Common and rare variant association analyses in amyotrophic lateral sclerosis identify 15 risk loci with distinct genetic architectures and neuron-specific biology

Wouter van Rheenen, Rick A A van der Spek, Mark K Bakker, Joke J F A van Vugt, Paul J Hop, Ramona A J Zwamborn, Niek de Klein, Harm-Jan Westra, Olivier B Bakker, Patrick Deelen, Gemma Shireby, Eilis Hannon, Matthieu Moisse, Denis Baird, Restuadi Restuadi, Egor Dolzhenko, Annelot M Dekker, Klara Gawor, Henk-Jan Westeneng, Gijs H P Tazelaar, Kristel R van Eijk, Maarten Kooyman, Ross P Byrne, Mark Doherty, Mark Heverin, Ahmad Al Khleifat, Alfredo Iacoangeli, Aleksey Shatunov, Nicola Ticozzi, Johnathan Cooper-Knock, Bradley N Smith, Marta Gromicho, Siddharthan Chandran, Suvankar Pal, Karen E Morrison, Pamela J Shaw, John Hardy, Richard W Orrell, Michael Sendtner, Thomas Meyer, Nazli Başak, Anneke J van der Kooi, Antonia Ratti, Isabella Fogh, Cinzia Gellera, Giuseppe Lauria, Stefania Corti, Cristina Cereda, Daisy Sproviero, Sandra D'Alfonso, Gianni Sorarù, Gabriele Siciliano, Massimiliano Filosto, Alessandro Padovani, Adriano Chiò, Andrea Calvo, Cristina Moglia, Maura Brunetti, Antonio Canosa, Maurizio Grassano, Ettore Beghi, Elisabetta Pupillo, Giancarlo Logroscino, Beatrice Nefussy, Alma Osmanovic, Angelica Nordin, Yossef Lerner, Michal Zabari, Marc Gotkine, Robert H Baloh, Shaughn Bell, Patrick Vourc'h, Philippe Corcia, Philippe Couratier, Stéphanie Millecamps, Vincent Meininger, François Salachas, Jesus S Mora Pardina, Abdelilah Assialioui, Ricardo Rojas-García, Patrick A Dion, Jay P Ross, Albert C Ludolph, Jochen H Weishaupt, David Brenner, Axel Freischmidt, Gilbert Bensimon, Alexis Brice, Alexandra Durr, Christine A M Payan, Safa Saker-Delye, Nicholas W Wood, Simon Topp, Rosa Rademakers, Lukas Tittmann, Wolfgang Lieb, Andre Franke, Stephan Ripke, Alice Braun, Julia Kraft, David C Whiteman, Catherine M Olsen, Andre G Uitterlinden, Albert Hofman, Marcella Rietschel, Sven Cichon, Markus M Nöthen, Philippe Amouyel, SLALOM Consortium, PARALS Consortium, SLAGEN Consortium, SLAP Consortium, Bryan J Traynor, Andrew B Singleton, Miguel Mitne Neto, Ruben J Cauchi, Roel A Ophoff, Martina Wiedau-Pazos, Catherine Lomen-Hoerth, Vivianna M van Deerlin, Julian Grosskreutz, Annekathrin Roediger, Nayana Gaur, Alexander Jörk, Tabea Barthel, Erik Theele, Benjamin Ilse, Beatrice Stubendorff, Otto W Witte, Robert Steinbach, Christian A Hübner, Caroline Graff, Lev Brylev, Vera Fominykh, Vera Demeshonok, Anastasia Ataulina, Boris Rogelj, Blaž Koritnik, Janez Zidar, Metka Ravnik-Glavač, Damjan Glavač, Zorica Stević, Vivian Drory, Monica Povedano, Ian P Blair, Matthew C Kiernan, Beben Benyamin, Robert D Henderson, Sarah Furlong, Susan Mathers, Pamela A McCombe, Merrilee Needham, Shyuan T Ngo, Garth A Nicholson, Roger Pamphlett, Dominic B Rowe, Frederik J Steyn, Kelly L Williams, Karen A Mather, Perminder S Sachdev, Anjali K Henders, Leanne Wallace, Mamede de Carvalho, Susana Pinto, Susanne Petri, Markus Weber, Guy A Rouleau, Vincenzo Silani, Charles J Curtis, Gerome Breen, Jonathan D Glass, Robert H Brown Jr, John E Landers, Christopher E Shaw, Peter M Andersen, Ewout J N Groen, Michael A van Es, R Jeroen Pasterkamp, Dongsheng Fan, Fleur C Garton, Allan F McRae, George Davey Smith, Tom R Gaunt, Michael A Eberle, Jonathan Mill, Russell L McLaughlin, Orla Hardiman, Kevin P Kenna, Naomi R Wray, Ellen Tsai, Heiko Runz, Lude Franke, Ammar Al-Chalabi, Philip Van Damme, Leonard H van den Berg, Jan H Veldink, Giancarlo Comi, Nilo Riva, Christian Lunetta, Francesca Gerardi, Maria Sofia Cotelli, Fabrizio Rinaldi, Luca Chiveri, Maria Cristina Guaita, Patrizia Perrone, Mauro Ceroni, Luca Diamanti, Carlo Ferrarese, Lucio Tremolizzo, Maria Luisa Delodovici, Giorgio Bono, Antonio Canosa, Umberto Manera, Rosario Vasta, Alessandro Bombaci, Federico Casale, Giuseppe Fuda, Paolina Salamone, Barbara Iazzolino, Laura Peotta, Paolo Cugnasco, Giovanni De Marco, Maria Claudia Torrieri, Francesca Palumbo, Salvatore Gallone, Marco Barberis, Luca Sbaiz, Salvatore Gentile, Alessandro Mauro, Letizia Mazzini, Fabiola De Marchi, Lucia Corrado, Sandra D'Alfonso, Antonio Bertolotto, Maurizio Gionco, Daniela Leotta, Enrico Odddenino, Daniele Imperiale, Roberto Cavallo, Pietro Pignatta, Marco De Mattei, Claudio Geda, Diego Maria Papurello, Graziano Gusmaroli, Cristoforo Comi, Carmelo Labate, Luigi Ruiz, Delfina Ferrandi, Eugenia Rota, Marco Aguggia, Nicoletta Di Vito, Piero Meineri, Paolo Ghiglione, Nicola Launaro, Michele Dotta, Alessia Di Sapio, Guido Giardini, Cinzia Tiloca, Silvia Peverelli, Franco Taroni, Viviana Pensato, Barbara Castellotti, Giacomo P Comi, Roberto Del Bo, Mauro Ceroni, Stella Gagliardi, Lucia Corrado, Letizia Mazzini, Flavia Raggi, Costanza