Genetic Landscape of the ACE2 Coronavirus Receptor

Background: SARS-CoV-2, the causal agent of COVID-19, enters human cells using the ACE2 (angiotensin-converting enzyme 2) protein as a receptor. ACE2 is thus key to the infection and treatment of the coronavirus. ACE2 is highly expressed in the heart and respiratory and gastrointestinal tracts, playing important regulatory roles in the cardiovascular and other biological systems. However, the genetic basis of the ACE2 protein levels is not well understood. Methods: We have conducted the largest genome-wide association meta-analysis of plasma ACE2 levels in >28 000 individuals of the SCALLOP Consortium (Systematic and Combined Analysis of Olink Proteins). We summarize the cross-sectional epidemiological correlates of circulating ACE2. Using the summary statistics–based high-definition likelihood method, we estimate relevant genetic correlations with cardiometabolic phenotypes, COVID-19, and other human complex traits and diseases. We perform causal inference of soluble ACE2 on vascular disease outcomes and COVID-19 severity using mendelian randomization. We also perform in silico functional analysis by integrating with other types of omics data. Results: We identified 10 loci, including 8 novel, capturing 30% of the heritability of the protein. We detected that plasma ACE2 was genetically correlated with vascular diseases, severe COVID-19, and a wide range of human complex diseases and medications. An X-chromosome cis–protein quantitative trait loci–based mendelian randomization analysis suggested a causal effect of elevated ACE2 levels on COVID-19 severity (odds ratio, 1.63 [95% CI, 1.10–2.42]; P=0.01), hospitalization (odds ratio, 1.52 [95% CI, 1.05–2.21]; P=0.03), and infection (odds ratio, 1.60 [95% CI, 1.08–2.37]; P=0.02). Tissue- and cell type–specific transcriptomic and epigenomic analysis revealed that the ACE2 regulatory variants were enriched for DNA methylation sites in blood immune cells. Conclusions: Human plasma ACE2 shares a genetic basis with cardiovascular disease, COVID-19, and other related diseases. The genetic architecture of the ACE2 protein is mapped, providing a useful resource for further biological and clinical studies on this coronavirus receptor.

[1]  Bjarni V. Halldórsson,et al.  Large-scale integration of the plasma proteome with genetics and disease , 2021, Nature Genetics.

[2]  I. Grundberg,et al.  Proximity Extension Assay in Combination with Next-Generation Sequencing for High-throughput Proteome-wide Analysis , 2021, Molecular & cellular proteomics : MCP.

[3]  P. Marques,et al.  Known Mutations at the Cause of Alpha-1 Antitrypsin Deficiency an Updated Overview of SERPINA1 Variation Spectrum , 2021, The application of clinical genetics.

[4]  K. To,et al.  Soluble ACE2-mediated cell entry of SARS-CoV-2 via interaction with proteins related to the renin-angiotensin system , 2021, Cell.

[5]  C. Mueller,et al.  Influence of renin‐angiotensin‐aldosterone system inhibitors on plasma levels of angiotensin‐converting enzyme 2 , 2021, ESC heart failure.

[6]  J. Marchini,et al.  Genome-wide analysis in 756,646 individuals provides first genetic evidence that ACE2 expression influences COVID-19 risk and yields genetic risk scores predictive of severe disease , 2020, medRxiv.

[7]  Grant D. Huang,et al.  Actionable druggable genome-wide Mendelian randomization identifies repurposing opportunities for COVID-19 , 2020, Nature Medicine.

[8]  S. Yusuf,et al.  Plasma ACE2 and risk of death or cardiometabolic diseases: a case-cohort analysis , 2020, The Lancet.

[9]  D. Seiffert,et al.  Clinical and Proteomic Correlates of Plasma ACE2 (Angiotensin-Converting Enzyme 2) in Human Heart Failure , 2020, Hypertension.

[10]  S. Yusuf,et al.  Angiotensin-converting enzyme 2 (ACE2) levels in relation to risk factors for COVID-19 in two large cohorts of patients with atrial fibrillation , 2020, European heart journal.

