Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jul 3.
Published in final edited form as: Neuron. 2024 May 2;112(13):2142–2156.e5. doi: 10.1016/j.neuron.2024.04.002

Genome-sequence analyses identify novel risk loci for multiple system atrophy

Ruth Chia 1,*, Anindita Ray 2,*, Zalak Shah 2, Jinhui Ding 3, Paola Ruffo 1,4, Masashi Fujita 5, Vilas Menon 5, Sara Saez-Atienzar 1, Paolo Reho 2, Karri Kaivola 2, Ronald L Walton 6, Regina H Reynolds 7,8,9, Ramita Karra 1, Shaimaa Sait 2, Fulya Akcimen 1, Monica Diez-Fairen 10, Ignacio Alvarez 10, Alessandra Fanciulli 11, Nadia Stefanova 11, Klaus Seppi 11, Susanne Duerr 11, Fabian Leys 11, Florian Krismer 11, Victoria Sidoroff 11, Alexander Zimprich 12, Walter Pirker 13, Olivier Rascol 14, Alexandra Foubert-Samier 15, Wassilios G Meissner 15,16,17, François Tison 15,16, Anne Pavy-Le Traon 18, Maria Teresa Pellecchia 19, Paolo Barone 20, Maria Claudia Russillo 20, Juan Marín-Lahoz 21,22,23, Jaime Kulisevsky 21,22, Soraya Torres 22, Pablo Mir 24,25,26, Maria Teresa Periñán 24,27, Christos Proukakis 28, Viorica Chelban 29,30, Lesley Wu 29, Yee Y Goh 29, Laura Parkkinen 31, Michele T Hu 32, Christopher Kobylecki 33, Jennifer A Saxon 34,35, Sara Rollinson 36, Emily Garland 37, Italo Biaggioni 37, Irene Litvan 38, Ileana Rubio 38, Roy N Alcalay 39,40, Kimberly T Kwei 39, Steven J Lubbe 41, Qinwen Mao 42,43, Margaret E Flanagan 42,44,45, Rudolph J Castellani 42, Vikram Khurana 46,47,48, Alain Ndayisaba 46,11, Andrea Calvo 49, Gabriele Mora 50, Antonio Canosa 49, Gianluca Floris 51, Ryan C Bohannan 52, Anni Moore 3, Lucy Norcliffe-Kaufmann 53, Jose-Alberto Palma 53, Horacio Kaufmann 53, Changyoun Kim 54, Michiyo Iba 54, Eliezer Masliah 54, Ted M Dawson 55,56,57,58, Liana S Rosenthal 55, Alexander Pantelyat 55, Marilyn S Albert 55, Olga Pletnikova 59,60, Juan C Troncoso 59, Jon Infante 61, Carmen Lage 61, Pascual Sánchez-Juan 61,62, Geidy E Serrano 63, Thomas G Beach 63, Pau Pastor 64,65, Huw R Morris 66, Diego Albani 67, Jordi Clarimon 68,69, Gregor K Wenning 70,, John A Hardy 9,71,72,73,74, Mina Ryten 7,8, Eric Topol 75, Ali Torkamani 75, Adriano Chiò 49,76,77, David A Bennett 78, Philip L De Jager 5, Philip A Low 79, Wolfgang Singer 79, William P Cheshire 80, Zbigniew K Wszolek 80, Dennis W Dickson 6,#, Bryan J Traynor 1,55,81,#, J Raphael Gibbs 3,#, Clifton L Dalgard 82,#, Owen A Ross 6,83,#, Henry Houlden 29,30,#, Sonja W Scholz 2,55,84,#
PMCID: PMC11223971  NIHMSID: NIHMS1986964  PMID: 38701790

Summary

Multiple system atrophy (MSA) is an adult-onset, sporadic synucleinopathy characterized by parkinsonism, cerebellar ataxia, and dysautonomia. The genetic architecture of MSA is poorly understood, and treatments are limited to supportive measures. Here, we performed a comprehensive analysis of whole-genome sequence data from 888 European-ancestry MSA cases and 7,128 controls to systematically investigate the genetic underpinnings of this understudied neurodegenerative disease. We identified four significantly associated risk loci using a genome-wide association study approach. Transcriptome-wide association analyses prioritized USP38-DT, KCTD7, and lnc-KCTD7-2 as novel susceptibility genes for MSA within these loci, and single-nucleus RNA sequence analysis found that the associated variants acted as cis-expression quantitative trait loci for multiple genes across neuronal and glial cell types. In conclusion, this study highlights the role of genetic determinants in the pathogenesis of MSA, and the publicly available data from this study represent a valuable resource for investigating synucleinopathies.

Keywords: multiple system atrophy (MSA), whole genome sequencing, genome-wide association study (GWAS), transcriptome-wide association study (TWAS), gene-burden analysis, colocalization, pathway analysis, repeat expansion mapping

Graphical Abstract

graphic file with name nihms-1986964-f0001.jpg

eTOC

Chia et al. comprehensively analyzed genome sequence data from patients with multiple system atrophy (MSA) and controls. The study identified four novel risk loci associated with MSA and prioritized significantly associated genes (USP38-DT, KCTD7, and lnc-KCTD7-2) within these loci. This initiative's data constitute a valuable resource for the research community.

Introduction

The three main synucleinopathies – neurological conditions characterized by abnormal α-synuclein protein aggregates – are Parkinson’s disease, Lewy body dementia, and multiple system atrophy (MSA).1 Considerable progress has been made in unraveling the genetic architecture of Parkinson’s disease and Lewy body dementia.2,3 In contrast, the molecular causes of MSA are poorly understood due to its rarity in the community (~15,000 patients in the United States), its sporadic nature, the heterogeneous clinical manifestations, and the possibility of mimic syndromes.4 Consequently, MSA remains the least understood member within the synucleinopathy triad.

Clinically, MSA is classified as a sporadic, adult-onset neurodegenerative disease that presents with variable combinations of parkinsonism, cerebellar ataxia, pyramidal signs, and dysautonomia.5 The mean age at disease onset is 55 years, and most patients die within six to ten years.6 Pathologically, MSA is defined by widespread neuronal loss and gliosis, with the deposition of fibrillar α-synuclein in oligodendroglial cells that spreads throughout the brain using prion-like mechanisms.7 As for most neurodegenerative diseases, no disease-modifying therapies are available, and treatments are directed toward managing the patient's symptoms.

Identifying genetic risk loci is at the heart of efforts to understand the pathogenesis of MSA and inform translational efforts. Candidate gene studies have implicated risk variants within the SNCA, GBA1, MAPT, and COQ2 genes, though replicating these loci has been challenging.8-16 Here, we attempted to fill this critical knowledge gap by generating a whole genome sequencing dataset and analyzing it to discover risk variants driving the risk of MSA. Our genome-wide association studies (GWAS) and transcriptomic analyses identified novel risk genes. We further examined the genetic architecture of MSA by mapping pathogenic repeat expansions and investigating rare, damaging variants. Importantly, we provide a valuable resource to stimulate and advance research in this understudied, fatal neurodegenerative disease.

Results

Genome-sequence data from 888 patients diagnosed with MSA and 7,128 control subjects were included in the analysis after quality control (see Figure S1 for a workflow diagram).

GWAS identifies novel MSA risk loci on chromosomes 4q31.21 and 15q23

We conducted a GWAS of ~ 9.2 million variants with a minor allele frequency (MAF) of 1% or higher to identify genetic loci associated with MSA. In Figure 1A, we show the Manhattan plot for the genome-wide analysis using the additive model, and Table 1 provides details of the most associated variants. Under this model, we identified a significantly associated locus on chromosome 4q31.21 (rs55894236, odds ratio (OR) = 1.37, 95% confidence interval (CI) = 1.23–1.52, p-value = 1.43 × 10−8). The index variant was within the first intron of the GAB1 gene (see Figure S2 for a regional association plot). We discovered a second locus on chromosome 15q23 near the RNA gene lnc-LRRC49-3 (rs142721461, OR = 2.30, 95% CI = 1.71–3.08, p-value = 3.40 × 10−8). The quantile-quantile (QQ) plot showed no notable population stratification (additive model λ1000 = 1.045; Figure S3), and a conditional analysis based on the index variants of these two loci found no additional signals (Figure S4).

Figure 1. Genome-wide and transcriptome-wide association study results in MSA.

Figure 1.

(A) Composite figure showing the additive MSA GWAS model (upper panel) with the corresponding TWAS results (lower panel) in 888 cases and 7,128 controls (MAF > 1%). The x-axis denotes the chromosomal position in hg38, and the y-axis shows the association p-values on a negative-log10 scale. Each dot represents a variant (GWAS) or a transcript (TWAS). The TWAS results were generated using GTEx gene expression data (version 8) for the hippocampus (additive model) and the caudate (recessive model). Red dots indicate genome-wide significant variants, while orange dots are sub-significant signals. A red dashed line indicates the Bonferroni threshold for genome-wide significance (5.0 × 10−8 for the GWAS, 1.38 × 10−5 for the TWAS in the hippocampus, and 6.77 × 10−6 for the TWAS in the caudate). The blue dashed line denotes the threshold for declaring variants to be subsignificant (5.0 × 10−7). The gene(s) closest to the index variant at each locus in the GWAS is listed. In the TWAS plot, the dot with a black diamond outline indicates a colocalization posterior prior probability hypothesis H4 > 0.80. Panel (B) shows the recessive MSA GWAS model with the corresponding TWAS results in the caudate.

Table 1.

Top association signals in the MSA GWAS using additive or recessive models

Chr. Position
(SNP ID)
Nearest Gene EA/OA EAF
Cases/Controls
OR 95% CI p-value
Additive GWAS model
4 143,370,884
(rs55894236)
GAB1 C/T 0.44/0.39 1.37 1.23–1.52 1.43 × 10−8
15 70,380,309
(rs142721461)
lnc-LRRC49-3 A/C 0.04/0.02 2.30 1.71–3.08 3.40 × 10−8
Recessive GWAS model
5 168,050,511
(rs77075949)
TENM2 T/TC 0.08/0.07 7.40 3.74–14.67 9.73 × 10−9
7 66,699,548
(rs11766262)
RABGEF1 C/T 0.63/0.59 1.95 1.55–2.46 1.65 × 10−8

The gene in closest proximity to the index variant at each locus is represented. The chromosomal position is shown according to hg38. The genome-wide significance was defined as a p-value < 5.00 × 10−8. The effect allele is defined as the allele associated with an increased risk of disease (i.e., odds ratio > 1.0). Abbreviations: Chr., chromosome; EA, effect allele; OA, other allele; EAF, effect allele frequency; OR, odds ratio; 95% CI, 95% confidence interval.

