Detailed stratified GWAS analysis for severe COVID-19 in four European populations

Abstract

Given the highly variable clinical phenotype of Coronavirus disease 2019 (COVID-19), a deeper analysis of the host genetic contribution to severe COVID-19 is important to improve our understanding of underlying disease mechanisms. Here, we describe an extended genome-wide association meta-analysis of a well-characterized cohort of 3255 COVID-19 patients with respiratory failure and 12 488 population controls from Italy, Spain, Norway and Germany/Austria, including stratified analyses based on age, sex and disease severity, as well as targeted analyses of chromosome Y haplotypes, the human leukocyte antigen region and the SARS-CoV-2 peptidome. By inversion imputation, we traced a reported association at 17q21.31 to a ~0.9-Mb inversion polymorphism that creates two highly differentiated haplotypes and characterized the potential effects of the inversion in detail. Our data, together with the 5th release of summary statistics from the COVID-19 Host Genetics Initiative including non-Caucasian individuals, also identified a new locus at 19q13.33, including NAPSA, a gene which is expressed primarily in alveolar cells responsible for gas exchange in the lung.

© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.

Figures

Figure 1
Figure 1
Forest plot of candidates from the in-depth stratified analysis. The plots show the variants chr3:45823240, chr3:45848457:C:T, chr17:46142465:T:A which were significantly associated with age (interaction P-value [P_FDR(I) < 0.05]) when comparing age groups ≤60 and >60. We additionally show variants chr19:4717660:A:G and chr21:33242905:T:C with insignificant, though strong trends for association with age. ORs and their respective 95% confidence intervals (CIs) are visualized for each of our four cohorts separately. The size of the dots indicates the size of the respective cohort (N). The OR value is displayed in addition as a numerical value. Only cohorts in which N > 50 in both cases and controls are shown. The headers are built as follows: gene variant id (chr:pos(hg38):allele)—rs-id—effect allele */**. * indicates a variant that was observed as associated from data in this study; **indicates a variant that was observed as associated in the release 5 of the COVID-19 HGI.
Figure 2
Figure 2
Association of the 17q21.31 locus with severe COVID-19 with respiratory failure. (A) Regional association plot showing the variant most strongly associated with severe COVID-19 (rs1819040, purple diamond) a ~ 0.9 Mb inversion polymorphism at 17q21.21 (56) (white line with blue rectangles representing the variable segmental duplication (SD) blocks at the breakpoints), and the large credible set obtained by statistical fine-mapping including 2178 SNPs in high LD (median [LD] = 0.97) with the inversion (Supplementary Material, Table S7). Pairwise LD values (r2) with lead variant rs1819040 were calculated from merged Italian, Spanish, German/Austrian and Norwegian GWAS main anlysis datasets. The dotted line indicates the genome-wide significance threshold (P = 5 × 10−8). Below, organization of the 17q21.31 inversion genomic region, with the extended haplotypes associated with each orientation (H1 and H2) shown as red and blue arrows, respectively, and breakpoint SDs as dark rectangles. Protein-coding genes for which the inversion is a lead eQTL in at least one GTEx tissue are shown as pointed rectangles indicating the direction of transcription. (B) Forest plot and extended meta-analysis of our first discovery analysis and the COVID-19 HGI release 5 analysis B2 dataset (Material and Methods) of the association between severe COVID-19 and the 17q21.31 inversion based on the presence relative to the absence of the inversion haplotype H2. We visualized the ORs and their 95% confidence invervals (CIs) across all analysed cohorts of the main analysis and the COVID-19 HGI release 5 analysis B2 data. In this analysis, the overlap between the main analysis cohort and the COVID-19 HGI data was excluded from the main analysis cohort. The OR value is displayed in addition as a numerical value. The size of the dots indicates the size of the respective cohort (N). (C) Phenome-wide association study (PheWAS) results for the 17q21.31 inversion allele H2 showing only potentially COVID-19-related phenotypes from the GWAS Catalog (P = 10−7) grouped by disease categories using different colors. The effect direction of known SNP-trait associations from the corresponding GWAS is shown using triangles pointing upward (increase) and downward (decrease), whereas dots represent unknown effect direction. Phenotypes shown were selected according to previously reported COVID-19 links with lung damage, blood cell alterations and exacerbated immune response, as well as some potential co-morbidities. The whole list of phenotypic associations is included in Supplementary Material, Table S11.