[11]  Barbara B. Shih,et al.  Genetic mechanisms of critical illness in COVID-19 , 2020, Nature.

[12]  D. Gurwitz,et al.  Ethnic differences in alpha‐1 antitrypsin deficiency allele frequencies may partially explain national differences in COVID‐19 fatality rates , 2020, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[13]  N. Samani,et al.  Genetic Associations With Plasma Angiotensin Converting Enzyme 2 Concentration , 2020, Circulation.

[14]  S. Vermeire,et al.  Intestinal Receptor of SARS-CoV-2 in Inflamed IBD Tissue Seems Downregulated by HNF4A in Ileum and Upregulated by Interferon Regulating Factors in Colon , 2020, Journal of Crohn's & colitis.

[15]  E. Argañaraz,et al.  Alpha‐1‐antitrypsin: A possible host protective factor against Covid‐19 , 2020, Reviews in medical virology.

[16]  A. Magis,et al.  Plasma levels of soluble ACE2are associated with sex, Metabolic Syndrome, and its biomarkers in a large cohort, pointing to a possible mechanism for increased severity in COVID-19 , 2020, Critical Care.

[17]  A. d’Arminio Monforte,et al.  ACE2 gene variants may underlie interindividual variability and susceptibility to COVID-19 in the Italian population , 2020, European Journal of Human Genetics.

[18]  Geng Chen,et al.  Systematic profiling of ACE2 expression in diverse physiological and pathological conditions for COVID‐19/SARS‐CoV‐2 , 2020, Journal of cellular and molecular medicine.

[19]  Y. Pawitan,et al.  High-definition likelihood inference of genetic correlations across human complex traits , 2020, Nature Genetics.

[20]  M. Luizon,et al.  Functional prediction and frequency of coding variants in human ACE2 at binding sites with SARS‐CoV‐2 spike protein on different populations , 2020, Journal of medical virology.

[21]  M. Karlsson,et al.  Age and sex differences in soluble ACE2 may give insights for COVID-19 , 2020, Critical Care.

[22]  The COVID-19 Host Genetics Initiative The COVID-19 Host Genetics Initiative, a global initiative to elucidate the role of host genetic factors in susceptibility and severity of the SARS-CoV-2 virus pandemic , 2020, European Journal of Human Genetics.

[23]  P. Ponikowski,et al.  Circulating plasma concentrations of angiotensin-converting enzyme 2 in men and women with heart failure and effects of renin–angiotensin–aldosterone inhibitors , 2020, European heart journal.

[24]  Fang Li,et al.  Cell entry mechanisms of SARS-CoV-2 , 2020, Proceedings of the National Academy of Sciences.

[25]  Daniel Wrapp,et al.  Site-specific glycan analysis of the SARS-CoV-2 spike , 2020, Science.

[26]  Fabian J Theis,et al.  SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes , 2020, Nature Medicine.

[27]  Martin Stahl,et al.  Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2 , 2020, Cell.

[28]  K. Yuen,et al.  Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2 , 2020, Cell.

[29]  Mushtaq Hussain,et al.  Structural variations in human ACE2 may influence its binding with SARS‐CoV‐2 spike protein , 2020, Journal of medical virology.

[30]  G. Herrler,et al.  SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor , 2020, Cell.

[31]  Andrea Marzi,et al.  Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses , 2020, Nature Microbiology.

[32]  S. Yusuf,et al.  ACE and Type 2 Diabetes Risk: A Mendelian Randomization Study , 2020, Diabetes Care.

[33]  J. Luban SARS-CoV-2 , 2020 .

[34]  Ceshi Chen,et al.  The roles of TNFAIP2 in cancers and infectious diseases , 2018, Journal of cellular and molecular medicine.

[35]  Valeriia Haberland,et al.  The MR-Base platform supports systematic causal inference across the human phenome , 2018, eLife.

