
allele
Previous studies This study (discovery phase)


Frequency
of reference
allele
Reported
association
Frequency
of reference
allele
Imputation
quality
(RSQ)
Meta-analysis (all
frontotemporal
dementia)
Behavioural
frontotemporal
dementia
Semantic dementia Progressive
nonfluent aphasia
Frontotemporal
dementia/motor
neuron disease






p value
(joint)
Odds
ratio
p value Odds
ratio
(95% CI)
p value Odds
ratio
(95% CI)
p value Odds
ratio
(95% CI)
p value Odds
ratio
(95% CI)
p value Odds
ratio
(95% CI)
Chromosome 17

Progressive supranuclear palsy or corticobasal degeneration40
  MAPT
    rs8070723 (44081064) A G 0·950 (stage 1); 0·940 (stage 2) 1·50×10−11 5·46 0·765 0·8400 2·80×10−4 1·201 (1·09–1·32) 3·14×10−3 1·201 (1·06–1·36) 4·34×10−1 1·103 (0·86–1·41) 8·72×10−3 1·471 (1·10−1·97) 5·82×10−1 1·091 (0·80–1·49)
    rs242557 (44019712) G A 0·470 (stage 1); 0·500 (stage 2) 4·20×10−70 0·51 0·634 0·5246 4·82×10−3 0·853 (1·05–1·31) 1·27×10−2 0·841 (0·73–0·96) 3·20×10−1 0·867 (0·65–1·15) 2·02×10−1 0·815 (0·59–1·11) 8·23×10−1 0·961 (0·68–1·36)
Chromosome 19
Alzheimer’s disease41
  TOMM40/APOE
    rs2075650 (45395619) G A 0·150 1·80×10−157 2·53 0·141 0·9978 8·81×10−7 1·304 (0·69–0·85) 1·37×10−6 1·383 (0·63–0·82) 3·64×10− 1·326 (0·58–0·98) 8·69×10−1 0·976 (0·76–1·38) 2·06×10−1 1·252 (0·56–1·13)

Chromosome 9

Amyotrophic lateral sclerosis39
  C9orf72/MOB3B
    rs3849942 (27543281) A G 0·260 1·01×10−8 1·20 0·253 0·9996 4·38×10−4 1·166 (1·07–1·27) 7·38×10−3 1·155 (0·78–0·96) 9·89×10−1 1·010 (0·80–1·25) 9·03×10−1 0·990 (0·79–1·31) 2·12×10−6 1·957 (0·39–0·68)

Chromosome 7

FTLD-TDP17
  TMEM106B
    rs1990622 (12283787) A G 0·679 1·08×10−11 1·64 0·600 0·9588 7·88×10−2 1·080 (0·99–1·16) 5·85×10−3 1·144 (1·04–1·26) 8·36×10−1 0·978 (0·80–1·20) 8·98×10−1 0·985 (0·79–1·23) 3·11×10−1 0·876 (0·68–1·13)
    rs6966915 (12265988) C T 0·679 1·63×10−11 1·64 0·596 0·9675 1·21×10−1 1·070 (0·87–1·02) 5·74×10−3 1·144 (1·04–1·26) 5·27×10−1 0·936 (0·76–1·15) 7·26×10−1 0·961 (0·77–1·20) 3·62×10−1 0·888 (0·69–1·14)
    rs1020004 (12255778) T C 0·767 5·00×10−11 1·66 0·693 0·9538 4·59×10−1 1·030 (0·95–1·12) 5·71×10−2 1·104 (1·00–1·22) 8·53×10−1 0·980 (0·79–1·21) 5·00×10−1 0·921 (0·72–1·17) 1·20×10−1 0·805 (0·61–1·06)

Chromosome 6

Multiple sclerosis43,44
  RAB38
    rs1386330 (87819427) C T 0·130 2·00×10−6 NA 0·141 0·9694 3·35×10−1 1·050 (0·85–1·06) 6·09×10−1 1·040 (0·84–1·10) 7·60×10−1 1·040 (0·72–1·27) 6·97×10−1 1·060 (0·68–1·29) 3·00×10−1 0·829 (0·58–1·18)
HLA-DRA
    rs3129871 (32406342) A G 0·230 8·94×10−81 1·99 0·131 0·9734 4·80×10−2 1·122 (1·00–1·26) 2·10×10−1 1·095 (0·79–1·05) 1·25×10−1 1·254 (0·60–1·06) 5·02×10−1 1·120 (0·64–1·24) 6·10×10−1 1·105 (0·61–1·33)
Parkinson’s disease45,46
  HLA-DRA
    rs3129871 (32406342) A C 0·504 5·70×10−15 1·72 0·337 0·9379 3·43×10−1 0·961 (0·88–1·04) 3·15×10−1 0·949 (0·95–1·16) 4·94×10−1 1·078 (0·75–1·15) 8·24×10−1 0·974 (0·81–1·30) 2·72×10−1 0·859 (0·89–1·53)
    rs3129882 (32409530) G A 0·450 1·90×10−10 1·26 0·456 0·9992 3·36×10−2 1·086 (0·85–0·99) 3·27×10−2 1·106 (0·82–0·99) 7·52×10−1 1·033 (0·79–1·18) 5·74×10−1 1·065 (0·75–1·17) 7·07×10−1 1·049 (0·74–1·22)

NA=. FLTD-TDP=frontotemporal lobar degeneration with TDP43-positive inclusions. RSQ=

For the MAPT locus (PSP/CBD), the SNPs rs242557 (effect allele G) and rs8070723 (effect allele A)40 were significantly associated only within the entire cohort and in the behavioural FTD and progressive nonfluent aphasia subtypes (rs8070723 only; table 5). The effect was small in our study although in the same direction as in the GWAS for progressive supranuclear palsy (5·4640 vs about 1·2–1·4 in our study; table 5). These results might have arisen because we excluded all known chromosome 17 mutation carriers and because tau pathology is a less common feature of sporadic FTD.

