Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Genome-wide association analyses identify new risk variants and the genetic architecture of amyotrophic lateral sclerosis

Abstract

To elucidate the genetic architecture of amyotrophic lateral sclerosis (ALS) and find associated loci, we assembled a custom imputation reference panel from whole-genome-sequenced patients with ALS and matched controls (n = 1,861). Through imputation and mixed-model association analysis in 12,577 cases and 23,475 controls, combined with 2,579 cases and 2,767 controls in an independent replication cohort, we fine-mapped a new risk locus on chromosome 21 and identified C21orf2 as a gene associated with ALS risk. In addition, we identified MOBP and SCFD1 as new associated risk loci. We established evidence of ALS being a complex genetic trait with a polygenic architecture. Furthermore, we estimated the SNP-based heritability at 8.5%, with a distinct and important role for low-frequency variants (frequency 1–10%). This study motivates the interrogation of larger samples with full genome coverage to identify rare causal variants that underpin ALS risk.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Comparison of imputation accuracy.
Figure 2: Meta-analysis and LMM associations.
Figure 3: Partitioned heritability.

Similar content being viewed by others

References

  1. Hardiman, O., van den Berg, L.H. & Kiernan, M.C. Clinical diagnosis and management of amyotrophic lateral sclerosis. Nat. Rev. Neurol. 7, 639–649 (2011).

    Article  CAS  Google Scholar 

  2. Al-Chalabi, A. et al. An estimate of amyotrophic lateral sclerosis heritability using twin data. J. Neurol. Neurosurg. Psychiatry 81, 1324–1326 (2010).

    Article  CAS  Google Scholar 

  3. van Es, M.A. et al. Genome-wide association study identifies 19p13.3 (UNC13A) and 9p21.2 as susceptibility loci for sporadic amyotrophic lateral sclerosis. Nat. Genet. 41, 1083–1087 (2009).

    Article  CAS  Google Scholar 

  4. Laaksovirta, H. et al. Chromosome 9p21 in amyotrophic lateral sclerosis in Finland: a genome-wide association study. Lancet Neurol. 9, 978–985 (2010).

    Article  CAS  Google Scholar 

  5. Shatunov, A. et al. Chromosome 9p21 in sporadic amyotrophic lateral sclerosis in the UK and seven other countries: a genome-wide association study. Lancet Neurol. 9, 986–994 (2010).

    Article  CAS  Google Scholar 

  6. DeJesus-Hernandez, M. et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9orf72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256 (2011).

    Article  CAS  Google Scholar 

  7. Renton, A.E. et al. A hexanucleotide repeat expansion in C9orf72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72, 257–268 (2011).

    Article  CAS  Google Scholar 

  8. Fogh, I. et al. A genome-wide association meta-analysis identifies a novel locus at 17q11.2 associated with sporadic amyotrophic lateral sclerosis. Hum. Mol. Genet. 23, 2220–2231 (2014).

    Article  CAS  Google Scholar 

  9. 1000 Genomes Project Consortium. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

  10. Genome of the Netherlands Consortium. Whole-genome sequence variation, population structure and demographic history of the Dutch population. Nat. Genet. 46, 818–825 (2014).

  11. Yang, J., Zaitlen, N.A., Goddard, M.E., Visscher, P.M. & Price, A.L. Advantages and pitfalls in the application of mixed-model association methods. Nat. Genet. 46, 100–106 (2014).

    Article  Google Scholar 

  12. Bulik-Sullivan, B.K. et al. LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat. Genet. 47, 291–295 (2015).

    Article  CAS  Google Scholar 

  13. Höglinger, G.U. et al. Identification of common variants influencing risk of the tauopathy progressive supranuclear palsy. Nat. Genet. 43, 699–705 (2011).

    Article  Google Scholar 

  14. Irwin, D.J. et al. Myelin oligodendrocyte basic protein and prognosis in behavioral-variant frontotemporal dementia. Neurology 83, 502–509 (2014).