A recessive GWAS identifies two additional loci at chromosomes 5q34 and 7q11.21

We also performed a GWAS under a recessive model, as prior research suggested a recessive inheritance pattern within MSA families.8,17 This analysis identified a locus on chromosome 5q34 within the TENM2 gene (rs77075949, OR = 7.40, 95% CI = 3.74–14.67, p-value = 9.73 × 10−9; Figure 1B, Table 1). A second associated locus was located on the long arm of chromosome 7, within RABGEF1 and near the KCTD7 gene, and significantly associated with MSA (rs11766262, OR = 1.95, 95% CI = 1.55–2.46, p-value = 1.65 × 10−8; see Figure 1B for the Manhattan plot; Table 1). Regional association and QQ plots for the recessive model (λ1000 = 0.97) are shown in Figure S2 and Figure S3. A conditional analysis based on the index variants found no additional signals (Figure S4).

Rare variant analyses identify enrichment of missense mutations in KCTD7

Next, we examined our whole genome sequence data for evidence of damaging mutations in candidate genes that included GBA1, SNCA, MAPT, COQ2, KCTD7, GAB1, and TENM2. To do this, we used gene-level sequence kernel association-optimized (SKAT-O) tests of missense and loss-of-function mutations with a MAF threshold of ≤ 1% and a minor allele count ≥ 2.18 Although genome-wide significance was not achieved due to the relatively small cohort size, we found that rare missense mutations in KCTD7 were nominally associated with the risk of developing MSA (p-value = 7.9 × 10−3, Table 2) when the analysis was limited to the previously implicated genes. The variants identified in canonical and non-canonical transcripts of the KCTD7 gene are listed in Table S1, and their distribution relative to the gene and protein structure is shown in Figure S5. No statistically significant associations were identified in other tested candidate genes or in a genome-wide gene-based analysis (Figure S6).

Table 2.

Rare variant association analysis

Gene Mutation Type SKAT-O p-value
GBA1 Loss-of-function -
Missense 0.69
SNCA Loss-of-function -
Missense -
MAPT Loss-of-function -
Missense 0.91
COQ2 Loss-of-function -
Missense 1.00
RABGEF1 Loss-of-function -
Missense -
KCTD7 Loss-of-function -
Missense 7.9 × 10−3
GAB1 Loss-of-function -
Missense 0.47
TENM2 Loss-of-function 0.25
Missense 0.68

SKAT-O test results for rare missense and loss-of-function variants in the gene-set analysis (MAF < 0.01, minor allele count ≥ 2, minor transcript count ≥ 2). Abbreviation: SKAT-O, sequence kernel association test - optimized.

Robustness of the GWAS signals

We tested the associations' robustness by performing leave-one-out analyses in the overall cohort. These tests showed the same directions of effect at the four GWAS loci (GAB1, lnc-LRRC49-3, TENM2, RABGEF1), demonstrating the strength of the association signals (Figure S7). We also performed sensitivity analyses by performing GWAS using only the pathologically confirmed subset of the MSA cases (n = 468 cases versus 7,128 controls). Although none of the loci achieved genome-wide significance in this investigation due to the smaller cohort size, the sensitivity evaluation demonstrated the same directions of effect and overall robustness of our findings (GAB1: rs55894236:C, OR = 1.42, 95% CI = 1.23–1.64, p-value = 1.23 × 10−6; lnc-LRRC49-3: rs142721461:A, OR = 2.12, 95% CI = 1.44–3.11, p-value = 1.21 × 10−4; TENM2: rs77075949:T, OR = 7.70, 95% CI = 3.35–17.71, p-value = 1.56 × 10−6; RABGEF1: rs11766262:C, OR = 1.89, 95% CI = 1.39–2.56, p-value = 4.31× 10−6).

Transcriptome-wide association analyses implicate additional RNA transcripts

We performed a transcriptome-wide association study (TWAS) using bulk RNA sequence data from the Genotype-Tissue Expression (GTEx) portal (https://www.gtexportal.org). We evaluated a broad range of CNS regions (n = 13) due to the multisystem nature of the disease and the widespread distribution of neuropathological changes observed in MSA brains at autopsy.19 This analysis identified transcripts associated with MSA (see Figures 1A and 1B, lower panels). At the 4q31.21 locus, the TWAS prioritized USP38-DT, a long non-coding RNA (lncRNA) located 293 kilobases (kb) upstream of GAB1. Lower expression of USP38-DT was predicted to increase the risk for MSA (rs300925, p-value within hippocampus = 4.38 × 10−6, Z = −4.59, Table 3). The risk allele of the lead GWAS SNP (rs55894236-C) in this locus was similarly associated with decreased expression of USP38-DT in the GTEx bulk brain samples (Figure 2A).

Table 3.

Transcriptome-wide association analysis results in MSA

GTEx Tissue Gene eQTL ID EA Locus Z-scores p-value PPH4
eQTL GWAS TWAS
Hippocampus USP38-DT rs300925 C 4q31.21 −4.20 3.89 −4.59 4.38 × 10−6 0.84
Caudate lnc-KCTD7 rs6958520 C 7q11.21 −10.32 5.42 −5.42 5.96 × 10−8 0.99
Cortex lnc-KCTD7 rs6958520 C 7q11.21 −10.81 5.42 −5.11 3.26 × 10−7 0.99
Cerebellar hemisphere lnc-KCTD7 rs10215516 A 7q11.21 −9.87 5.45 −5.08 3.70 × 10−7 0.99
Hippocampus lnc-KCTD7 rs10215516 A 7q11.21 −10.07 5.45 −5.05 4.44 × 10−7 0.99
Frontal cortex (BA9) lnc-KCTD7 rs10215516 A 7q11.21 −10.17 5.45 −4.99 6.04 × 10−7 0.99
Hippocampus KCTD7 rs10215516 A 7q11.21 −6.31 5.45 −4.95 7.47 × 10−7 0.99
Amygdala lnc-KCTD7 rs17566701 C 7q11.21 −8.79 5.44 −4.93 8.05 × 10−7 0.99
Anterior cingulate lnc-KCTD7 rs6958520 C 7q11.21 −9.72 5.42 −4.90 9.58 × 10−7 0.99
Putamen lnc-KCTD7 rs6958520 C 7q11.21 −9.37 5.42 −4.89 1.01 × 10−6 0.99
Cervical spinal cord KCTD7 rs6958520 C 7q11.21 −8.24 5.42 −4.87 1.11 × 10−6 0.99
Substantia nigra lnc-KCTD7 rs6958520 C 7q11.21 −7.81 5.42 −4.83 1.38 × 10−6 0.99
Amygdala KCTD7 rs10215516 A 7q11.21 −4.97 5.45 −4.66 3.23 × 10−6 0.98
Nucleus accumbens lnc-KCTD7 rs6958520 C 7q11.21 −10.67 5.42 −4.54 5.69 × 10−6 0.99

Significant eQTLs with coloc PPH4 values ≥ 0.8 and permutation p-values < 0.05 are listed. The eQTL ID refers to the best eQTL in a given locus. Abbreviations: EA, effect allele; GTEx, Genotype-Tissue Expression project (https://gtexportal.org); GWAS, genome-wide association study; PPH4, posterior probability of hypothesis 4; TWAS, transcriptome-wide association study.

Figure 2. Bulk RNA-sequencing and colocalization analyses of the MSA loci.

Figure 2.

(A) The effect of the 4q31.21 locus index variant, rs55894236-C allele, on USP38-DT expression in brain tissues from the GTEx Consortium is shown. Error bars indicate the standard error. (B) The effect of the 7q11.21 locus index variant, rs11766262-C allele, on KCTD7 expression is depicted. (C) Summary of significant colocalization signals (PPH4 > 0.80) for transcripts at the 4q31.21 and 7q11.21 loci across the GTEx brain tissues. (D) Schematic summary of the GWAS, TWAS, colocalization, and SKAT-O results at the four MSA risk loci. The red squares depict a significant analysis result for the listed gene. The numbers in the red squares show the number of tissues that had a significant colocalization PPH4 signal. Abbreviations: GWAS, genome-wide association study; TWAS, transcriptome-wide association study; coloc, colocalization analysis; PPH4, posterior probability of hypothesis four; and SKAT-O, sequence kernel association test – optimized.

At the 7q11.21 locus, our TWAS analysis identified a long non-coding RNA (lnc-KCTD7-2) as being associated with MSA in ten brain regions (Table 3); decreased expression of lnc-KCTD7-2 was predicted to increase disease risk with the most prominent association being observed in the caudate (rs6958520, p-value = 5.96 × 10−8, Z = −5.42). We also found evidence of association for the KCTD7 transcript in three brain regions, including the amygdala, the hippocampus, and the cervical spinal cord. Similar to lnc-KCTD7-2, decreased expression of KCTD7 was predicted to increase MSA risk across these regions (for example, rs10215516 in the amygdala, p-value = 3.23 × 10−6, Z = −4.66, Table 3). The risk allele of the lead GWAS SNP (rs11766262-C) in this locus was similarly associated with decreased expression of KCTD7 in the GTEx bulk brain samples (Figure 2B).

Colocalization analysis nominates genes in the pathogenesis of MSA

Next, we determined which genes within each locus might be driving susceptibility to MSA. To do this, we used cis-expression quantitative trait locus (eQTL) data generated using bulk brain expression data in the GTEx project (version 8). We performed colocalization tests on the SNPs with a p-value < 1.00 × 10−4 within each GWAS locus to estimate the probability that a given variant is associated with both disease risk and gene expression.20 This approach prioritized USP38-DT as the likely causal gene in the 4q31.21 locus, as it had a high posterior probability in the hippocampus (rs300925: PPH4 = 0.84), a brain region known to be affected in MSA (Figure 2C). It also prioritized KCTD7 and lnc-KCTD7-2 as the likely causal genes in the 7q11.21 locus, based on high posterior probability values across multiple brain regions (Figure 2C, Table 3). No genes were prioritized by the colocalization analysis in the 5q34 and 15q23 loci. A summary of the GWAS, TWAS, colocalization, and rare variant analyses is provided for each locus in Figure 2D.

Cell type-specific expression

We also explored the cell type expression of these genes using single-nucleus RNA sequencing (snRNA-Seq) expression data generated from sorted CNS cell types for the ROS/MAP brain autopsy collection (n = 424 healthy subjects).21 In the 4q31.21 locus, the lead GWAS SNP (rs55894236:C) was associated with decreased expression of GAB1 in excitatory neurons (β = −0.26, p-value = 4.7 × 10−28; Figure 3A) and inhibitory neurons (β = −0.079, p-value = 9.1 × 10−3). Cell type-specific data were not available for the USP38-DT long noncoding RNA transcript. However, we noted a transcript within this locus, INPP4B, that is regulated by USP38-DT. INPP4B that has been previously implicated in Alzheimer’s disease.22 We found that rs55894236:C increases the expression of INPP4B in astrocytes (β = 0.28, p-value = 1.0 × 10−7), oligodendrocytes (β = 0.37, p-value = 4.7 × 10−13), and oligodendrocyte precursor cells (β = 0.42, p-value = 5.9 × 10−13), suggesting that it may also play a role in increasing susceptibility for MSA (Figure 3B).