Simoncini, Annalisa Lo Gerfo, Maurizio Inghilleri, Alessandra Ferlini, Isabella L Simone, Bruno Passarella, Vito Guerra, Stefano Zoccolella, Cecilia Nozzoli, Ciro Mundi, Maurizio Leone, Michele Zarrelli, Filippo Tamma, Francesco Valluzzi, Gianluigi Calabrese, Giovanni Boero, Augusto Rini, Wouter van Rheenen, Rick A A van der Spek, Mark K Bakker, Joke J F A van Vugt, Paul J Hop, Ramona A J Zwamborn, Niek de Klein, Harm-Jan Westra, Olivier B Bakker, Patrick Deelen, Gemma Shireby, Eilis Hannon, Matthieu Moisse, Denis Baird, Restuadi Restuadi, Egor Dolzhenko, Annelot M Dekker, Klara Gawor, Henk-Jan Westeneng, Gijs H P Tazelaar, Kristel R van Eijk, Maarten Kooyman, Ross P Byrne, Mark Doherty, Mark Heverin, Ahmad Al Khleifat, Alfredo Iacoangeli, Aleksey Shatunov, Nicola Ticozzi, Johnathan Cooper-Knock, Bradley N Smith, Marta Gromicho, Siddharthan Chandran, Suvankar Pal, Karen E Morrison, Pamela J Shaw, John Hardy, Richard W Orrell, Michael Sendtner, Thomas Meyer, Nazli Başak, Anneke J van der Kooi, Antonia Ratti, Isabella Fogh, Cinzia Gellera, Giuseppe Lauria, Stefania Corti, Cristina Cereda, Daisy Sproviero, Sandra D'Alfonso, Gianni Sorarù, Gabriele Siciliano, Massimiliano Filosto, Alessandro Padovani, Adriano Chiò, Andrea Calvo, Cristina Moglia, Maura Brunetti, Antonio Canosa, Maurizio Grassano, Ettore Beghi, Elisabetta Pupillo, Giancarlo Logroscino, Beatrice Nefussy, Alma Osmanovic, Angelica Nordin, Yossef Lerner, Michal Zabari, Marc Gotkine, Robert H Baloh, Shaughn Bell, Patrick Vourc'h, Philippe Corcia, Philippe Couratier, Stéphanie Millecamps, Vincent Meininger, François Salachas, Jesus S Mora Pardina, Abdelilah Assialioui, Ricardo Rojas-García, Patrick A Dion, Jay P Ross, Albert C Ludolph, Jochen H Weishaupt, David Brenner, Axel Freischmidt, Gilbert Bensimon, Alexis Brice, Alexandra Durr, Christine A M Payan, Safa Saker-Delye, Nicholas W Wood, Simon Topp, Rosa Rademakers, Lukas Tittmann, Wolfgang Lieb, Andre Franke, Stephan Ripke, Alice Braun, Julia Kraft, David C Whiteman, Catherine M Olsen, Andre G Uitterlinden, Albert Hofman, Marcella Rietschel, Sven Cichon, Markus M Nöthen, Philippe Amouyel, SLALOM Consortium, PARALS Consortium, SLAGEN Consortium, SLAP Consortium, Bryan J Traynor, Andrew B Singleton, Miguel Mitne Neto, Ruben J Cauchi, Roel A Ophoff, Martina Wiedau-Pazos, Catherine Lomen-Hoerth, Vivianna M van Deerlin, Julian Grosskreutz, Annekathrin Roediger, Nayana Gaur, Alexander Jörk, Tabea Barthel, Erik Theele, Benjamin Ilse, Beatrice Stubendorff, Otto W Witte, Robert Steinbach, Christian A Hübner, Caroline Graff, Lev Brylev, Vera Fominykh, Vera Demeshonok, Anastasia Ataulina, Boris Rogelj, Blaž Koritnik, Janez Zidar, Metka Ravnik-Glavač, Damjan Glavač, Zorica Stević, Vivian Drory, Monica Povedano, Ian P Blair, Matthew C Kiernan, Beben Benyamin, Robert D Henderson, Sarah Furlong, Susan Mathers, Pamela A McCombe, Merrilee Needham, Shyuan T Ngo, Garth A Nicholson, Roger Pamphlett, Dominic B Rowe, Frederik J Steyn, Kelly L Williams, Karen A Mather, Perminder S Sachdev, Anjali K Henders, Leanne Wallace, Mamede de Carvalho, Susana Pinto, Susanne Petri, Markus Weber, Guy A Rouleau, Vincenzo Silani, Charles J Curtis, Gerome Breen, Jonathan D Glass, Robert H Brown Jr, John E Landers, Christopher E Shaw, Peter M Andersen, Ewout J N Groen, Michael A van Es, R Jeroen Pasterkamp, Dongsheng Fan, Fleur C Garton, Allan F McRae, George Davey Smith, Tom R Gaunt, Michael A Eberle, Jonathan Mill, Russell L McLaughlin, Orla Hardiman, Kevin P Kenna, Naomi R Wray, Ellen Tsai, Heiko Runz, Lude Franke, Ammar Al-Chalabi, Philip Van Damme, Leonard H van den Berg, Jan H Veldink, Giancarlo Comi, Nilo Riva, Christian Lunetta, Francesca Gerardi, Maria Sofia Cotelli, Fabrizio Rinaldi, Luca Chiveri, Maria Cristina Guaita, Patrizia Perrone, Mauro Ceroni, Luca Diamanti, Carlo Ferrarese, Lucio Tremolizzo, Maria Luisa Delodovici, Giorgio Bono, Antonio Canosa, Umberto Manera, Rosario Vasta, Alessandro Bombaci, Federico Casale, Giuseppe Fuda, Paolina Salamone, Barbara Iazzolino, Laura Peotta, Paolo Cugnasco, Giovanni De Marco, Maria Claudia Torrieri, Francesca Palumbo, Salvatore Gallone, Marco Barberis, Luca Sbaiz, Salvatore Gentile, Alessandro Mauro, Letizia Mazzini, Fabiola De Marchi, Lucia Corrado, Sandra D'Alfonso, Antonio Bertolotto, Maurizio Gionco, Daniela Leotta, Enrico Odddenino, Daniele Imperiale, Roberto Cavallo, Pietro Pignatta, Marco De Mattei, Claudio Geda, Diego Maria Papurello, Graziano Gusmaroli, Cristoforo Comi, Carmelo Labate, Luigi Ruiz, Delfina Ferrandi, Eugenia Rota, Marco Aguggia, Nicoletta Di Vito, Piero Meineri, Paolo Ghiglione, Nicola Launaro, Michele Dotta, Alessia Di Sapio, Guido Giardini, Cinzia Tiloca, Silvia Peverelli, Franco Taroni, Viviana Pensato, Barbara Castellotti, Giacomo P Comi, Roberto Del Bo, Mauro Ceroni, Stella Gagliardi, Lucia Corrado, Letizia Mazzini, Flavia Raggi, Costanza Simoncini, Annalisa Lo Gerfo, Maurizio Inghilleri, Alessandra Ferlini, Isabella L Simone, Bruno Passarella, Vito Guerra, Stefano Zoccolella, Cecilia Nozzoli, Ciro Mundi, Maurizio Leone, Michele Zarrelli, Filippo Tamma, Francesco Valluzzi, Gianluigi Calabrese, Giovanni Boero, Augusto Rini