[36]  Jimok Yoon,et al.  The MICALs are a Family of F-actin Dismantling Oxidoreductases Conserved from Drosophila to Humans , 2018, Scientific Reports.

[37]  Erdogan Taskesen,et al.  Functional mapping and annotation of genetic associations with FUMA , 2017, Nature Communications.

[38]  J. Danesh,et al.  Efficiency and safety of varying the frequency of whole blood donation (INTERVAL): a randomised trial of 45 000 donors , 2017, The Lancet.

[39]  Robert M. Maier,et al.  Causal associations between risk factors and common diseases inferred from GWAS summary data , 2017, Nature Communications.

[40]  R. McPherson,et al.  TRIB1 is a positive regulator of hepatocyte nuclear factor 4-alpha , 2017, Scientific Reports.

[41]  K. Burns,et al.  Measurement of Angiotensin Converting Enzyme 2 Activity in Biological Fluid (ACE2) , 2017, Methods in molecular biology.

[42]  Stephen Burgess,et al.  PhenoScanner: a database of human genotype–phenotype associations , 2016, Bioinform..

[43]  Tom R. Gaunt,et al.  LD Hub: a centralized database and web interface to perform LD score regression that maximizes the potential of summary level GWAS data for SNP heritability and genetic correlation analysis , 2016, bioRxiv.

[44]  D. Rader,et al.  Tribbles-1 regulates hepatic lipogenesis through posttranscriptional regulation of C/EBPα. , 2015, The Journal of clinical investigation.

[45]  Michael Q. Zhang,et al.  Integrative analysis of 111 reference human epigenomes , 2015, Nature.

[46]  Douglas J. Kojetin,et al.  REV-ERB and ROR nuclear receptors as drug targets , 2014, Nature Reviews Drug Discovery.

[47]  O. Jahn,et al.  TMPRSS2 and ADAM17 Cleave ACE2 Differentially and Only Proteolysis by TMPRSS2 Augments Entry Driven by the Severe Acute Respiratory Syndrome Coronavirus Spike Protein , 2013, Journal of Virology.

[48]  Kavaljit H. Chhabra,et al.  The transcription factor HNF1α induces expression of angiotensin-converting enzyme 2 (ACE2) in pancreatic islets from evolutionarily conserved promoter motifs. , 2013, Biochimica et biophysica acta.

[49]  R. Stockley,et al.  Recent advances in α-1-antitrypsin deficiency-related lung disease , 2013, Expert review of respiratory medicine.

[50]  P. Visscher,et al.  Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits , 2012, Nature Genetics.

[51]  Wei Wang,et al.  Genomics Meets Glycomics—The First GWAS Study of Human N-Glycome Identifies HNF1α as a Master Regulator of Plasma Protein Fucosylation , 2010, PLoS genetics.

[52]  Yun Li,et al.  METAL: fast and efficient meta-analysis of genomewide association scans , 2010, Bioinform..

[53]  N. Hooper,et al.  Circulating Activities of Angiotensin-Converting Enzyme, Its Homolog, Angiotensin-Converting Enzyme 2, and Neprilysin in a Family Study , 2006, Hypertension.

[54]  N. Hooper,et al.  Tumor Necrosis Factor-α Convertase (ADAM17) Mediates Regulated Ectodomain Shedding of the Severe-acute Respiratory Syndrome-Coronavirus (SARS-CoV) Receptor, Angiotensin-converting Enzyme-2 (ACE2) , 2005, Journal of Biological Chemistry.

[55]  D. Averill,et al.  Upregulation of Angiotensin-Converting Enzyme 2 After Myocardial Infarction by Blockade of Angiotensin II Receptors , 2004, Hypertension.

[56]  Nigel M Hooper,et al.  ACE2: from vasopeptidase to SARS virus receptor , 2004, Trends in Pharmacological Sciences.

[57]  JoAnn E. Manson,et al.  Design of the Women's Health Initiative clinical trial and observational study. The Women's Health Initiative Study Group. , 1998, Controlled clinical trials.