    Article  CAS  Google Scholar 

  15. Cirulli, E.T. et al. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science 347, 1436–1441 (2015).

    Article  CAS  Google Scholar 

  16. Freischmidt, A. et al. Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia. Nat. Neurosci. 18, 631–636 (2015).

    Article  CAS  Google Scholar 

  17. Skol, A.D., Scott, L.J., Abecasis, G.R. & Boehnke, M. Joint analysis is more efficient than replication-based analysis for two-stage genome-wide association studies. Nat. Genet. 38, 209–213 (2006).

    Article  CAS  Google Scholar 

  18. Nicolae, D.L. et al. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 6, e1000888 (2010).

    Article  Google Scholar 

  19. Ramasamy, A. et al. Genetic variability in the regulation of gene expression in ten regions of the human brain. Nat. Neurosci. 17, 1418–1428 (2014).

    Article  CAS  Google Scholar 

  20. Wray, N.R. et al. Pitfalls of predicting complex traits from SNPs. Nat. Rev. Genet. 14, 507–515 (2013).

    Article  CAS  Google Scholar 

  21. Johnston, C.A. et al. Amyotrophic lateral sclerosis in an urban setting: a population based study of inner city London. J. Neurol. 253, 1642–1643 (2006).

    Article  Google Scholar 

  22. Lee, S.H. et al. Estimating the proportion of variation in susceptibility to schizophrenia captured by common SNPs. Nat. Genet. 44, 247–250 (2012).

    Article  CAS  Google Scholar 

  23. Pers, T.H. et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nat. Commun. 6, 5890 (2015).

    Article  CAS  Google Scholar 

  24. Ramakrishnan, N.A., Drescher, M.J. & Drescher, D.G. The SNARE complex in neuronal and sensory cells. Mol. Cell. Neurosci. 50, 58–69 (2012).

    Article  CAS  Google Scholar 

  25. Ferraiuolo, L., Kirby, J., Grierson, A.J., Sendtner, M. & Shaw, P.J. Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat. Rev. Neurol. 7, 616–630 (2011).

    Article  CAS  Google Scholar 

  26. Lai, C.K. et al. Functional characterization of putative cilia genes by high-content analysis. Mol. Biol. Cell 22, 1104–1119 (2011).

    Article  CAS  Google Scholar 

  27. Ma, X., Peterson, R. & Turnbull, J. Adenylyl cyclase type 3, a marker of primary cilia, is reduced in primary cell culture and in lumbar spinal cord in situ in G93A SOD1 mice. BMC Neurosci. 12, 71 (2011).

    Article  Google Scholar 

  28. Krohn, K. et al. Immunochemical characterization of a novel mitochondrially located protein encoded by a nuclear gene within the DFNB8/10 critical region on 21q22.3. Biochem. Biophys. Res. Commun. 238, 806–810 (1997).

    Article  CAS  Google Scholar 

  29. Fang, X. et al. The NEK1 interactor, C21orf2, is required for efficient DNA damage repair. Acta Biochim. Biophys. Sin. (Shanghai) 47, 834–841 (2015).

    Article  CAS  Google Scholar 

  30. Vérièpe, J., Fossouo, L. & Parker, J.A. Neurodegeneration in C. elegans models of ALS requires TIR-1/Sarm1 immune pathway activation in neurons. Nat. Commun. 6, 7319 (2015).

    Article  Google Scholar 

  31. Delaneau, O. et al. Integrating sequence and array data to create an improved 1000 Genomes Project haplotype reference panel. Nat. Commun. 5, 3934 (2014).

    Article  CAS  Google Scholar 

  32. Howie, B., Marchini, J. & Stephens, M. Genotype imputation with thousands of genomes. G3 (Bethesda) 1, 457–470 (2011).

