1
|
Yuan Y, Zheng X, Zhang W, Ren Z, Liang B. A cross-tissue transcriptome-wide association study identifies novel susceptibility genes for atrial fibrillation. J Arrhythm 2025; 41:e70097. [PMID: 40416952 PMCID: PMC12099065 DOI: 10.1002/joa3.70097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/09/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
Background Atrial fibrillation (AF), the most common cardiac arrhythmia, has been linked to numerous loci identified by genome-wide association studies (GWAS). However, the causal genes and underlying mechanisms remain unclear. Methods We conducted a cross-tissue transcriptome-wide association studies (TWAS) using the unified test for molecular signatures (UTMOST), integrating genetic data from the FinnGen R11 cohort (287 805 individuals) with gene expression profiles from the genotype-tissue expression (GTEx) project. To enhance reliability, we applied functional summary-based imputation (FUSION), fine-mapping of causal gene sets (FOCUS), and multi-marker analysis of GenoMic annotation (MAGMA) for gene prioritization, followed by Mendelian randomization (MR) and colocalization analyses. GeneMANIA was used to explore gene functions. Results By integrating four TWAS approaches, this study identified five novel susceptibility genes significantly associated with AF risk. MR analysis further revealed that the gene expression levels of FKBP7, CEP68, and CAMK2D were positively associated with AF risk, while SPATS2L exhibited a significant protective effect. Colocalization analysis demonstrated that CEP68 and SPATS2L share causal variants with AF. Through comprehensive evaluation of multidimensional functional annotations and existing biological evidence, this study highlighted SPATS2L and CEP68 as potential functional candidate genes in AF pathogenesis. Conclusions This cross-tissue TWAS identified five novel AF susceptibility genes (CAMK2D, SPAST2L, CEP68, FKBP7, and SHRMOO3). Elevated expression of FKBP7, CEP68, and CAMK2D increases AF risk, while SPATS2L showed a protective effect, with colocalization analysis implicating CEP68 and SPATS2L as prioritized candidates. The integration of multi-omics approaches effectively unravels AF's genetic mechanisms.
Collapse
Affiliation(s)
- Yalin Yuan
- Shanxi Medical UniversityTaiyuanShanxiChina
| | - Xin Zheng
- Shanxi Medical UniversityTaiyuanShanxiChina
| | | | - Zhaoyu Ren
- Shanxi Medical UniversityTaiyuanShanxiChina
| | - Bin Liang
- Department of Cardiovascular MedicineSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| |
Collapse
|
2
|
Kadagandla S, Gunamalai L, Lee D, Kapoor A. Identification and Functional Assessment of Candidate Causal Cis-Regulatory Variants Underlying Electrocardiographic QT Interval GWAS Loci. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025:e005032. [PMID: 40421528 DOI: 10.1161/circgen.124.005032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/17/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Identifying causal variants among tens or hundreds of associated variants at each locus in genome-wide association studies (GWAS) is challenging. As the vast majority of GWAS variants are noncoding, sequence variation at cis-regulatory elements (CREs) affecting transcriptional expression of specific genes is a widely accepted molecular hypothesis. Following this hypothesis, combined with the observation that open chromatin is a universal hallmark of all types of CREs, we aimed to identify candidate causal cis-regulatory variants underlying QT interval GWAS loci. METHODS Common variants in high linkage disequilibrium with genome-wide significant variants were identified using variant call format tools. Genome-wide maps of cardiac putative CREs were generated by MACS2-based peak calling in human cardiac left ventricular DNase I sequencing and Assay for Transposase-Accessible Chromatin using sequencing data sets (n=13). Variant-CRE overlap was performed using custom tracks in the Table Browser tool at the UCSC Genome Browser. Luciferase reporter-based enhancer assays for variant-centered test elements were performed in mouse HL1 cardiomyocyte cells. Reporter activities of allelic pairs were compared using a Student t test. RESULTS At a dozen GWAS loci, selected for higher effect sizes and better understanding of the likely causal genes, we identified all genome-wide significant variants (n=1401) and included all common variants (minor allele frequency >1%) in high linkage disequilibrium (r2>0.9) with them as candidate variants (n=3482). Candidate variants were filtered for overlap with cardiac left ventricular putative CREs to identify candidate causal cis-regulatory variants (n=476), which were further assessed for being a known cardiac expression quantitative trait locus variant as additional functional evidence (n=243). Functional evaluation of a subset of seven candidate variants by luciferase reporter-based enhancer assays in HL1 cells using variant-centered test elements led to the identification of 6 enhancer variants with significant allelic differences. CONCLUSIONS These efforts have generated a comprehensive set of candidate causal variants expected to be enriched for cis-regulatory potential and thereby, explaining the observed genetic associations.
Collapse
Affiliation(s)
- Supraja Kadagandla
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (S.K., L.G., A.K.)
- Department of Biosciences, Rice University, Houston, TX (S.K.)
| | - Lavanya Gunamalai
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (S.K., L.G., A.K.)
| | - Dongwon Lee
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, MA (D.L.)
| | - Ashish Kapoor
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston (S.K., L.G., A.K.)
| |
Collapse
|
3
|
Gomaa AA, Zeid AM, Nagy IM. The role of genetic polymorphisms in KCNN2 in cardiovascular complications in patients with renal failure. Gene 2025; 944:149269. [PMID: 39884404 DOI: 10.1016/j.gene.2025.149269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/30/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Patients with end-stage renal disease (ESRD) are at a higher risk of cardiovascular (CV) complications and mortality compared to the general population. This study aimed to investigate the genetic polymorphisms of KCNN2, a key gene encoding a subtype of small-conductance calcium-activated potassium (SK) channels, which regulate an important SK current pathway potentially involved in the development of CV complications, particularly arrhythmias, in ESRD patients. A total of 169 ESRD patients were enrolled in this study. The patients were divided into two groups based on the presence of CV complications: Group I, consisting of 84 patients without CV complications, and Group II, comprising 85 patients with CV complications. Twelve tagging single nucleotide polymorphisms (tSNPs) in KCNN2 were examined. Polymerase chain reaction (PCR) was performed, and genotyping was correlated with CV complications in each group. The TC and CC genotypes of rs10076582, and the GT and TT genotypes of rs11738819 in the KCNN2 gene, were associated with an increased risk of CV complications in ESRD patients. After adjusting for potential risk factors, these associations remained significant. Additionally, KCNN2 haplotypes with the allele combinations GGCCCTCCGAG and AGTCCTCCGGT were significantly associated with a higher risk of CV complications in ESRD patients. In conclusion, our findings report that specific genetic polymorphisms in the KCNN2 gene, particularly the rs10076582 and rs11738819 variants, as well as GGCCCTCCGAG and AGTCCTCCGGT haplotypes, are significantly associated with an increased risk of cardiovascular complications in ESRD patients. These genetic markers may serve as potential biomarkers for identifying individuals at high risk of cardiovascular complications in this vulnerable population.
Collapse
Affiliation(s)
- Azza A Gomaa
- Internal Medicine Department Menofia University Menofia Egypt.
| | - Amany M Zeid
- Clinical Pathology Department Menofia University Menofia Egypt.
| | - Ibrahim M Nagy
- Medicinal Chemistry Department Menofia University Menofia Egypt.
| |
Collapse
|
4
|
Wada Y, Blair MA, Strickland TL, Laudeman JA, Kim K, Harvey ML, Solus JF, Fountain DF, Knollmann BC, Shoemaker MB, Kannankeril PJ, Roden DM. Increased L-type calcium current causes action potential prolongation in Jervell and Lange-Nielsen syndrome and is a drug target. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.20.25324224. [PMID: 40166542 PMCID: PMC11957160 DOI: 10.1101/2025.03.20.25324224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background KCNQ1 loss of function variants are thought to cause type 1 long QT syndrome by reducing I Ks. However, we have recently reported that pharmacologic block of I Ks in human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) produced minimal increases in action potential duration at 90% repolarization (APD90), while genetic loss of KCNQ1 markedly prolonged APD90. We sought here to define mechanisms underlying APD prolongation by genetic loss of KCNQ1. Methods We studied iPSC-CMs from population controls, an isogenic KCNQ1 knock out (KO) line created by a homozygous edit for the R518X loss of function variant, and 2 unrelated patients with the Jervell and Lange-Nielsen syndrome (JLN) due to compound heterozygosity for loss of function KCNQ1 variants. Results In both JLN and the KCNQ1-KO lines, I Ks was absent, APD90 was markedly prolonged, and L-type Ca channel (LTCC) current (I Ca-L) was significantly increased, 2-3-fold, compared to the control cells with no change in kinetics or gating. RNA-sequencing identified 298 and 584 genes that were up- and down-regulated, respectively, by KCNQ1-KO compared to the isogenic control cells. Gene ontology analysis identified down-regulation of 6 Ca2+ channel negative regulatory genes (p=0.0002, FDR=0.02), and in knockdown experiments in wild-type iPSC-CMs, three of these, CBARP, FKBP1B, and RRAD, increased I Ca-L, and RRAD increased APD90. A therapeutic low concentration (1 μM) of the Ca channel antagonist diltiazem significantly shortened APD90 in the two JLN cell lines and in KCNQ1-KO cells. A single low dose of intravenous diltiazem in one of the JLN patients shortened QTc. Conclusions These data further support the concept that delayed repolarization in JLN cannot be explained solely by loss of I Ks. Our findings demonstrate that KCNQ1 mutations lead to down-regulation of Ca2+ channel inhibitory genes, with resultant increased I Ca-L that underlies delayed repolarization in JLN. We further propose that diltiazem can be repurposed for treatment of patients with JLN.
Collapse
Affiliation(s)
- Yuko Wada
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Arrhythmia Research and Therapeutic, Vanderbilt University Medical Center, Nashville, TN
| | - Marcia A. Blair
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | | | - Julie A. Laudeman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Kyungsoo Kim
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Arrhythmia Research and Therapeutic, Vanderbilt University Medical Center, Nashville, TN
| | - M. Lorena Harvey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joseph F. Solus
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Darlene F. Fountain
- Department of Pediatrics, Monroe Carell Jr. Children’s Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN
| | - Bjorn C. Knollmann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Arrhythmia Research and Therapeutic, Vanderbilt University Medical Center, Nashville, TN
| | - M. Benjamin Shoemaker
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Arrhythmia Research and Therapeutic, Vanderbilt University Medical Center, Nashville, TN
| | - Prince J. Kannankeril
- Vanderbilt Center for Arrhythmia Research and Therapeutic, Vanderbilt University Medical Center, Nashville, TN
- Department of Pediatrics, Monroe Carell Jr. Children’s Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN
| | - Dan M. Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Arrhythmia Research and Therapeutic, Vanderbilt University Medical Center, Nashville, TN
- Departments of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
5
|
Mulvey JF, Meyer EL, Svenningsen MS, Lundby A. Integrating -Omic Technologies across Modality, Space, and Time to Decipher Remodeling in Cardiac Disease. Curr Cardiol Rep 2025; 27:74. [PMID: 40116972 PMCID: PMC11928419 DOI: 10.1007/s11886-025-02226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
PURPOSE OF REVIEW Despite significant efforts to understand pathophysiological processes underlying cardiac diseases, the molecular causes for the most part remain unresolved. Rapid advancements in -omics technologies, and their application in cardiac research, offer new insight into cardiac remodeling in disease states. This review aims to provide an accessible overview of recent advances in omics approaches for studying cardiac remodeling, catering to readers without extensive prior expertise. RECENT FINDINGS We provide a methodologically focused overview of current methods for performing transcriptomics and proteomics, including their extensions for single-cell and spatial measurements. We discuss approaches to integrate data across modalities, resolutions and time. Key recent applications within the cardiac field are highlighted. Each -omics modality can provide insight, yet each existing experimental method has technical or conceptual limitations. Integrating data across multiple modalities can leverage strengths and mitigate weaknesses, ultimately enhancing our understanding of cardiac pathophysiology.