Abstract

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with a lifetime risk of one in 350 people and an unmet need for disease-modifying therapies. We conducted a cross-ancestry genome-wide association study (GWAS) including 29,612 patients with ALS and 122,656 controls, which identified 15 risk loci. When combined with 8,953 individuals with whole-genome sequencing (6,538 patients, 2,415 controls) and a large cortex-derived expression quantitative trait locus (eQTL) dataset (MetaBrain), analyses revealed locus-specific genetic architectures in which we prioritized genes either through rare variants, short tandem repeats or regulatory effects. ALS-associated risk loci were shared with multiple traits within the neurodegenerative spectrum but with distinct enrichment patterns across brain regions and cell types. Of the environmental and lifestyle risk factors obtained from the literature, Mendelian randomization analyses indicated a causal role for high cholesterol levels. The combination of all ALS-associated signals reveals a role for perturbations in vesicle-mediated transport and autophagy and provides evidence for cell-autonomous disease initiation in glutamatergic neurons.

Conflict of interest statement

J.H.V. has sponsored research agreements with Biogen. L.H.v.d.B. receives personal fees from Cytokinetics outside of the submitted work. A.A.-C. has served on scientific advisory boards for Mitsubishi Tanabe Pharma, Orion Pharma, Biogen, Lilly, GSK, Apellis, Amylyx and Wave Therapeutics. A. Chiò. serves on scientific advisory boards for Mitsubishi Tanabe, Roche, Biogen, Denali and Cytokinetics. J.E.L. is a member of the scientific advisory board for Cerevel Therapeutics, a consultant for ACI Clinical LLC sponsored by Biogen, Inc. or Ionis Pharmaceuticals, Inc. J.E.L. is also a consultant for Perkins Coie LLP and may provide expert testimony. The remaining authors declare no competing interests related to this work.

© 2021. The Author(s).

Figures

Fig. 1. Manhattan plot of cross-ancestry meta-analysis.
Fig. 1. Manhattan plot of cross-ancestry meta-analysis.
Genome-wide association statistics obtained by IVW meta-analysis of the stratified SAIGE logistic mixed model regression. The y axis corresponds to two-tailed −log10 (Pvalues); the x axis corresponds to genomic coordinates (GRCh37). The horizontal dashed line reflects the threshold for calling genome-wide significant SNPs (P = 5 × 10−8). Color coding and gene labels reflect those prioritized by the gene-prioritization analysis. Labels in bold indicate genes with known highly pathogenic mutations for ALS. SAIGE = Scalable and Accurate Implementation of Generalized mixed model software package. Source data
Fig. 2. Genetic modifier analyses.
Fig. 2. Genetic modifier analyses.
a, Cox proportional HRs for genome-wide significant SNPs (brown, n = 15), PRSs (red, n = 2) and rare variant burden in ALS-risk genes (pink, n = 7) on survival (months) tested in 6,095 patients with ALS. Estimated HRs are displayed with error bars corresponding to 95% CIs. Higher HRs correspond to shorter survival times. b, Effect estimates from a linear regression model of age at onset (years) in 6,095 patients with ALS. Lower effect estimates correspond to a younger age at onset. Effect estimates from linear regression are displayed with error bars corresponding to 95% CIs. The risk-increasing allele for ALS corresponds to the effect allele for both survival and age-at-onset analyses. Source data
Fig. 3. Shared genetic risk between ALS…
Fig. 3. Shared genetic risk between ALS and neurodegenerative diseases.
a, Genetic correlation analysis. Genetic correlation was estimated with LDSC between each pair of neurodegenerative diseases (ALS, AD, CBD, PD, PSP and FTD). Correlations marked with an asterisk reached nominal statistical significance (PALS,AD = 0.01, PALS,PD = 0.01, PALS,PSP = 0.0001, PPSP,PD = 0.002). b, SNP associations of ALS lead SNPs or LD proxies in neurodegenerative diseases. The association with ALS is shown at the top. Effective sample size is shown on the left. Posterior probabilities of the same causal SNP affecting two diseases were estimated through colocalization analysis and are highlighted as connections. Source data
Fig. 4. Tissue and cell type enrichment…
Fig. 4. Tissue and cell type enrichment analysis.