For the TOMM40/APOE locus (Alzheimer’s disease), the SNP rs2075650 (effect allele G) (table 5). Several Alzheimer’s disease GWASs reported association with the minor allele of this SNP with ORs greater than 2·5,41 but in our study the OR was about 1·3 (table 5). This suggestive association might be indicative of clinical overlap between patients with clinically diagnosed FTD and those with Alzheimer’s disease.42

For the TMEM106B locus (FTLD-TDP), we assessed the three associated SNPs reported by Van Deerlin and colleagues (rs1990622, effect allele A; rs6966915, effect allele C; rs1020004, effect allele T).17 All achieved modest p values in the entire dataset with lowest p values in the range of 10−2–10−3 only in the behavioural FTD subtype (table 5). Van Deerlin and colleagues’ study17 was done on samples from patients with autopsy-confirmed FTLD-TDP, whereas our cohort is mainly clinically defined. Additionally, the previous study included many GRN mutation carriers, who frequently present with behavioural FTD;17 in our study, GRN mutation carriers were excluded. Biochemical evidence has suggested that TMEM106B is directly related to GRN metabolism,13 thus we regard our data as a limited replication of the original finding.

Finally, the RAB38 locus previously showed suggestive association in multiple sclerosis,43 but the HLA locus was reported to associate with multiple sclerosis,44,45 Parkinson’s disease,19,46 and Alzheimer’s disease.47 None of the SNPs reported in these studies, and which were assessed in our dataset (table 5),4346 showed association with FTD, probably suggesting that different risk haplotype sub-structures at the same loci associate with distinctive phenotypes.

Discussion

FTD is characterised by a broad range of clinical manifestations, differential pathological signatures, and substantial genetic variability, which imply a complex disease mechanisms.15 In the search for novel disease risk loci associated with FTD we have done an extensive GWAS on a large cohort of mainly clinically diagnosed FTD samples from patients of European ancestry. Several limitations might apply to this study. In view of the phenotype heterogeneity of FTD, and considering that it is a rare neurodegenerative disorder,2 testing the hypothesis “common variant – common disease” for diseases of this kind is challenging and clearly benefits from large sample sizes. Additionally, our findings might indicate association with specific loci without necessarily implying causality; low heritability due to common variability can also apply. However, the QQ plots and associated λ values (appendix) conformed to GWAS standards, lending support to our findings.

We included samples from more than 3500 patients and, thus, we know of no larger GWAS for FTD. We have identified two novel potential loci for FTD: 11q14, encompassing RAB38/CTSC, was suggestive for the behavioural FTD subtype, and 6p21.3, encompassing the HLA locus was statistically significant for the entire cohort.

RAB3848 encodes the transmembrane protein RAB38, which is expressed in the thyroid, in elements of the immune system, and in the brain. From a functional perspective, RAB38 has been shown to mediate protein trafficking to lysosomal-related organelles and maturation of phagosomes (panel).49,50 CTSC is a lysosomal cysteine-proteinase that participates in the activation of serine proteinases in immune and inflammatory cells that are involved in immune and inflammatory processes including phagocytosis of pathogens and local activation and deactivation of inflammatory factors (Online Mendelian Inheritance in Man [OMIM] number 602365). The SNP rs302652 at the RAB38/CTSC locus shows an eQTL in monocytes38 associated with decreased expression of RAB38, possibly indicating that a decreased function of RAB38 might be the mechanism by which the association at this locus is mediated. Both RAB38 and CTSC are implicated in lysosomal biology and an association with lysosomal and autophagic processes in FTD was previously suggested in two studies of GRN51 and TMEM106B.52 A role for autophagy has also been shown in Parkinson’s disease.53 Our findings will need to be replicated in other FTD cohorts in follow-up studies (eg, fine-mapping studies) to lend support to the proposal that lysosomal biology and autophagy might be involved in the aetiology of FTD.54

The genetic association that we identified with the HLA locus supports the idea of a link between FTD and the immune system. Our mQTL data showed that risk at this locus is associated with cis-changes in methylation levels of HLA-DRA in the frontal cortex. HLA associations have been previously reported in Alzheimer’s disease,47 Parkinson’s disease,19,46 and multiple sclerosis.44,45 Additionally, a general involvement of the innate and the adaptive immune responses has been suggested in the pathogenesis of neurodegenerative diseases,55,56 lending supporting to the idea that the immune system plays an important part within the spectrum of neurological disorders.

Future studies should aim to replicate our findings and, in so doing, elucidate the functional basis of FTD. Additionally, our data indicate that common pathways and processes might underlie different forms of neurodegenerative disorders, including Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and FTD. Exploring the possibility of developing therapeutic measures targeting general damage responses could hold promise—after replication and validation of our findings—for the development and implementation of treatment options for these neurological disorders, including FTD.