Collapse
Affiliation(s)
- John F Mulvey
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emily L Meyer
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Skjoldan Svenningsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Lee D, Gunamalai L, Kannan J, Vickery K, Yaacov O, Onuchic-Whitford AC, Chakravarti A, Kapoor A. Massively parallel reporter assays identify functional enhancer variants at QT interval GWAS loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642686. [PMID: 40161821 PMCID: PMC11952420 DOI: 10.1101/2025.03.11.642686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Genome-wide association studies (GWAS) have identified >30 loci with multiple common noncoding variants explaining interindividual electrocardiographic QT interval (QTi) variation. Of the many types of noncoding functional elements, here we sought to identify transcriptional enhancers with sequence variation and their cognate transcription factors (TFs) that alter the expression of proximal cardiac genes to affect QTi variation. We used massively parallel reporter assays (MPRA) in mouse cardiomyocyte HL-1 cells to screen for functional enhancer variants among 1,018 QTi-associated GWAS variants that overlap candidate cardiac enhancers across 31 loci. We identified 445 GWAS variant-containing enhancers of which 79 showed significant allelic difference in enhancer activity across 21 GWAS loci, with multiple enhancer variants per locus. Of these, we predicted differential binding by cardiac TFs, including AP-1, ATF-1, GATA2, MEF2, NKX2.5, SRF and TBX5 which are known to play key roles in development and homeostasis, at 49 enhancer variants. Finally, we used expression quantitative trait locus mapping and predicted promoter-enhancer contacts to identify 14 candidate target genes through analyses of 36 enhancer variants at 16 loci. This study provides strong evidence for 14 cardiac genes, 10 of them novel, impacting on QTi variation, beyond explaining observed genetic associations.
Collapse
Affiliation(s)
- Dongwon Lee
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Lavanya Gunamalai
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jeerthi Kannan
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Kyla Vickery
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Or Yaacov
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY, USA
| | - Ana C. Onuchic-Whitford
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Renal division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Aravinda Chakravarti
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY, USA
| | - Ashish Kapoor
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
7
|
Khan N, Shaar A, Kunji K, Khan A, Elshrif M, Bashir M, Ali MT, Al Haj Zen A, Kiryluk K, Nemer G, Fahed AC, Saad M. Genome-Wide Association Study for Resting Electrocardiogram in the Qatari Population Identifies 6 Novel Genes and Validates Novel Polygenic Risk Scores. J Am Heart Assoc 2025; 14:e038341. [PMID: 40008532 DOI: 10.1161/jaha.124.038341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/19/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Electrocardiography is one of the most valuable noninvasive diagnostic tools in determining the presence of many cardiovascular diseases. Genetic factors are important in determining ECG abnormalities and their link to cardiovascular diseases. Genome-wide association studies and polygenic risk scores (PRSs) have been conducted for various ECG traits such as QT interval and QRS duration. However, these studies mainly focused on cohorts of European descent. METHODS In this cohort study, genome-wide association studies for 6 ECG traits (RR, PR, corrected QT interval [QTc], QRS, JT, and P wave duration) were conducted in a Middle Eastern cohort from the Qatar Precision Health Institute, comprising 13 827 subjects with whole-genome sequence data. Middle Eastern PRSs were developed using clumping and thresholding, and their performance was compared with 26 published PRSs. Genetic predisposition to long QT syndrome was explored using rare variant analysis. RESULTS Seventy-four independent loci were obtained with genome-wide significance across the 6 traits (P<5×10-8). Of the 74 loci, 67 (90.5%) were previously reported, and 7 loci (9.5%) were novel and contained 6 genes: STAC and CSMD1 for PR, ANK1 and NCOA2 for QRS, LSP1 for QTc, and MKLN1 for P wave duration. All 26 published PRSs showed good performance in our cohort. PGS002276 showed the best performance for QTc (R2=0.059, P=4.83×10-185), PGS002166 showed the best performance for QRS (R2=0.024, P=1.53×10-75), and PGS000905 showed the best performance for PR (R2=0.053, P=2.57×10-165). Some of these PRSs were associated with cardiovascular diseases. For example, PGS003500, a QTc PRS, was significantly associated with cardiomyopathy (odds ratio per 1 SD=1.58 [95% CI, 1.23-2.01]; P=2.42×10-4). Middle Eastern PRSs substantially outperformed published PRSs and did not perform well in the UK Biobank data. Ten pathogenic variants, including 3 that are specific to Qatari individuals, were observed in 17 long QT syndrome genes and were carried by 19 individuals. The QTc average was larger for mutation carriers (415.6±23.5 versus 402.3±18.5 in noncarriers). Five-year follow-up data did not show a significant change in ECG patterns, regardless of mutation status and PRS values. Four of 2302 individuals had prolonged QTc intervals over the 2 time points. CONCLUSIONS In this first genome-wide association study for ECG traits in the Middle East using whole-genome sequence data, 7 novel loci (6 genes) were identified. Published PRSs performed well, but newly developed Middle Eastern-specific PRSs performed the best. Novel variants in long QT syndrome genes were observed for the first time in Qatari individuals. Follow-up data did not show significant changes in ECG patterns.
Collapse
Affiliation(s)
- Nahin Khan
- Qatar Computing Research Institute, Hamad Bin Khalifa University Doha Qatar
| | - Abdullah Shaar
- Qatar Computing Research Institute, Hamad Bin Khalifa University Doha Qatar
| | - Khalid Kunji
- Qatar Computing Research Institute, Hamad Bin Khalifa University Doha Qatar
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University New York NY USA
| | - Mohamed Elshrif
- Qatar Computing Research Institute, Hamad Bin Khalifa University Doha Qatar
| | | | | | - Ayman Al Haj Zen
- College of Health and Life Sciences, Hamad Bin Khalifa University Doha Qatar
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University New York NY USA
| | - Georges Nemer
- College of Health and Life Sciences, Hamad Bin Khalifa University Doha Qatar
| | - Akl C Fahed
- Cardiovascular Research Center Massachusetts General Hospital, Harvard Medical School Boston MA USA
- Cardiovascular Disease Initiative Broad Institute of Harvard and MIT Cambridge MA USA
| | - Mohamad Saad
- Qatar Computing Research Institute, Hamad Bin Khalifa University Doha Qatar
| |
Collapse
|
8
|
Arora A, Zareba W, Woosley RL, Klimentidis YC, Patel IY, Quan SF, Wendel C, Shamoun F, Guerra S, Parthasarathy S, Patel SI. Genetic QT score as a predictor of sudden cardiac death in participants with sleep-disordered breathing in the UK Biobank. J Clin Sleep Med 2025; 21:549-557. [PMID: 39589075 PMCID: PMC11874099 DOI: 10.5664/jcsm.11474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024]
Abstract
STUDY OBJECTIVES The goal of this study was to evaluate the association between a polygenic risk score (PRS) for QT prolongation (QTc-PRS), corrected QT intervals (QTc) and sudden cardiac death (SCD) in participants enrolled in the UK Biobank with and without sleep-disordered breathing (SDB). METHODS The QTc-PRS was calculated using allele copy number and previously reported effect estimates for each single nuclear polymorphism. Competing-risk regression models adjusting for age, sex, body mass index, QT prolonging medication, race, and comorbid cardiovascular conditions were used for SCD analyses. RESULTS A total of 500,584 participants were evaluated (56.5 ± 8 years, 54% female, 1.4% diagnosed with sleep apnea). A higher QTc-PRS was independently associated with the increased QTc interval duration (P < .0001). The mean QTc for the top QTc-PRS quintile was 15 msec longer than the bottom quintile (P < .001). SDB was found to be an effect modifier in the relationship between QTc-PRS and SCD. The adjusted hazard ratio per 5-unit change in QTc-PRS for SCD was 1.64 (95% confidence interval 1.16-2.31, P = .005) among those with SDB and 1.04 (95% confidence interval 0.95-1.14, P = .44) among those without SDB (P for interaction = .01). Black participants with SDB had significantly elevated adjusted risk of SCD (hazard ratio = 9.6, 95% confidence interval 1.24-74, P = .03). CONCLUSIONS In the UK Biobank population, the QTc-PRS was associated with SCD among participants with SDB but not among those without SDB, indicating that SDB is a significant modifier of the genetic risk. Black participants with SDB had a particularly high risk of SCD. CITATION Arora A, Zareba W, Woosley RL, et al. Genetic QT score as a predictor of sudden cardiac death in participants with sleep-disordered breathing in the UK Biobank. J Clin Sleep Med. 2025;21(3):549-557.
Collapse
Affiliation(s)
- Amit Arora
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
| | - Wojciech Zareba
- Division of Cardiology, University of Rochester Medical Center, Rochester, New York
| | - Raymond L. Woosley
- Division of Clinical Data Analytics and Decision Support, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | - Yann C. Klimentidis
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
| | - Imran Y. Patel
- El Rio Health, Tucson, Arizona
- UAHS Center for Sleep and Circadian Sciences, University of Arizona, Tucson, Arizona
- Division of Pulmonary, Allergy, Critical Care Medicine and Sleep Medicine, University of Arizona College of Medicine – Tucson, Tucson, Arizona
| | - Stuart F. Quan
- UAHS Center for Sleep and Circadian Sciences, University of Arizona, Tucson, Arizona
- Division of Pulmonary, Allergy, Critical Care Medicine and Sleep Medicine, University of Arizona College of Medicine – Tucson, Tucson, Arizona
| | - Christopher Wendel
- UAHS Center for Sleep and Circadian Sciences, University of Arizona, Tucson, Arizona
| | | | - Stefano Guerra
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona
- Division of Pulmonary, Allergy, Critical Care Medicine and Sleep Medicine, University of Arizona College of Medicine – Tucson, Tucson, Arizona
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona
| | - Sairam Parthasarathy
- UAHS Center for Sleep and Circadian Sciences, University of Arizona, Tucson, Arizona
- Division of Pulmonary, Allergy, Critical Care Medicine and Sleep Medicine, University of Arizona College of Medicine – Tucson, Tucson, Arizona
| | - Salma I. Patel
- UAHS Center for Sleep and Circadian Sciences, University of Arizona, Tucson, Arizona
- Division of Pulmonary, Allergy, Critical Care Medicine and Sleep Medicine, University of Arizona College of Medicine – Tucson, Tucson, Arizona
- The University of Arizona College of Health Sciences, Tucson, Arizona
| |
Collapse
|
9
|
Isaacs A, Zeemering S, Winters J, Batlle M, Bidar E, Boukens B, Casadei B, Chua W, Crijns HJGM, Fabritz L, Guasch E, Hatem SN, Hermans B, Kääb S, Kawczynski M, Maesen B, Maessen J, Mont L, Sinner MF, Wakili R, Verheule S, Kirchhof P, Schotten U, Stoll M. Lateral Atrial Expression Patterns Provide Insights into Local Transcription Disequilibrium Contributing to Disease Susceptibility. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004594. [PMID: 39846178 DOI: 10.1161/circgen.124.004594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 11/19/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Transcriptional dysregulation, possibly affected by genetic variation, contributes to disease etiology. Due to dissimilarities in development, function, and remodeling during disease progression, transcriptional differences between the left atrium (LA) and right atrium (RA) may provide insight into diseases such as atrial fibrillation. METHODS Lateral differences in atrial transcription were evaluated in CATCH ME (Characterizing Atrial fibrillation by Translating its Causes into Health Modifiers in the Elderly) using a 2-stage discovery and replication design. The design took advantage of the availability of 32 paired samples, for which both LA and RA tissue were obtained, as a discovery cohort, and 98 LA and 69 RA unpaired samples utilized as a replication cohort. RESULTS A total of 714 transcripts were identified and replicated as differentially expressed (DE) between LA and RA, as well as 98 exons in 55 genes. Approximately 50% of DE transcripts were colocated with another frequently correlated DE transcript (PFDR ≤0.05 for 579 regions). These "transcription disequilibrium" blocks contained examples including side-specific differential exon usage, such as the PITX2 locus, where ENPEP showed evidence of differential exon usage. Analysis of this region in conjunction with BMP10 identified rs9790621 as associated with ENPEP transcription in LA, while rs7687878 was associated with BMP10 expression in RA. In RA, BMP10 and ENPEP were strongly correlated in noncarriers, which was attenuated in risk-allele carriers, where BMP10 and PITX2 expression were strongly correlated. CONCLUSIONS These results significantly expand knowledge of the intricate, tissue-specific transcriptional landscape in human atria, including DE transcripts and side-specific isoform expression. Furthermore, they suggest the existence of blocks of transcription disequilibrium influenced by genetics.