Figure 3
Figure 3
Expression analysis of the most plausible candidate genes associated with the 17q.21.31 and 19q.13.33 loci in organ tissues and COVID-19 relevant cell types. (A) GTEx tissue-specific expression QTL (eQTL, upper panel) and splicing QTL (sQTL, middle panel) effects of the 17q21.31 and 19q.13.33 loci on selected candidate genes as well as expression of these genes in GTEx (20) tissues (lower panel). The direction of the normalized eQTL and sQTL effect size (NES) of the lead SNP rs1405655 and the inversion tagger rs62055540 in perfect LD with the inversion is represented by color intensities, and statistical significance by dot size. Black rectangles indicate genes for which the expression colocalizes (regional probability > 0.9) with GWAS loci in a given human tissue from the GTEx dataset. Heatmap displays gene-wise centered median by tissue expression values (represented by color intensities), showing in which tissues candidate genes are mostly enriched. (B) Expression levels of candidate genes in scRNA-seq datasets from healthy upper airways (nasal, bronchi) and lung (parenchyma) cells (47) and adult human brain cells from recently deceased, non-diseased donors (48). Figure displays log-normalized mean expression (represented by color) and fraction of cells expressing those genes (indicated by dot size). Processed and cell-type-annotated gene expression levels from studies were retrieved from COVID-19 Cell Atlas (49). (C) The figure shows differential expression of candidate genes in lung cells of COVID-19 patients compared with healthy controls. Log2 fold change (log2FC) values are presented as color gradient. Nominal P-values in −log10 scale are shown proportionally to dot size. Black-bordered circles indicate significantly differentially expressed genes after FDR correction. Results were obtained from pseudo-bulk differential expression analysis by Delorey et al. (21). More detailed figures are shown in Supplementary Material, Figures S11, S12 and S15.

References

    1. Booth, A., Reed, A.B., Ponzo, S., Yassaee, A., Aral, M., Plans, D., Labrique, A. and Mohan, D. (2021) Population risk factors for severe disease and mortality in COVID-19: a global systematic review and meta-analysis. PLoS One, 16, e0247461.
    1. Wong, C.K.H., Wong, J.Y.H., Tang, E.H.M., Au, C.H. and Wai, A.K.C. (2020) Clinical presentations, laboratory and radiological findings, and treatments for 11,028 COVID-19 patients: a systematic review and meta-analysis. Sci. Rep., 10, 19765.
    1. Peckham, H., de Gruijter, N.M., Raine, C., Radziszewska, A., Ciurtin, C., Wedderburn, L.R., Rosser, E.C., Webb, K. and Deakin, C.T. (2020) Male sex identified by global COVID-19 meta-analysis as a risk factor for death and ITU admission. Nat. Commun., 11, 6317.
    1. Hu, Y., Sun, J., Dai, Z., Deng, H., Li, X., Huang, Q., Wu, Y., Sun, L. and Xu, Y. (2020) Prevalence and severity of corona virus disease 2019 (COVID-19): a systematic review and meta-analysis. J. Clin. Virol., 127, 104371.
    1. Severe Covid-19 GWAS Group (2020) Genomewide association study of severe Covid-19 with respiratory failure. N. Engl. J. Med., 383, 1522–1534.
    1. Pairo-Castineira, E., Clohisey, S., Klaric, L., Bretherick, A.D., Rawlik, K., Pasko, D., Walker, S., Parkinson, N., Fourman, M.H., Russell, C.D. et al. (2021) Genetic mechanisms of critical illness in COVID-19. Nature, 591, 92–98.
    1. COVID-19 Host Genetics Initiative, C (2021) Mapping the human genetic architecture of COVID-19. Nature, 600, 472–477.
    1. Velavan, T.P., Pallerla, S.R., Rüter, J., Augustin, Y., Kremsner, P.G., Krishna, S. and Meyer, C.G. (2021) Host genetic factors determining COVID-19 susceptibility and severity. EBioMedicine, 72, 103629.