a, Enrichment of tissues and brain regions included in GTEx version 8 illustrates a brain-specific enrichment pattern in ALS, similar to that in PD but contrasting with that in AD. Tissues and brain regions displayed are those significantly enriched in ALS or PD, tissues previously reported in AD and tissues of specific interest for ALS (spinal cord, tibial nerve and muscle). Color represents the enrichment coefficient, and size indicates two-sided −log10 (P-values) of enrichment obtained by the linear regression model in the MAGMA gene property analysis. b, Cell type enrichment analyses indicate neuron-specific enrichment for glutamatergic neurons. In ALS, no enrichment was found for microglia or other non-neuronal cell types, contrasting with the pattern observed in AD. Color represents the enrichment coefficient, and size indicates two-sided −log10 (P-values) of enrichment obtained by the linear regression model in the MAGMA gene property analysis. Statistically significant enrichments after correction for multiple testing over all tissues (n = 54), cell types (n = 7) and neurons (n = 3) with FDR < 0.05 are marked with an asterisk. Cx, cortex; GABA, γ-aminobutyric acid; OPCs, oligodendrocyte progenitor cells. Source data
Fig. 5. Causal inference of total cholesterol…
Fig. 5. Causal inference of total cholesterol levels and years of schooling in ALS.
a, MR results for ALS and total cholesterol levels. Results for the five different MR methods for two different P-value cutoffs for SNP instrument selection are presented. In total, 83 and 178 SNPs were used as instruments at cutoffs of P < 5 × 10−8 and P < 5 × 10−5, respectively. All methods show a consistent positive effect for an increased risk of ALS with higher total cholesterol levels. There is no evidence for reverse causality. Point estimates for MR are presented with error bars reflecting 95% CIs. b, MR results for ALS and years of schooling. In total, 306 and 681 SNPs were used as instruments at cutoffs of P < 5 × 10−8 and P < 5 × 10−5. Point estimates for MR are presented, with error bars reflecting 95% CIs. Statistically significant effects with a two-sided P-value passing Bonferroni correction for multiple testing over all tested traits (n = 22), instrument P-value cutoffs (n = 2) and MR methods (n = 5) are marked with an asterisk (total cholesterol, Pweighted median = 0.0003 and Pweighted median = 0.0007 for cutoffs at P < 5 × 10−8 and P < 5 × 10−5, respectively; years of schooling, PIVW = 0.0002 at the cutoff of P < 5 × 10−5). Here, SNP outliers were not removed for instrument selection. Z, genetic instrument; bxy, estimated causal effect for an increase of 1 s.d. in genetically predicted exposure. Source data
Extended Data Fig. 1. Manhattan plot in…
Extended Data Fig. 1. Manhattan plot in European ancestries GWAS.
Genome-wide association statistics obtained by inverse-variance weighted meta-analysis of the stratified SAIGE logistic mixed model regression in European ancestry cohorts. Y-axis corresponds to the two-tailed -log10(P-value), x-axis corresponds to the genomic coordinates (GRCh37). Loci containing a genome-wide significant SNP are highlighted in red. SNP IDs are the top associated SNPs in each locus. The dotted horizontal line reflects the threshold for genome-wide significance (P = 5 × 10−8). Source data
Extended Data Fig. 2. Annotation specific heritability…
Extended Data Fig. 2. Annotation specific heritability enrichment.
Enrichment of SNP-based heritability was calculated with LD-score regression. Grey dashed line represents no enrichment (enrichment = 1). Error bars denote standard error of enrichment estimate. Nominal statistically significant enrichment estimates (two-sided P Conserved_LindbladToh P = 6.5 × 10−5, SuperEnhancer_Hnisz P = 0.014, TFBS_ENCODE P = 0.017, H3K4me1_peaks_Trynka P = 0.018, Coding_UCSC P = 0.028, H3K9ac_Trynka P = 0.037). The category Conserved_LindbladToh was significant after Bonferroni correction for multiple testing across all categories (N = 28). Due to the regression framework in LDSC, enrichment estimates < 0 are possible (with large standard errors). Source data
Extended Data Fig. 3. PRS stratified by…
Extended Data Fig. 3. PRS stratified by rare variant carrier status.
Distribution of PRS in controls and ALS patients with or without one or more rare variants in ALS risk genes. There was no statistically significant difference in PRS between ALS patients with and without rare variants in ALS risk genes (labeled as gene_mut or gene_wt respectively). In total, 5,112 ALS patients and 2,132 controls from stratum 6 with whole-genome sequencing data available were included. For SOD1, TARDBP, FUS, NEK1, TBK1, and CFAP410, rare variants were included according to the model that yielded the strongest association in the rare variant burden association analyses. For C9orf72, patients with the pathogenic hexanucleotide repeat expansion were compared to those without the expansion. The ‘any ALS gene’ groups all patients together with a rare variant in any of the ALS risk genes. P-values for difference in PRS were derived by two-tailed logistic regression. The number of ALS patients carrying a rare variant per gene is denoted in the corresponding panel. Intervals for boxplots: center = median, box = lower and upper quartile, hinges = median ± 2 * IQR, IQR = interquartile range. Source data
Extended Data Fig. 4. NEK1 repeat distribution.
Extended Data Fig. 4. NEK1 repeat distribution.
The frequency of STR alleles in ALS cases and controls are shown. A repeat length of 11 and longer was used as the optimal threshold for disease-associated genotype. The P-value was calculated by Firth logistic regression and FDR correction over all possible thresholds. Y-axis shows the allele frequency of repeat lengths. Repeat position on GRCh37, and repeat motif are shown. Source data
Extended Data Fig. 5. Genetic correlations between…
Extended Data Fig. 5. Genetic correlations between brain diseases.