Collapse
Affiliation(s)
- Aaron Isaacs
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Maastricht Center for Systems Biology (A.I.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Stef Zeemering
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Joris Winters
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Montserrat Batlle
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
| | - Elham Bidar
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Bas Boukens
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Barbara Casadei
- NIHR Oxford Biomedical Research Center, John Radcliffe Hospital, University of Oxford, United Kingdom (B.C.)
| | - Winnie Chua
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
| | - Harry J G M Crijns
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Cardiology (H.J.G.M.C.), Maastricht University Medical Centre, the Netherlands
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
- University Center of Cardiovascular Sciences, University Medical Center Hamburg Eppendorf, Germany (L.F.)
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
| | - Eduard Guasch
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
- Clinic Barcelona, Universitat de Barcelona, Spain (E.G., L.M.)
| | - Stephane N Hatem
- INSERM UMRS 1166, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne University, Paris, France (S.N.H.)
| | - Ben Hermans
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Stefan Kääb
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
- University Heart and Vascular Center, University Hospital Hamburg Eppendorf, Germany (S.K., M.F.S.)
- Department of Cardiology, University Hospital of Munich, Germany (S.K., M.F.S.)
| | - Michal Kawczynski
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Bart Maesen
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Jos Maessen
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Lluis Mont
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
- Clinic Barcelona, Universitat de Barcelona, Spain (E.G., L.M.)
| | - Moritz F Sinner
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
- University Heart and Vascular Center, University Hospital Hamburg Eppendorf, Germany (S.K., M.F.S.)
- Department of Cardiology, University Hospital of Munich, Germany (S.K., M.F.S.)
| | - Reza Wakili
- Department of Medicine and Cardiology, Goethe University, Frankfurt, Germany (R.W.)
- German Center for Cardiovascular Research DZHK, Rhine-Main (R.W.)
| | - Sander Verheule
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany (P.K.)
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
| | - Ulrich Schotten
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Monika Stoll
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Biochemistry (M.S.), Maastricht University, the Netherlands
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Germany (M.S.)
| |
Collapse
|
10
|
Emerson JI, Shi W, Paredes-Larios J, Walker WG, Hutton JE, Cristea IM, Marzluff WF, Conlon FL. X-Chromosome-Linked miRNAs Regulate Sex Differences in Cardiac Physiology. Circ Res 2025; 136:258-275. [PMID: 39772608 PMCID: PMC11781965 DOI: 10.1161/circresaha.124.325447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Males and females exhibit distinct anatomic and functional characteristics of the heart, predisposing them to specific disease states. METHODS We identified microRNAs (miRNAs/miR) with sex-differential expression in mouse hearts. RESULTS Four conserved miRNAs are present in a single locus on the X-chromosome and are expressed at higher levels in females than males. We show miRNA, miR-871, is responsible for decreased expression of the protein SRL (sarcalumenin) in females. SRL is involved in calcium signaling, and we show it contributes to differences in electrophysiology between males and females. miR-871 overexpression mimics the effects of the cardiac physiology of conditional cardiomyocyte-specific Srl-null mice. Inhibiting miR-871 with an antagomir in females shortened ventricular repolarization. The human orthologue of miR-871, miR-888, coevolved with the SRL 3' untranslated region and regulates human SRL. CONCLUSIONS These data highlight the importance of sex-differential miRNA mechanisms in mediating sex-specific functions and their potential relevance to human cardiac diseases.
Collapse
Affiliation(s)
- James I. Emerson
- Department of Biochemistry & Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Wei Shi
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jose Paredes-Larios
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William G. Walker
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Josiah E. Hutton
- Department of Molecular Biology, Princeton University, Lew Thomas Laboratory, Princeton, NJ 08544, USA
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Lew Thomas Laboratory, Princeton, NJ 08544, USA
| | - William F. Marzluff
- Department of Biochemistry & Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Frank L. Conlon
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Science, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
11
|
Gunamalai L, Singh P, Berg B, Shi L, Sanchez E, Smith A, Breton G, Bedford MT, Balciunas D, Kapoor A. Functional characterization of QT interval associated SCN5A enhancer variants identify combined additive effects. HGG ADVANCES 2025; 6:100358. [PMID: 39354714 PMCID: PMC11532988 DOI: 10.1016/j.xhgg.2024.100358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/03/2024] Open
Abstract
Several empirical and theoretical studies suggest the presence of multiple enhancers per gene that collectively regulate gene expression, and that common sequence variation impacting on the activities of these enhancers is a major source of inter-individual gene expression variability. However, for the vast majority of genes, enhancers and the underlying regulatory variation remains unknown. Even for the genes with well-characterized enhancers, the nature of the combined effects from multiple enhancers and their variants, when known, on gene expression regulation remains unexplored. Here, we have evaluated the combined effects from five SCN5A enhancers and their regulatory variants that are known to collectively correlate with SCN5A cardiac expression and underlie QT interval association in the general population. Using small deletions centered at the regulatory variants in episomal reporter assays in a mouse cardiomyocyte cell line, we demonstrate that the variants and their flanking sequences play critical role in individual enhancer activities, likely being a transcription factor (TF) binding site. By oligonucleotide-based pulldown assays on predicted TFs, we identify the TFs likely driving allele-specific enhancer activities. Using all 32 possible allelic synthetic constructs in reporter assays, representing the five bi-allelic enhancers, we demonstrate combined additive effects on overall enhancer activities. Using transient enhancer assays in zebrafish embryos we demonstrate that four elements act as enhancers in vivo. Together, these studies uncover the TFs driving the enhancer activities of QT interval associated SCN5A regulatory variants, reveal the additive effects from allelic combinations of these regulatory variants, and prove their potential to act as enhancers in vivo.
Collapse
Affiliation(s)
- Lavanya Gunamalai
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Parul Singh
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brian Berg
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Leilei Shi
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ernesto Sanchez
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Alexa Smith
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ghislain Breton
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mark T Bedford
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Darius Balciunas
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Ashish Kapoor
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Kornetova EG, Galkin SA, Mednova IA, Tiguntsev VV, Paderina DZ, Ivanova SA. [The association of NOS1AP gene polymorphisms with the duration of the QT interval in patients with schizophrenia receiving antipsychotic therapy]. Zh Nevrol Psikhiatr Im S S Korsakova 2025; 125:98-104. [PMID: 39930683 DOI: 10.17116/jnevro202512501198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To study the associations of NOS1AP gene polymorphisms with the duration of the QT interval (QTc) in patients with schizophrenia receiving antipsychotics. MATERIAL AND METHODS One hundred and sixty-eight patients (78 men and 90 women) with an established diagnosis of schizophrenia were examined. The patients received basic antipsychotic therapy. At the time of admission to the hospital, a standard 12-lead electrocardiogram recording was performed. The calculation of the QTc interval was carried out using the Bazett formula. Three NOS1AP polymorphisms (rs12029454, rs10494366 and rs12143842) were selected for genotyping. RESULTS Spearman's correlation revealed a significant relationship between CPZeq and the duration of the QTc interval in the group of men (rs=0.262; p=0.021; pBonf=0.042). CPZeq was positively correlated with the duration of the QTc interval in men with AA and AG rs12029454 (rs=0.717; p=0.00025; pbonf=0.0015), GG and GT rs10494366 (rs=0.381; p=0.008; pbonf=0.048), TT and CT rs12143842 (rs=0.389; p=0.003; pbonf=0.018) genotypes. We did not find any significant correlations in the group of women. CONCLUSION Genetic variants rs12029454, rs10494366 and rs12143842 of the NOS1AP gene are associated with the duration of the QTc interval in men with schizophrenia in response to antipsychotics. The AA/AG rs12029454, GG/GT rs10494366 and TT/CT rs12143842 genotypes in men contribute to the prolongation of the QTc interval and the development of undesirable cardiovascular disorders against the background of antipsychotic therapy.
Collapse
Affiliation(s)
- E G Kornetova
- Mental Health Research Institute - Tomsk National Research Medical Center Russian Academy of Science, Tomsk, Russia
| | - S A Galkin
- Mental Health Research Institute - Tomsk National Research Medical Center Russian Academy of Science, Tomsk, Russia
| | - I A Mednova
- Mental Health Research Institute - Tomsk National Research Medical Center Russian Academy of Science, Tomsk, Russia
| | - V V Tiguntsev
- Mental Health Research Institute - Tomsk National Research Medical Center Russian Academy of Science, Tomsk, Russia
| | - D Z Paderina
- Mental Health Research Institute - Tomsk National Research Medical Center Russian Academy of Science, Tomsk, Russia
| | - S A Ivanova
- Mental Health Research Institute - Tomsk National Research Medical Center Russian Academy of Science, Tomsk, Russia
| |
Collapse
|
13
|
Lopez-Medina AI, Campos-Staffico AM, Chahal CAA, Jacoby JP, Volkers I, Berenfeld O, Luzum JA. Polygenic risk score for drug-induced long QT syndrome: independent validation in a real-world patient cohort. Pharmacogenet Genomics 2025; 35:45-56. [PMID: 39470415 PMCID: PMC11543509 DOI: 10.1097/fpc.0000000000000548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
OBJECTIVE Drug-induced long QT syndrome (diLQTS) is an adverse reaction from over 150 FDA-approved medications, posing the risk of triggering torsades de pointes and sudden death. While common genetic variants may modestly impact QT interval individually, their collective effect can significantly amplify risk of diLQTS. Consequently, this study aimed to validate a polygenic risk score (PRS) for diLQTS previously proposed by Strauss et al . METHODS A retrospective cohort study was conducted utilizing patients from the Michigan Genomics Initiative prescribed 27 high-risk QT-prolonging drugs and an ECG during the prescription. The primary outcome was marked prolongation of the QTc interval (either >60 ms change from baseline or >500 ms absolute value) during treatment with a high-risk QT-prolonging drug. RESULTS The primary outcome occurred in 12.0% of n = 6070 self-reported White, 12.4% of 558 African American, and 8.2% of 110 Asian patients. The PRS significantly associated with diLQTS in White patients [adjusted odds ratio = 1.44 (95% CI: 1.09-1.89); P = 0.009]. However the study lacked sufficient statistical power to confirm the PRS as a risk factor in African Americans [adjusted odds ratio = 2.18 (95% CI: 0.98-5.49); P = 0.073] and Asians [adjusted odds ratio = 3.21 (95% CI: 0.69-16.87); P = 0.139] due to smaller sample sizes in these groups. CONCLUSION The previously published PRS for diLQTS was validated in a large, real-world cohort, demonstrating its potential as a tool for identifying high-risk patients. Incorporating this PRS into routine clinical practice could enable proactive measures to prevent life-threatening diLQTS.