Figure 3. Single-nucleus RNA-sequence analyses of common variants at the 4q31.21 and 7q11.21 loci.

Figure 3.

(A) Single-nucleus RNA-sequence analyses identified cis-eQTLs for rs55894236 for GAB1 in excitatory neurons, inhibitory neurons, and microglia (p-value threshold < 0.05). This SNP was also a cis-eQTL for INPP4B in astrocytes, oligodendrocytes, oligodendrocyte precursor cells, and inhibitory cells (B). Additionally, we identified a cis-eQTL for rs11766262 regulating KCTD7 expression in oligodendroglia, microglia, and excitatory neurons (C). Abbreviations: Ast, astrocytes; End, endocytes; Exc, excitatory neurons; Inh, inhibitory neurons; Mic, microglia; Oli, oligodendroglia; OPC, oligodendroglia precursor cells.

In the 7q11.21 locus, the lead GWAS SNP (rs11766262:C) was associated with markedly decreased expression of KCTD7 in oligodendrocytes (β = −0.64, p-value = 4.7 × 10−29) and, to a lesser extent, in microglia (β = −0.19, p-value = 1.7 × 10−5). In contrast, the same SNP was associated with mildly increased expression of KCTD7 in excitatory neurons (β = 0.1, p-value = 2.3 × 10−3; Figure 3C). Cell-type-specific data were not available for the lnc-KCTD7-2 transcript.

Repeat expansion mapping in MSA identifies rare pathogenic allele carriers

We used the ExpansionHunter Targeted tool (version 5) to map repeat elements in ten genes known to carry pathogenic repeat expansions (AR, ATN1, ATXN1, ATXN2, ATXN3, C9orf72, DMPK, FMR1, FXN, and HTT). This analysis identified 8 (0.9%) MSA cases with pathogenic expansions in the genes ATXN1, ATXN3, HTT, and AR (Table S2). Remarkably, seven of these cases had pathologically confirmed MSA, arguing against mimic syndromes as an alternative cause of their neurological syndrome. The observation of the repeat expansions in our MSA cases may reflect the relatively high prevalence of these alleles in the general population, an observation corroborated by their frequent occurrence among our control cohort (n = 20 controls [0.66%] carried a pathogenic expansion; Table S2).23,24

Previously nominated genetic loci were not associated with MSA

We investigated our GWAS data for common variation in loci previously reported to be associated with MSA, including the COQ2,8 MAPT,13 SNCA,12,25 ZIC1-ZIC4,26 and PLA2G4C loci.27 None of the tested SNPs in these loci surpassed the Bonferroni threshold for multiple testing (Figure S8). The most associated SNP was rs242557, located within the first intron of the MAPT gene on the long arm of chromosome 17, with a p-value of 0.049, which is far from genome-wide or even regional Bonferroni-adjusted significance (Table S3). We, therefore, lack evidence that common genetic variation in these loci plays a major role in MSA risk in the European/Northern American population. We also used the Manta algorithm to examine our whole genome sequence data for evidence of SNCA duplications or triplications; none of the cases (n = 888) carried a specific structural variant of this gene.

Gene-set enrichment analysis identifies a pathway associated with MSA

Pathway enrichment analysis was performed based on the GWAS summary statistics. We discovered a significant pathway under the recessive model, namely 3’-5’ DNA helicase activity, also known as ATP-dependent DNA helicase activity (GO:0003678, number of genes = 15, p-value = 3.05 × 10−5, FDR-adjusted p-value = 0.0495). None of the other pathways achieved significance under the additive or recessive models after correction for multiple testing.

Discussion

Our analyses of whole genome sequence data illustrate the impact of common and rare variants in MSA, a fatal neurodegenerative disease. Specifically, our GWAS identified several novel loci associated with MSA risk, and gene-burden tests implicated KCTD7 in this synucleinopathy. Functional mapping using TWAS and colocalization analyses also revealed changes in the USP38-DT, KCTD7, and lnc-KCTD7-2 transcripts at these risk loci, connecting their expression to disease risk. Our pathway analysis implicated 3’−5’ DNA helicase activity in the MSA pathogenesis, which has not been previously implicated in the disease. Our investigations highlight the value of unbiased data-driven evaluations, which may open new avenues for future exploration. These analytical approaches were chosen as these databases already exist, and, indeed, they complemented each other. Finally, we showed that MSA is characterized, at least in part, by a primary molecular deficit localized within oligodendrocytes, corroborating the converging evidence from preclinical models and post-mortem studies indicating that MSA is a primary oligodendrogliopathy.4,28

We identified a new locus on the long arm of chromosome 4 using an additive GWAS model (Figure 1A). Functional mapping using TWAS and colocalization analysis narrowed the candidate genes in this region to USP38-DT (Figure 1A, Figure 3). USP38-DT is a ubiquitously expressed, validated long non-coding RNA, a transcript class that can upregulate or decrease the expression of genes via cis- or trans-mechanisms.29 Interestingly, this lncRNA regulates the expression of the nearby INPP4B (https://lncipedia.org), an Alzheimer’s disease-related gene involved in lysosomal homeostasis and the autophagic clearance of protein aggregates.30 Additionally, in cellular studies, overexpression of INPP4B was necessary for α-synuclein-mediated endocytosis,31 likely due to its activity in phosphorylating the phosphoinositol membrane lipids for micropinocytosis.32

Our transcriptome-wide analysis of MSA identified a significant association of USP38-DT at the chromosome 4q31.21 locus in the hippocampus rather than the cerebellum, brainstem, and basal ganglia, which are the more prominently affected brain regions in autopsies. Nevertheless, the hippocampus is known to be affected in MSA, reflecting the widespread neuropathology associated with the disease. Indeed, prominent pathological hippocampal changes were observed in a large autopsy series of MSA,33 and cognitive impairment in patients correlated with this hippocampal involvement.34,35 Further work will be needed to examine the phenotype correlation of these transcripts. However, our findings already highlight the power of genomics to uncover novel results when applied in a broad empirical manner.

Besides USP38-DT, the location of the GWAS association signal within the 4q31.21 locus also points to the GAB1 gene as a potential candidate underlying the molecular pathomechanism. The scaffolding protein encoded by GAB1 regulates oligodendrocyte development,36,37 a cell type that is particularly affected in MSA.28 Conditional deletions of Gab1 in a murine model have been shown to impair myelination by affecting oligodendrocyte progenitor cell differentiation.37 Interestingly, patients with MSA show early myelin dysfunction and relocation of myelin proteins,38 making GAB1 a plausible risk gene. GAB1 has also been shown to be involved in Parkinson’s disease,39 and a key paralog of this gene, GAB2, is the principal activator of phosphatidylinositol-3 kinase that has been implicated in Alzheimer's disease.40 Our genomic data prompt us to speculate how disruption of this locus may increase the risk for disease; a preliminary model suggests complex interactions involving multiple genes within the chromosome 4q31.21 locus may lead to MSA.41 Despite this complexity, these observations could have therapeutic implications, as antisense oligonucleotides targeting USP38-DT, INPP4B, or GAB1 in the CNS are worth exploring as a means to slowing symptom progression in patients with MSA.

A second GWAS risk locus, located on chromosome 15q23, was detected under the additive model. This association signal was located within a distal enhancer-like signature (ENCODE accession #: EH38E1774799) and downstream of the RNA gene lnc-LRRC49-3. However, the TWAS analysis found no significant expression changes at this locus (Figure 1). Further work is therefore needed to pinpoint a possible molecular mechanism associated with this risk locus.

Under a recessive GWAS model, we detected a significant MSA risk locus within the TENM2 gene on chromosome 5q34 (Figure 1). TENM2 encodes the teneurin transmembrane protein 2, which is involved in neuronal migration,42 calcium-mediated signaling,43 cell-cell adhesion,44 and retrograde trans-synaptic signaling.45 However, no TWAS signal was identified at this locus, and more work is needed to understand the molecular mechanism by which genetic variation is associated with susceptibility for MSA.

We detected a second risk locus under the recessive model, located on chromosome 7q11.21 within RABGEF1 and downstream of KCTD7 (Figure 1). Our TWAS and colocalization analyses prioritized both KCTD7 and lnc-KCTD7-2 as the genes within this region most likely contributing to the pathogenesis of MSA. Gene burden analysis also found a nominally significant enrichment of missense mutations in KCTD7 in MSA cases (Table 2). Although this association was not significant at the genome-wide level, it does provide additional evidence supporting its role in MSA pathogenesis. Like other neurodegenerative disease loci, such as GBA2, GRN46, and LRRK247, where common and rare variants have been implicated, KCTD7 may be pleomorphic in MSA. It also remains possible that the expression of other transcripts in this locus is affected and contributes to the pathogenesis of MSA.

KCTD7 is a member of the potassium channel tetramerization domain-containing protein family that is highly expressed in the cerebellum and modulates neuron excitability.34 Mutations in this gene have already been linked to a severe neurodegenerative disease called progressive myoclonic epilepsy, type 3 (OMIM: 611726); this syndrome manifests with intractable myoclonic seizures before the age of two, developmental regression, and truncal ataxia, a clinical feature that is also frequently observed among patients with MSA.48

Intriguingly, KCTD7 was recently found to regulate calpains, a group of non-lysosomal cysteine proteases, by inducing ubiquitination.49 Loss of this KCTD7-induced ubiquitination leads to calpain hyperactivation, aberrant cleavage of downstream targets, and caspase-3 activation.49 CRISPR/Cas9-mediated knockout of Kctd7 in mice phenotypically recapitulated human KCTD7 deficiency and resulted in calpain hyperactivation, behavioral impairments, and neurodegeneration; these phenotypes were largely prevented by pharmacological inhibition of calpains.49 Overall, our genomic data implicate a novel molecular mechanism in the pathogenesis of MSA. Therapeutic strategies targeting malfunctions of calpains are also under development50 and, based on our work, may be appropriate for therapeutic development in MSA.

We used a recessive inheritance model to identify the association of KCTD7 in MSA; intriguingly, this locus was not detected under the additive model. The importance of evaluating non-additive inheritance models is increasingly recognized for complex traits,51 such as obesity,52 type 2 diabetes,53 and autoimmune diseases.54 Examining the recessive model may be particularly beneficial for age-related illnesses like MSA, where recessive loci with reduced penetrance may contribute.17 Such traits appear to occur sporadically within the population as they rarely recur within families. Despite this, recessive alleles are easier to map from fewer affected individuals, provided the appropriate model is deployed.55 Overall, our data reinforce the relevance of adopting the recessive model in GWAS studies and highlight the contribution of recessive variants to late-onset neurological diseases.