    1. Namkoong, H., Edahiro, R., Fukunaga, K., Shirai, Y., Sonehara, K., Tanaka, H., Lee, H., Hasegawa, T., Kanai, M., Naito, T. et al. (2021) Japan COVID-19 task force: a nation-wide consortium to elucidate host genetics of COVID-19 pandemic in Japan. medRxiv. 2021.05.17.21256513; 10.1101/2021.05.17.2125651.
    1. Li, Y., Ke, Y., Xia, X., Wang, Y., Cheng, F., Liu, X., Jin, X., Li, B., Xie, C., Liu, S. et al. (2021) Genome-wide association study of COVID-19 severity among the Chinese population. Cell Discov., 7, 76.
    1. Arora, J., McLaren, P.J., Chaturvedi, N., Carrington, M., Fellay, J. and Lenz, T.L. (2019) HIV peptidome-wide association study reveals patient-specific epitope repertoires associated with HIV control. Proc. Natl. Acad. Sci. U. S. A., 116, 944–949.
    1. Zhou, W., Nielsen, J.B., Fritsche, L.G., Dey, R., Gabrielsen, M.E., Wolford, B.N., LeFaive, J., VandeHaar, P., Gagliano, S.A., Gifford, A. et al. (2018) Efficiently controlling for case-control imbalance and sample relatedness in large-scale genetic association studies. Nat. Genet., 50, 1335–1341.
    1. Willer, C.J., Li, Y. and Abecasis, G.R. (2010) METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics, 26, 2190–2191.
    1. McGuire, D., Jiang, Y., Liu, M., Weissenkampen, J.D., Eckert, S., Yang, L., Chen, F., Liu, M., Jiang, Y., Wedow, R. et al. (2021) Model-based assessment of replicability for genome-wide association meta-analysis. Nat. Commun., 12, 1964.
    1. Benner, C., Spencer, C.C.A., Havulinna, A.S., Salomaa, V., Ripatti, S. and Pirinen, M. (2016) FINEMAP: efficient variable selection using summary data from genome-wide association studies. Bioinformatics, 32, 1493–1501.
    1. Nakanishi, T., Pigazzini, S., Degenhardt, F., Cordioli, M., Butler-Laporte, G., Maya-Miles, D., Nafría-Jiménez, B., Bouysran, Y., Niemi, M., Palom, A. et al. (2021) Age-dependent impact of the major common genetic risk factor for COVID-19 on severity and mortality. J. Clin. Invest., 131, e152386.
    1. Stefansson, H., Helgason, A., Thorleifsson, G., Steinthorsdottir, V., Masson, G., Barnard, J., Baker, A., Jonasdottir, A., Ingason, A., Gudnadottir, V.G. et al. (2005) A common inversion under selection in Europeans. Nat. Genet., 37, 129–137.
    1. Howie, B., Marchini, J. and Stephens, M. (2011) Genotype imputation with thousands of genomes. G3, 1, 457–470.
    1. MacArthur, J., Bowler, E., Cerezo, M., Gil, L., Hall, P., Hastings, E., Junkins, H., McMahon, A., Milano, A., Morales, J. et al. (2017) The new NHGRI-EBI catalog of published genome-wide association studies (GWAS catalog). Nucleic Acids Res., 45, D896–D901.
    1. Delorey, T.M., Ziegler, C.G.K., Heimberg, G., Normand, R., Yang, Y., Segerstolpe, Å., Abbondanza, D., Fleming, S.J., Subramanian, A., Montoro, D.T. et al. (2021) COVID-19 tissue atlases reveal SARS-CoV-2 pathology and cellular targets. Nature, 595, 107–113.
    1. Quach, H., Rotival, M., Pothlichet, J., Loh, Y.H.E., Dannemann, M., Zidane, N., Laval, G., Patin, E., Harmant, C., Lopez, M. et al. (2016) Genetic adaptation and Neandertal admixture shaped the immune system of human populations. Cell, 167, 643–656.e17.
    1. Mankelow, T.J., Singleton, B.K., Moura, P.L., Stevens-Hernandez, C.J., Cogan, N.M., Gyorffy, G., Kupzig, S., Nichols, L., Asby, C., Pooley, J. et al. (2021) Blood group type a secretors are associated with a higher risk of COVID-19 cardiovascular disease complications. eJHaem, 2, 175–187.