Collapse
Affiliation(s)
- Ana I Lopez-Medina
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States
| | | | - Choudhary Anwar A Chahal
- Center for Inherited Cardiovascular Diseases, WellSpan Health, Lancaster, PA, USA. Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA. Department of Cardiology, Barts Heart Centre, London, UK, Queen Mary University of London, London, UK
| | - Juliet P. Jacoby
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States
| | - Isabella Volkers
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States
| | - Omer Berenfeld
- Center for Arrhythmia Research, Departments of Internal Medicine – Cardiology, Biomedical Engineering, and Applied Physics. University of Michigan, Ann Arbor, MI, United States
| | - Jasmine A. Luzum
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
14
|
Lukas E, van de Weijer M, Bergstedt J, Bezzina CR, Treur JL. Causal inference in the field of arrhythmia: An introduction to mendelian randomization. Heart Rhythm 2025; 22:203-216. [PMID: 39019383 DOI: 10.1016/j.hrthm.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024]
Abstract
Mendelian randomization (MR) uses genetic variants associated with an exposure (eg, high blood pressure) as instrumental variables to test causal effects on an outcome (eg, atrial fibrillation [AF]). By leveraging the random assortment of genetic variants during gamete formation, MR reduces biases like confounding and reverse causation. We screened 391 papers, examining 277 that applied MR to investigate arrhythmia and, in others, cardiovascular traits, lifestyle, behavioral traits, and body composition. Our analysis focused on MR studies of arrhythmia and cardiovascular traits. Key findings highlight high systolic blood pressure, low resting heart rate, elevated cardiac troponin I levels, coronary artery disease, and heart failure as risk factors for AF, whereas AF itself increases heart failure risk. As genetic data become more accessible, MR's relevance grows. Sensitivity analyses and integrating MR with other methodologies in a triangulation framework enhance the robustness of causal inferences by navigating different biases.
Collapse
Affiliation(s)
- Eva Lukas
- Genetic Epidemiology, Department of Psychiatry, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Margot van de Weijer
- Genetic Epidemiology, Department of Psychiatry, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Jacob Bergstedt
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; European Reference Network for rare, low prevalence and complex diseases of the heart: ERN GUARD-Heart
| | - Jorien L Treur
- Genetic Epidemiology, Department of Psychiatry, Amsterdam UMC, University of Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Bentestuen MS, Weis CN, Jeppesen CB, Thiele LS, Thirstrup JP, Cordero-Solorzano J, Jensen HK, Starnawska A, Hauser AS, Gasse C. Pharmacogenomic markers associated with drug-induced QT prolongation: a systematic review. Pharmacogenomics 2025; 26:53-72. [PMID: 40116580 PMCID: PMC11988217 DOI: 10.1080/14622416.2025.2481025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025] Open
Abstract
AIM To systematically assess clinical studies involving patients undergoing drug therapy, comparing different genotypes to assess the relationship with changes in QT intervals, with no limitations on study design, setting, population, dosing regimens, or duration. METHODS This systematic review followed PRISMA guidelines and a pre-registered protocol. Clinical human studies on PGx markers of diQTP were identified, assessed using standardized tools, and categorized by design. Gene associations were classified as pharmacokinetic or pharmacodynamic. Identified genes underwent pathway enrichment analyses. Drugs were classified by third-level Anatomical Therapeutic Chemical (ATC) codes. Descriptive statistics were computed by study category and drug classes. RESULTS Of 4,493 reports, 84 studies were included, identifying 213 unique variants across 42 drug classes, of which 10% were replicated. KCNE1-Asp85Asn was the most consistent variant. Most findings (82%) were derived from candidate gene studies, suggesting bias toward known markers. The diQTP-associated genes were mainly linked to "cardiac conduction" and "muscle contraction" pathways (false discovery rate = 4.71 × 10-14). We also found an overlap between diQTP-associated genes and congenital long QT syndrome genes. CONCLUSION Key genes, drugs, and pathways were identified, but few consistent PGx markers emerged. Extensive, unbiased studies with diverse populations are crucial to advancing the field. REGISTRATION A protocol was pre-registered at PROSPERO under registration number CRD42022296097. DATA DEPOSITION Data sets generated by this review are available at figshare: DOI: 10.6084/m9.figshare.27959616.
Collapse
Affiliation(s)
- Marlene Schouby Bentestuen
- Psychosis Research Unit, Aarhus University Hospital Psychiatry, Aarhus, Denmark
- Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Christian Noe Weis
- Department of Forensic Psychiatry, Aarhus University Hospital Psychiatry, Aarhus, Denmark
| | | | - Liv Swea Thiele
- Department of Affective Disorders, Aarhus University Hospital Psychiatry, Aarhus, Denmark
| | - Janne Pia Thirstrup
- Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
- Department of Affective Disorders, Aarhus University Hospital Psychiatry, Aarhus, Denmark
- Department of Biomedicine, Health, Aarhus University, Aarhus, Denmark
| | - Juan Cordero-Solorzano
- Department of Biomedicine, Health, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, CGPM, and Center for Integrative Sequencing, iSEQ, Aarhus, Denmark
| | - Henrik Kjærulf Jensen
- Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart: ERN GUARD‐Heart, Aarhus, Denmark
| | - Anna Starnawska
- Department of Biomedicine, Health, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, CGPM, and Center for Integrative Sequencing, iSEQ, Aarhus, Denmark
| | - Alexander Sebastian Hauser
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christiane Gasse
- Psychosis Research Unit, Aarhus University Hospital Psychiatry, Aarhus, Denmark
- Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
- Department of Affective Disorders, Aarhus University Hospital Psychiatry, Aarhus, Denmark
| |
Collapse
|
16
|
Mousavi Z, Arvanitis M, Duong T, Brody JA, Battle A, Sotoodehnia N, Shojaie A, Arking DE, Bader JS. Prioritization of causal genes from genome-wide association studies by Bayesian data integration across loci. PLoS Comput Biol 2025; 21:e1012725. [PMID: 39774334 PMCID: PMC11741684 DOI: 10.1371/journal.pcbi.1012725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 01/17/2025] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
MOTIVATION Genome-wide association studies (GWAS) have identified genetic variants, usually single-nucleotide polymorphisms (SNPs), associated with human traits, including disease and disease risk. These variants (or causal variants in linkage disequilibrium with them) usually affect the regulation or function of a nearby gene. A GWAS locus can span many genes, however, and prioritizing which gene or genes in a locus are most likely to be causal remains a challenge. Better prioritization and prediction of causal genes could reveal disease mechanisms and suggest interventions. RESULTS We describe a new Bayesian method, termed SigNet for significance networks, that combines information both within and across loci to identify the most likely causal gene at each locus. The SigNet method builds on existing methods that focus on individual loci with evidence from gene distance and expression quantitative trait loci (eQTL) by sharing information across loci using protein-protein and gene regulatory interaction network data. In an application to cardiac electrophysiology with 226 GWAS loci, only 46 (20%) have within-locus evidence from Mendelian genes, protein-coding changes, or colocalization with eQTL signals. At the remaining 180 loci lacking functional information, SigNet selects 56 genes other than the minimum distance gene, equal to 31% of the information-poor loci and 25% of the GWAS loci overall. Assessment by pathway enrichment demonstrates improved performance by SigNet. Review of individual loci shows literature evidence for genes selected by SigNet, including PMP22 as a novel causal gene candidate.
Collapse
Affiliation(s)
- Zeinab Mousavi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Marios Arvanitis
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - ThuyVy Duong
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Ali Shojaie
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Dan E. Arking
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Joel S. Bader
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
Bukaeva A, Ershova A, Kharlap M, Kiseleva A, Kutsenko V, Sotnikova E, Divashuk M, Pokrovskaya M, Garbuzova E, Blokhina A, Kopylova O, Zotova E, Petukhova A, Zharikova A, Ramensky V, Zaicenoka M, Vyatkin Y, Meshkov A, Drapkina O. The Yield of Genetic Testing and Putative Genetic Factors of Disease Heterogeneity in Long QT Syndrome Patients. Int J Mol Sci 2024; 25:11976. [PMID: 39596046 PMCID: PMC11593843 DOI: 10.3390/ijms252211976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Genetic overdiagnosis of long QT syndrome (LQTS) becomes a critical concern due to the high clinical significance of DNA diagnosis. Current guidelines for LQTS genetic testing recommend a limited scope and strict referral based on the Schwartz score. Nevertheless, LQTS may be underdiagnosed in patients with borderline phenotypes. We aimed to evaluate the total yield of rare variants in cardiac genes in LQTS patients. The cohort of 82 patients with LQTS referral diagnosis underwent phenotyping, Schwartz score counting, and exome sequencing. We assessed known LQTS genes for diagnostics, as per guidelines, and a broader set of genes for research. Diagnostic testing yield reached 75% in index patients; all causal variants were found in KCNQ1, KCNH2, and SCN5A genes. Research testing of 248 heart-related genes achieved a 50% yield of molecular diagnosis in patients with a low Schwartz score (<3.5). In patients with LQTS-causing variants, each additional rare variant in heart-related genes added 0.94 points to the Schwartz score (p value = 0.04), reflecting the more severe disease in such patients than in those with causal variants but without additional findings. We conclude that the current LQTS genetic diagnosis framework is highly specific but may lack sensitivity for patients with a Schwartz score <3.5. Improving referral criteria for these patients could enhance DNA diagnosis. Also, our results suggest that additional variants in cardiac genes may affect the severity of the disease in the carriers of LQTS-causing variants, which may aid in identifying new modifier genes.