We found eight MSA patients with pathogenic repeat expansions in disease-related genes, making up less than 1% of the cohort. This rate is similar to that in the general population,23,24,56 suggesting the patients carried the genetic risk variants for two neurological diseases coincidentally. Alternatively, these pathogenic repeat expansions may produce phenotypic syndromes indistinguishable from MSA. There is a growing awareness that mutations in one gene can lead to different neuro-psychiatric syndromes.23 Indeed, the eight patients’ diagnoses were verified by medical record review, and seven had classical MSA features on post-mortem evaluation, ruling out mimic syndromes as an alternative diagnosis. Regardless, screening for these mutations should be considered part of the initial MSA evaluation, especially because new treatments targeting these loci are emerging.57-60

Our study had limitations. First, although our cohort constituted a large genome-sequence dataset generated for MSA, the sample size was relatively small by genomic standards, limiting our power to detect common genetic variants of small effect size. Second, our study only included individuals of European ancestry, as this was the population in which large cohorts of MSA cases and matching control data were readily available; thus, our findings may not be generalizable to the non-European populations.61 Third, the clinical diagnosis of MSA can be challenging, and some of the clinically diagnosed cases could have been mimic syndromes arising from other diseases. To minimize this possibility, we only included patients who fulfilled consensus criteria for clinically probable disease5 and prioritized whole genome sequencing of pathologically confirmed cases.

An additional limitation of our study was the need for a replication cohort. MSA is rare in the general population, making it challenging to collect large numbers of cases. This is an implicit obstacle to identifying the genetic causes of any rare disease. We hope that future studies involving larger cohorts can help us further understand MSA's genetic etiology; indeed, we have made the summary statistics publicly available with that goal in mind. In the meantime, researchers in the rare disease space must rely on orthogonal evidence to confirm the validity of their findings.62 In our example, we discovered that genes linked to MSA were expressed in oligodendroglia, and KCTD7 mutations were already known to cause a juvenile neurodegenerative disorder. We also performed leave-one-out analyses and a sensitivity analysis using only pathologically confirmed cases, demonstrating the robustness of the detected signals.

Conclusion

Our genomic analyses identified four novel risk loci for MSA, a rare and fatal adult-onset neurodegenerative disease. Our discoveries begin to unravel the missing genetic etiology of this understudied member of the synucleinopathy triad. We created a foundational genomic resource that can be systematically investigated to unravel the architecture of MSA. In this way, our study advances the understanding of MSA’s pathogenesis and paves the way for modeling the disease and developing targeted treatments.

STAR Methods

Resource availability

Lead Contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Sonja W. Scholz (sonja.scholz@nih.gov).

Materials Availability

The study did not generate any new unique reagents.

Data and Code Availability

The summary statistics from the additive and recessive GWAS models have been deposited in the GWAS catalog (https://www.ebi.ac.uk/gwas/). The individual-level sequence data for a subset of the MSA genomes (n = 683 cases) reported in this paper will be available upon publication in dbGaP (accession number: phs001963). Public data sharing was not feasible for the remaining 205 MSA genomes; access to these data will be granted to qualified researchers via appropriate collaboration agreements. The TOPMed control genome data are available in dbGaP (accession numbers: phs001662.v2.p1, phs00974.v5.p4, phs000951.v5.p5). The control genome data from 1,980 subjects from the DementiaSeq project are available in dbGaP (accession number: phs001963), and the remaining control genomes are available upon request from the Wellderly study team (contact: atorkama@scripps.edu). The programming code used in this paper is available at https://zenodo.org/records/10723069.

Experimental Model and Subject Details

Study cohorts

The study workflow is depicted in Figure S1. The cohort included 3,978 participants of European ancestry (958 MSA cases and 3,021 neurologically healthy controls). MSA cases were recruited across twenty North American and European sites and consortia to create a genomic resource for MSA research (see Table S4 for a list of the participating sites). MSA cases were diagnosed with clinically probable (n = 416 [47%]) or pathologically definite disease (n = 468 [53%]) according to the Gilman consensus criteria.5 The control subjects were obtained from the DementiaSeq project (dbGaP accession number: phs001963) and selected based on a lack of evidence of cognitive decline in their clinical history and no neurological deficits on neurological examination.2 The pathologically confirmed control individuals had no evidence of significant neurodegenerative disease on histopathological examination. We additionally obtained whole genome sequence data from 5,963 European-ancestry convenience controls from the TOPMed consortium (dbGaP accession numbers: phs001662.v2.p1, phs00974.v5.p4, phs000951.v5.p5). The demographic characteristics of the cohorts are summarized in Table 4. The appropriate institutional review boards of participating institutions approved the study, and informed consent was obtained from all subjects or their surrogate decision-makers according to the Declaration of Helsinki.

Table 4.

Characteristics of study participants

Variable Cases Controls
Neurologically Healthy
Controls
TOPMed Controls
Number 888 3,018 4,110
Female Sex, n (%) 416 (47%) 1,612 (53%) 2,129 (52%)
Diagnostic Category
    Clinically Probable MSA, n (%) 420 (47%) - -
    Definite MSA, n (%) 468 (53%) - -
Clinical Subtype
    MSA-P, n (%) 202 (23%) - -
    MSA-C, n (%) 127 (14%) - -
    Not available, n (%) 559 (63%) - -
Mean Age (yrs, range) 64 (38–91)* 77 (16–110) 62 (20–93)
*

Age information was not available for 116 MSA cases.

Data generation and preprocessing

Whole genome sequencing:

PCR-free libraries from genomic DNA samples were constructed using the Illumina TruSeq chemistry, according to the manufacturer's protocol. Whole genome sequencing was performed on an Illumina NovaSeq sequencer using 150 base pair, paired-end cycles (version 2.5 chemistry, Illumina). The control subjects were previously sequenced on an Illumina HiSeq X Ten using the same parameters, as described elsewhere.2 The mean sequencing coverage of the samples was 35.85 (range, 18.34–70.75, see Figure S9).

Sequence alignment and variant calling:

The whole genome sequence data were aligned to reference genome build hg38 and processed on the Google Cloud Platform, according to GATK (2016) Best Practices.63 Variants were called by a combination of the publicly available GATK Best Practices and another workflow for joint discovery and Variant Quality Score Recalibration by the Broad Institute (https://github.com/gatk-workflows/broad-prod-wgs-germline-snps-indels). All genome sequence data were processed using a uniform pipeline. The convenience control genomes obtained from the TOPMed consortium were called separately and merged with the study data for quality control checks (Figure S1).

Quality control:

A workflow diagram of the quality control steps is shown in Figure S1. The cohort consisted of a discovery dataset (n = 958 MSA cases and 3,021 controls) and a convenience control dataset from TOPMed (n = 5,963 controls). For sample-level quality control of the discovery dataset, we removed genomes based on the following criteria: (1) failed library preparations or sequencing, (2) abnormal heterozygosity (F-statistic outside of the −0.15 to 0.15 range), (3) low call rate (≤ 95%), (4) sex check failure (i.e., a discrepancy between reported sex and genotypic sex), (5) non-European ancestry (based on principal component analysis when compared to Hapmap3 data; Figure S3), (6) duplicate samples (pi-hat statistic > 0.8), (7) related samples (pi-hat statistic > 0.125), and (8) cases in whom the final diagnosis was changed. For variant-level quality control of the discovery dataset, we excluded variants based on the following criteria: (1) spanning deletions, (2) minor allele frequencies (MAFs) significantly different in controls from reported frequencies in the NHLBI TransOmics TOPMed database (freeze 5b; www.nhlbiwgs.org) or gnomAD (version 3.1.2; https://gnomad.broadinstitute.org), (3) a significant departure from Hardy-Weinberg equilibrium in the control cohort (p-value ≤ 1 × 10−6), (4) non-autosomal variants (X, Y, mitochondrial DNA), (5) non-random missingness between cases and controls (excluding variants with p-value < 1 × 10−4), (6) haplotype-based non-random missingness (excluding variants with p-value ≤ 1 × 10−4), (7) variants with a high missingness rate (i.e., ≥ 5%), (8) variants mapping to variable, diversity, and joining (VDJ) recombination sites, and variants in the centromeric regions ± 10 kb (due to poor sequence alignment and incomplete resolution of the reference genome assembled at these sites), (9) variants failing gnomAD filters (version 3.1.2; https://gnomad.broadinstitute.org), and (10) variants with poor sequence alignment. A total of 888 MSA cases and 3,018 controls from the discovery dataset were included in downstream analyses. Of note, for rare variant analyses only this jointly called discovery dataset was used. Following the quality control steps of the discovery dataset, we merged the data with the convenience control genomes from TOPMed and applied the same sample- and variant-level quality control steps. Additionally, we excluded variants that had significantly different minor allele frequencies between the discovery control genomes and the TOPMed control genomes (excluding variants with per chromosome FDR-corrected p-value ≤ 0.05). The final dataset included 91,594,360 variants in 888 cases and 7,128 controls, which were used for downstream common variant analyses (Figure S1).

Quantification and Statistical Analysis

Genome-wide association analyses (GWAS)

The GWAS was performed in 888 MSA cases and 7,128 controls using the PLINK toolset (version 2.0).64 We applied additive and recessive logistic regression models using an MAF threshold of > 1% (based on allele frequency estimates in the MSA cases). We determined the relevant genetic principal components in FlashPCA (version 2.0) and applied the step function in R Mass package (R version 3.5.2; https://www.R-project.org/) to calculate the number of principal components required for population substructure correction.65 Based on this analysis, we included sex and seven principal components as covariates in our GWAS study. The Bonferroni threshold for genome-wide significance was 5 × 10−8, and variants achieving a p-value less than or equal to 5.0 × 10−7 were considered subsignificant. The effect allele was defined as the allele associated with an increased risk of disease (i.e., odds ratio > 1.0).66 All of the genes located within a 1 Mb upstream and downstream of each gene were included in transcriptomic analyses to ensure the detection of ancillary signals.

For conditional analyses on GWAS loci identified in the additive and recessive models, we additionally included the respective index variants in the covariates (Figure S4). To demonstrate the robustness of GWAS signals, we performed leave-one-out analyses by withholding samples based on their institutional source; there were twenty-four cohorts from the twenty different institutions at which samples were collected, meaning that twenty-four separate GWAS analyses were performed (Figure S7). A sensitivity analysis was performed in a subset of pathologically confirmed MSA cases (n = 468) and healthy controls (n = 7,128) under both additive and recessive models, with sex and seven principal components included as covariates.

Transcriptome-wide association study (TWAS)

Tissue-specific expression was predicted based on the GWAS summary statistics from the additive and the recessive models by transcriptome-wide association analyses (TWAS). To do so, we obtained gene expression data from the Genotype-Tissue Expression project (GTEx, version 8; https://gtexportal.org). To explore a gene’s association with disease, a transcriptome-wide imputation was achieved using the FUSION pipeline,67 where the precomputed gene expression weights obtained from the GTEx data for thirteen brain regions were considered. These regions included: (i) the amygdala, (ii) anterior cingulate cortex (BA24), (iii) caudate, (iv) cerebellar hemisphere, (v) cerebellum, (vi) cervical spinal cord (C-1), (vii) cortex, (viii) frontal cortex (BA9), (ix) hippocampus, (x) hypothalamus, (xi) nucleus accumbens, (xii) putamen, and (xiii) substantia nigra. The significant association threshold was defined as 0.05 divided by the number of genes in GTEx (version 8) in the thirteen types of brain regions. This threshold ranged from 2.17 × 10−5 in the substantia nigra to 6.77 × 10−6 in the cerebellum due to the variable number of genes expressed in each tissue. Variants achieving a p-value ten-fold higher than the significance threshold were considered subsignificant.