    Article  Google Scholar 

Download references

Acknowledgements

The work of the contributing groups was supported by various grants from governmental and charitable bodies. Details are provided in the Supplementary Note.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

A.V., N.T., K.L., B.R., K.V., M.R.-G., B.K., J.Z., L.L., L.D.G., S.M., F.S., V.M., M.d.C., S. Pinto, J.S.M., R.R.-G., M.P., S. Chandran, S. Colville, R.S., K.E.M., P.J.S., J.H., R.W.O., A. Pittman, K.S., P.F., A. Malaspina, S.T., S. Petri, S. Abdulla, C.D., M.S., T. Meyer, R.A.O., K.A.S., M.W.-P., C.L.-H., V.M.V.D., J.Q.T., L.E., L. McCluskey, A.N.B., Y.P., T. Meitinger, P.L., M.R.-B., C.R.A., C. Maurel, G. Bensimon, B.L., A.B., C.A.M.P., S.S.-D., A.D., N.W.W., L.T., W.L., A.F., M.R., S. Cichon, M.M.N., P.A., C. Tzourio, J.-F.D., A.G.U., F.R., K.E., A.H., C. Curtis, H.M.B., A.J.v.d.K., M.d.V., A.G., M.W., C.E.S., B.N.S., O.P., C. Cereda, R.D.B., G.P.C., S.D'A., C.B., G.S., L. Mazzini, V.P., C.G., C. Tiloca, A.R., A. Calvo, C. Moglia, M.B., S. Arcuti, R.C., C.Z., C.L., S. Penco, N.R., A. Padovani, M.F., B.M., R.J.S., PARALS Registry, SLALOM Group, SLAP Registry, FALS Sequencing Consortium, SLAGEN Consortium, NNIPPS Study Group, I.B., G.A.N., D.B.R., R.P., M.C.K., J.G., O.W.W., T.R., B.S., I.K., C.A.H., P.N.L., F.C., A. Chìo, E.B., E.P., R.T., G.L., J.P., A.C.L., J.H.W., W.R., P.V.D., L.F., T.P., R.H.B., J.D.G., J.E.L., O. Hardiman, P.M.A., P.C., P.V., V.S., M.A.v.E., A.A.-C., L.H.v.d.B. and J.H.V. were involved in phenotyping, sample collection and management. W.v.R., A.S., A.M.D., R.L.M., F.P.D., R.A.A.v.d.S., P.T.C.v.D., G.H.P.T., M.K., A.M.B., W.S., A.R.J., K.P.K., I.F., A.V., N.T., R.D.S., W.J.B., A.V., K.V., M.R.-G., B.K., L.L., S. Abdulla, K.S., E.P., F.P.D., J.M., C. Curtis, G. Breen, A.A.-C. and J.H.V. prepared DNA and performed SNP array hybridizations. W.v.R., S.L.P., K.P.K., K.L., A.M.D., P.T.C.v.D., G.H.P.T., K.R.v.E., P.I.W.d.B. and J.H.V. were involved in the next-generation sequencing analyses. W.v.R., K.R.v.E., A. Menelaou, P.I.W.d.B., A.A.-C. and J.H.V. performed the imputation. W.v.R., A.S., F.P.D., R.L.M., S.L.P., S.d.J., I.F., N.T., W.S., A.R.J., K.P.K., K.R.v.E., K.S., H.M.B., P.I.W.d.B., M.A.v.E., C.M.L., G. Breen, A.A.-C., L.H.v.d.B. and J.H.V. performed GWAS analyses. W.v.R., A.M.D., R.A.A.v.d.S., R.L.M., C.R.A., M.K., A.M.B., R.D.S., E.P.M., J.A.F., C. Tunca, H.H., K.Z., P.C., P.V. and J.H.V. performed the replication analyses. W.v.R., A.S., R.L.M., M.R.R., J.Y., N.R.W., P.M.V., C.M.L., A.A.-C. and J.H.V. performed polygenic risk scoring and heritability analyses. S.d.J., U.V., L.F., T.H.P., W.v.R., O. Harschnitz, G. Breen, R.J.P. and J.H.V. performed biological pathway analyses. U.V., L.F., W.v.R. and J.H.V. performed eQTL analyses. W.v.R., A.S., A.A.-C., L.H.v.d.B. and J.H.V. prepared the manuscript with contributions from all authors. A.A.-C., L.H.v.d.B. and J.H.V. directed the study.