Correlation matrix for genetic correlation estimates obtained from bivariate LD score regression. Colors correspond to genetic correlation estimates. Strongest clusters appear between neurodegenerative diseases and within the psychiatric traits. ALS = amyotrophic lateral sclerosis, FTD = frontotemporal dementia, PSP progressive supranuclear palsy, PD = Parkinson’s disease, CBD = corticobasal degeneration, AD = (clinically diagnosed) Alzheimer’s disease, MS = multiple sclerosis, IS = ischemic stroke (any), ICH = intracerebral hemorrhage, IA = intracranial aneurysm (any), AN = anorexia nervosa, OCD = obsessive compulsive disorder, Anxiety = anxiety disorder (score), PTSD = post-traumatic stress disorder, MDD = major depressive disorder, BIP = bipolar disorder, SCZ = schizophrenia, TS = Tourette’s syndrome, ASD = autism spectrum disorder, ADHD = attention-deficit hyperactivity disorder. Source data
Extended Data Fig. 6. Colocalization signals.
Extended Data Fig. 6. Colocalization signals.
Loci were selected for colocalization analysis when the top associated SNP was associated with any neurodegenerative disease at 5 × 10−5. For ALS, the European ancestries meta-analysis was used. Bayesian posterior probabilities for a shared variant driving risk of both traits (PPH4) are reported below locus names. Colors reflect LD between the variant and top associated SNP. Source data
Extended Data Fig. 7. Colocalization analysis with…
Extended Data Fig. 7. Colocalization analysis with FTD subtypes.
Top associated SNPs in the ALS GWAS were selected for colocalization analysis between ALS and FTD subtypes using COLOC. In the top panel, point height is the two-sided -log10(P-value) of the top-associated SNP in the ALS GWAS. In the bottom panel, association P-values of these SNPs with FTD subtypes are shown by color. The Bayesian posterior probability for a shared causal variant between traits (PPH4) is depicted by a connection between points. Source data
Extended Data Fig. 8. Tissue and cell-type…
Extended Data Fig. 8. Tissue and cell-type enrichment analyses for all brain diseases.
Tissue (a) and cell-type (b) enrichment for all included brain diseases obtained from two-sided MAGMA linear regression. Only brain diseases with exome-wide significant gene-based MAGMA associations (P < 2.7 × 10−6) were suitable for tissue and cell-type enrichment analyses. The color represents enrichment coefficient and size indicates two-sided -log10(P-value) of enrichment obtained by the linear regression model in the MAGMA gene-property analysis. Due to the large number of significant genes in the gene-based MAGMA analyses for schizophrenia, bipolar disorder and multiple sclerosis the enrichment P-values were truncated at P < 1.0 × 10−5. ALS = amyotrophic lateral sclerosis, PD = Parkinson’s disease, AD = Alzheimer’s disease, ADHD = attention-deficit hyperactivity disorder, ASD = autism spectrum disorder, TS = Tourette’s syndrome, SCZ = schizophrenia, BIP = bipolar disorder, MDD = major depressive disorder, PTSD = post-traumatic stress disorder, Anxiety = anxiety disorder (score), AN = anorexia nervosa, IA intracranial aneurysm (any), IS = ischemic stroke, MS = multiple sclerosis, Cx = cortex, OPC = oligodendrocyte progenitor cells. Source data
Extended Data Fig. 9. Cell-type enrichment analysis…
Extended Data Fig. 9. Cell-type enrichment analysis in mice.
Cell-type enrichment analysis using the DropViz single-cell RNA sequencing dataset obtained from mice. Similar to the cell-type enrichment analyses there is neuron-specific enrichment in ALS and Parkinson’s disease. In Alzheimer’s disease microglia are the most enriched cell-types. The color represents enrichment coefficient and size indicates two-sided -log10(P-value) of enrichment obtained by the linear regression model in the MAGMA gene-property analysis. Statistically significant enrichments after correction for multiple testing with a false discovery rate (FDR) < 0.05 are marked with an asterisk. ALS = amyotrophic lateral sclerosis, PD = Parkinson’s disease, AD = Alzheimer’s disease, Cx = cortex. Source data
Extended Data Fig. 10. Human phenotype ontology…
Extended Data Fig. 10. Human phenotype ontology term enrichment.
Downstreamer enrichment analyses were performed using the multi-tissue and brain-specific co-expression matrix to identify co-regulated ALS-genes. The distribution of enrichment statistics (Z-scores) for all Human phenotype ontology (HPO) terms are plotted per HPO parent branch. The multi-tissue analysis indicates enrichment for the neurology parent branch ‘abnormality of the nervous system’ (dark-red), although no term passes the Bonferroni threshold for multiple testing. The brain-specific analysis illustrates stronger enrichment for the neurology parent branch. In total, 58 HPO terms pass the threshold for multiple testing of which 42 are defined within the ‘abnormality of the nervous system’ branch. Source data

References

    1. van Es MA, et al. Amyotrophic lateral sclerosis. Lancet. 2017;390:2084–2098.
    1. Al-Chalabi A, van den Berg LH, Veldink JH. Gene discovery in amyotrophic lateral sclerosis: implications for clinical management. Nat. Rev. Neurol. 2017;13:96–104.
    1. Trabjerg BB, et al. ALS in Danish registries: heritability and links to psychiatric and cardiovascular disorders. Neurol. Genet. 2020;6:e398.
    1. Ryan M, Heverin M, McLaughlin RL, Hardiman O. Lifetime risk and heritability of amyotrophic lateral sclerosis. JAMA Neurol. 2019;76:1367–1374.
    1. Byrne S, Elamin M, Bede P, Hardiman O. Absence of consensus in diagnostic criteria for familial neurodegenerative diseases. J. Neurol. Neurosurg. Psychiatry. 2012;83:365–367.