Colocalization analyses and gene prioritization

We used the COLOC function within the FUSION package67 to test the hypothesis that an MSA risk variant colocalized with an eQTL variant in bulk RNA-seq data obtained from the GTEx project (version 8). For the four genome-wide significant loci in the GWAS (4q31.21, 5q34, 7q11.21, and 15q23), we extracted all SNPs with a p-value < 1×10−4 for colocalization analysis to evaluate the probability of the MSA loci and eQTL sharing a single causal variant for each region. In each eQTL dataset, we extracted the associations for the SNP-gene pairs within that range and tested for colocalization.20 A colocalization posterior prior probability hypothesis 4 (PPH4) ≥ 80% and a permutation p-value < 0.05 was considered evidence for an eQTL-GWAS association that could substantially influence both the expression and the GWAS trait in that region for disease.

Cell type-specific expression analysis

We evaluated the expression of SNPs and nominated risk genes identified through GWAS and TWAS in a single-nucleus RNA-seq dataset generated using the Religious Orders Study/Memory and Aging Project (ROS/MAP) cohort.21 The ROS/MAP data were derived from 424 dorsolateral prefrontal cortexes of individuals of advanced age using the 10x Genomics Single Cell 3’ kit, as described elsewhere.21 Sequencing reads were processed, and the unique molecule identifier count matrix was generated using Cell Ranger software (version 6.0.0, 10x Genomics). The cell types were classified by clustering cells by gene expression using the R package Seurat (version 4).68 The “pseudobulk” gene expression matrix was constructed by aggregating unique molecule identifier counts of the same cell type of the same donor and normalizing them to the log2 counts-per-million-reads-mapped values. Sample genotyping was performed by whole genome sequencing followed by GATK processing. The cis-eQTLs were mapped using Matrix-eQTL (version 2.3) for single nucleotide polymorphisms within 1 megabase of the transcription start sites.69

Gene-based, rare variant association analyses

A gene-based SKAT-O analyses of missense and loss-of-function mutations were conducted to determine the difference in the aggregate burden of rare coding variants in the MSA cases (n = 888) versus healthy controls (n = 3,018). All variants were annotated in Variant Effect Predictor (VEP; version 101),70 with the ‘LoFtee’ plugin to annotate high-confidence loss-of-function variants using the default parameters. The variants were filtered using an MAF threshold of ≤ 1% and an MAC of ≥ 2. We then performed a SKAT-O analysis of filtered and annotated variants in RVTESTS (version 2.1.0)61, including sex and five principal components as covariates. We used a genome-wide significance threshold of 3.03 × 10−6 (= 0.05/16,507 genes). From the genome-wide data, we extracted the values for eight genes (COQ2, GBA1, MAPT, SNCA, GAB1, RABGEF1, KCTD7, and TENM2) implicated in MSA and used a gene-wide significance threshold of 0.006 (= 0.05/8) to test for significant enrichment of coding mutations.

Repeat expansion analysis in short-read genomes from MSA cases and controls

As repeat expansion diseases can occasionally mimic the clinical features of MSA, we assessed the frequency of pathogenic repeat expansions in our MSA case-control whole genome sequence data (n = 888 cases and 3,018 controls) using the ExpansionHunter Targeted tool (version 5; Illumina).71 This tool has been validated for measuring repeat expansions in ten known disease genes (AR, ATN1, ATXN1, ATXN2, ATXN3, C9orf72, DMPK, FMR1, FXN, and HTT).23 Pathogenic repeat expansions were validated manually by visualization in the Repeat Expansion Viewer (REViewer; Illumina).72,73

Structural variant evaluation in the MSA cohort

We used the Manta algorithm to detect the structural variants (i.e., duplications) within the SNCA locus on chromosome 4q22.1 in the 888 MSA cases and the 3,018 neurologically healthy subjects.74 This analysis used default settings and focused on the region defined by the SNCA gene [chr4:89,724,099-89,837,161]. The result files were merged with bcftools,75 and missing genotypes were set to reference homozygotes.

Candidate gene analyses

GWAS data were analyzed for evidence of association in six genes previously reported to be associated with MSA, including COQ2, MAPT, SNCA, ZIC1, ZIC4, and PLA2G4C.8,12,13,25-27 The analysis was performed by subsetting gene regions from post-quality control variant files and testing for association using a generalized logistic regression model as described above.

Pathway analyses

Gene-set enrichment analyses were performed in MAGMA (version 1.10). The primary analysis used the binary PLINK files to annotate all the variants to genes if they were within the genic boundaries of 1.5 kb upstream and downstream. This was followed by a gene analysis, where the summary statistics from both the additive and recessive GWAS models were used to generate different gene-level metrics. Using the ‘mean’ test statistics (snp-wise = mean) across all the genes, pathway enrichment evaluation was performed using 13,159 gene sets from MSigDB (https://www.gsea-msigdb.org/gsea/msigdb; version 7.5.1) to identify potential pathways associated with MSA.76

Supplementary Material

1

Key Resources Table

REAGENT/RESOURCE SOURCE IDENTIFIER
Biological Samples
Human cerebellar brain tissue and/or whole blood Comprehensive list of study sites where samples were collected is listed in the Table S1 of this paper N/A
Critical Commercial Assays
Maxwell RSC Tissue DNA Kit Promega Catalog # AS1610
PicoGreen dsDNA assay Thermo Fisher Catalog # P7589
TruSeq PCR-free Library Prep Kit Illumina Catalog # 20015963
HiSeq X Ten Reagent Kit (v.2.5 chemistry) Illumina Catalog # FC-502-2501
Deposited Data
Human reference genome NCBI build 38, GRCh38 Genome Reference Consortium https://www.ncbi.nlm/nih.gov/grc/human
RRID:SCR_006553
Individual-level whole genome sequence data from neurologically healthy, aged controls DementiaSeq dbGAP
(www.ncbi.nlm.nih.gov/gap/)
Accession # phs001963
RRID: SCR_002709
Individual-level whole genome sequence data from neurologically healthy, aged controls Wellderly cohort Available upon request Contact: Dr. Ali Torkamani (atorkama@scripps.edu)
TOPMed control genome data TOPMed consortium Available on dbGaP
(www.ncbi.nlm.nih.gov/gap/)
Accession # phs001662.v2.p1, phs00974.v5.p4, phs000951.v5.p5)
RRID: SCR_002709
Gene expression data GTEx (v.8) https://gtexportal.org/home/ RRID: SCR_013042
Individual-level, whole genome sequencing data from MSA cases This paper dbGAP
(www.ncbi.nlm.nih.gov/gap/)
Accession #: phs001963
RRID: SCR_002709
MSA GWAS summary statistics This paper GWAS catalog: www.ebi.ac.uk/gwas/
Software and Algorithms
GATK Broad Institute https://gatk.broadinstitute.org/
RRID:SCR_001876
Pipeline Standardization CCDG https://github.com/CCDG/Pipeline-Standardization/blob/master/PipelineStandard.md
prod-wgs-germline-snps-indels Broad Institute https://github.com/gatk-workflows/broad-prod-wgs-germline-snps-indels
PLINK (v.2.0) Chang C.C. et al. (2015) https://www.cog-genomics.org/plink/2.0/
RRID:SCR_001757
FlashPCA (v.2.0) Abraham G. et al. (2017) https://github.com/gabraham/flashpca
RRID:SCR_021680
R (v.3.5.2) R core team https://www.r-project.org/
RRID: SCR_001905
SAMtools Li and Durbin (2009) https://samtools.sourceforge.net/
RRID:SCR_002105
BCFtools (v.1.16.1) Danecek P. et al. (2021) https://samtools.github.io/bcftools/bcftools.html
RRID:SCR_005227
FUSION Gusev A. et al. (2016) https://github.com/gusevlab/fusion_twas
Cell Ranger (v.6.0.0) 10X Genomics https://support.10xgenomics.com/single-cell-gene-expression/software/overview/welcome
RRID:SCR_017344
Seurat (v.4.3.0) Stuart T. et al. (2019) https://github.com/satijalab/seurat
RRID:SCR_016341
Matrix eQTL Shabalin, A.A. et al. (2012) http://www.bios.unc.edu/research/genomic_software/Matrix_eQTL/
Ensembl Variant Effect Predictor (VEP, v.101) Ensembl https://useast.ensembl.org/info/docs/tools/vep/index.html
RRID:SCR_007931
Loss-Of-Function Transcript Effect Estimator (LOFTEE) Karczewski K.J. et al. (2020) https://github.com/konradjk/loftee
RVTests (v.2.1.0) Zhan X. et al. (2016) https://github.com/zhanxw/rvtests
RRID:SCR_007639
MAGMA (v.1.10) de Leeuw C. et al. (2015) https://ctg.cncr.nl/software/magma
Gene set pathway analysis Molecular Signatures Database (MSigDB), v7.5.1 https://www.gsea-msigdb.org/gsea/msigdb/human/genesets.jsp
RRID:SCR_016863
ExpansionHunter Targeted (v.5) Dolzhenko E. et al. (2017) https://github.com/Illumina/ExpansionHunter
Repeat Expansion Viewer (REViewer, v.0.2.7) Dolzhenko E. et al. (2021) https://github.com/Illumina/REViewer
MSA genomic analyses code This paper https://zenodo.org/records/10723069

Highlights.