Collapse
Affiliation(s)
- Anna Bukaeva
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Alexandra Ershova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Maria Kharlap
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Anna Kiseleva
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Vladimir Kutsenko
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Evgeniia Sotnikova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Mikhail Divashuk
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
- All-Russia Research Institute of Agricultural Biotechnology, 127550 Moscow, Russia
| | - Maria Pokrovskaya
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Elizaveta Garbuzova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Anastasia Blokhina
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Oksana Kopylova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Evgenia Zotova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Anna Petukhova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Anastasia Zharikova
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Vasily Ramensky
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia
- MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Marija Zaicenoka
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Yuri Vyatkin
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
- MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Alexey Meshkov
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| | - Oxana Drapkina
- National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia; (A.E.); (M.K.); (A.K.); (V.K.); (M.D.); (M.P.); (E.G.); (A.B.); (A.P.); (A.Z.); (V.R.); (M.Z.); (Y.V.); (A.M.); (O.D.)
| |
Collapse
|
18
|
Young WJ, van der Most PJ, Bartz TM, Bos MM, Biino G, Duong T, Foco L, Lominchar JT, Müller‐Nurasyid M, Nardone GG, Pecori A, Ramirez J, Repetto L, Schramm K, Shen X, van Duijvenboden S, van Heemst D, Weiss S, Yao J, Benjamins J, Alonso A, Spedicati B, Biggs ML, Brody JA, Dörr M, Fuchsberger C, Gögele M, Guo X, Ikram MA, Jukema JW, Kääb S, Kanters JK, Lifelines Cohort Study, Lin HJ, Linneberg A, Nauck M, Nolte IM, Pianigiani G, Santin A, Soliman EZ, Tesolin P, Vaccargiu S, Waldenberger M, van der Harst P, Verweij N, Arking DE, Concas MP, De Grandi A, Girotto G, Grarup N, Kavousi M, Mook‐Kanamori DO, Navarro P, Orini M, Padmanabhan S, Pattaro C, Peters A, Pirastu M, Pramstaller PP, Heckbert SR, Sinner M, Snieder H, Völker U, Wilson JF, Gauderman WJ, Lambiase PD, Sotoodehnia N, Tinker A, Warren HR, Noordam R, Munroe PB. Genome-Wide Interaction Analyses of Serum Calcium on Ventricular Repolarization Time in 125 393 Participants. J Am Heart Assoc 2024; 13:e034760. [PMID: 39206732 PMCID: PMC11646519 DOI: 10.1161/jaha.123.034760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Ventricular repolarization time (ECG QT and JT intervals) is associated with malignant arrhythmia. Genome-wide association studies have identified 230 independent loci for QT and JT; however, 50% of their heritability remains unexplained. Previous work supports a causal effect of lower serum calcium concentrations on longer ventricular repolarization time. We hypothesized calcium interactions with QT and JT variant associations could explain a proportion of the missing heritability. METHODS AND RESULTS We performed genome-wide calcium interaction analyses for QT and JT intervals. Participants were stratified by their calcium level relative to the study distribution (top or bottom 20%). We performed a 2-stage analysis (genome-wide discovery [N=62 532] and replication [N=59 861] of lead variants) and a single-stage genome-wide meta-analysis (N=122 393, [European ancestry N=117 581, African ancestry N=4812]). We also calculated 2-degrees of freedom joint main and interaction and 1-degree of freedom interaction P values. In 2-stage and single-stage analyses, 50 and 98 independent loci, respectively, were associated with either QT or JT intervals (2-degrees of freedom joint main and interaction P value <5×10-8). No lead variant had a significant interaction result after correcting for multiple testing and sensitivity analyses provided similar findings. Two loci in the single-stage meta-analysis were not reported previously (SPPL2B and RFX6). CONCLUSIONS We have found limited support for an interaction effect of serum calcium on QT and JT variant associations despite sample sizes with suitable power to detect relevant effects. Therefore, such effects are unlikely to explain a meaningful proportion of the heritability of QT and JT, and factors including rare variation and other environmental interactions need to be considered.
Collapse
Affiliation(s)
- William J. Young
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- Barts Heart CentreSt Bartholomew’s Hospital, Barts Health NHS TrustLondonUnited Kingdom
| | - Peter J. van der Most
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Department of Biostatistics and MedicineUniversity of WashingtonSeattleWAUSA
| | - Maxime M. Bos
- Department of EpidemiologyErasmus MC University Medical CenterRotterdamNetherlands
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of ItalyPaviaItaly
| | - ThuyVy Duong
- Department of Genetic MedicineMcKusick‐Nathans Institute, Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Luisa Foco
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
| | - Jesus T. Lominchar
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Martina Müller‐Nurasyid
- German Research Center for Environmental HealthInstitute of Genetic Epidemiology, Helmholtz Zentrum MünchenNeuherbergGermany
- IBE, Faculty of Medicine, LMU MunichMunichGermany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg UniversityMainzGermany
| | | | - Alessandro Pecori
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”TriesteItaly
| | - Julia Ramirez
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- Aragon Institute of Engineering Research, University of ZaragozaSpain
- Centro de Investigación Biomédica en Red—Bioingeniería, Biomateriales y NanomedicinaZaragozaSpain
| | - Linda Repetto
- Centre for Global Health ResearchUsher Institute, University of EdinburghScotland
| | - Katharina Schramm
- German Research Center for Environmental HealthInstitute of Genetic Epidemiology, Helmholtz Zentrum MünchenNeuherbergGermany
- IBE, Faculty of Medicine, LMU MunichMunichGermany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Xia Shen
- Centre for Global Health ResearchUsher Institute, University of EdinburghScotland
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan UniversityGuangzhouChina
| | - Stefan van Duijvenboden
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- Institute of Cardiovascular Sciences, University of College LondonLondonUnited Kingdom
- Nuffield Department of Population HealthUniversity of OxfordUnited Kingdom
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and GeriatricsLeiden University Medical CenterLeidenThe Netherlands
| | - Stefan Weiss
- DZHK (German Centre for Cardiovascular Research), partner site GreifswaldGreifswaldGermany
- Interfaculty Institute for Genetics and Functional Genomics; Department of Functional GenomicsUniversity Medicine GreifswaldGreifswaldGermany
| | - Jie Yao
- Department of PediatricsThe Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Jan‐Walter Benjamins
- Department of CardiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Alvaro Alonso
- Department of EpidemiologyRollins School of Public Health, Emory UniversityAtlantaGAUSA
| | - Beatrice Spedicati
- Department of Medicine, Surgery and Health SciencesUniversity of TriesteItaly
| | - Mary L. Biggs
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWAUSA
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWAUSA
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Research), partner site GreifswaldGreifswaldGermany
- Department of Internal Medicine B—Cardiology, Pneumology, Infectious Diseases, Intensive Care MedicineUniversity Medicine GreifswaldGreifswaldGermany
| | - Christian Fuchsberger
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
- Department of BiostatisticsUniversity of Michigan School of Public HealthAnn ArborMIUSA
- Center for Statistical GeneticsUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Martin Gögele
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor‐UCLA Medical CenterTorranceCAUSA
- Department of PediatricsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - M. Arfan Ikram
- Department of EpidemiologyErasmus MC University Medical CenterRotterdamNetherlands
| | - J. Wouter Jukema
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
- Netherlands Heart InstituteUtrechtThe Netherlands
| | - Stefan Kääb
- Department of CardiologyUniversity Hospital, LMU MunichMunichGermany
- DZHK (German Centre for Cardiovascular Research), partner site: Munich Heart AllianceMunichGermany
| | - Jørgen K. Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical SciencesUniversity of CopenhagenDenmark
| | | | - Henry J. Lin
- Department of PediatricsThe Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
- Department of PediatricsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Pediatrics/Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Allan Linneberg
- Center for Clinical Research and PreventionBispebjerg and Frederiksberg Hospital, The Capital RegionCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Matthias Nauck
- DZHK (German Centre for Cardiovascular Research), partner site GreifswaldGreifswaldGermany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine GreifswaldGreifswaldGermany
| | - Ilja M. Nolte
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Giulia Pianigiani
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”TriesteItaly
| | - Aurora Santin
- Department of Medicine, Surgery and Health SciencesUniversity of TriesteItaly
| | - Elsayed Z. Soliman
- Epidemiological Cardiology Research Center (EPICARE)Wake Forest School of MedicineWinston SalemUSA
| | - Paola Tesolin
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”TriesteItaly
| | - Simona Vaccargiu
- Institute for Genetic and Biomedical Research, National Research Council of ItalyCagliariItaly
| | - Melanie Waldenberger
- DZHK (German Centre for Cardiovascular Research), partner site: Munich Heart AllianceMunichGermany
- Research Unit Molecular EpidemiologyInstitute of Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental HealthNeuherbergGermany
| | - Pim van der Harst
- Department of CardiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Department of Cardiology, Heart and Lung DivisionUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Niek Verweij
- Department of CardiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Dan E. Arking
- Department of Genetic MedicineMcKusick‐Nathans Institute, Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Maria Pina Concas
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”TriesteItaly
| | - Alessandro De Grandi
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health SciencesUniversity of TriesteItaly
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Maryam Kavousi
- Department of EpidemiologyErasmus MC University Medical CenterRotterdamNetherlands
| | - Dennis O. Mook‐Kanamori
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Public Health and Primary CareLeiden University Medical CenterLeidenThe Netherlands
| | - Pau Navarro
- MRC Human Genetics UnitInstitute of Genetics and Cancer, University of EdinburghScotland
| | - Michele Orini
- Barts Heart CentreSt Bartholomew’s Hospital, Barts Health NHS TrustLondonUnited Kingdom
- Institute of Cardiovascular Sciences, University of College LondonLondonUnited Kingdom
| | | | - Cristian Pattaro
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
| | - Annette Peters
- German Research Center for Environmental HealthInstitute of Genetic Epidemiology, Helmholtz Zentrum MünchenNeuherbergGermany
- IBE, Faculty of Medicine, LMU MunichMunichGermany
- DZHK (German Centre for Cardiovascular Research), partner site: Munich Heart AllianceMunichGermany
| | - Mario Pirastu
- Institute for Genetic and Biomedical Research, Sassari Unit, National Research Council of ItalySassariItaly
| | - Peter P. Pramstaller
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
- Department of NeurologyUniversity of LübeckGermany
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
| | - Mortiz Sinner
- Department of CardiologyUniversity Hospital, LMU MunichMunichGermany
- DZHK (German Centre for Cardiovascular Research), partner site: Munich Heart AllianceMunichGermany
| | - Harold Snieder
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Uwe Völker
- DZHK (German Centre for Cardiovascular Research), partner site GreifswaldGreifswaldGermany
- Interfaculty Institute for Genetics and Functional Genomics; Department of Functional GenomicsUniversity Medicine GreifswaldGreifswaldGermany
| | - James F. Wilson
- Centre for Global Health ResearchUsher Institute, University of EdinburghScotland
- MRC Human Genetics UnitInstitute of Genetics and Cancer, University of EdinburghScotland
| | - W. James Gauderman
- Department of population and public health sciencesUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Pier D. Lambiase
- Barts Heart CentreSt Bartholomew’s Hospital, Barts Health NHS TrustLondonUnited Kingdom
- Institute of Cardiovascular Sciences, University of College LondonLondonUnited Kingdom
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of MedicineUniversity of WashingtonSeattleWAUSA
| | - Andrew Tinker
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- NIHR Barts Biomedical Research CentreBarts and The London Faculty of Medicine and Dentistry, Queen Mary University of LondonUnited Kingdom
| | - Helen R. Warren
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- NIHR Barts Biomedical Research CentreBarts and The London Faculty of Medicine and Dentistry, Queen Mary University of LondonUnited Kingdom
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and GeriatricsLeiden University Medical CenterLeidenThe Netherlands
| | - Patricia B. Munroe
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- NIHR Barts Biomedical Research CentreBarts and The London Faculty of Medicine and Dentistry, Queen Mary University of LondonUnited Kingdom
| |
Collapse
|
19
|
Pastika L, Sau A, Patlatzoglou K, Sieliwonczyk E, Ribeiro AH, McGurk KA, Khan S, Mandic D, Scott WR, Ware JS, Peters NS, Ribeiro ALP, Kramer DB, Waks JW, Ng FS. Artificial intelligence-enhanced electrocardiography derived body mass index as a predictor of future cardiometabolic disease. NPJ Digit Med 2024; 7:167. [PMID: 38918595 PMCID: PMC11199586 DOI: 10.1038/s41746-024-01170-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
The electrocardiogram (ECG) can capture obesity-related cardiac changes. Artificial intelligence-enhanced ECG (AI-ECG) can identify subclinical disease. We trained an AI-ECG model to predict body mass index (BMI) from the ECG alone. Developed from 512,950 12-lead ECGs from the Beth Israel Deaconess Medical Center (BIDMC), a secondary care cohort, and validated on UK Biobank (UKB) (n = 42,386), the model achieved a Pearson correlation coefficient (r) of 0.65 and 0.62, and an R2 of 0.43 and 0.39 in the BIDMC cohort and UK Biobank, respectively for AI-ECG BMI vs. measured BMI. We found delta-BMI, the difference between measured BMI and AI-ECG-predicted BMI (AI-ECG-BMI), to be a biomarker of cardiometabolic health. The top tertile of delta-BMI showed increased risk of future cardiometabolic disease (BIDMC: HR 1.15, p < 0.001; UKB: HR 1.58, p < 0.001) and diabetes mellitus (BIDMC: HR 1.25, p < 0.001; UKB: HR 2.28, p < 0.001) after adjusting for covariates including measured BMI. Significant enhancements in model fit, reclassification and improvements in discriminatory power were observed with the inclusion of delta-BMI in both cohorts. Phenotypic profiling highlighted associations between delta-BMI and cardiometabolic diseases, anthropometric measures of truncal obesity, and pericardial fat mass. Metabolic and proteomic profiling associates delta-BMI positively with valine, lipids in small HDL, syntaxin-3, and carnosine dipeptidase 1, and inversely with glutamine, glycine, colipase, and adiponectin. A genome-wide association study revealed associations with regulators of cardiovascular/metabolic traits, including SCN10A, SCN5A, EXOG and RXRG. In summary, our AI-ECG-BMI model accurately predicts BMI and introduces delta-BMI as a non-invasive biomarker for cardiometabolic risk stratification.