    1. Cirulli ET, et al. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science. 2015;347:1436–1441.
    1. Freischmidt A, et al. Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia. Nat. Neurosci. 2015;18:631–636.
    1. Kenna KP, et al. NEK1 variants confer susceptibility to amyotrophic lateral sclerosis. Nat. Genet. 2016;48:1037–1042.
    1. Brenner D, et al. NEK1 mutations in familial amyotrophic lateral sclerosis. Brain. 2016;139:e28.
    1. Majounie E, et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol. 2012;11:323–330.
    1. Nicolas A, et al. Genome-wide analyses identify KIF5A as a novel ALS gene. Neuron. 2018;97:1268–1283.
    1. van Es MA, et al. Genome-wide association study identifies 19p13.3 (UNC13A) and 9p21.2 as susceptibility loci for sporadic amyotrophic lateral sclerosis. Nat. Genet. 2009;41:1083–1087.
    1. Laaksovirta H, et al. Chromosome 9p21 in amyotrophic lateral sclerosis in Finland: a genome-wide association study. Lancet Neurol. 2010;9:978–985.
    1. van Rheenen W, et al. Genome-wide association analyses identify new risk variants and the genetic architecture of amyotrophic lateral sclerosis. Nat. Genet. 2016;48:1043–1048.
    1. Benyamin B, et al. Cross-ethnic meta-analysis identifies association of the GPX3–TNIP1 locus with amyotrophic lateral sclerosis. Nat. Commun. 2017;8:611.
    1. Nakamura R, et al. A multi-ethnic meta-analysis identifies novel genes, including ACSL5, associated with amyotrophic lateral sclerosis. Commun. Biol. 2020;3:526.
    1. DeJesus-Hernandez M, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72:245–256.
    1. Renton AE, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS–FTD. Neuron. 2011;72:257–268.
    1. Diekstra FP, et al. C9orf72 and UNC13A are shared risk loci for amyotrophic lateral sclerosis and frontotemporal dementia: a genome-wide meta-analysis. Ann. Neurol. 2014;76:120–133.
    1. Chen JA, et al. Joint genome-wide association study of progressive supranuclear palsy identifies novel susceptibility loci and genetic correlation to neurodegenerative diseases. Mol. Neurodegener. 2018;13:41.
    1. McCarthy S, et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat. Genet. 2016;48:1279–1283.
    1. Iacoangeli A, et al. Genome-wide meta-analysis finds the ACSL5–ZDHHC6 locus is associated with ALS and links weight loss to the disease genetics. Cell Rep. 2020;33:108323.
    1. Võsa U, et al. Large-scale cis- and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat. Genet. 2021;53:1300–1310.
    1. de Klein, N. et al. Brain expression quantitative trait locus and network analysis reveals downstream effects and putative drivers for brain-related diseases. Preprint at bioRxiv10.1101/2021.03.01.433439 (2021).
    1. Pidsley R, et al. Critical evaluation of the Illumina MethylationEPIC BeadChip microarray for whole-genome DNA methylation profiling. Genome Biol. 2016;17:208.
    1. Shireby GL, et al. Recalibrating the epigenetic clock: implications for assessing biological age in the human cortex. Brain. 2020;143:3763–3775.
    1. Hannon E, et al. An integrated genetic–epigenetic analysis of schizophrenia: evidence for co-localization of genetic associations and differential DNA methylation. Genome Biol. 2016;17:176.
    1. Fang X, et al. The NEK1 interactor, C21ORF2, is required for efficient DNA damage repair. Acta Biochim. Biophys. Sin. 2015;47:834–841.
    1. Brown, A.-L. et al. Common ALS/FTD risk variants in UNC13A exacerbate its cryptic splicing and loss upon TDP-43 mislocalization. Preprint at bioRxiv10.1101/2021.04.02.438170 (2021).
    1. Ma, X. R. et al. TDP-43 represses cryptic exon inclusion in FTD/ALS gene UNC13A. Preprint at bioRxiv10.1101/2021.04.02.438213 (2021).
    1. Jansen IE, et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet. 2019;51:404–413.
    1. Leeuw CA, de Mooij JM, Heskes T, Posthuma D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput. Biol. 2015;11:e1004219.
    1. Watanabe K, Taskesen E, van Bochoven A, Posthuma D. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun. 2017;8:1826.
    1. Watanabe K, Umićević Mirkov M, de Leeuw CA, van den Heuvel MP, Posthuma D. Genetic mapping of cell type specificity for complex traits. Nat. Commun. 2019;10:3222.
    1. Deelen P, et al. Improving the diagnostic yield of exome-sequencing by predicting gene–phenotype associations using large-scale gene expression analysis. Nat. Commun. 2019;10:2837.
    1. Hop, P. J. et al. Genome-wide study of DNA methylation in amyotrophic lateral sclerosis identifies differentially methylated loci and implicates metabolic, inflammatory and cholesterol pathways. Preprint at medRxiv10.1101/2021.03.12.21253115 (2021).
    1. Davies NM, Holmes MV, Smith GD. Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. BMJ. 2018;362:k601.
    1. Bowden J, et al. Improving the visualization, interpretation and analysis of two-sample summary data Mendelian randomization via the radial plot and radial regression. Int. J. Epidemiol. 2018;47:1264–1278.
    1. Munafò MR, Tilling K, Taylor AE, Evans DM, Davey Smith G. Collider scope: when selection bias can substantially influence observed associations. Int. J. Epidemiol. 2018;47:226–235.
    1. Watanabe Y, et al. An amyotrophic lateral sclerosis-associated mutant of C21ORF2 is stabilized by NEK1-mediated hyperphosphorylation and the inability to bind FBXO3. iScience. 2020;23:101491.
    1. Wood AR, et al. Defining the role of common variation in the genomic and biological architecture of adult human height. Nat. Genet. 2014;46:1173–1186.
    1. Luo Y, et al. Exploring the genetic architecture of inflammatory bowel disease by whole-genome sequencing identifies association at ADCY7. Nat. Genet. 2017;49:186–192.
    1. Kathiresan S, et al. Six new loci associated with blood low-density lipoprotein cholesterol, high-density lipoprotein cholesterol or triglycerides in humans. Nat. Genet. 2008;40:189–197.
    1. Saez-Atienzar S, et al. Genetic analysis of amyotrophic lateral sclerosis identifies contributing pathways and cell types. Sci. Adv. 2021;7:eabd9036.