Figure 1. Manhattan plots identifying regions with genome–wide significant associations.

Figure 1

(A) Manhattan plot for the entire dataset of the discovery phase depicts the associated regionat 6p21.3. Single–nucleotide polymorphisms (SNPs) with smallest p values and their location within or in proximity of the nearest genes are shown. (B) Manhattan plot for the behavioural frontotemporal dementia set in the discovery phase depicts the associated region at 11q14.

Figure 2. Manhattan plots identifying regions with genome–wide significant associations.

Figure 2

SNPs with smallest p values and their location within or in proximity of the nearest genes are shown.Manhattan plots for semantic dementia (C), progressive nonfluent aphasia (D), and motor neurone disease (E) frontotemporal dementia sets.

Panel: Research in context.

Systematic review

We searched PubMed for the most relevant research articles and review articles on frontotemporal dementia using the following terms: “FTD” and “genetics”, and “FTD” and “review”.1,4,5,817,54 We compared our results to several previously published genome-wide association studies. We identified only one directly relevant study that investigated a pathologically defined subtype of frontotemporal dementia (frontotemporal lobar degeneration with TDP43-positive inclusions; FTLD-TDP).17 The other studies were of related diseases such as amyotrophic lateral sclerosis,39 Alzheimer’s disease,41,47 progressive supranuclear palsy and corticobasal degeneration,40 multiple sclerosis,4345 and Parkinson’s disease.19,46

Interpretation

To the best of our knowledge, ours is the first genome-wide association study in samples from patients with clinical frontotemporal dementia. In view of the complexity and heterogeneity of the disease, mutations in only three main genes—MAPT, GRN, and C9orf72—have been associated with frontotemporal dementia, and these explain only a small proportion of cases. Most importantly, little is known about the mechanisms involved in the development of this disorder. Our findings suggest that common variability in loci that point to immune processes, and possibly to lysosomal biology and autophagy, are involved in the pathobiology of the disease. These findings provide a basis for future replication and functional studies.