Collapse
Affiliation(s)
- Libor Pastika
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Arunashis Sau
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Cardiology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Ewa Sieliwonczyk
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Laboratory of Medical Sciences, Imperial College London, London, United Kingdom
| | - Antônio H Ribeiro
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Kathryn A McGurk
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Laboratory of Medical Sciences, Imperial College London, London, United Kingdom
| | - Sadia Khan
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Chelsea and Westminster NHS Foundation Trust, London, United Kingdom
| | - Danilo Mandic
- Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - William R Scott
- MRC Laboratory of Medical Sciences, Imperial College London, London, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Laboratory of Medical Sciences, Imperial College London, London, United Kingdom
| | - Nicholas S Peters
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Cardiology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Antonio Luiz P Ribeiro
- Department of Internal Medicine, Faculdade de Medicina, and Telehealth Center and Cardiology Service, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniel B Kramer
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jonathan W Waks
- Harvard-Thorndike Electrophysiology Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Fu Siong Ng
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
- Department of Cardiology, Imperial College Healthcare NHS Trust, London, United Kingdom.
- Chelsea and Westminster NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
20
|
Simon ST, Lin M, Trinkley KE, Aleong R, Rafaels N, Crooks KR, Reiter MJ, Gignoux CR, Rosenberg MA. A polygenic risk score for the QT interval is an independent predictor of drug-induced QT prolongation. PLoS One 2024; 19:e0303261. [PMID: 38885227 PMCID: PMC11182491 DOI: 10.1371/journal.pone.0303261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 04/23/2024] [Indexed: 06/20/2024] Open
Abstract
Drug-induced QT prolongation (diLQTS), and subsequent risk of torsade de pointes, is a major concern with use of many medications, including for non-cardiac conditions. The possibility that genetic risk, in the form of polygenic risk scores (PGS), could be integrated into prediction of risk of diLQTS has great potential, although it is unknown how genetic risk is related to clinical risk factors as might be applied in clinical decision-making. In this study, we examined the PGS for QT interval in 2500 subjects exposed to a known QT-prolonging drug on prolongation of the QT interval over 500ms on subsequent ECG using electronic health record data. We found that the normalized QT PGS was higher in cases than controls (0.212±0.954 vs. -0.0270±1.003, P = 0.0002), with an unadjusted odds ratio of 1.34 (95%CI 1.17-1.53, P<0.001) for association with diLQTS. When included with age and clinical predictors of QT prolongation, we found that the PGS for QT interval provided independent risk prediction for diLQTS, in which the interaction for high-risk diagnosis or with certain high-risk medications (amiodarone, sotalol, and dofetilide) was not significant, indicating that genetic risk did not modify the effect of other risk factors on risk of diLQTS. We found that a high-risk cutoff (QT PGS ≥ 2 standard deviations above mean), but not a low-risk cutoff, was associated with risk of diLQTS after adjustment for clinical factors, and provided one method of integration based on the decision-tree framework. In conclusion, we found that PGS for QT interval is an independent predictor of diLQTS, but that in contrast to existing theories about repolarization reserve as a mechanism of increasing risk, the effect is independent of other clinical risk factors. More work is needed for external validation in clinical decision-making, as well as defining the mechanism through which genes that increase QT interval are associated with risk of diLQTS.
Collapse
Affiliation(s)
- Steven T. Simon
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Meng Lin
- Colorado Center for Personalized Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Katy E. Trinkley
- Department of Clinical Pharmacy, School of Pharmacy, University of Colorado, Aurora, CO, United States of America
| | - Ryan Aleong
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Nicholas Rafaels
- Colorado Center for Personalized Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Kristy R. Crooks
- Colorado Center for Personalized Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Michael J. Reiter
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Christopher R. Gignoux
- Colorado Center for Personalized Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Michael A. Rosenberg
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| |
Collapse
|
21
|
Yu J, Ren W, Yuan J, Liu R, Ma L, Tang S, Pang Y. Identification of urine biomarkers predictive of prolonged QTc interval in multidrug-resistant tuberculosis patients treated with bedaquiline. Front Pharmacol 2024; 15:1362544. [PMID: 38873419 PMCID: PMC11169739 DOI: 10.3389/fphar.2024.1362544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
The most frequent adverse event associated with bedaquiline (BDQ) is the QTc interval prolongation; however, there was no biomarkers that could be used to predict the occurrence of QTc prolongation in BDQ-treated patients. In this study, we employed the ultra-high performance liquid chromatography-MS/MS (UHPLC-MS/MS) to generate metabolic profiling for the discovery of potential predictive urine biomarkers of QTc prolongation in these patients. Untargeted metabolomic technique was used to concentrate the differential metabolic pathway, and targeted metabolomic technique was subsequently performed to identify predictive biomarkers for QTc prolongation. A total of 45 rifampicin-resistant TB (RR-TB) and multidrug-resistant TB (MDR-TB) patients were enrolled in our study, including 15 RR/MDR-TB patients with QTc interval prolongation (QIP) and 30 RR/MDR-TB patients with QTc interval un-prolongations (QIU). Untargeted technique revealed that the lipid metabolism was the most differential metabolic pathway between two groups. Further targeted technique identified four differential metabolites, including betaine, LPE (18:2), LPE (20:3), and LPE (20:4). The combined analysis of metabolisms revealed that the combined use of LPE (20:3) and LPE (20:4) had the best performance for predicting the occurrence of QTc prolongation in TB patients, yielding a sensitivity of 87.4% and a specificity of 78.5%. In addition, with the progression of BDQ treatment, the LPEs exhibited persistent difference in the BDQ-treated TB patients experiencing QTc interval prolongation. In conclusion, our data demonstrate that the combined use of LPE (20:3) and LPE (20:4) yields promising performance for predicting the occurrence of QTc interval prolongation in BDQ-treated patients.
Collapse
Affiliation(s)
- Jiajia Yu
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Weicong Ren
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Jinfeng Yuan
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Rongmei Liu
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Liping Ma
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Shenjie Tang
- Clinical Center on Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Pang
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
22
|
Garbutt TA, Wang Z, Wang H, Ma H, Ruan H, Dong Y, Xie Y, Tan L, Phookan R, Stouffer J, Vedantham V, Yang Y, Qian L, Liu J. Epigenetic Regulation of Cardiomyocyte Maturation by Arginine Methyltransferase CARM1. Circulation 2024; 149:1501-1515. [PMID: 38223978 PMCID: PMC11073921 DOI: 10.1161/circulationaha.121.055738] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/19/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND During the neonatal stage, the cardiomyocyte undergoes a constellation of molecular, cytoarchitectural, and functional changes known collectively as cardiomyocyte maturation to increase myocardial contractility and cardiac output. Despite the importance of cardiomyocyte maturation, the molecular mechanisms governing this critical process remain largely unexplored. METHODS We leveraged an in vivo mosaic knockout system to characterize the role of Carm1, the founding member of protein arginine methyltransferase, in cardiomyocyte maturation. Using a battery of assays, including immunohistochemistry, immuno-electron microscopy imaging, and action potential recording, we assessed the effect of loss of Carm1 function on cardiomyocyte cell growth, myofibril expansion, T-tubule formation, and electrophysiological maturation. Genome-wide transcriptome profiling, H3R17me2a chromatin immunoprecipitation followed by sequencing, and assay for transposase-accessible chromatin with high-throughput sequencing were used to investigate the mechanisms by which CARM1 (coactivator-associated arginine methyltransferase 1) regulates cardiomyocyte maturation. Finally, we interrogated the human syntenic region to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks for single-nucleotide polymorphisms associated with human heart diseases. RESULTS We report that mosaic ablation of Carm1 disrupts multiple aspects of cardiomyocyte maturation cell autonomously, leading to reduced cardiomyocyte size and sarcomere thickness, severe loss and disorganization of T tubules, and compromised electrophysiological maturation. Genomics study demonstrates that CARM1 directly activates genes that underlie cardiomyocyte cytoarchitectural and electrophysiological maturation. Moreover, our study reveals significant enrichment of human heart disease-associated single-nucleotide polymorphisms in the human genomic region syntenic to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks. CONCLUSIONS This study establishes a critical and multifaceted role for CARM1 in regulating cardiomyocyte maturation and demonstrates that deregulation of CARM1-dependent cardiomyocyte maturation gene expression may contribute to human heart diseases.
Collapse
Affiliation(s)
- Tiffany A. Garbutt
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zhenhua Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cardiovascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Ma
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Present address: Department of Cardiology, 2 Affiliated Hospital, School of Medicine, Zhejiang University. Hangzhou 310009, China
| | - Hongmei Ruan
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yanhan Dong
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yifang Xie
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Lianmei Tan
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ranan Phookan
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joy Stouffer
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Vasanth Vedantham
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
23
|
Caruso E, Farruggio S, Guccione P. Calmodulin mutation in long QT syndrome 15 associated with congenital heart defects further complicated by a functional 2:1 atrioventricular block: Management from foetal life to postpartum. Indian Pacing Electrophysiol J 2024; 24:150-154. [PMID: 38281621 PMCID: PMC11143731 DOI: 10.1016/j.ipej.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 12/27/2023] [Accepted: 01/17/2024] [Indexed: 01/30/2024] Open
Abstract
We report a long QT syndrome 15 whose diagnosis was suspected during foetal life and confirmed at birth and was associated with congenital heart disease. Genetic testing revealed a rare mutation associated with the CALM2 gene. At 23 weeks of gestation, severe foetal sinus bradycardia (∼100 bpm) was detected. In the third trimester, the foetus developed severe right ventricular hypertrophy. At birth, the electrocardiogram showed a long QT interval of 640 ms, and after 1 hour, the newborn showed functional 2:1 atrioventricular block at ventricular rate of 50 bpm. After further pharmacological therapies, epicardial wires were surgically implanted for transient pacing in VVI mode at 90 bpm. Echocardiogram showed aneurysmatic left atrial appendage, dilated right segments, hypertrophied right ventricle, ostium secundum type atrial septal defect, and muscular ventricular septal defect. At two weeks of postpartum, a permanent dual-chamber pacemaker was implanted in the DDD mode and the patient was discharged with a prescription of beta-blockers and calcium therapy.