  • Generation of a foundational genomic resource in multiple system atrophy

  • GWAS identifies novel risk loci at GAB1, lnc-LRRC49-3, TENM2, and RABGEF1

  • Functional genomics implicates USP38-DT, KCTD7, and lnc-KCTD7-2 within these loci

  • Gene burden analysis identifies nominal enrichment of rare missense mutations in KCTD7

Acknowledgments

We thank the contributors who collected the samples and data used in this study and the patients and their families whose help and participation made this work possible. We thank the members of the Laboratory of Neurogenetics and the Neurodegenerative Diseases Research Unit (NIH) for their collegial support and technical assistance. This study used DNA samples and clinical data from the NINDS Repository at Coriell (www.coriell.org). We are grateful to the NIH NeuroBioBank for the provision of tissue samples. The ROS/MAP study was supported by the National Institute on Aging (RF1 AG057474, U01 AG061356). The study used tissue samples and data from the Johns Hopkins Morris K. Udall Center of Excellence for Parkinson’s Disease Research (NIH P50 NS38377). We thank the Banner Sun Health Research Institute Brain and Body Donation Program of Sun City, Arizona, for providing human biological materials. The Brain and Body Donation Program has been supported by the National Institute of Neurological Disorders and Stroke (U24 NS072026 National Brain and Tissue Resource for Parkinson’s Disease and Related Disorders), the National Institute on Aging (P30 AG19610 and P30 AG072980, Arizona Alzheimer’s Disease Center), the Arizona Department of Health Services (contract 211002, Arizona Alzheimer’s Center), the Arizona Biomedical Research Commission (contracts 4001, 0011, 05-901 and 1001 to the Arizona Parkinson’s Disease Consortium) and the Michael J. Fox Foundation for Parkinson’s Research. The Columbia Parkinsonism Brain Bank is funded by the Parkinson’s Foundation. The Genotype-Tissue Expression (GTEx) Project was supported by the Common Fund of the Office of the Director of the National Institutes of Health, and by NCI, NHGRI, NHLBI, NIA, NIMH, and NINDS. Biospecimens used in this article were obtained from the Northwestern Movement Disorders Center (MDC) Biorepository. As such, the investigators within MDC Biorepository contributed to the design and implementation of the MDC Biorepository and/or provided data and collected biospecimens but did not participate in the analysis or writing of this report. MDS Biorepository investigators include Rizwan Akhtar, MD, PhD, Tanya Simuni, MD; Dimitri Krainc, MD, PhD; Puneet Opal, MD, PhD; Steven Lubbe, PhD; Niccolo Mencacci, MD, PhD; Joanna Blackburn, MD; and Lisa Kinsley, MS, CGC. For up-to-date information on the study, visit http://www.parkinsons.northwestern.edu/research/clinical-trials/mdc-biorepository.html. This work was supported by the NUgene Project at Northwestern University. We thank the contributors who collected the samples used in this study and the patients whose help and participation made NUgene and this work possible. Several authors of this publication are members of the European Reference Network for Rare Neurological Diseases – Project ID No 739510. We acknowledge the Oxford Brain Bank, supported by Brains for Dementia Research (BDR) (Alzheimer Society and Alzheimer Research UK) and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC). V.K and A.N. acknowledge funding from the MSA Coalition, Barbara Bloom Ranson Fund for MSA Research at Brigham and Women’s Hospital, Brigham Research Institute’s Director’s Transformative Award and NIH 1R01NS109209-01A1. Molecular data for the Trans-Omics in Precision Medicine (TOPMed) Program was supported by the National Heart, Lung and Blood Institute (NHLB). Genome sequencing for “NHLBI TOPMed: Lung Tissue Research Consortium (LTRC)” was performed at Broad Genomics and Northwest Genomics Center. Core support including centralized genomic read mapping and genotype calling, along with variant quality metrics and filtering were provided by the TOPMed Informatics Research Center (3R01HL-117626-02S1; contract HHSN268201800002I). Core support, including phenotype harmonization, data management, sample identity QC, and general program coordination were provided by the TOPMed Data Coordinating Center (R01HL-120393; U01HL-120393; contract HHSN268201800001I). We gratefully acknowledge the studies and participants who provided biological samples and data for TOPMed. This study utilized data provided by the Lung Tissue Research Consortium (LTRC) supported by the National Heart, Lung, and Blood Institute (NHLBI). This research was supported in part by the Intramural Research Program of the National Institutes of Health (the National Institute on Aging and the National Institute of Neurological Disorders and Stroke; project numbers: 1ZIAAG000935 and 1ZIANS003154). This study used the computational resources of the NIH HPC Biowulf cluster (http://hpc.nih.gov).

Footnotes

Publisher's Disclaimer: This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of Interests

T.G.B. is a consultant for Aprinoia Therapeutics, Vivid Genomics, and Avid Radiopharmaceutical, and is a scientific advisory board member for Vivid Genomics. J.A.H., H.R.M., S.P.-B., and B.J.T. and H.R.M. hold US, EU, and Canadian patents on the clinical testing and therapeutic intervention for the hexanucleotide repeat expansion of C9orf72. B.J.T. and S.W.S receive research support from Cerevel Therapeutics. B.J.T. is an editorial and advisory board member for Brain, eClinicalMedicine, Journal of Neurology, Neurosurgery, and Psychiatry, and Neurobiology of Aging. H.R.M. reports paid consultancy from Biogen, Biohaven, Lundbeck, UCB, and Denali, as well as lecture fees and honoraria from the Wellcome Trust and the Movement Disorders Society. H.R.M. received research grants from Parkinson’s UK, Cure Parkinson’s Trust, PSP Association, CBD Solutions, Drake Foundation, and the Medical Research Council. H.K. is Editor-in-Chief of Clinical Autonomic Research, serves as PI of a clinical trial sponsored by Biogen MA Inc. (TRACK MSA, S19-01846), received consultancy fees from Lilly USA LLC, Biohaven Pharmaceuticals Inc, Takeda Pharmaceutical Company Ltd, Ono Pharma UK Ltd, Lundbeck LLC, and Theravance Biopharma US Inc. A.F. reports royalties from Springer Verlag, speaker fees and honoraria from Therevance Biopharma, GE Health Care, Broadview Ventures, Austrian Autonomic Society, Stopp-HSP, Elsevier, and research grants from the FWF-Austrian Science Fund, Medical University of Innsbruck, US MSA Coalition, Dr. Johannes and Hertha Tuba Foundation and Austrian Exchange Program, outside of the present work. J.A.P. is an editorial board member of Movement Disorders, Parkinsonism & Related Disorders, BMC Neurology, and Clinical Autonomic Research. I.B. received consultancy fees from Theravance Biopharma US Inc., Amenal Pharmaceutics, Regeneron Pharmaceuticals, Takeda Pharmaceutics, and Neurawell Therapeutics. S.W.S. serves on the scientific advisory board of the Lewy Body Dementia Association and the Multiple System Atrophy Coalition. S.W.S. is an editorial board member for the Journal of Parkinson’s Disease and JAMA Neurology. A.P. serves on the board of directors for CurePSP, has received research grants from the National Institutes of Health and the Michael J. Fox Foundation, and has received consultancy fees from AbbVie Inc., Biogen Inc., SciNeuro Pharmaceuticals, Ono Pharma, and Ferrer Internacional, S.A. A.T. serves on the scientific advisory board for Vivid Genomics. R.H.R. is currently employed by CoSyne Therapeutics; all work performed for this publication was performed on her own time and not as a part of her duties as an employee. ZKW is partially supported by the NIH/NIA and NIH/NINDS (1U19AG063911, FAIN: U19AG063911), Mayo Clinic Center for Regenerative Medicine, the gifts from the Donald G. and Jodi P. Heeringa Family, the Haworth Family Professorship in Neurodegenerative Diseases fund, and The Albertson Parkinson's Research Foundation. He serves as PI or Co-PI on Biohaven Pharmaceuticals, Inc. (BHV4157-206) and Vigil Neuroscience, Inc. (VGL101-01.002, VGL101-01.201, PET tracer development protocol, Csf1r biomarker and repository project, and ultra-high field MRI in the diagnosis and management of CSF1R-related adult-onset leukoencephalopathy with axonal spheroids and pigmented glia) projects/grants. He serves as Co-PI of the Mayo Clinic APDA Center for Advanced Research and as an external advisory board member for Vigil Neuroscience, Inc., and as a consultant on neurodegenerative medical research for Eli Lilli & Company. F.K. received personal fees from Institut de Recherches Internationales Servier, Takeda Pharmaceuticals, Sanofi, Teva, Vial, and the Austrian Society of Neurology in the past 12 months, and he has ongoing grant support from the Austrian Science Fund (FWF) and the National Institutes of Health outside of the submitted work. W.G. M. has received fees for editorial activities with Elsevier and has served as an advisor for Lundbeck, Biohaven, Roche, Alterity, Servier, Inhibikase, Takeda, and Teva. All other authors report no competing interests.