Acknowledgments

We received intramural funding from the National Institute of Neurological Disorders and Stroke (NINDS) and National Institute on Aging (NIA), the Wellcome/MRC Centre on Parkinson’s disease, Alzheimer’s Research UK (ARUK, Grant ARUK-PG2012-18), and by the office of the Dean of the School of Medicine, Department of Internal Medicine, at Texas Tech University Health Sciences Center. We thank Mike Hubank and Kerra Pearce at the Genomic core facility at the Institute of Child Health (ICH), UCL, for assisting RF in doing Illumina genotyping experiments (FTD-GWAS genotyping). The work done by the North American Brain Expression Consortium (NABEC) was supported in part by the Intramural Research Program of the National Institute on Aging, National Institutes of Health, part of the US Department of Health and Human Services (project number ZIA AG000932-04), and by a Research Grant from the Department of Defense (W81XWH-09-2-0128). Work done by the UK Brain Expression Consortium (UKBEC) was supported by the MRC through the MRC Sudden Death Brain Bank (CS), by a Project Grant (G0901254 to JH and MW), and by a Fellowship award (G0802462 to MR). DT was supported by the King Faisal Specialist Hospital and Research Centre, Saudi Arabia. Computing facilities used at King’s College London were supported by the National Institute for Health Research (NIHR) Biomedical Research Centre based at Guy’s and St Thomas’ NHS Foundation Trust and King’s College London. We thank AROS Applied Biotechnology AS company laboratories and Affymetrix for their valuable input. JBJK was supported by the National Health and Medical Resarch Council (NHMRC), Australia (project grants 510217 and 1005769). CDS was supported by NHMRC (project grants 630428 and 1005769). PRS was supported by NHMRC (project grants 510217 and 1005769) and acknowledges that DNA samples were prepared by Genetic Repositories Australia, supported by NHMRC Enabling Grant 401184. GMH was supported by NHMRC Research Fellowship 630434, Project Grant 1029538, and Program Grant 1037746. JRH was supported by the Australian Research Council Federation Fellowship, NHMRC Project Grant 1029538 and NHMRC Program Grant 1037746. OP was supported by NHMRC Career Development Fellowship 1022684, Project Grant 1003139. IH, AR, and MB acknowledge the patients and controls who participated in this project and the Trinitat Port-Carbó and her family who are supporting Fundació ACE research programmes. CC was supported by Grant P30-NS069329-01 and acknowledges that the recruitment and clinical characterisation of research participants at Washington University were supported by NIH P50 AG05681, P01 AG03991, and P01 AG026276. LB and GB were supported by the Ricerca Corrente, Italian Ministry of Health. RG was supported by Fondazione CARIPLO 2009-2633, Ricerca Corrente, Italian Ministry of Health. GF was supported by Fondazione CARIPLO 2009–2633. ES was supported by the Italian Ministry of Health. CF was supported by Fondazione Cariplo. MS was supported from the Italian Ministry of Health (Ricerca Corrente). MLW was supported by Government funding of clinical research within NHS Sweden (ALF). KN was supported by Thure Carlsson Foundation. CN was supported by Swedish Alzheimer Fund. IRAM and GYRH were supported by CIHR (grant 74580) PARF (grant C06-01). JG was supported by the NINDS intramural research funds for FTD research. CMM was supported by Medical Research Council UK, Brains for Dementia Research, Alzheimer’s Society, Alzheimer’s Research UK, National Institutes for Health Research, Department of Health, and Yvonne Mairy Bequest, and acknowledges that tissue samples made available for this study were provided by the Newcastle Brain Tissue Resource, which was funded in part by grants G0400074 and G1100540 from the UK MRC, the Alzheimer’s Research Trust and Alzheimer’s Society through the Brains for Dementia Research Initiative and an NIHR Biomedical Research Centre Grant in Ageing and Health, and NIHR Biomedical Research Unit in Lewy Body Disorders. CMM was supported by the UK Department of Health and Medical Research Council and the Research was supported by the National Institute for Health Research Newcastle Biomedical Research Centre based at Newcastle Hospitals Foundation Trust and Newcastle University and acknowledges that the views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health. JA was supported by MRC, Dunhill Medical Trust, and Alzheimer’s Research UK. TDG was supported by Wellcome Trust Senior Clinical Fellow. IGM was supported by NIHR Biomedical Research Centre and Unit on Ageing Grants and acknowledges the National Institute for Health Research Newcastle Biomedical Research Centre based at Newcastle Hospitals Foundation Trust and Newcastle University. The views expressed are those of the authors and not necessarily those of the NHS, the NIHR, or the Department of Health. AJT was supported by Medical Research Council, Alzheimer’s Society, Alzheimer’s Research UK, and the National Institutes for Health Research. EJ was supported by NIHR and Newcastle Biomedical Research Centre. PP, CR, SOC, and EA were supported partially by FIMA (Foundation for Applied Medical Research). PP acknowledges Manuel Seijo-Martínez (Department of Neurology, Hospital do Salnés, Pontevedra, Spain) and Ramon Rene, Jordi Gascon, and Jaume Campdelacreu (Department of Neurology, Hospital de Bellvitge, Barcelona, Spain) for providing FTD DNA samples. RP, JDS, PA, and AK were supported by the German Federal Ministry of Education and Research (BMBF; grant number FKZ 01GI1007A—German FTLD consortium). IR was supported by Ministero dell’Istruzione, dell’Università e della Ricerca (MIUR) of Italy. PStGH was supported by the Canadian Institutes of Health Research, Wellcome Trust, Ontario Research Fund. FT was supported by the Italian Ministry of Health (ricerca corrente) and MIUR grant RBAP11FRE9. GR and GG were supported by the Italian Ministry of Health (ricerca corrente). JBR was supported by Cambridge NIHR Biomedical Research Centre and Wellcome Trust (088324). JU, JC, and SM were supported by the MRC Prion Unit core funding and acknowledge MRC UK, UCLH Biomedical Research Centre, and Queen Square Dementia BRU. SM thanks John Beck, Tracy Campbell, Gary Adamson, Ron Druyeh, Jessica Lowe, and Mark Poulter. AD thanks Benedikt Bader, Manuela Neumann, Sigrun Roeber, Thomas Arzberger, and Hans Kretzschmar. VMVD and JQT were supported by grants AG032953, AG017586 and AG010124. MG was supported by Grants AG032953, AG017586, AG010124, and NS044266. VMVD thanks EunRan Suh for assistance with sample handling and Elisabeth McCarty-Wood for help in selection of patients. JQT thanks Terry Schuck, John Robinson, and Kevin Raible for assistance with neuropathological assessment of patients. CVB and the Antwerp site were in part funded by the MetLife Foundation Award for Medical Research (to CVB), the Belgian Science Policy Office Interuniversity Attraction Poles programme; the Foundation for Alzheimer Research (SAO-FRA); the Medical Foundation Queen Elisabeth; the Flemish Government Methusalem Excellence award (to CVB); the Research Foundation Flanders (FWO); and the University of Antwerp Research Fund. JvdZ holds a postdoctoral fellowship of the FWO. CVB and the Antwerp site authors thanks neurologists S Engelborghs, PP De Deyn, A Sieben, and Rik Vandenberghe and neuropathologist JJ Martin for the clinical and pathological diagnoses. Isabelle Leber and Alexis Brice were supported by the programme Investissements d’avenir ANR-10-IAIHU-06 and acknowledges the contribution of The French Research Network on FTLD/FTLD-ALS for the contribution in samples collection. BN, SS, SB, and IP were supported by Prin 2010-prot.2010PWNJXK; Cassa di Rispario di Firenze e Cassa di Risparmio di Pistoia e Pescia. JEN was supported by the Novo Nordisk Foundation, Denmark. MR was supported by the German National Genome Network (NGFN) and the German Ministry for Education and Research Grant Number 01GS0465. JDR, MNR, NCF, and JDW were supported by an MRC programme grant, the NIHR Queen Square Dementia Biomedical Research Unit and the Leonard Wolfson Experimental Neurology Centre. MGS was supported by MRC grant n G0301152, Cambridge Biomedical Research Centre, and thanks K Westmore for extracting DNA. HM was supported by the Motor Neuron Disease Association (Grant 6057). RR was supported by P50 AG016574, R01 NS080882, R01 NS065782, P50 NS72187, and the Consortium for Frontotemporal Dementia. DWD was supported by P50NS072187, P50AG016574, State of Florida Alzheimer Disease Initiative, and CurePSP Inc. NRGR, JEP, RCP, DK, and BFB were supported by P50 AG016574. KAJ was supported by R01 AG037491. WWS was supported by NIH AG023501, AG019724, Consortium for Frontotemporal Dementia Research. BLM was supported by P50AG023501, P01AG019724, Consortium for FTD Research. HR was supported by AG032306. JCvS was supported by Stichting Dioraphte Foundation (11 02 03 00), Nuts Ohra Foundation (0801-69), Hersenstichting Nederland (BG 2010-02), and Alzheimer Nederland. CG and HHC acknowledge families, patients, clinicians including Inger Nennesmo and Vesna Jelic, Laura Fratiglioni for control samples and Jenny Björkström, Håkan Thonberg, Charlotte Forsell, Anna-Karin Lindström, and Lena Lilius for sample handling. CG was supported by Swedish Brain Power (SBP), the Strategic Research Programme in Neuroscience at Karolinska Institutet (StratNeuro), the regional agreement on medical training and clinical research (ALF) between Stockholm County Council and Karolinska Institutet, Swedish Alzheimer Foundation, Swedish Research Council, Karolinska Institutet PhD-student funding, King Gustaf V, and Queen Victoria’s Free Mason Foundation. FP, AR, VD, and FL acknowledge Labex DISTALZ. RF acknowledges the help and support of June Howard at the Texas Tech University Health Sciences Center Office of Sponsored Programs for tremendous help in managing Material Transfer Agreement at TTUHSC.