Collapse
Affiliation(s)
- Elio Caruso
- Mediterranean Pediatric Cardiology Center "Bambino Gesù", San Vincenzo Hospital, Taormina, ME, Italy
| | - Silvia Farruggio
- Mediterranean Pediatric Cardiology Center "Bambino Gesù", San Vincenzo Hospital, Taormina, ME, Italy.
| | - Paolo Guccione
- Mediterranean Pediatric Cardiology Center "Bambino Gesù", San Vincenzo Hospital, Taormina, ME, Italy
| |
Collapse
|
24
|
Pironet A, Vandewiele F, Vennekens R. Exploring the role of TRPM4 in calcium-dependent triggered activity and cardiac arrhythmias. J Physiol 2024; 602:1605-1621. [PMID: 37128952 DOI: 10.1113/jp283831] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023] Open
Abstract
Cardiac arrhythmias pose a major threat to a patient's health, yet prove to be often difficult to predict, prevent and treat. A key mechanism in the occurrence of arrhythmias is disturbed Ca2+ homeostasis in cardiac muscle cells. As a Ca2+-activated non-selective cation channel, TRPM4 has been linked to Ca2+-induced arrhythmias, potentially contributing to translating an increase in intracellular Ca2+ concentration into membrane depolarisation and an increase in cellular excitability. Indeed, evidence from genetically modified mice, analysis of mutations in human patients and the identification of a TRPM4 blocking compound that can be applied in vivo further underscore this hypothesis. Here, we provide an overview of these data in the context of our current understanding of Ca2+-dependent arrhythmias.
Collapse
Affiliation(s)
- Andy Pironet
- Laboratory of Ion Channel Research, VIB Centre for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frone Vandewiele
- Laboratory of Ion Channel Research, VIB Centre for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, VIB Centre for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Ryan T, Roberts JD. Stem cell models of inherited arrhythmias. NATURE CARDIOVASCULAR RESEARCH 2024; 3:420-430. [PMID: 39196215 DOI: 10.1038/s44161-024-00451-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/29/2024] [Indexed: 08/29/2024]
Abstract
Inherited arrhythmias are a heterogeneous group of conditions that confer risk of sudden death. Many inherited arrhythmias have been linked to pathogenic genetic variants that result in ion channel dysfunction, although current genetic testing panels fail to identify variants in many patients, potentially secondary to their underlying substrates being oligogenic or polygenic. Here we review the current state of knowledge surrounding the cellular mechanisms of inherited arrhythmias generated from stem cell models with a focus on integrating genetic and mechanistic data. The utility and limitations of human induced pluripotent stem cell models in disease modeling and drug development are also explored with a particular focus on examples of pharmacogenetics and precision medicine. We submit that progress in understanding inherited arrhythmias is likely to be made by using human induced pluripotent stem cells to model probable polygenic cases as well as to interrogate the diverse and potentially complex molecular networks implicated by genome-wide association studies.
Collapse
Affiliation(s)
- Tammy Ryan
- McMaster University, Hamilton, Ontario, Canada.
| | - Jason D Roberts
- McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute and Hamilton Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
26
|
Kadagandla S, Kapoor A. Identification of candidate causal cis -regulatory variants underlying electrocardiographic QT interval GWAS loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.584880. [PMID: 38585875 PMCID: PMC10996567 DOI: 10.1101/2024.03.13.584880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Identifying causal variants among tens or hundreds of associated variants at each locus mapped by genome-wide association studies (GWAS) of complex traits is a challenge. As vast majority of GWAS variants are noncoding, sequence variation at cis -regulatory elements affecting transcriptional expression of specific genes is a widely accepted molecular hypothesis. Following this cis -regulatory hypothesis and combining it with the observation that nucleosome-free open chromatin is a universal hallmark of all types of cis -regulatory elements, we aimed to identify candidate causal regulatory variants underlying electrocardiographic QT interval GWAS loci. At a dozen loci, selected for higher effect sizes and a better understanding of the likely causal gene, we identified and included all common variants in high linkage disequilibrium with the GWAS variants as candidate variants. Using ENCODE DNase-seq and ATAC-seq from multiple human adult cardiac left ventricle tissue samples, we generated genome-wide maps of open chromatin regions marking putative regulatory elements. QT interval associated candidate variants were filtered for overlap with cardiac left ventricle open chromatin regions to identify candidate causal cis -regulatory variants, which were further assessed for colocalizing with a known cardiac GTEx expression quantitative trait locus variant as additional evidence for their causal role. Together, these efforts have generated a comprehensive set of candidate causal variants that are expected to be enriched for cis -regulatory potential and thereby, explaining the observed genetic associations.
Collapse
|
27
|
Gunamalai L, Singh P, Berg B, Shi L, Sanchez E, Smith A, Breton G, Bedford MT, Balciunas D, Kapoor A. Functional characterization of QT interval associated SCN5A enhancer variants identify combined additive effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584440. [PMID: 38559211 PMCID: PMC10979898 DOI: 10.1101/2024.03.11.584440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Several empirical and theoretical studies suggest presence of multiple enhancers per gene that collectively regulate gene expression, and that common sequence variation impacting on the activities of these enhancers is a major source of inter-individual variability in gene expression. However, for vast majority of genes, enhancers and the underlying regulatory variation remains unknown. Even for the genes with well-characterized enhancers, the nature of the combined effects from multiple enhancers and their variants, when known, on gene expression regulation remains unexplored. Here, we have evaluated the combined effects from five SCN5A enhancers and their regulatory variants that are known to collectively correlate with SCN5A cardiac expression and underlie QT interval association in the general population. Using small deletions centered at the regulatory variants in episomal reporter assays in a mouse cardiomyocyte cell line we demonstrate that the variants and their flanking sequences play critical role in individual enhancer activities, likely being a transcription factor (TF) binding site. By performing oligonucleotide-based pulldown assays on predicted TFs we identify the TFs likely driving allele-specific enhancer activities. Using all 32 possible allelic synthetic constructs in reporter assays, representing the five biallelic enhancers in tandem in their genomic order, we demonstrate combined additive effects on overall enhancer activities. Using transient enhancer assays in developing zebrafish embryos we demonstrate the four out the five enhancer elements act as enhancers in vivo . Together, these studies extend the previous findings to uncover the TFs driving the enhancer activities of QT interval associated SCN5A regulatory variants, reveal the additive effects from allelic combinations of these regulatory variants, and prove their potential to act as enhancers in vivo .
Collapse
|
28
|
Kabakov AY, Roder K, Bronk P, Turan NN, Dhakal S, Zhong M, Lu Y, Zeltzer ZA, Najman-Licht YB, Karma A, Koren G. E3 ubiquitin ligase rififylin has yin and yang effects on rabbit cardiac transient outward potassium currents (I to) and corresponding channel proteins. J Biol Chem 2024; 300:105759. [PMID: 38367666 PMCID: PMC10945274 DOI: 10.1016/j.jbc.2024.105759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 02/19/2024] Open
Abstract
Genome-wide association studies have reported a correlation between a SNP of the RING finger E3 ubiquitin protein ligase rififylin (RFFL) and QT interval variability in humans (Newton-Cheh et al., 2009). Previously, we have shown that RFFL downregulates expression and function of the human-like ether-a-go-go-related gene potassium channel and corresponding rapidly activating delayed rectifier potassium current (IKr) in adult rabbit ventricular cardiomyocytes. Here, we report that RFFL also affects the transient outward current (Ito), but in a peculiar way. RFFL overexpression in adult rabbit ventricular cardiomyocytes significantly decreases the contribution of its fast component (Ito,f) from 35% to 21% and increases the contribution of its slow component (Ito,s) from 65% to 79%. Since Ito,f in rabbits is mainly conducted by Kv4.3, we investigated the effect of RFFL on Kv4.3 expressed in HEK293A cells. We found that RFFL overexpression reduced Kv4.3 expression and corresponding Ito,f in a RING domain-dependent manner in the presence or absence of its accessory subunit Kv channel-interacting protein 2. On the other hand, RFFL overexpression in Kv1.4-expressing HEK cells leads to an increase in both Kv1.4 expression level and Ito,s, similarly in a RING domain-dependent manner. Our physiologically detailed rabbit ventricular myocyte computational model shows that these yin and yang effects of RFFL overexpression on Ito,f, and Ito,s affect phase 1 of the action potential waveform and slightly decrease its duration in addition to suppressing IKr. Thus, RFFL modifies cardiac repolarization reserve via ubiquitination of multiple proteins that differently affect various potassium channels and cardiac action potential duration.
Collapse
Affiliation(s)
- Anatoli Y Kabakov
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Karim Roder
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Peter Bronk
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Nilüfer N Turan
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Saroj Dhakal
- Physics Department and Center for Interdisciplinary Research on Complex Systems, Northeastern University, Boston, Massachusetts, USA
| | - Mingwang Zhong
- Physics Department and Center for Interdisciplinary Research on Complex Systems, Northeastern University, Boston, Massachusetts, USA
| | - Yichun Lu
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Zachary A Zeltzer
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Yonatan B Najman-Licht
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Alain Karma
- Physics Department and Center for Interdisciplinary Research on Complex Systems, Northeastern University, Boston, Massachusetts, USA
| | - Gideon Koren
- Division of Cardiology, Department of Medicine, Cardiovascular Research Center, Rhode Island Hospital, The Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA.
| |
Collapse
|
29
|
Zhang M, Hillegass WB, Yu X, Majumdar S, Daryl Pollard J, Jackson E, Knudson J, Wolfe D, Kato GJ, Maher JF, Mei H. Genetic variants and effect modifiers of QT interval prolongation in patients with sickle cell disease. Gene 2024; 890:147824. [PMID: 37741592 DOI: 10.1016/j.gene.2023.147824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Sickle cell disease (SCD) is a common inherited blood disorder among African Americans (AA), with premature mortality which has been associated with prolongation of the heart rate-corrected QT interval (QTc), a known risk factor for sudden cardiac death. Although numerous genetic variants have been identified as contributors to QT interval prolongation in the general population, their impact on SCD patients remains unclear. This study used an unweighted polygenic risk score (PRS) to validate the previously identified associations between SNPs and QTc interval in SCD patients, and to explore possible interactions with other factors that prolong QTc interval in AA individuals with SCD. METHODS In SCD patients, candidate genetic variants associated with the QTc interval were genotyped. To identify any risk SNPs that may be correlated with QTc interval prolongation, linear regression was employed, and an unweighted PRS was subsequently constructed. The effect of PRS on the QTc interval was evaluated using linear regression, while stratification analysis was used to assess the influence of serum alanine transaminase (ALT), a biomarker for liver disease, on the PRS effect. We also evaluated the PRS with the two subcomponents of QTc, the QRS and JTc intervals. RESULTS Out of 26 candidate SNPs, five risk SNPs were identified for QTc duration under the recessive model. For every unit increase in PRS, the QTc interval prolonged by 4.0 ms (95% CI: [2.0, 6.1]; p-value: <0.001) in the additive model and 9.4 ms in the recessive model (95% CI: [4.6, 14.1]; p-value: <0.001). Serum ALT showed a modification effect on PRS-QTc prolongation under the recessive model. In the normal ALT group, each PRS unit increased QTc interval by 11.7 ms (95% CI: [6.3, 17.1]; p-value: 2.60E-5), whereas this effect was not observed in the elevated ALT group (0.9 ms; 95% CI: [-7.0, 8.8]; p-value: 0.823). CONCLUSION Several candidate genetic variants are associated with QTc interval prolongation in SCD patients, and serum ALT acts as a modifying factor. The association of a CPS1 gene variant in both QTc and JTc duration adds to NOS1AP as evidence of involvement of the urea cycle and nitric oxide metabolism in cardiac repolarization in SCD. Larger replication studies are needed to confirm these findings and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Mengna Zhang
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - William B Hillegass
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Xue Yu
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Suvankar Majumdar
- Division of Hematology, Children's National Hospital, Washington, DC, USA
| | - J Daryl Pollard
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Erin Jackson
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jarrod Knudson
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Douglas Wolfe
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Gregory J Kato
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Joseph F Maher
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Internal Medicine/Cancer Genetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA.