Corresponding authors

Correspondence to Ammar Al-Chalabi or Jan H Veldink.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

A list of members appears in the Supplementary Note.

Integrated supplementary information

Supplementary Figure 1 Coverage distribution for reference panel.

(a) Average coverage for all non-reference bases for all samples in the custom reference panel. An average coverage of 43.7 reads per base was achieved. (b) Percentage of non-reference bases covered by at least 5 (blue), 10 (yellow) or 20 (red) reads. Histograms include all individuals, before quality control.

Source data

Supplementary Figure 2 Comparison between meta-analysis and linear mixed-model results.

Regressing the –log10 (P values) derived from the linear mixed model on the meta-analysis P values yielded a slope below the diagonal (β = 0.86), implying no overall inflation of the test statistic. Strongly associated SNPs, however, deviated from the regression line representing the increased power of a linear mixed model in comparison to a meta-analysis for association testing.

Supplementary Figure 3 Quantile–quantile plots.

(a,b) Meta-analysis (a) and linear mixed model (b). For presentation purposes, P values <5 × 10−8 are plotted at 5 × 10−8.

Supplementary Figure 4 Replication results.

Forest plot for the inverse-variance-weighted, fixed-effect meta-analysis of the discovery phase and replication cohorts. OR, odds ratio; CI, confidence interval.

Supplementary Figure 5 Fine-mapping of the C21orf2 locus.

(a) Associations for all SNPs analyzed in the GWAS in the C21orf2 locus where only rs75087725 reaches genome-wide significance. (b) LD as defined by |D′| (absolute D-prime value) for all variants obtained from whole-genome sequencing data of the custom reference panel. Possible causal variants for driving factors of the GWAS association were considered when they met the following criteria: (i) risk allele frequency difference between cases and controls exceeding 0.4% (risk allele frequency difference for rs75087725 = 0.8%), (ii) rs75087725 as the best genotyped GWAS tag for the variant and (iii) minor allele with a risk-increasing effect on ALS. (c) No such variant was in LD with rs75087725. (d) LD defined by R2 is very sparse. (e) C21orf2 is the only gene in this locus with an increased burden of rare nonsynonymous variants in the sequencing data of our custom reference panel, indicating that C21orf2 is indeed the ALS risk gene.

Source data

Supplementary Figure 6 C21orf2 rare variant burden.

Summary of the rare (MAF < 0.05) nonsynonymous and loss-of-function mutations in the canonical transcript of C21orf2. Conditioning on the SNP found to be associated in the GWAS (rs75087725, p.V58L; gray), there was an increased burden of nonsynonymous and loss-of-function mutations among ALS cases (PT5 = 9.2 × 10−5, PT1 = 0.01). Odds ratios (calculated by counting alleles in cases and controls per stratum, unadjusted for principal components, combined in a Cochran–Mantel–Haenszel test) are 1.63 and 1.48 for T5 and T1 burden, respectively. The two loss-of-function mutations observed in cases are colored red.

Supplementary Figure 7 cis-eQTL regional plots for six genome-wide-significant loci.

Highlighted are cis-eQTLs acquired from several resources (genes in green) and brain cis-eQTLs (genes in black). Stranded RNA-seq data for one fetal brain (3,000-bp sliding window) are shown on a separate track. For the MOBP region, there was an eQTL effect for MOBP (P = 7.12 × 10−18), but the effect SNP, rs1707953, did not show any association with ALS in our GWAS (P = 0.74). Further details of highlighted brain cis-eQTLs and non-brain cis-eQTLs are given in Supplementary Table 10 and the Supplementary Data Set. eQTL annotation and LD data are shown only for SNPs present in the 1000 Genomes Project p1v3 CEU population.