    1. Yamanaka K, et al. Mutant SOD1 in cell types other than motor neurons and oligodendrocytes accelerates onset of disease in ALS mice. Proc. Natl Acad. Sci. USA. 2008;105:7594–7599.
    1. Ralph GS, et al. Silencing mutant SOD1 using RNAi protects against neurodegeneration and extends survival in an ALS model. Nat. Med. 2005;11:429–433.
    1. Blokhuis AM, Groen EJN, Koppers M, van den Berg LH, Pasterkamp RJ. Protein aggregation in amyotrophic lateral sclerosis. Acta Neuropathol. 2013;125:777–794.
    1. Seelen M, et al. Prior medical conditions and the risk of amyotrophic lateral sclerosis. J. Neurol. 2014;261:1949–1956.
    1. Bandres-Ciga S, et al. Shared polygenic risk and causal inferences in amyotrophic lateral sclerosis. Ann. Neurol. 2019;85:470–481.
    1. Armon C. Smoking is a cause of ALS. High LDL-cholesterol levels? Unsure. Ann. Neurol. 2019;85:465–469.
    1. Turner MR, Wotton C, Talbot K, Goldacre MJ. Cardiovascular fitness as a risk factor for amyotrophic lateral sclerosis: indirect evidence from record linkage study. J. Neurol. Neurosurg. Psychiatry. 2012;83:395–398.
    1. Singh R, et al. Autophagy regulates lipid metabolism. Nature. 2009;458:1131–1135.
    1. Koga H, Kaushik S, Cuervo AM. Altered lipid content inhibits autophagic vesicular fusion. FASEB J. 2010;24:3052–3065.
    1. Fraldi A, et al. Lysosomal fusion and SNARE function are impaired by cholesterol accumulation in lysosomal storage disorders. EMBO J. 2010;29:3607–3620.
    1. Barbero-Camps E, et al. Cholesterol impairs autophagy-mediated clearance of amyloid β while promoting its secretion. Autophagy. 2018;14:1129–1154.
    1. Brooks, B. R., Miller, R. G., Swash, M. & Munsat, T. L. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Other Motor Neuron Disord. 1, 293–299 (2000).
    1. Zhou W, et al. Efficiently controlling for case–control imbalance and sample relatedness in large-scale genetic association studies. Nat. Genet. 2018;50:1335–1341.
    1. Willer CJ, Li Y, Abecasis GR. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics. 2010;26:2190–2191.
    1. Bulik-Sullivan BK, et al. LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat. Genet. 2015;47:291–295.
    1. Brown, B. C. et al. Transethnic genetic-correlation estimates from summary statistics. Am. J. Hum. Genet. 99, 76–88 (2016).
    1. Yang J, et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 2012;44:369–375.
    1. Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 2011;88:76–82.
    1. Project MinE ALS Sequencing Consortium. Project MinE: study design and pilot analyses of a large-scale whole-genome sequencing study in amyotrophic lateral sclerosis. Eur. J. Hum. Genet. 26, 1537–1546 (2018).
    1. Spek RAAvander, et al. The project MinE databrowser: bringing large-scale whole-genome sequencing in ALS to researchers and the public. Amyotroph. Lateral Scler. Frontotemporal Degener. 2019;20:432–440.
    1. Genovese G, et al. Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia. Nat. Neurosci. 2016;19:1433–1441.
    1. Cingolani P, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff. Fly. 2012;6:80–92.
    1. Vaser R, Adusumalli S, Leng SN, Sikic M, Ng PC. SIFT missense predictions for genomes. Nat. Protoc. 2016;11:1–9.
    1. Adzhubei IA, et al. A method and server for predicting damaging missense mutations. Nat. Methods. 2010;7:248–249.
    1. Chun S, Fay JC. Identification of deleterious mutations within three human genomes. Genome Res. 2009;19:1553–1561.
    1. Schwarz JM, Cooper DN, Schuelke M, Seelow D. MutationTaster2: mutation prediction for the deep-sequencing age. Nat. Methods. 2014;11:361–362.
    1. Reva B, Antipin Y, Sander C. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 2011;39:e118.
    1. Choi Y, Chan AP. PROVEAN web server: a tool to predict the functional effect of amino acid substitutions and indels. Bioinformatics. 2015;31:2745–2747.
    1. Dolzhenko E, et al. Detection of long repeat expansions from PCR-free whole-genome sequence data. Genome Res. 2017;27:1895–1903.
    1. Dolzhenko E, et al. ExpansionHunter Denovo: a computational method for locating known and novel repeat expansions in short-read sequencing data. Genome Biol. 2020;21:102.
    1. Mousavi N, Shleizer-Burko S, Yanicky R, Gymrek M. Profiling the genome-wide landscape of tandem repeat expansions. Nucleic Acids Res. 2019;47:e90.
    1. Wu Y, et al. Integrative analysis of omics summary data reveals putative mechanisms underlying complex traits. Nat. Commun. 2018;9:918.
    1. Zhu Z, et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat. Genet. 2016;48:481–487.
    1. Barbeira AN, et al. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat. Commun. 2018;9:1825.
    1. Gusev A, et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 2016;48:245–252.
    1. Hannon E, et al. Leveraging DNA-methylation quantitative-trait loci to characterize the relationship between methylomic variation, gene expression, and complex traits. Am. J. Hum. Genet. 2018;103:654–665.
    1. Hop PJ, et al. Genome-wide identification of genes regulating DNA methylation using genetic anchors for causal inference. Genome Biol. 2020;21:220.
    1. McLean CY, et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 2010;28:495–501.
    1. Wei T, et al. CpGtools: a Python package for DNA methylation analysis. Bioinformatics. 2021;37:1598–1599.
    1. Zeng J, et al. Signatures of negative selection in the genetic architecture of human complex traits. Nat. Genet. 2018;50:746–753.
    1. Lloyd-Jones LR, et al. Improved polygenic prediction by Bayesian multiple regression on summary statistics. Nat. Commun. 2019;10:5086.
    1. Kunkle BW, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat. Genet. 2019;51:414–430.
    1. Nalls MA, et al. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: a meta-analysis of genome-wide association studies. Lancet Neurol. 2019;18:1091–1102.