CVB and MC are inventors on patent applications for GRN and C9orf72. PRS receives speaker fees from Janssen pharmaceutical. RR receives research support from the NIH (R01 NS080882, R01 NS065782, R01 AG026251, R01 NS076471, and P50 AG16574), the ALS Therapy Alliance, and the Consortium for Frontotemporal Degeneration Research, honoraria for lectures or educational activities not funded by industry. RR serves on the medical advisory board of the Association for Frontotemporal Degeneration and the board of directors of the International Society for Frontotemporal Dementia, and holds a patent on methods to screen for the hexanucleotide repeat expansion in the C9ORF72 gene. DWD serves on the editorial boards of the American Journal of Pathology, Journal of Neuropathology and Experimental Neurology, Brain Pathology, Neurobiology of Aging, Journal of Neurology, Neurosurgery, and Psychiatry, Annals of Neurology, and Neuropathology. DWD is supported by NIH grants (P50 AG16574, P50 NS72187, P01 AG03949), the Mangurian Foundation, CurePSP, and the Robert E Jacoby Professorship for Alzheimer’s Research. NRGR is on the Scientific Advisory Board for Codman, TauRzx multicenter study, Consultation for CYTOX. RCP chairs a Data Monitoring Committee for Pfizer and Janssen Alzheimer Immunotherapy, and is a consultant for GE Healthcare and Elan Pharmaceuticals. RCP receives royalties from Oxford University Press for Mild Cognitive Impairment. DK is Deputy Editor for Neurology. DK has served on a data safety monitoring board for Lilly Pharmaceuticals, as a consultant to TauRx, was an investigator in clinical trials sponsored by Baxter, Elan Pharmaceuticals, and Forest Pharmaceuticals in the past 2 years and receives research support from the NIH. BFB has served as an investigator for clinical trials sponsored by Cephalon Inc, Allon Pharmaceuticals, and GE Healthcare. BFB receives royalties from the publication of a book entitled Behavioral Neurology Of Dementia (Cambridge Medicine, 2009). BFB has received honoraria from the American Academy of Neurology. BFB serves on the Scientific Advisory Board of the Tau Consortium. BFB receives research support from the National Institute on Aging (P50 AG016574, U01 AG006786, RO1 AG032306, RO1 AG041797) and the Mangurian Foundation. BLM is on the Board Membership of The Larry L Hillblom Foundation, The John Douglas French Foundation, The Tau Consortium, Sagol School of Neuroscience Tel Aviv University. BLM holds consultancy for Tau Rx lts—Chair, Scientific Advisory Board bvFTD Trial Allon Therapeutics—Steering Committee AL-108-231 Study, Bristol-Myers Squibb-Advisory Board, Progressive Supranuclear Palsy (PSP), Neurology Scientific Advisory Board Meeting Siemens Molecular Imaging, and Eli Lilly US Alzheimer’s Disease Advisory Board, and receives royalties from Cambridge University Press Guilford Publications Inc, Neurocase.

