Genome Medicine (May 2024)

High-throughput functional mapping of variants in an arrhythmia gene, KCNE1, reveals novel biology

  • Ayesha Muhammad,
  • Maria E. Calandranis,
  • Bian Li,
  • Tao Yang,
  • Daniel J. Blackwell,
  • M. Lorena Harvey,
  • Jeremy E. Smith,
  • Zerubabell A. Daniel,
  • Ashli E. Chew,
  • John A. Capra,
  • Kenneth A. Matreyek,
  • Douglas M. Fowler,
  • Dan M. Roden,
  • Andrew M. Glazer

DOI
https://doi.org/10.1186/s13073-024-01340-5
Journal volume & issue
Vol. 16, no. 1
pp. 1 – 20

Abstract

Read online

Abstract Background KCNE1 encodes a 129-residue cardiac potassium channel (I Ks) subunit. KCNE1 variants are associated with long QT syndrome and atrial fibrillation. However, most variants have insufficient evidence of clinical consequences and thus limited clinical utility. Methods In this study, we leveraged the power of variant effect mapping, which couples saturation mutagenesis with high-throughput sequencing, to ascertain the function of thousands of protein-coding KCNE1 variants. Results We comprehensively assayed KCNE1 variant cell surface expression (2554/2709 possible single-amino-acid variants) and function (2534 variants). Our study identified 470 loss- or partial loss-of-surface expression and 574 loss- or partial loss-of-function variants. Of the 574 loss- or partial loss-of-function variants, 152 (26.5%) had reduced cell surface expression, indicating that most functionally deleterious variants affect channel gating. Nonsense variants at residues 56–104 generally had WT-like trafficking scores but decreased functional scores, indicating that the latter half of the protein is dispensable for protein trafficking but essential for channel function. 22 of the 30 KCNE1 residues (73%) highly intolerant of variation (with > 70% loss-of-function variants) were in predicted close contact with binding partners KCNQ1 or calmodulin. Our functional assay data were consistent with gold standard electrophysiological data (ρ = − 0.64), population and patient cohorts (32/38 presumed benign or pathogenic variants with consistent scores), and computational predictors (ρ = − 0.62). Our data provide moderate-strength evidence for the American College of Medical Genetics/Association of Molecular Pathology functional criteria for benign and pathogenic variants. Conclusions Comprehensive variant effect maps of KCNE1 can both provide insight into I Ks channel biology and help reclassify variants of uncertain significance.

Keywords