    1. Hammer, M. (2002) A nomenclature system for the tree of human Y-chromosomal binary haplogroups. Genome Res., 12, 339–348.
    1. Jiang, X., Holmes, C. and McVean, G. (2021) The impact of age on genetic risk for common diseases. PLoS Genet., 17, e1009723.
    1. Ward, H.E. and Nicholas, T.E. (1984) Alveolar type I and type II cells. Aust. NZ J. Med., 14, 731–734.
    1. Melms, J.C., Biermann, J., Huang, H., Wang, Y., Nair, A., Tagore, S., Katsyv, I., Rendeiro, A.F., Amin, A.D., Schapiro, D. et al. (2021) A molecular single-cell lung atlas of lethal COVID-19. Nature, 595, 114–119.
    1. Islam, A.B.M.M.K. and Khan, M.A.A.K. (2020) Lung transcriptome of a COVID-19 patient and systems biology predictions suggest impaired surfactant production which may be druggable by surfactant therapy. Sci. Rep., 10, 19395.
    1. Wu, L., Zhu, J., Liu, D., Sun, Y. and Wu, C. (2021) An integrative multiomics analysis identifies putative causal genes for COVID-19 severity. Genet. Med., 23, 2076–2086.
    1. American Journal of Respiratory and Critical Care Medicine; 203, 2021. This seems to be from an online abstract submission. 203.1_MeetingAbstracts.A3765.
    1. Astle, W.J., Elding, H., Jiang, T., Allen, D., Ruklisa, D., Mann, A.L., Mead, D., Bouman, H., Riveros-Mckay, F., Kostadima, M.A. et al. (2016) The allelic landscape of human blood cell trait variation and links to common complex disease. Cell, 167, 1415–1429.e19.
    1. Sakornsakolpat, P., Prokopenko, D., Lamontagne, M., Reeve, N.F., Guyatt, A.L., Jackson, V.E., Shrine, N., Qiao, D., Bartz, T.M., Kim, D.K. et al. (2019) Genetic landscape of chronic obstructive pulmonary disease identifies heterogeneous cell-type and phenotype associations. Nat. Genet., 51, 494–505.
    1. Tantisira, K.G., Lazarus, R., Litonjua, A.A., Klanderman, B. and Weiss, S.T. (2008) Chromosome 17: association of a large inversion polymorphism with corticosteroid response in asthma. Pharmacogenet. Genomics, 18, 733–737.
    1. den Dekker, A.D., Davis, F.M., Joshi, A.D., Wolf, S.J., Allen, R., Lipinski, J., Nguyen, B., Kirma, J., Nycz, D., Bermick, J. et al. (2020) TNF-α regulates diabetic macrophage function through the histone acetyltransferase MOF. JCI Insight, 5, e132306.
    1. Cursano, S., Battaglia, C.R., Urrutia-Ruiz, C., Grabrucker, S., Schön, M., Bockmann, J., Braumüller, S., Radermacher, P., Roselli, F., Huber-Lang, M. et al. (2020) A CRHR1 antagonist prevents synaptic loss and memory deficits in a trauma-induced delirium-like syndrome. Mol. Psychiatry., 26, 3778–3794.
    1. Thompson, S.B., Sandor, A.M., Lui, V., Chung, J.W., Waldman, M.M., Long, R.A., Estin, M.L., Matsuda, J.L., Friedman, R.S. and Jacobelli, J. (2020) Formin-like 1 mediates effector t cell trafficking to inflammatory sites to enable t cell-mediated autoimmunity. elife, 9. 10.7554/eLife.58046.
    1. Pickett, B.E., Greer, D.S., Zhang, Y., Stewart, L., Zhou, L., Sun, G., Gu, Z., Kumar, S., Zaremba, S., Larsen, C.N. et al. (2012) Virus pathogen database and analysis resource (ViPR): a comprehensive bioinformatics database and analysis resource for the coronavirus research community. Viruses, 4, 3209–3226.