Footnotes

See Online for appendix

Contributors

JH, PM, ABS, MAN, RF, and JDR designed the study. JDR, RF and JH did the clinical quality checks. RF coordinated sample collection, received samples at UCL and TTUHSC, and did material quality control for discovery and replication phases. DGH received samples at NIH and coordinated material quality control at NIH. JDR, JBJK, CDS, PRS, WSB, JRH, GMH, OP, LB, ET, EH, IH, AR, MB BB, AP, LB, GB, RG, GF, DG, ES, CF, MS, JC, AL, RB, MLW, KN, CN, IRAM, GYRH, DMAM, JG, CMM, JA, TDG, IGM, AJT, PP, EDH, EMW, AB, EJ, MCT, PP, CR, SOC, EA, RP, JDS, PA, AK, IR, ER, LP, ER, PStGH, ER, GR, FT, GG, JBR, JCMS, JU, JC, SM, AD, VMVD, MG, JQT, JvdZ, TVL, CVB, WD, MC, SFC, ILB, AB, DH, VG, MV, BN, SS, SB, IP, JEN, LEH, MR, BI, MM, GG, SP, WG, MNR, NCF, JDW, MGS, HM, PR, PH, JSS, AG, AR, SR, ACB, RM, FF, CC, LB, MA, MG, MEC, NS, RR, MB, DWD, JEP, NRGR, RCP, DK, KAJ, BFB, WWS, BLM, AMK, HR, JCvS, EGPD, HS, YALP, PS, GL, RC, VN, AAP, MF, AP, GM, PS, HHC, CG, FP, AR, VD, FL, DK, LF, and SPB collected and characterised samples. MK was responsible for genotyping at ICH. JH, PM, ABS, and SPB provided funding for this study. JH, PM, and ABS supervised the study. MAN did statistical and association analyses. RF, MAN, and JH analysed and interpreted the data. AR helped in the interpretation of the e/mQTL data. RF, MAN, JH, and PM wrote the first draft of the paper. All other co-authors participated in preparation of the paper by reading and commenting on drafts before submission.

Declaration of interests

RF, DGH, MAN, JDR, AR, JBJK, CDS, WSB, GMH, JRH, OP, LB, ET, EH, IH, AR, MB, BB, AP, CC, NJC, LB, GB, RG, GF, DG, CF, MS, ES, JC, AL, RB, MLW, KN, CN, IRAM, GYRH, DMAM, JG, CMM, JA, TDG, IGM, AJT, PP, EDH, EMW, AB, EJ, MCT, PP, CR, SOC, EA, RP, JDS, PA, AK, IR, ER, LP, ER, PStGH, GR, FT, GG, JBR, JCMS, JU, JC, SM, AD, VMVD, MG, JQT, JvdZ, WD, TVL, SFC, ILB, DH, VG, MV, AB, BN, SS, SB, IP, JEN, LEH, MR, MM, BI, GG, SP, WG, MNR, NCF, JDW, MGS, HRM, PR, PH, JSS, SR, AR, AG, ACB, RM, FF, CC, LB, MA, MG, MEC, NS, MB, KAJ, JEP, WWS, AMK, HR, JCvS, EGPD, HS, YALP, PS, GL, RC, VN, AAP, MF, AP, GM, PS, MK, HHC, CG, FP, AR, VD, FL, DK, LF, SPB, JH, PM, and ABS declare no competing interests.