Source data

Supplementary Figure 8 Polygenic risk scores.

(a) Polygenic risk score analyses where nine cohorts were used as targets. Best predictions were made when including the six genome-wide-significant SNPs from the C9orf72 and UNC13A loci only. (b) Polygenic risk score analyses excluding all variants on chromosome 9. Increased polygenic risk score predictions were made when including more variants by lowering the P-value threshold. Note that the overall prediction accuracy is lower than when SNPs on chromosome 9 were included.

Source data

Supplementary Figure 9 Partitioned heritability excluding candidate loci.

(a) SNPs in the C9orf72 locus (within 1 Mb of rs3849943 and r2 >0.2) were excluded from heritability estimates. (b) SNPs within 1 Mb of the top associated SNP and r2 >0.2 for all loci exceeding genome-wide significance were excluded for heritability estimates. In both instances, most heritability was explained by low-frequency variants (MAF < 0.1).

Source data

Supplementary Figure 10 DEPICT biological pathway analysis.

The top ten terms from Gene Ontology, KEGG and Reactome pathways that are most enriched for genes tagged by SNPs in the GWAS are displayed. Different thresholds of significance were used to select SNPs for the DEPICT analyses. The lengths of the bars correspond to the nominal significance levels, the black line indicates the P-value threshold of 0.05 and color corresponds to FDR, determined by 200 permutations. When all SNPs with a P value <1 × 10−4 in the linear mixed-model analysis were included, it identifies the Gene Ontology category SNAP receptor (SNARE) activity as the only significantly enriched term after correction for multiple testing.

Supplementary Figure 11 Population outlier removal.

(a) Example of stratum sNL2 projected onto the first two principal components calculated on HapMap 3 individuals. Individuals of non-European ancestry (±10 s.d. from the HapMap CEU mean on PC1–PC4) were removed. (b) Subsequent removal of samples deviating by more than 4 s.d. from the stratum mean eigenvalues on PC1 and PC2 (HapMap 3). (c,d) Individuals remaining after removing population outliers resulting in a homogenous European sample where the Scandinavian individuals (mainly Finnish), as expected, depart from the other strata.

Source data

Supplementary Figure 12 Population structure of the custom reference panel.

Individuals projected onto the first two principal components calculated on HapMap 3 individuals. NL, Netherlands; Ir, Ireland; It, Italy; No, Norway; Sp, Spain; Sw, Sweden; US, United States.

Source data

Supplementary Figure 13 C21orf2 rare variant analysis quality control.

Quantile–quantile plots for the single-SNV regression of chromosome 21 including the first ten principal components as covariates are displayed on the left. Principal-components plots for each stratum are shown on the right. Population stratification was successfully corrected for, resulting in well-behaved burden tests without evidence for overall inflation of the test statistic.

Source data

Supplementary Figure 14 Genetic relationship matrix distribution.

(a) Diagonal values of the SNP-based GRM. (b) Distribution of the off-diagonal values representing the relatedness between samples. Pairs of individuals whose relatedness exceeded 0.05 were excluded from the heritability estimations. GRM, genetic relationship matrix.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–14, Supplementary Tables 1–20 and Supplementary Note. (PDF 4545 kb)

Supplementary Data Set: Brain cis-eQTLs.

Overlap with previously identified brain cis-eQTLs for all SNPs in the genome-wide-significant loci. (XLSX 88 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

van Rheenen, W., Shatunov, A., Dekker, A. et al. Genome-wide association analyses identify new risk variants and the genetic architecture of amyotrophic lateral sclerosis. Nat Genet 48, 1043–1048 (2016). https://doi.org/10.1038/ng.3622

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3622

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing