Genome-wide association meta-analysis of spontaneous coronary artery dissection identifies risk variants and genes related to artery integrity and tissue-mediated coagulation

David Adlam, Takiy-Eddine Berrandou, Adrien Georges, Christopher P Nelson, Eleni Giannoulatou, Joséphine Henry, Lijiang Ma, Montgomery Blencowe, Tamiel N Turley, Min-Lee Yang, Sandesh Chopade, Chris Finan, Peter S Braund, Ines Sadeg-Sayoud, Siiri E Iismaa, Matthew L Kosel, Xiang Zhou, Stephen E Hamby, Jenny Cheng, Lu Liu, Ingrid Tarr, David W M Muller, Valentina d'Escamard, Annette King, Liam R Brunham, Ania A Baranowska-Clarke, Stéphanie Debette, Philippe Amouyel, Jeffrey W Olin, Snehal Patil, Stephanie E Hesselson, Keerat Junday, Stavroula Kanoni, Krishna G Aragam, Adam S Butterworth, CARDIoGRAMPlusC4D, MEGASTROKE, International Stroke Genetics Consortium (ISGC) Intracranial Aneurysm Working Group, Marysia S Tweet, Rajiv Gulati, Nicolas Combaret, DISCO register, Daniella Kadian-Dodov, Jonathan M Kalman, Diane Fatkin, Aroon D Hingorani, Jacqueline Saw, Tom R Webb, Sharonne N Hayes, Xia Yang, Santhi K Ganesh, Timothy M Olson, Jason C Kovacic, Robert M Graham, Nilesh J Samani, Nabila Bouatia-Naji, Mark K Bakker, Ynte M Ruigrok, David Adlam, Takiy-Eddine Berrandou, Adrien Georges, Christopher P Nelson, Eleni Giannoulatou, Joséphine Henry, Lijiang Ma, Montgomery Blencowe, Tamiel N Turley, Min-Lee Yang, Sandesh Chopade, Chris Finan, Peter S Braund, Ines Sadeg-Sayoud, Siiri E Iismaa, Matthew L Kosel, Xiang Zhou, Stephen E Hamby, Jenny Cheng, Lu Liu, Ingrid Tarr, David W M Muller, Valentina d'Escamard, Annette King, Liam R Brunham, Ania A Baranowska-Clarke, Stéphanie Debette, Philippe Amouyel, Jeffrey W Olin, Snehal Patil, Stephanie E Hesselson, Keerat Junday, Stavroula Kanoni, Krishna G Aragam, Adam S Butterworth, CARDIoGRAMPlusC4D, MEGASTROKE, International Stroke Genetics Consortium (ISGC) Intracranial Aneurysm Working Group, Marysia S Tweet, Rajiv Gulati, Nicolas Combaret, DISCO register, Daniella Kadian-Dodov, Jonathan M Kalman, Diane Fatkin, Aroon D Hingorani, Jacqueline Saw, Tom R Webb, Sharonne N Hayes, Xia Yang, Santhi K Ganesh, Timothy M Olson, Jason C Kovacic, Robert M Graham, Nilesh J Samani, Nabila Bouatia-Naji, Mark K Bakker, Ynte M Ruigrok

Abstract

Spontaneous coronary artery dissection (SCAD) is an understudied cause of myocardial infarction primarily affecting women. It is not known to what extent SCAD is genetically distinct from other cardiovascular diseases, including atherosclerotic coronary artery disease (CAD). Here we present a genome-wide association meta-analysis (1,917 cases and 9,292 controls) identifying 16 risk loci for SCAD. Integrative functional annotations prioritized genes that are likely to be regulated in vascular smooth muscle cells and artery fibroblasts and implicated in extracellular matrix biology. One locus containing the tissue factor gene F3, which is involved in blood coagulation cascade initiation, appears to be specific for SCAD risk. Several associated variants have diametrically opposite associations with CAD, suggesting that shared biological processes contribute to both diseases, but through different mechanisms. We also infer a causal role for high blood pressure in SCAD. Our findings provide novel pathophysiological insights involving arterial integrity and tissue-mediated coagulation in SCAD and set the stage for future specific therapeutics and preventions.

Conflict of interest statement

D.A. has received kind support from AstraZeneca (for gene sequencing in patients with SCAD) and grant funding from AstraZeneca for unrelated research. He has also received research funding from Abbott Vascular to support a clinical research fellow and has undertaken consultancy for General Electric to support research funds. He holds unrelated patents EP3277337A1 and PCT/GB2017/050877. The remaining authors declare no competing interests.

© 2023. The Author(s).

Figures

Fig. 1. GWAS meta-analysis main association results…
Fig. 1. GWAS meta-analysis main association results and gene prioritization at-risk loci.
a, Manhattan plot representation of SNP-based association meta-analysis in SCAD. The x axis shows the genomic coordinates and the y axis shows the −log10[P value] obtained by two-sided Wald test. SNPs located around genome-wide significant signals (±500 kb) are highlighted. The labels show the rsIDs for the lead SNPs, with newly identified loci in red and previously known loci in black. The dashed red line represents genome-wide significance (P = 5 × 10−8) and the gray line suggestive association (P = 10−5). b, Summary of the strategy for the annotation of gene prioritization. The dots indicate genes fulfilling one of the following eight criteria: (1) colocalization of SCAD association signal and eQTL association in the aorta, coronary artery, tibial artery, fibroblasts or whole blood samples (GTEx version 8 release); (2) a TWAS hit in any of the above-mentioned tissues; (3) a cardiovascular (CV) phenotype in the gene knockout mouse; (4) existing evidence of gene function in cardiovascular disease (CVD) pathophysiology in humans; (5) the gene is an eGene for a nearby lead SNP in the above-mentioned GTEx tissues; (6) Hi-C evidence for a promoter of the gene in a chromatin loop from human aorta tissue that includes variants from the credible set of causal variants; (7) the closest gene upstream or downstream from the lead SNP; or (8) variants in the credible set of causal variants map in the gene. Criteria 1 and 2 (blue dots) were given a tenfold weighted score over criteria 3–8. Genes with the most criteria were prioritized in each locus and are shown here.
Fig. 2. Enrichment of SCAD SNPs in…
Fig. 2. Enrichment of SCAD SNPs in open chromatin regions from arterial cells and genetically predicted expression changes of nearby genes.
a, Top, representation of the fold-enrichment of SCAD SNPs (top y axis) and enrichment P value (log scale; bottom y axis) among the open chromatin regions of seven single-cell subclusters contributing to >1% of cells in artery tissue. The SCAD 95% credible set of causal SNPs and their linkage disequilibrium proxies were matched to random pools of neighboring SNPs using the GREGOR package. Enrichment represents the ratio of the number of SCAD SNPs overlapping open chromatin regions over the average number of matched SNPs overlapping the same regions. P values were evaluated by binomial one-sided test, with greater enrichment as the alternative hypothesis. The bottom dashed line represents significance (P < 0.05) after adjustment for 105 subclusters. Higher opacity is used to identify significant associations (adjusted P < 0.05). Bottom, composition of artery tissues relative to 105 single-cell subclusters, as determined by snATAC-seq in 30 adult tissues. Only subclusters representing >1% of cells from either the aorta or tibial artery were represented. b, Representation of the SCAD TWAS z score for each prioritized gene in GWAS loci. The point shape indicates the tissue used in the TWAS association. The point color distinguishes genes located at different loci. The absence of a symbol indicates that the gene did not show significant heritability based on the eQTL data in the corresponding tissue. TWAS P values were calculated by two-tailed z test against a null distribution calculated by permutation for each gene or tissue. Higher opacity is used to identify significant associations (Bonferroni adjusted P < 0.05), corresponding to a z score of >4.8 or <−4.8 (dashed gray lines).
Fig. 3. Colocalization and genetic correlation of…
Fig. 3. Colocalization and genetic correlation of SCAD genetic association with cardiovascular diseases and traits.
a, Heatmap representing the colocalization of SCAD signals with GWAS analysis of the following cardiovascular diseases or traits: cervical artery dissection (CeAD), multifocal FMD, migraine, blood pressure (SBP and DBP), LDL cholesterol blood concentration, hemoglobin concentration (HGB), any stroke (AS), intracranial aneurysm (IA) and CAD. The tile color represents the H4 coefficient of approximate Bayes factor (ABF) colocalization (that is, the posterior probability of the two traits sharing one causal variant at the locus (PP.H4.ABF; 0–1)) multiplied by the sign of colocalization (+1 if both traits have the same risk or higher mean allele and −1 if opposite allele)). b, Forest plot representing genetic correlations with SCAD. The Rho coefficient of genetic correlation (rg), obtained using linkage disequilibrium score regression, is represented on the x axis (center of the error bar). The range of each bar represents the 95% CI. Unadjusted P values obtained by two-sided Wald test for genetic correlations are indicated. Asterisks indicate significance after Bonferroni correction for testing 26 traits (P < 1.9 × 10−3) (Supplementary Table 10).
Fig. 4. Mendelian randomization associations between main…
Fig. 4. Mendelian randomization associations between main cardiovascular risk factors and SCAD or CAD.
a,b, Forest plots representing Mendelian randomization associations between cardiovascular risk factors and SCAD (ncases = 1,917; ncontrols = 9,292) (a) or CAD (ncases = 181,522; ncontrols = 984,168) (b). Association estimates (β; center of the error bars) obtained from Mendelian randomization analyses using the IVW method are represented on the x axis. The range of each bar represents the 95% CI. Unadjusted P values from the associations obtained by two-sided Wald test are indicated. n = 340,159 (SBP), 340,162 (DBP), 359,983 (BMI), 315,133 (HDL), 343,621 (LDL), 343,992 (triglycerides (TG)), 164,638 cases and 195,068 controls (smoking (SMK)) and 74,124 cases and 824,006 controls (type 2 diabetes (T2D)). The asterisks indicate significance after Bonferroni correction for testing nine traits (P < 5.6 × 10−3) (Supplementary Table 13).

References

    1. Vogel B, et al. The Lancet women and cardiovascular disease Commission: reducing the global burden by 2030. Lancet. 2021;397:2385–2438.
    1. Adlam D, Alfonso F, Maas A, Vrints C, Writing Committee. European Society of Cardiology, acute cardiovascular care association, SCAD study group: a position paper on spontaneous coronary artery dissection. Eur. Heart J. 2018;39:3353–3368.
    1. Collet JP, et al. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Eur. Heart J. 2021;42:1289–1367.
    1. Hayes SN, et al. Spontaneous coronary artery dissection: current state of the science: a scientific statement from the American Heart Association. Circulation. 2018;137:e523–e557.
    1. Hayes SN, et al. Spontaneous coronary artery dissection: JACC state-of-the-art review. J. Am. Coll. Cardiol. 2020;76:961–984.
    1. Ibanez B, et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: the Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC) Eur. Heart J. 2018;39:119–177.
    1. Kim ESH. Spontaneous coronary-artery dissection. N. Engl. J. Med. 2020;383:2358–2370.
    1. Margaritis M, et al. Vascular histopathology and connective tissue ultrastructure in spontaneous coronary artery dissection: pathophysiological and clinical implications. Cardiovasc. Res. 2022;118:1835–1848.
    1. Kok SN, et al. Prevalence and clinical factors of migraine in patients with spontaneous coronary artery dissection. J. Am. Heart Assoc. 2018;7:e010140.
    1. Combaret N, et al. National French registry of spontaneous coronary artery dissections: prevalence of fibromuscular dysplasia and genetic analyses. EuroIntervention. 2021;17:508–515.
    1. Persu A, et al. Prevalence and disease spectrum of extracoronary arterial abnormalities in spontaneous coronary artery dissection. JAMA Cardiol. 2022;7:159–166.
    1. Prasad M, et al. Prevalence of extracoronary vascular abnormalities and fibromuscular dysplasia in patients with spontaneous coronary artery dissection. Am. J. Cardiol. 2015;115:1672–1677.
    1. Saw J, Ricci D, Starovoytov A, Fox R, Buller CE. Spontaneous coronary artery dissection: prevalence of predisposing conditions including fibromuscular dysplasia in a tertiary center cohort. JACC Cardiovasc. Interv. 2013;6:44–52.
    1. Jackson R, et al. Distinct and complementary roles for alpha and beta isoenzymes of PKC in mediating vasoconstrictor responses to acutely elevated glucose. Br. J. Pharmacol. 2016;173:870–887.
    1. Erdmann J, Kessler T, Munoz Venegas L, Schunkert H. A decade of genome-wide association studies for coronary artery disease: the challenges ahead. Cardiovasc. Res. 2018;114:1241–1257.
    1. Amrani-Midoun A, Adlam D, Bouatia-Naji N. Recent advances on the genetics of spontaneous coronary artery dissection. Circ. Genom. Precis. Med. 2021;14:e003393.
    1. Adlam D, et al. Association of the PHACTR1/EDN1 genetic locus with spontaneous coronary artery dissection. J. Am. Coll. Cardiol. 2019;73:58–66.
    1. Saw J, et al. Chromosome 1q21.2 and additional loci influence risk of spontaneous coronary artery dissection and myocardial infarction. Nat. Commun. 2020;11:4432.
    1. Turley TN, et al. Susceptibility locus for pregnancy-associated spontaneous coronary artery dissection. Circ. Genom. Precis. Med. 2021;14:e003398.
    1. Turley TN, et al. Identification of susceptibility loci for spontaneous coronary artery dissection. JAMA Cardiol. 2020;5:929–938.
    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. Speed D, Balding DJ. SumHer better estimates the SNP heritability of complex traits from summary statistics. Nat. Genet. 2019;51:277–284.
    1. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74.
    1. Zhang K, et al. A single-cell atlas of chromatin accessibility in the human genome. Cell. 2021;184:5985–6001.e19.
    1. Jung I, et al. A compendium of promoter-centered long-range chromatin interactions in the human genome. Nat. Genet. 2019;51:1442–1449.
    1. Finan C, et al. The druggable genome and support for target identification and validation in drug development. Sci. Transl. Med. 2017;9:eaag1166.
    1. Bennett BJ, et al. Genetic architecture of atherosclerosis in mice: a systems genetics analysis of common inbred strains. PLoS Genet. 2015;11:e1005711.
    1. GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science. 2020;369:1318–1330.
    1. Zhu J, et al. An integrative genomics approach to the reconstruction of gene networks in segregating populations. Cytogenet. Genome Res. 2004;105:363–374.
    1. Saw J, et al. Canadian spontaneous coronary artery dissection cohort study: in-hospital and 30-day outcomes. Eur. Heart J. 2019;40:1188–1197.
    1. Zhu Z, et al. Causal associations between risk factors and common diseases inferred from GWAS summary data. Nat. Commun. 2018;9:224.
    1. Carss KJ, et al. Spontaneous coronary artery dissection: insights on rare genetic variation from genome sequencing. Circ. Genom. Precis. Med. 2020;13:e003030.
    1. Nakashima T, et al. Prognostic impact of spontaneous coronary artery dissection in young female patients with acute myocardial infarction: a report from the Angina Pectoris-Myocardial Infarction Multicenter Investigators in Japan. Int. J. Cardiol. 2016;207:341–348.
    1. Clare R, et al. Characteristics and clinical outcomes of patients with spontaneous coronary artery dissection. J. Am. Heart Assoc. 2019;8:e012570.
    1. Georges A, et al. Genetic investigation of fibromuscular dysplasia identifies risk loci and shared genetics with common cardiovascular diseases. Nat. Commun. 2021;12:6031.
    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. Kobayashi Y, et al. Mice lacking hypertension candidate gene ATP2B1 in vascular smooth muscle cells show significant blood pressure elevation. Hypertension. 2012;59:854–860.
    1. Saw J, et al. Spontaneous coronary artery dissection: clinical outcomes and risk of recurrence. J. Am. Coll. Cardiol. 2017;70:1148–1158.
    1. Moons AH, Levi M, Peters RJ. Tissue factor and coronary artery disease. Cardiovasc. Res. 2002;53:313–325.
    1. Fan D, Kassiri Z. Biology of tissue inhibitor of metalloproteinase 3 (TIMP3), and its therapeutic implications in cardiovascular pathology. Front. Physiol. 2020;11:661.
    1. Aragam KG, et al. Discovery and systematic characterization of risk variants and genes for coronary artery disease in over a million participants. Nat. Genet. 2022;54:1803–1815.
    1. Steffensen LB, Rasmussen LM. A role for collagen type IV in cardiovascular disease? Am. J. Physiol. Heart Circ. Physiol. 2018;315:H610–H625.
    1. Schmidt EM, et al. GREGOR: evaluating global enrichment of trait-associated variants in epigenomic features using a systematic, data-driven approach. Bioinformatics. 2015;31:2601–2606.
    1. Gusev A, et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 2016;48:245–252.
    1. Loh PR, et al. Reference-based phasing using the Haplotype Reference Consortium panel. Nat. Genet. 2016;48:1443–1448.
    1. Das S, et al. Next-generation genotype imputation service and methods. Nat. Genet. 2016;48:1284–1287.
    1. Chang CC, et al. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience. 2015;4:7.
    1. Willer CJ, Li Y, Abecasis GR. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics. 2010;26:2190–2191.
    1. Myers TA, Chanock SJ, Machiela MJ. LDlinkR: an R package for rapidly calculating linkage disequilibrium statistics in diverse populations. Front. Genet. 2020;11:157.
    1. Davis CA, et al. The Encyclopedia of DNA Elements (ENCODE): data portal update. Nucleic Acids Res. 2018;46:D794–D801.
    1. Miller CL, et al. Integrative functional genomics identifies regulatory mechanisms at coronary artery disease loci. Nat. Commun. 2016;7:12092.
    1. Feng J, Liu T, Qin B, Zhang Y, Liu XS. Identifying ChIP-seq enrichment using MACS. Nat. Protoc. 2012;7:1728–1740.
    1. Afgan E, et al. The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2018 update. Nucleic Acids Res. 2018;46:W537–W544.
    1. Lawrence M, Gentleman R, Carey V. rtracklayer: an R package for interfacing with genome browsers. Bioinformatics. 2009;25:1841–1842.
    1. Freese NH, Norris DC, Loraine AE. Integrated genome browser: visual analytics platform for genomics. Bioinformatics. 2016;32:2089–2095.
    1. Bult CJ, et al. Mouse Genome Database (MGD) 2019. Nucleic Acids Res. 2019;47:D801–D806.
    1. Pinero J, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 2020;48:D845–D855.
    1. Mendez D, et al. ChEMBL: towards direct deposition of bioassay data. Nucleic Acids Res. 2019;47:D930–D940.
    1. Burgess S, Small DS, Thompson SG. A review of instrumental variable estimators for Mendelian randomization. Stat. Methods Med. Res. 2017;26:2333–2355.

Source: PubMed

3
Sottoscrivi