REFERENCES

  • 1.McCann H, Stevens CH, Cartwright H, and Halliday GM (2014). alpha-Synucleinopathy phenotypes. Parkinsonism Relat Disord 20 Suppl 1, S62–67. 10.1016/S1353-8020(13)70017-8. [DOI] [PubMed] [Google Scholar]
  • 2.Chia R, Sabir MS, Bandres-Ciga S, Saez-Atienzar S, Reynolds RH, Gustavsson E, Walton RL, Ahmed S, Viollet C, Ding J, et al. (2021). Genome sequencing analysis identifies new loci associated with Lewy body dementia and provides insights into its genetic architecture. Nat Genet 53, 294–303. 10.1038/s41588-021-00785-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S, Chang D, Tan M, Kia DA, Noyce AJ, Xue A, et al. (2019). Identification of novel risk loci, causal insights, and heritable risk for Parkinson's disease: a meta-analysis of genome-wide association studies. Lancet Neurol 18, 1091–1102. 10.1016/S1474-4422(19)30320-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Poewe W, Stankovic I, Halliday G, Meissner WG, Wenning GK, Pellecchia MT, Seppi K, Palma JA, and Kaufmann H (2022). Multiple system atrophy. Nat Rev Dis Primers 8, 56. 10.1038/s41572-022-00382-6. [DOI] [PubMed] [Google Scholar]
  • 5.Gilman S, Wenning GK, Low PA, Brooks DJ, Mathias CJ, Trojanowski JQ, Wood NW, Colosimo C, Durr A, Fowler CJ, et al. (2008). Second consensus statement on the diagnosis of multiple system atrophy. Neurology 71, 670–676. 10.1212/01.wnl.0000324625.00404.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Fanciulli A, and Wenning GK (2015). Multiple-system atrophy. N Engl J Med 372, 1375–1376. 10.1056/NEJMc1501657. [DOI] [PubMed] [Google Scholar]
  • 7.Watts JC, Giles K, Oehler A, Middleton L, Dexter DT, Gentleman SM, DeArmond SJ, and Prusiner SB (2013). Transmission of multiple system atrophy prions to transgenic mice. Proc Natl Acad Sci U S A 110, 19555–19560. 10.1073/pnas.1318268110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Multiple-System Atrophy Research, C. (2013). Mutations in COQ2 in familial and sporadic multiple-system atrophy. N Engl J Med 369, 233–244. 10.1056/NEJMoa1212115. [DOI] [PubMed] [Google Scholar]
  • 9.Ronchi D, Di Biase E, Franco G, Melzi V, Del Sorbo F, Elia A, Barzaghi C, Garavaglia B, Bergamini C, Fato R, et al. (2016). Mutational analysis of COQ2 in patients with MSA in Italy. Neurobiol Aging 45, 213 e211–213 e212. 10.1016/j.neurobiolaging.2016.05.022. [DOI] [PubMed] [Google Scholar]
  • 10.Ross OA, Vilarino-Guell C, Wszolek ZK, Farrer MJ, and Dickson DW (2010). Reply to: SNCA variants are associated with increased risk of multiple system atrophy. Ann Neurol 67, 414–415. 10.1002/ana.21786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Sailer A, Scholz SW, Nalls MA, Schulte C, Federoff M, Price TR, Lees A, Ross OA, Dickson DW, Mok K, et al. (2016). A genome-wide association study in multiple system atrophy. Neurology 87, 1591–1598. 10.1212/WNL.0000000000003221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Scholz SW, Houlden H, Schulte C, Sharma M, Li A, Berg D, Melchers A, Paudel R, Gibbs JR, Simon-Sanchez J, et al. (2009). SNCA variants are associated with increased risk for multiple system atrophy. Ann Neurol 65, 610–614. 10.1002/ana.21685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Vilarino-Guell C, Soto-Ortolaza AI, Rajput A, Mash DC, Papapetropoulos S, Pahwa R, Lyons KE, Uitti RJ, Wszolek ZK, Dickson DW, et al. (2011). MAPT H1 haplotype is a risk factor for essential tremor and multiple system atrophy. Neurology 76, 670–672. 10.1212/WNL.0b013e31820c30c1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wernick AI, Walton RL, Koga S, Soto-Beasley AI, Heckman MG, Gan-Or Z, Ren Y, Rademakers R, Uitti RJ, Wszolek ZK, et al. (2020). GBA variation and susceptibility to multiple system atrophy. Parkinsonism Relat Disord 77, 64–69. 10.1016/j.parkreldis.2020.06.007. [DOI] [PubMed] [Google Scholar]
  • 15.Yun JY, Lee WW, Lee JY, Kim HJ, Park SS, and Jeon BS (2010). SNCA variants and multiple system atrophy. Ann Neurol 67, 554–555. 10.1002/ana.21889. [DOI] [PubMed] [Google Scholar]
  • 16.Zhao Q, Yang X, Tian S, An R, Zheng J, and Xu Y (2016). Association of the COQ2 V393A variant with risk of multiple system atrophy in East Asians: a case-control study and meta-analysis of the literature. Neurol Sci 37, 423–430. 10.1007/s10072-015-2414-8. [DOI] [PubMed] [Google Scholar]
  • 17.Hara K, Momose Y, Tokiguchi S, Shimohata M, Terajima K, Onodera O, Kakita A, Yamada M, Takahashi H, Hirasawa M, et al. (2007). Multiplex families with multiple system atrophy. Arch Neurol 64, 545–551. 10.1001/archneur.64.4.545. [DOI] [PubMed] [Google Scholar]
  • 18.Lee S, Emond MJ, Bamshad MJ, Barnes KC, Rieder MJ, Nickerson DA, Team, N.G.E.S.P.-E.L.P., Christiani DC, Wurfel MM, and Lin X (2012). Optimal unified approach for rare-variant association testing with application to small-sample case-control whole-exome sequencing studies. Am J Hum Genet 91, 224–237. 10.1016/j.ajhg.2012.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Campese N, Fanciulli A, Stefanova N, Haybaeck J, Kiechl S, and Wenning GK (2021). Neuropathology of multiple system atrophy: Kurt Jellinger;s legacy. J Neural Transm (Vienna) 128, 1481–1494. 10.1007/s00702-021-02383-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Farrell K, Humphrey J, Chang T, Zhao Y, Leung YY, Kuksa PP, Patil V, Lee W-P, Kuzma AB, Valladares O, et al. (2023). Genetic, transcriptomic, histological, and biochemical analysis of progressive supranuclear palsy implicates glial activation and novel risk genes. BioRxiv. https://doi.org/ 10.1101/2023.11.09.565552. [DOI] [Google Scholar]
  • 21.Fujita M, Gao Z, Zeng L, McCabe C, White CC, Ng B, Green GS, Rozenblatt-Rosen O, Phillips D, Amir-Zilberstein L, et al. (2024). Cellsubtype specific effects of genetic variation in the Alzheimer’s disease brain. Nat. Genet 56, 605–614. 10.1038/s41588-024-01685-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hadar A, Milanesi E, Squassina A, Niola P, Chillotti C, Pasmanik-Chor M, Yaron O, Martasek P, Rehavi M, Weissglas-Volkov D, et al. (2016). RGS2 expression predicts amyloid-beta sensitivity, MCI and Alzheimer's disease: genome-wide transcriptomic profiling and bioinformatics data mining. Transl Psychiatry 6, e909. 10.1038/tp.2016.179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Dewan R, Chia R, Ding J, Hickman RA, Stein TD, Abramzon Y, Ahmed S, Sabir MS, Portley MK, Tucci A, et al. (2021). Pathogenic Huntingtin Repeat Expansions in Patients with Frontotemporal Dementia and Amyotrophic Lateral Sclerosis. Neuron 109, 448–460 e444. 10.1016/j.neuron.2020.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zanovello M, Ibanez K, Brown AL, Sivakumar P, Bombaci A, Santos L, van Vugt J, Narzisi G, Karra R, Scholz SW, et al. (2023). Unexpected frequency of the pathogenic AR CAG repeat expansion in the general population. Brain 146, 2723–2729. 10.1093/brain/awad050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Al-Chalabi A, Durr A, Wood NW, Parkinson MH, Camuzat A, Hulot JS, Morrison KE, Renton A, Sussmuth SD, Landwehrmeyer BG, et al. (2009). Genetic variants of the alpha-synuclein gene SNCA are associated with multiple system atrophy. PLoS One 4, e7114. 10.1371/journal.pone.0007114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hopfner F, Tietz AK, Ruf VC, Ross OA, Koga S, Dickson D, Aguzzi A, Attems J, Beach T, Beller A, et al. (2022). Common Variants Near ZIC1 and ZIC4 in Autopsy-Confirmed Multiple System Atrophy. Mov Disord 37, 2110–2121. 10.1002/mds.29164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Nakahara Y, Mitsui J, Date H, Porto KJ, Hayashi Y, Yamashita A, Kusakabe Y, Matsukawa T, Ishiura H, Yasuda T, et al. (2023). Genome-wide association study identifies a new susceptibility locus in PLA2G4C for Multiple System Atrophy. medRxiv. 10.1101/2023.05.02.23289328. [DOI] [Google Scholar]
  • 28.Wenning GK, Stefanova N, Jellinger KA, Poewe W, and Schlossmacher MG (2008). Multiple system atrophy: a primary oligodendrogliopathy. Ann Neurol 64, 239–246. 10.1002/ana.21465. [DOI] [PubMed] [Google Scholar]
  • 29.Statello L, Guo CJ, Chen LL, and Huarte M (2021). Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol 22, 96–118. 10.1038/s41580-020-00315-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Rodgers SJ, Jones EI, Arumugam S, Hamila SA, Danne J, Gurung R, Eramo MJ, Nanayakkara R, Ramm G, McGrath MJ, and Mitchell CA (2022). Endosome maturation links PI3Kalpha signaling to lysosome repopulation during basal autophagy. EMBO J 41, e110398. 10.15252/embj.2021110398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Schechter M, Atias M, Abd Elhadi S, Davidi D, Gitler D, and Sharon R (2020). alpha-Synuclein facilitates endocytosis by elevating the steady-state levels of phosphatidylinositol 4,5-bisphosphate. J Biol Chem 295, 18076–18090. 10.1074/jbc.RA120.015319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Maekawa M, Terasaka S, Mochizuki Y, Kawai K, Ikeda Y, Araki N, Skolnik EY, Taguchi T, and Arai H (2014). Sequential breakdown of 3-phosphorylated phosphoinositides is essential for the completion of macropinocytosis. Proc Natl Acad Sci U S A 111, E978–987. 10.1073/pnas.1311029111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ando T, Riku Y, Akagi A, Miyahara H, Hirano M, Ikeda T, Yabata H, Koizumi R, Oba C, Morozumi S, et al. (2022). Multiple system atrophy variant with severe hippocampal pathology. Brain Pathol 32, e13002. 10.1111/bpa.13002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Koga S, Parks A, Uitti RJ, van Gerpen JA, Cheshire WP, Wszolek ZK, and Dickson DW (2017). Profile of cognitive impairment and underlying pathology in multiple system atrophy. Mov Disord 32, 405–413. 10.1002/mds.26874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Miki Y, Foti SC, Hansen D, Strand KM, Asi YT, Tsushima E, Jaunmuktane Z, Lees AJ, Warner TT, Quinn N, et al. (2020). Hippocampal alpha-synuclein pathology correlates with memory impairment in multiple system atrophy. Brain 143, 1798–1810. 10.1093/brain/awaa126. [DOI] [PubMed] [Google Scholar]
  • 36.Qian X, Wang H, Wang Y, Chen J, Guo X, and Deng H (2020). Enhanced Autophagy in GAB1-Deficient Vascular Endothelial Cells Is Responsible for Atherosclerosis Progression. Front Physiol 11, 559396. 10.3389/fphys.2020.559396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhou L, Shao CY, Xie YJ, Wang N, Xu SM, Luo BY, Wu ZY, Ke YH, Qiu M, and Shen Y (2020). Gab1 mediates PDGF signaling and is essential to oligodendrocyte differentiation and CNS myelination. Elife 9. 10.7554/eLife.52056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Song YJ, Lundvig DM, Huang Y, Gai WP, Blumbergs PC, Hojrup P, Otzen D, Halliday GM, and Jensen PH (2007). p25alpha relocalizes in oligodendroglia from myelin to cytoplasmic inclusions in multiple system atrophy. Am J Pathol 171, 1291–1303. 10.2353/ajpath.2007.070201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhu B, Park J, Coffey S, Hsu I, Lam T, Gopal P, Ginsberg SD, Wang J, Su C, Zhao H, et al. (2022). Single-cell transcriptomic and proteomic analysis of Parkinson’s disease brains. BioRxiv doi: 10.1101/2022.02.14.480397. [DOI] [Google Scholar]
  • 40.Schjeide BM, Hooli B, Parkinson M, Hogan MF, DiVito J, Mullin K, Blacker D, Tanzi RE, and Bertram L (2009). GAB2 as an Alzheimer disease susceptibility gene: follow-up of genomewide association results. Arch Neurol 66, 250–254. 10.1001/archneurol.2008.552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Leys F, Eschlbock S, Campese N, Mahlknecht P, Peball M, Goebel G, Sidoroff V, Granata R, Bonifati V, Zschocke J, et al. (2022). Family History for Neurodegeneration in Multiple System Atrophy: Does it Indicate Susceptibility? Mov Disord 37, 2310–2312. 10.1002/mds.29202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Del Toro D, Carrasquero-Ordaz MA, Chu A, Ruff T, Shahin M, Jackson VA, Chavent M, Berbeira-Santana M, Seyit-Bremer G, Brignani S, et al. (2020). Structural Basis of Teneurin-Latrophilin Interaction in Repulsive Guidance of Migrating Neurons. Cell 180, 323–339 e319. 10.1016/j.cell.2019.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Silva JP, Lelianova VG, Ermolyuk YS, Vysokov N, Hitchen PG, Berninghausen O, Rahman MA, Zangrandi A, Fidalgo S, Tonevitsky AG, et al. (2011). Latrophilin 1 and its endogenous ligand Lasso/teneurin-2 form a high-affinity transsynaptic receptor pair with signaling capabilities. Proc Natl Acad Sci U S A 108, 12113–12118. 10.1073/pnas.1019434108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Beckmann J, Schubert R, Chiquet-Ehrismann R, and Muller DJ (2013). Deciphering teneurin domains that facilitate cellular recognition, cell-cell adhesion, and neurite outgrowth using atomic force microscopy-based single-cell force spectroscopy. Nano Lett 13, 2937–2946. 10.1021/nl4013248. [DOI] [PubMed] [Google Scholar]
  • 45.Zhang X, Lin PY, Liakath-Ali K, and Sudhof TC (2022). Teneurins assemble into presynaptic nanoclusters that promote synapse formation via postsynaptic non-teneurin ligands. Nat Commun 13, 2297. 10.1038/s41467-022-29751-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fernandez MV, Kim JH, Budde JP, Black K, Medvedeva A, Saef B, Deming Y, Del-Aguila J, Ibanez L, Dube U, et al. (2017). Analysis of neurodegenerative Mendelian genes in clinically diagnosed Alzheimer Disease. PLoS Genet 13, e1007045. 10.1371/journal.pgen.1007045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Cookson MR (2015). LRRK2 Pathways Leading to Neurodegeneration. Curr Neurol Neurosci Rep 15, 42. 10.1007/s11910-015-0564-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Van Bogaert P, Azizieh R, Desir J, Aeby A, De Meirleir L, Laes JF, Christiaens F , and Abramowicz MJ (2007). Mutation of a potassium channel-related gene in progressive myoclonic epilepsy. Ann Neurol 61, 579–586. 10.1002/ana.21121. [DOI] [PubMed] [Google Scholar]
  • 49.Sharma J, Mulherkar S, Chen UI, Xiong Y, Bajaj L, Cho BK, Goo YA, Leung HE, Tolias KF, and Sardiello M (2023). Calpain activity is negatively regulated by a KCTD7-Cullin-3 complex via non-degradative ubiquitination. Cell Discov 9, 32. 10.1038/s41421-023-00533-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ono Y, Saido TC, and Sorimachi H (2016). Calpain research for drug discovery: challenges and potential. Nat Rev Drug Discov 15, 854–876. 10.1038/nrd.2016.212. [DOI] [PubMed] [Google Scholar]
  • 51.Guindo-Martinez M, Amela R, Bonas-Guarch S, Puiggros M, Salvoro C, Miguel-Escalada I, Carey CE, Cole JB, Rueger S, Atkinson E, et al. (2021). The impact of non-additive genetic associations on age-related complex diseases. Nat Commun 12, 2436. 10.1038/s41467-021-21952-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wood AR, Tyrrell J, Beaumont R, Jones SE, Tuke MA, Ruth KS, consortium, G., Yaghootkar H, Freathy RM, Murray A, et al. (2016). Variants in the FTO and CDKAL1 loci have recessive effects on risk of obesity and type 2 diabetes, respectively. Diabetologia 59, 1214–1221. 10.1007/s00125-016-3908-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Moltke I, Grarup N, Jorgensen ME, Bjerregaard P, Treebak JT, Fumagalli M, Korneliussen TS, Andersen MA, Nielsen TS, Krarup NT, et al. (2014). A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes. Nature 512, 190–193. 10.1038/nature13425. [DOI] [PubMed] [Google Scholar]
  • 54.Goyette P, Boucher G, Mallon D, Ellinghaus E, Jostins L, Huang H, Ripke S, Gusareva ES, Annese V, Hauser SL, et al. (2015). High-density mapping of the MHC identifies a shared role for HLA-DRB1*01:03 in inflammatory bowel diseases and heterozygous advantage in ulcerative colitis. Nat Genet 47, 172–179. 10.1038/ng.3176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mani A, Meraji SM, Houshyar R, Radhakrishnan J, Mani A, Ahangar M, Rezaie TM, Taghavinejad MA, Broumand B, Zhao H, et al. (2002). Finding genetic contributions to sporadic disease: a recessive locus at 12q24 commonly contributes to patent ductus arteriosus. Proc Natl Acad Sci U S A 99, 15054–15059. 10.1073/pnas.192582999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gardiner SL, Boogaard MW, Trompet S, de Mutsert R, Rosendaal FR, Gussekloo J, Jukema JW, Roos RAC, and Aziz NA (2019). Prevalence of Carriers of Intermediate and Pathological Polyglutamine Disease-Associated Alleles Among Large Population-Based Cohorts. JAMA Neurol 76, 650–656. 10.1001/jamaneurol.2019.0423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tabrizi SJ, Leavitt BR, Landwehrmeyer GB, Wild EJ, Saft C, Barker RA, Blair NF, Craufurd D, Priller J, Rickards H, et al. (2019). Targeting Huntingtin Expression in Patients with Huntington's Disease. N Engl J Med 380, 2307–2316. 10.1056/NEJMoa1900907. [DOI] [PubMed] [Google Scholar]
  • 58.O'Callaghan B, Hofstra B, Handler HP, Kordasiewicz HB, Cole T, Duvick L, Friedrich J, Rainwater O, Yang P, Benneyworth M, et al. (2020). Antisense Oligonucleotide Therapeutic Approach for Suppression of Ataxin-1 Expression: A Safety Assessment. Mol Ther Nucleic Acids 21, 1006–1016. 10.1016/j.omtn.2020.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hauser S, Helm J, Kraft M, Korneck M, Hubener-Schmid J, and Schols L (2022). Allele-specific targeting of mutant ataxin-3 by antisense oligonucleotides in SCA3-iPSC-derived neurons. Mol Ther Nucleic Acids 27, 99–108. 10.1016/j.omtn.2021.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lopez ER, Borschel WF, and Traynor BJ (2022). New antisense oligonucleotide therapies reach first base in ALS. Nat Med 28, 25–27. 10.1038/s41591-021-01629-7. [DOI] [PubMed] [Google Scholar]
  • 61.Zhan X, Hu Y, Li B, Abecasis GR, and Liu DJ (2016). RVTESTS: an efficient and comprehensive tool for rare variant association analysis using sequence data. Bioinformatics 32, 1423–1426. 10.1093/bioinformatics/btw079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Cassa CA, Akle S, Jordan DM, and Rosenfeld JA (2017). When "N of 2" is not enough: integrating statistical and functional data in gene discovery. Cold Spring Harb Mol Case Stud 3, a001099. 10.1101/mcs.a001099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, Jordan T, Shakir K, Roazen D, Thibault J, et al. (2013). From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics 43, 11 10 11–11 10 33. 10.1002/0471250953.bi1110s43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, and Lee JJ (2015). Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience 4, 7. 10.1186/s13742-015-0047-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Abraham G, Qiu Y, and Inouye M (2017). FlashPCA2: principal component analysis of Biobank-scale genotype datasets. Bioinformatics 33, 2776–2778. 10.1093/bioinformatics/btx299. [DOI] [PubMed] [Google Scholar]
  • 66.Winkler TW, Day FR, Croteau-Chonka DC, Wood AR, Locke AE, Magi R, Ferreira T, Fall T, Graff M, Justice AE, et al. (2014). Quality control and conduct of genome-wide association meta-analyses. Nat Protoc 9, 1192–1212. 10.1038/nprot.2014.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gusev A, Ko A, Shi H, Bhatia G, Chung W, Penninx BW, Jansen R, de Geus EJ, Boomsma DI, Wright FA, et al. (2016). Integrative approaches for large-scale transcriptome-wide association studies. Nat Genet 48, 245–252. 10.1038/ng.3506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, Hao Y, Stoeckius M, Smibert P, and Satija R (2019). Comprehensive Integration of Single-Cell Data. Cell 177, 1888–1902 e1821. 10.1016/j.cell.2019.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Shabalin AA (2012). Matrix eQTL: ultra fast eQTL analysis via large matrix operations. Bioinformatics 28, 1353–1358. 10.1093/bioinformatics/bts163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GR, Thormann A, Flicek P, and Cunningham F (2016). The Ensembl Variant Effect Predictor. Genome Biol 17, 122. 10.1186/s13059-016-0974-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dolzhenko E, van Vugt J, Shaw RJ, Bekritsky MA, van Blitterswijk M, Narzisi G, Ajay SS, Rajan V, Lajoie BR, Johnson NH, et al. (2017). Detection of long repeat expansions from PCR-free whole-genome sequence data. Genome Res 27, 1895–1903. 10.1101/gr.225672.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Dolzhenko E, Weisburd B, Ibanez K, Rajan-Babu IS, Anyansi C, Bennett MF, Billingsley K, Carroll A, Clamons S, Danzi MC, et al. (2022). REViewer: haplotype-resolved visualization of read alignments in and around tandem repeats. Genome Med 14, 84. 10.1186/s13073-022-01085-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ibanez K, Polke J, Hagelstrom RT, Dolzhenko E, Pasko D, Thomas ERA, Daugherty LC, Kasperaviciute D, Smith KR, Group, W.G.S.f.N.D., et al. (2022). Whole genome sequencing for the diagnosis of neurological repeat expansion disorders in the UK: a retrospective diagnostic accuracy and prospective clinical validation study. Lancet Neurol 21, 234–245. 10.1016/S1474-4422(21)00462-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chen X, Schulz-Trieglaff O, Shaw R, Barnes B, Schlesinger F, Kallberg M, Cox AJ, Kruglyak S, and Saunders CT (2016). Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications. Bioinformatics 32, 1220–1222. 10.1093/bioinformatics/btv710. [DOI] [PubMed] [Google Scholar]
  • 75.Danecek P, Bonfield JK, Liddle J, Marshall J, Ohan V, Pollard MO, Whitwham A, Keane T, McCarthy SA, Davies RM, and Li H (2021). Twelve years of SAMtools and BCFtools. Gigascience 10. 10.1093/gigascience/giab008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, and Tamayo P (2015). The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 1, 417–425. 10.1016/j.cels.2015.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

Data Availability Statement

The summary statistics from the additive and recessive GWAS models have been deposited in the GWAS catalog (https://www.ebi.ac.uk/gwas/). The individual-level sequence data for a subset of the MSA genomes (n = 683 cases) reported in this paper will be available upon publication in dbGaP (accession number: phs001963). Public data sharing was not feasible for the remaining 205 MSA genomes; access to these data will be granted to qualified researchers via appropriate collaboration agreements. The TOPMed control genome data are available in dbGaP (accession numbers: phs001662.v2.p1, phs00974.v5.p4, phs000951.v5.p5). The control genome data from 1,980 subjects from the DementiaSeq project are available in dbGaP (accession number: phs001963), and the remaining control genomes are available upon request from the Wellderly study team (contact: atorkama@scripps.edu). The programming code used in this paper is available at https://zenodo.org/records/10723069.

RESOURCES