References

  • 1.Neary D, Snowden JS, Gustafson L, et al. Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria. Neurology. 1998;51:1546–1554. doi: 10.1212/wnl.51.6.1546. [DOI] [PubMed] [Google Scholar]
  • 2.Rabinovici GD, Miller BL. Frontotemporal lobar degeneration: epidemiology, pathophysiology, diagnosis and management. CNS Drugs. 2010;24:375–398. doi: 10.2165/11533100-000000000-00000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rohrer JD, Warren JD. Phenotypic signatures of genetic frontotemporal dementia. Curr Opin Neurol. 2011;24:542–549. doi: 10.1097/WCO.0b013e32834cd442. [DOI] [PubMed] [Google Scholar]
  • 4.Rascovsky K, Hodges JR, Knopman D, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain. 2011;134:2456–2477. doi: 10.1093/brain/awr179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gorno–Tempini ML, Hillis AE, Weintraub S, et al. Classification of primary progressive aphasia and its variants. Neurology. 2011;76:1006–1014. doi: 10.1212/WNL.0b013e31821103e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mackenzie IR, Neumann M, Baborie A, et al. A harmonized classification system for FTLD–TDP pathology. Acta Neuropathol. 2011;122:111–113. doi: 10.1007/s00401-011-0845-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Halliday G, Bigio EH, Cairns NJ, Neumann M, Mackenzie IR, Mann DM. Mechanisms of disease in frontotemporal lobar degeneration: gain of function versus loss of function effects. Acta Neuropathol. 2012;124:373–382. doi: 10.1007/s00401-012-1030-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hutton M, Lendon CL, Rizzu P, et al. Association of missense and 5′–splice–site mutations in tau with the inherited dementia FTDP–17. Nature. 1998;393:702–705. doi: 10.1038/31508. [DOI] [PubMed] [Google Scholar]
  • 9.Baker M, Mackenzie IR, Pickering-Brown SM, et al. Mutations in progranulin cause tau–negative frontotemporal dementia linked to chromosome 17. Nature. 2006;442:916–919. doi: 10.1038/nature05016. [DOI] [PubMed] [Google Scholar]
  • 10.Cruts M, Gijselinck I, van der Zee J, et al. Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature. 2006;442:920–924. doi: 10.1038/nature05017. [DOI] [PubMed] [Google Scholar]
  • 11.DeJesus–Hernandez M, Mackenzie IR, Boeve BF, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72:245–256. doi: 10.1016/j.neuron.2011.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Renton AE, Majounie E, Waite A, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron. 2011;72:257–268. doi: 10.1016/j.neuron.2011.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ferrari R, Thumma A, Momeni P. eLS. Chichester: John Wiley & Sons, Ltd; 2013. Molecular Genetics of Frontotemporal Dementia. [Google Scholar]
  • 14.van der Zee J, Gijselinck I, Dillen L, et al. A Pan-European Study of the C9orf72 Repeat Associated with FTLD: Geographic Prevalence, Genomic Instability, and Intermediate Repeats. Hum mutat. 2013;34:363–373. doi: 10.1002/humu.22244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rohrer JD, Rosen HJ. Neuroimaging in frontotemporal dementia. Int Rev Psychiatry. 2013;25:221–229. doi: 10.3109/09540261.2013.778822. [DOI] [PubMed] [Google Scholar]
  • 16.Ling SC, Polymenidou M, Cleveland DW. Converging Mechanisms in ALS and FTD: Disrupted RNA and Protein Homeostasis. Neuron. 2013;79:416–438. doi: 10.1016/j.neuron.2013.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Van Deerlin VM, Sleiman PM, Martinez–Lage M, et al. Common variants at 7p21 are associated with frontotemporal lobar degeneration with TDP-43 inclusions. Nat Genet. 2010;42:234–239. doi: 10.1038/ng.536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Strong MJ, Grace GM, Freedman M, et al. Consensus criteria for the diagnosis of frontotemporal cognitive and behavioural syndromes in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2009;10:131–146. doi: 10.1080/17482960802654364. [DOI] [PubMed] [Google Scholar]
  • 19.International Parkinson Disease Genomics Consortium. Nalls MA, Plagnol V, et al. Imputation of sequence variants for identification of genetic risks for Parkinson’s disease: a meta-analysis of genome-wide association studies. Lancet. 2011;377:641–649. doi: 10.1016/S0140-6736(10)62345-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.1000 Genomes Project Consortium. Abecasis GR, Auton A, et al. An integrated map of genetic variation from 1092 human genomes. Nature. 2012;491:56–65. doi: 10.1038/nature11632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.International HapMap 3 Consortium. Altshuler DM, Gibbs RA, et al. Integrating common and rare genetic variation in diverse human populations. Nature. 2010;467:52–58. doi: 10.1038/nature09298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich D. Principal components analysis corrects for stratification in genome-wide association studies. Nature Gen. 2006;38:904–909. doi: 10.1038/ng1847. [DOI] [PubMed] [Google Scholar]
  • 23.Grove ML, Yu B, Cochran BJ, et al. Best Practices and Joint Calling of the HumanExome BeadChip: The CHARGE Consortium. PLoS One. 2013;8:e68095. doi: 10.1371/journal.pone.0068095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Millar T, Walker R, Arango JC, et al. Tissue and organ donation for research in forensic pathology: the MRC Sudden Death Brainand Tissue Bank. J Pathol. 2007;213:369–375. doi: 10.1002/path.2247. [DOI] [PubMed] [Google Scholar]
  • 25.Beach TG, Sue LI, Walker DG, et al. The Sun Health Research Institute Brain Donation Program: description and experience, 1987–2007. Cell Tissue Bank. 2008;9:229–245. doi: 10.1007/s10561-008-9067-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hawrylycz MJ, Lein ES, Guillozet–Bongaarts AL, et al. An anatomically comprehensive atlas of the adult human brain transcriptome. Nature. 2012;489:391–399. doi: 10.1038/nature11405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kang HJ, Kawasawa YI, Cheng F, et al. Spatio-temporal transcriptome of the human brain. Nature. 2011;478:483–489. doi: 10.1038/nature10523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Roth RB, Hevezi P, Lee J, et al. Gene expression analyses reveal molecular relationships among 20 regions of the human CNS. Neurogenetics. 2006;7:67–80. doi: 10.1007/s10048-006-0032-6. [DOI] [PubMed] [Google Scholar]