    1. Kirkpatrick, M. (2010) How and why chromosome inversions evolve. PLoS Biol., 8, e1000501.
    1. Wellenreuther, M. and Bernatchez, L. (2018) Eco-evolutionary genomics of chromosomal inversions. Trends Ecol. Evol., 33, 427–440.
    1. Ben Shachar, S., Barda, N., Manor, S., Israeli, S., Dagan, N., Carmi, S., Balicer, R., Zisser, B. and Louzoun, Y. (2021) MHC Haplotyping of SARS-CoV-2 patients: HLA subtypes are not associated with the presence and severity of COVID-19 in the Israeli population. J. Clin. Immunol., 41, 1154–1161.
    1. Purcell, S. PLINK 1.9. PLINK 1.9. .
    1. Purcell, S., Neale, B., Todd-Brown, K., Thomas, L., Ferreira, M.A., Bender, D., Maller, J., Sklar, P., de Bakker, P.I., Daly, M.J. and Sham, P.C. (2007) PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet., 81, 559–575.
    1. Price, A.L., Weale, M.E., Patterson, N., Myers, S.R., Need, A.C., Shianna, K.V., Ge, D., Rotter, J.I., Torres, E., Taylor, K.D.D. et al. (2008) Long-range LD can confound genome scans in admixed populations. Am. J. Hum. Genet., 83, 132–135.
    1. Auton, A., Brooks, L.D., Durbin, R.M., Garrison, E.P., Kang, H.M., Korbel, J.O., Marchini, J.L., McCarthy, S., McVean, G.A. and Abecasis, G.R. (2015) A global reference for human genetic variation. Nature, 526, 68–74.
    1. Abraham, G., Qiu, Y. and Inouye, M. (2017) FlashPCA2: principal component analysis of biobank-scale genotype datasets. Bioinformatics, 33, 2776–2778.
    1. Taliun, D., Harris, D.N., Kessler, M.D., Carlson, J., Szpiech, Z.A., Torres, R., Taliun, S.A.G., Corvelo, A., Gogarten, S.M., Kang, H.M. et al. (2021) Sequencing of 53,831 diverse genomes from the NHLBI TOPMed program. Nature, 590, 290–299.
    1. König, I.R., Loley, C., Erdmann, J. and Ziegler, A. (2014) How to include chromosome X in your genome-wide association study. Genet. Epidemiol., 38, 97–103.
    1. Viechtbauer, W. (2010) Conducting meta-analyses in R with the metafor. J. Stat. Softw., 36, 1–48.
    1. Antonacci, F., Kidd, J.M., Marques-Bonet, T., Ventura, M., Siswara, P., Jiang, Z. and Eichler, E.E. (2009) Characterization of six human disease-associated inversion polymorphisms. Hum. Mol. Genet., 18, 2555–2566.
    1. Steinberg, K.M., Antonacci, F., Sudmant, P.H., Kidd, J.M., Campbell, C.D., Vives, L., Malig, M., Scheinfeldt, L., Beggs, W., Ibrahim, M. et al. (2012) Structural diversity and African origin of the 17q21.31 inversion polymorphism. Nat. Genet., 44, 872–880.
    1. Puig, M., Lerga-Jaso, J., Giner-Delgado, C., Pacheco, S., Izquierdo, D., Delprat, A., Gayà-Vidal, M., Regan, J.F., Karlin-Neumann, G. and Cáceres, M. (2020) Determining the impact of uncharacterized inversions in the human genome by droplet digital PCR. Genome Res., 30, 724–735.
    1. Kwan, J.S.H., Li, M.X., Deng, J.E. and Sham, P.C. (2016) FAPI: fast and accurate P-value imputation for genome-wide association study. Eur. J. Hum. Genet., 24, 761–766.
    1. Foley, C.N., Staley, J.R., Breen, P.G., Sun, B.B., Kirk, P.D.W., Burgess, S. and Howson, J.M.M. (2021) A fast and efficient colocalization algorithm for identifying shared genetic risk factors across multiple traits. Nat. Commun., 12, 764.
    1. Zhang, T., Klein, A., Sang, J., Choi, J. and Brown, K.M. (2022) ezQTL: a web platform for interactive visualization and colocalization of quantitative trait loci and GWAS. Genomics Proteomics Bioinformatics. 25:S1672-0229(22)00069-9. doi: 10.1016/j.gpb.2022.05.004. Epub ahead of print. PMID: 35643189.