    1. Ferrari, R., Hernandez, D. G., Nalls, M. A. & Rohrer, J. D. Frontotemporal dementia and its subtypes: a genome-wide association study. Lancet Neurol. 13, 686–699 (2014).
    1. Kouri N, et al. Genome-wide association study of corticobasal degeneration identifies risk variants shared with progressive supranuclear palsy. Nat. Commun. 2015;6:7247.
    1. Marioni RE, et al. GWAS on family history of Alzheimer’s disease. Transl. Psychiatry. 2018;8:99.
    1. International Multiple Sclerosis Genetics Consortium. Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science. 2019;365:eaav7188.
    1. Malik R, et al. Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes. Nat. Genet. 2018;50:524–537.
    1. Woo D, et al. Meta-analysis of genome-wide association studies identifies 1q22 as a susceptibility locus for intracerebral hemorrhage. Am. J. Hum. Genet. 2014;94:511–521.
    1. Bakker MK, et al. Genome-wide association study of intracranial aneurysms identifies 17 risk loci and genetic overlap with clinical risk factors. Nat. Genet. 2020;52:1303–1313.
    1. Watson HJ, et al. Genome-wide association study identifies eight risk loci and implicates metabo–psychiatric origins for anorexia nervosa. Nat. Genet. 2019;51:1207–1214.
    1. International Obsessive Compulsive Disorder Foundation Genetics Collaborative (IOCDF-GC) and OCD Collaborative Genetics Association Studies (OCGAS). Revealing the complex genetic architecture of obsessive–compulsive disorder using meta-analysis. Mol. Psychiatry23, 1181–1188 (2018).
    1. Otowa T, et al. Meta-analysis of genome-wide association studies of anxiety disorders. Mol. Psychiatry. 2016;21:1391–1399.
    1. Nievergelt CM, et al. International meta-analysis of PTSD genome-wide association studies identifies sex- and ancestry-specific genetic risk loci. Nat. Commun. 2019;10:4558.
    1. Wray NR, et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat. Genet. 2018;50:668–681.
    1. Stahl EA, et al. Genome-wide association study identifies 30 loci associated with bipolar disorder. Nat. Genet. 2019;51:793–803.
    1. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature511, 421–427 (2014).
    1. Yu D, et al. Interrogating the genetic determinants of Tourette’s syndrome and other tic disorders through genome-wide association studies. Am. J. Psychiatry. 2019;176:217–227.
    1. Grove J, et al. Identification of common genetic risk variants for autism spectrum disorder. Nat. Genet. 2019;51:431–444.
    1. Demontis D, et al. Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nat. Genet. 2019;51:63–75.
    1. Giambartolomei C, et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 2014;10:e1004383.
    1. Darmanis S, et al. A survey of human brain transcriptome diversity at the single cell level. Proc. Natl Acad. Sci. USA. 2015;112:7285–7290.
    1. Hodge RD, et al. Conserved cell types with divergent features in human versus mouse cortex. Nature. 2019;573:61–68.
    1. Saunders A, et al. Molecular diversity and specializations among the cells of the adult mouse brain. Cell. 2018;174:1015–1030.
    1. Lamparter D, Marbach D, Rueedi R, Kutalik Z, Bergmann S. Fast and rigorous computation of gene and pathway scores from SNP-based summary statistics. PLoS Comput. Biol. 2016;12:e1004714.
    1. 1000 Genomes Project Consortium et al. A global reference for human genetic variation. Nature526, 68–74 (2015).
    1. Yengo L, et al. Meta-analysis of genome-wide association studies for height and body mass index in ∼700000 individuals of European ancestry. Hum. Mol. Genet. 2018;27:3641–3649.
    1. Lee JJ, et al. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat. Genet. 2018;50:1112–1121.
    1. Liu M, et al. Association studies of up to 1.2 million individuals yield new insights into the genetic etiology of tobacco and alcohol use. Nat. Genet. 2019;51:237–244.
    1. Sudlow C, et al. UK Biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 2015;12:e1001779.
    1. van der Harst P, Verweij N. Identification of 64 novel genetic loci provides an expanded view on the genetic architecture of coronary artery disease. Circ. Res. 2018;122:433–443.
    1. Evangelou E, et al. Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nat. Genet. 2018;50:1412–1425.
    1. Vuckovic D, et al. The polygenic and monogenic basis of blood traits and diseases. Cell. 2020;182:1214–1231.
    1. Ligthart S, et al. Genome analyses of >200,000 individuals identify 58 loci for chronic inflammation and highlight pathways that link inflammation and complex disorders. Am. J. Hum. Genet. 2018;103:691–706.
    1. Willer CJ, et al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 2013;45:1274–1283.
    1. Zeng P, Wang T, Zheng J, Zhou X. Causal association of type 2 diabetes with amyotrophic lateral sclerosis: new evidence from Mendelian randomization using GWAS summary statistics. BMC Med. 2019;17:225.
    1. Cragg JG, Donald SG. Testing identifiability and specification in instrumental variable models. Econ. Theory. 1993;9:222–240.
    1. Hemani, G. et al. The MR-Base platform supports systematic causal inference across the human phenome. eLife7, e34408 (2018).
    1. Smith GD, Davey Smith G, Hemani G. Mendelian randomization: genetic anchors for causal inference in epidemiological studies. Hum. Mol. Genet. 2014;23:R89–R98.
    1. Hemani G, Tilling K, Davey Smith G. Orienting the causal relationship between imprecisely measured traits using GWAS summary data. PLoS Genet. 2017;13:e1007081.
    1. Burgess S, Thompson SG. Multivariable Mendelian randomization: the use of pleiotropic genetic variants to estimate causal effects. Am. J. Epidemiol. 2015;181:251–260.
    1. Sanderson E, Davey Smith G, Windmeijer F, Bowden J. An examination of multivariable Mendelian randomization in the single-sample and two-sample summary data settings. Int. J. Epidemiol. 2019;48:713–727.

Source: PubMed

3
Abonnere