  • 29.Trabzuni D, Ryten M, Walker R, et al. Quality control parameters on a large dataset of regionally dissected human control brains for whole genome expression studies. J Neurochem. 2011;119:275–282. doi: 10.1111/j.1471-4159.2011.07432.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Irizarry RA, Hobbs B, Collin F, et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 2003;4:249–264. doi: 10.1093/biostatistics/4.2.249. [DOI] [PubMed] [Google Scholar]
  • 31.Ramasamy A, Trabzuni D, Gibbs JR, et al. Resolving the polymorphism-in-probe problem is critical for correct interpretation of expression QTL studies. Nucleic Acids Res. 2013;41:e88. doi: 10.1093/nar/gkt069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.International Parkinson’s Disease Genomics Consortium (IPDGC); Wellcome Trust Case Control Consortium 2 (WTCCC2) A two-stage meta-analysis identifies several new loci for Parkinson’s disease. PLoS Genet. 2011;7:e1002142. doi: 10.1371/journal.pgen.1002142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Li Y, Willer C, Sanna S, Abecasis G. Genotype imputation. Annu Rev Genomics Hum Genet. 2009;10:387–406. doi: 10.1146/annurev.genom.9.081307.164242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Li Y, Willer CJ, Ding J, Scheet P, Abecasis GR. MaCH: using sequence and genotype data to estimate haplotypes and unobserved genotypes. Genet Epidemiol. 2010;34:816–834. doi: 10.1002/gepi.20533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gibbs JR, van der Brug MP, Hernandez DG, et al. Abundant quantitative trait loci exist for DNA methylation and gene expression in human brain. PLoS Genet. 2010;6:e1000952. doi: 10.1371/journal.pgen.1000952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Barbosa-Morais NL, Dunning MJ, Samarajiwa SA, et al. A re-annotation pipeline for Illumina BeadArrays: improving the interpretation of gene expression data. Nucleic Acids Res. 2010;38:e1.7. doi: 10.1093/nar/gkp942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Shabalin AA. Matrix eQTL: Ultra fast eQTL analysis via large matrix operations. Bioinformatics. 2012;28:1353–1358. doi: 10.1093/bioinformatics/bts163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zeller T, Wild P, Szymczak S, et al. Genetics and beyond the transcriptome of human monocytes and disease susceptibility. PLoS One. 2010;5:e10693. doi: 10.1371/journal.pone.0010693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.van Es MA, Veldink JH, Saris CG, et al. Genome-wide association study identifies 19p13.3 (UNC13A) and 9p21.2 as susceptibility loci for sporadic amyotrophic lateral sclerosis. Nat Genet. 2009;41:1083–1087. doi: 10.1038/ng.442. [DOI] [PubMed] [Google Scholar]
  • 40.Hölinger GU, Melhem NM, Dickson DW, et al. Identification of common variants influencing risk of the tauopathy progressive supranuclear palsy. Nat Genet. 2011;43:699–705. doi: 10.1038/ng.859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Seshadri S, Fitzpatrick AL, Ikram MA, et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA. 2010;303:1832–1840. doi: 10.1001/jama.2010.574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.van der Zee J, Sleegers K, Van Broeckhoven C. Invited article: the Alzheimer disease-frontotemporal lobar degeneration spectrum. Neurology. 2008;71:1191–1197. doi: 10.1212/01.wnl.0000327523.52537.86. [DOI] [PubMed] [Google Scholar]
  • 43.Baranzini SE, Wang J, Gibson RA, et al. Genome wide association analysis of susceptibility and clinical phenotype in multiple sclerosis. Hum Mol Genet. 2009;18:767–778. doi: 10.1093/hmg/ddn388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.International Multiple Sclerosis Genetics Consortium. Hafler DA, Compston A, et al. Risk alleles for multiple sclerosis identified by a genomewide study. N Engl J Med. 2007;357:851–862. doi: 10.1056/NEJMoa073493. [DOI] [PubMed] [Google Scholar]
  • 45.Mero IL, Gustavsen MW, Sæther HS, et al. Oligoclonal band status in Scandinavian multiple sclerosis patients is associated with specific genetic risk alleles. PLoS One. 2013;8:e58352. doi: 10.1371/journal.pone.0058352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hamza TH, Zabetian CP, Tenesa A, et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat Genet. 2010;42:781–785. doi: 10.1038/ng.642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lambert JC, Ibrahim-Verbaas CA, Harold D, et al. Meta-analysis of 74046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013;45:1452–1458. doi: 10.1038/ng.2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Jäger D, Stockert E, Jäger E, et al. Serological cloning of a melanocyte rab guanosine 5-prime-triphosphate-binding protein and a chromosome condensation protein from a melanoma complementary DNA library. Cancer Res. 2000;60:3584–3359. [PubMed] [Google Scholar]
  • 49.Bultema JJ, Ambrosio AL, Burek CL, Di Pietro SM. BLOC-2, AP-3, and AP-1 proteins function in concert with Rab38 and Rab32 proteins to mediate protein trafficking to lysosome–related organelles. J Biol Chem. 2012;287:19550–19563. doi: 10.1074/jbc.M112.351908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Seto S, Tsujimura K, Koide Y. Rab GTPAses regulating phagosome maturation are differentially recruited to mycobacterial phagosomes. Traffic. 2011;12:407–420. doi: 10.1111/j.1600-0854.2011.01165.x. [DOI] [PubMed] [Google Scholar]
  • 51.Hu F, Padukkavidana T, Vægter CB, et al. Sortilin–mediated endocytosis determines levels of the frontotemporal dementia protein, progranulin. Neuron. 2010;68:654–667. doi: 10.1016/j.neuron.2010.09.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Brady OA, Zheng Y, Murphy K, Huang M, Hu F. The frontotemporal lobar degeneration risk factor, TMEM106B, regulates lysosomal morphology and function. Hum Mol Genet. 2013;22:685–695. doi: 10.1093/hmg/dds475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Westbroek W, Gustafson AM, Sidransky E. Exploring the link between glucocerebrosidase mutations and parkinsonism. Trends Mol Med. 2011;17:485–493. doi: 10.1016/j.molmed.2011.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hardy J1, Rogaeva E. Motor neuron disease and frontotemporal dementia: sometimes related, sometimes not. Exp Neurol. 2013 doi: 10.1016/j.expneurol.2013.11.006. pii: S0014–4886(13)00330–0. [DOI] [PubMed] [Google Scholar]
  • 55.Amor S, Woodroof N. Review series on immune responses in neurodegenerative diseases: innate and adaptive immune responses in neurodegeneration and repair. Immunology. 2013 [Google Scholar]
  • 56.Träger U, Tabrizi SJ. Peripheral inflammation in neurodegeneration. J Mol Med. 2013;91:673–681. doi: 10.1007/s00109-013-1026-0. [DOI] [PubMed] [Google Scholar]

RESOURCES