    1. Martínez-Fundichely, A., Casillas, S., Egea, R., Ràmia, M., Barbadilla, A., Pantano, L., Puig, M. and Cáceres, M. (2014) InvFEST, a database integrating information of polymorphic inversions in the human genome. Nucleic Acids Res., 42, D1027–D1032.
    1. Aguet, F., Barbeira, A.N., Bonazzola, R., Brown, A., Castel, S.E., Jo, B., Kasela, S., Kim-Hellmuth, S., Liang, Y., Oliva, M. et al. (2020) The GTEx consortium atlas of genetic regulatory effects across human tissues. Science (80-.), 369, 1318–1330.
    1. Frankish, A., Diekhans, M., Ferreira, A.M., Johnson, R., Jungreis, I., Loveland, J., Mudge, J.M., Sisu, C., Wright, J., Armstrong, J. et al. (2019) GENCODE reference annotation for the human and mouse genomes. Nucleic Acids Res., 47, D766–D773.
    1. Papatheodorou, I., Moreno, P., Manning, J., Fuentes, A.M.P., George, N., Fexova, S., Fonseca, N.A., Füllgrabe, A., Green, M., Huang, N. et al. (2020) Expression atlas update: from tissues to single cells. Nucleic Acids Res., 48, D77–D83.
    1. Stunnenberg, H.G., Abrignani, S., Adams, D., de Almeida, M., Altucci, L., Amin, V., Amit, I., Antonarakis, S.E., Aparicio, S., Arima, T. et al. (2016) The international human epigenome consortium: a Blueprint for scientific collaboration and discovery. Cell, 167, 1145–1149.
    1. Vieira Braga, F.A., Kar, G., Berg, M., Carpaij, O.A., Polanski, K., Simon, L.M., Brouwer, S., Gomes, T., Hesse, L., Jiang, J. et al. (2019) A cellular census of human lungs identifies novel cell states in health and in asthma. Nat. Med., 25, 1153–1163.
    1. Habib, N., Avraham-Davidi, I., Basu, A., Burks, T., Shekhar, K., Hofree, M., Choudhury, S.R., Aguet, F., Gelfand, E., Ardlie, K. et al. (2017) Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods, 14, 955–958.
    1. Sungnak, W., Huang, N., Bécavin, C., Berg, M., Queen, R., Litvinukova, M., Talavera-López, C., Maatz, H., Reichart, D., Sampaziotis, F. et al. (2020) SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat. Med., 26, 681–687.
    1. Wolf, F.A., Angerer, P. and Theis, F.J. (2018) SCANPY: large-scale single-cell gene expression data analysis. Genome Biol., 19, 15.
    1. Song, E., Zhang, C., Israelow, B., Lu-Culligan, A., Prado, A.V., Skriabine, S., Lu, P., El Weizman, O., Liu, F., Dai, Y. et al. (2021) Neuroinvasion of SARS-CoV-2 in human and mouse brain. J. Exp. Med., 218, e20202135. doi: 10.1084/jem.20202135.
    1. Finak, G., McDavid, A., Yajima, M., Deng, J., Gersuk, V., Shalek, A.K., Slichter, C.K., Miller, H.W., McElrath, M.J., Prlic, M., Linsley, P.S. and Gottardo, R. (2015) MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol., 16, 278.
    1. Degenhardt, F., Wendorff, M., Wittig, M., Ellinghaus, E., Datta, L.W., Schembri, J., Ng, S.C., Rosati, E., Hübenthal, M., Ellinghaus, D. et al. (2019) Construction and benchmarking of a multi-ethnic reference panel for the imputation of HLA class I and II alleles. Hum. Mol. Genet., 28, 2078–2092.
    1. Degenhardt, F., Mayr, G., Wendorff, M., Boucher, G., Ellinghaus, E., Ellinghaus, D., ElAbd, H., Rosati, E., Hübenthal, M., Juzenas, S. et al. (2021) Trans-ethnic analysis of the human leukocyte antigen region for ulcerative colitis reveals common disease signatures. Hum. Mol. Genet., 30, 356–369.

Source: PubMed

3
Abonner