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
30
|
Schulze-Bahr E, Dittmann S. Human Genetics of Cardiac Arrhythmias. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:1033-1055. [PMID: 38884768 DOI: 10.1007/978-3-031-44087-8_66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Inherited forms of cardiac arrhythmias mostly are rare diseases (prevalence <1:2000) and considered to be either "primary electrical heart disorders" due to the absence of structural heart abnormalities or "cardiac ion channel disorders" due to the myocellular structures involved. Precise knowledge of the electrocardiographic features of these diseases and their genetic classification will enable early disease recognition and prevention of cardiac events including sudden cardiac death.The genetic background of these diseases is complex and heterogeneous. In addition to the predominant "private character" of a mutation in each family, locus heterogeneity involving many ion channel genes for the same familial arrhythmia syndrome is typical. Founder pathogenic variants or mutational hot spots are uncommon. Moreover, phenotypes may vary and overlap even within the same family and mutation carriers. For the majority of arrhythmias, the clinical phenotype of an ion channel mutation is restricted to cardiac tissue, and therefore, the disease is nonsyndromic.Recent and innovative methods of parallel DNA analysis (so-called next-generation sequencing, NGS) will enhance further mutation and other variant detection as well as arrhythmia gene identification.
Collapse
Affiliation(s)
- Eric Schulze-Bahr
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany.
| | - Sven Dittmann
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany
| |
Collapse
|
31
|
Arora A, Zareba W, Woosley RL, Klimentidis YC, Patel IY, Quan SF, Wendel C, Shamoun F, Guerra S, Parthasarathy S, Patel SI. Genetic QT Score and Sleep Apnea as Predictors of Sudden Cardiac Death in the UK Biobank. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.07.23298237. [PMID: 37986981 PMCID: PMC10659512 DOI: 10.1101/2023.11.07.23298237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Introduction The goal of this study was to evaluate the association between a polygenic risk score (PRS) for QT prolongation (QTc-PRS), QTc intervals and mortality in patients enrolled in the UK Biobank with and without sleep apnea. Methods The QTc-PRS was calculated using allele copy number and previously reported effect estimates for each single nuclear polymorphism SNP. Competing-risk regression models adjusting for age, sex, BMI, QT prolonging medication, race, and comorbid cardiovascular conditions were used for sudden cardiac death (SCD) analyses. Results 500,584 participants were evaluated (56.5 ±8 years, 54% women, 1.4% diagnosed with sleep apnea). A higher QTc-PRS was independently associated with the increased QTc interval duration (p<0.0001). The mean QTc for the top QTc-PRS quintile was 15 msec longer than the bottom quintile (p<0.001). Sleep apnea was found to be an effect modifier in the relationship between QTc-PRS and SCD. The adjusted HR per 5-unit change in QTc-PRS for SCD was 1.64 (95% CI 1.16 - 2.31, p=0.005) among those with sleep apnea and 1.04 (95% CI 0.95 - 1.14, p=0.44) among those without sleep apnea (p for interaction =0.01). Black participants with sleep apnea had significantly elevated adjusted risk of SCD compared to White participants (HR=9.6, 95% CI 1.24 - 74, p=0.03). Conclusion In the UK Biobank population, the QTc-PRS was associated with SCD among participants with sleep apnea but not among those without sleep apnea, indicating that sleep apnea is a significant modifier of the genetic risk. Black participants with sleep apnea had a particularly high risk of SCD.
Collapse
|
32
|
Smith A, Auer D, Johnson M, Sanchez E, Ross H, Ward C, Chakravarti A, Kapoor A. Cardiac muscle-restricted partial loss of Nos1ap expression has limited but significant impact on electrocardiographic features. G3 (BETHESDA, MD.) 2023; 13:jkad208. [PMID: 37708408 PMCID: PMC10627271 DOI: 10.1093/g3journal/jkad208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 08/16/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023]
Abstract
Genome-wide association studies have identified sequence polymorphisms in a functional enhancer of the NOS1AP gene as the most common genetic regulator of QT interval and human cardiac NOS1AP gene expression in the general population. Functional studies based on in vitro overexpression in murine cardiomyocytes and ex vivo knockdown in zebrafish embryonic hearts, by us and others, have also demonstrated that NOS1AP expression levels can alter cellular electrophysiology. Here, to explore the role of NOS1AP in cardiac electrophysiology at an organismal level, we generated and characterized constitutive and heart muscle-restricted Nos1ap knockout mice to assess whether NOS1AP disruption alters the QT interval in vivo. Constitutive loss of Nos1ap led to genetic background-dependent variable lethality at or right before birth. Heart muscle-restricted Nos1ap knockout, generated using cardiac-specific alpha-myosin heavy chain promoter-driven tamoxifen-inducible Cre, resulted in tissue-level Nos1ap expression reduced by half. This partial loss of expression had no detectable effect on the QT interval or other electrocardiographic and echocardiographic parameters, except for a small but significant reduction in the QRS interval. Given that challenges associated with defining the end of the T wave on murine electrocardiogram can limit identification of subtle effects on the QT interval and that common noncoding NOS1AP variants are also associated with the QRS interval, our findings support the role of NOS1AP in regulation of the cardiac electrical cycle.
Collapse
Affiliation(s)
- Alexa Smith
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Dallas Auer
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Morgan Johnson
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ernesto Sanchez
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Holly Ross
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christopher Ward
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Aravinda Chakravarti
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Human Genetics and Genomics, New York University School of Medicine, New York, NY 10016, USA
| | - Ashish Kapoor
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
33
|
He J, Ding Y, Lin H, Liu X, Chen X, Shen W, Zhou S, Feng C, Wang M, Xia J, He N. Differential genome-wide associated variants and enriched pathways of ECG parameters among people with versus without HIV. AIDS 2023; 37:1871-1882. [PMID: 37418550 PMCID: PMC10481915 DOI: 10.1097/qad.0000000000003647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/09/2023]
Abstract
OBJECTIVES People with HIV (PWH) are more likely to develop ECG abnormalities. Substantial evidence exists for genetic contribution to ECG parameters among general population. However, whether and how would host genome associate with ECG parameters among PWH is unclear. Our research aims to analyze and compare genetic variants, mapped genes, and enriched pathways of ECG parameters among PWH and HIV-negative controls. DESIGN A cross-sectional study. METHOD We performed a large original genome-wide association study (GWAS) of ECG parameters among PWH ( n = 1730) and HIV-negative controls ( n = 3746). Genome-wide interaction analyses were also conducted. RESULTS A total of 18 novel variants were detected among PWH, six for PR interval including rs76345397 at ATL2 , 11 for QRS duration including rs10483994 at KCNK10 and rs2478830 at JCAD , and one for QTc interval (rs9815364). Among HIV-negative controls, we identified variants located at previously reported ECG-related genes ( SCN5A , CNOT1 ). Genetic variants had a significant interaction with HIV infection ( P < 5 × 10 -8 ), implying that HIV infection and host genome might jointly influence ECG parameters. Mapped genes for PR interval and QRS duration among PWH were enriched in the biological process of viral genome replication and host response to virus, respectively, whereas enriched pathways for PR interval among HIV-negative controls were in the cellular component of voltage-gated sodium channel complex. CONCLUSION The present GWAS indicated a distinctive impact of host genome on quantitative ECG parameters among PWH. Different from HIV-negative controls, host genome might influence the cardiac electrical activity by interfering with HIV viral infection, production, and latency among PWH.
Collapse
Affiliation(s)
- Jiayu He
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai
| | - Yingying Ding
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Haijiang Lin
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Taizhou City Center for Disease Control and Prevention, Zhejiang Province, China
| | - Xing Liu
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Xiaoxiao Chen
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Taizhou City Center for Disease Control and Prevention, Zhejiang Province, China
| | - Weiwei Shen
- Taizhou City Center for Disease Control and Prevention, Zhejiang Province, China
| | - Sujuan Zhou
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai
| | - Cheng Feng
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Miaochen Wang
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Jingjing Xia
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
| | - Na He
- Department of Epidemiology, School of Public Health, and Key Laboratory of Public Health Safety of Ministry of Education
- Yi-Wu Research Institute
- Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai
| |
Collapse
|
34
|
Tchou G, Ponce-Balbuena D, Liu N, Gore-Panter S, Hsu J, Liu F, Opoku E, Brubaker G, Schumacher SM, Moravec CS, Barnard J, Van Wagoner DR, Chung MK, Smith JD. Decreased FAM13B Expression Increases Atrial Fibrillation Susceptibility by Regulating Sodium Current and Calcium Handling. JACC Basic Transl Sci 2023; 8:1357-1378. [PMID: 38094680 PMCID: PMC10714175 DOI: 10.1016/j.jacbts.2023.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 04/17/2024]
Abstract
A specific genetic variant associated with atrial fibrillation risk, rs17171731, was identified as a regulatory variant responsible for controlling FAM13B expression. The atrial fibrillation risk allele decreases FAM13B expression, whose knockdown alters the expression of many genes in stem cell-derived cardiomyocytes, including SCN2B, and led to pro-arrhythmogenic changes in the late sodium current and Ca2+ cycling. Fam13b knockout mice had increased P-wave and QT interval duration and were more susceptible to pacing-induced arrhythmias vs control mice. FAM13B expression, its regulation, and downstream effects are potential targets for investigation of patient-specific therapeutics.
Collapse
Affiliation(s)
- Gregory Tchou
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Nana Liu
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Shamone Gore-Panter
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jeffrey Hsu
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Fang Liu
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Emmanuel Opoku
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Gregory Brubaker
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sarah M. Schumacher
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christine S. Moravec
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - John Barnard
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - David R. Van Wagoner
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mina K. Chung
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jonathan D. Smith
- Departments of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
35
|
Selewa A, Luo K, Wasney M, Smith L, Sun X, Tang C, Eckart H, Moskowitz IP, Basu A, He X, Pott S. Single-cell genomics improves the discovery of risk variants and genes of atrial fibrillation. Nat Commun 2023; 14:4999. [PMID: 37591828 PMCID: PMC10435551 DOI: 10.1038/s41467-023-40505-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
Genome-wide association studies (GWAS) have linked hundreds of loci to cardiac diseases. However, in most loci the causal variants and their target genes remain unknown. We developed a combined experimental and analytical approach that integrates single cell epigenomics with GWAS to prioritize risk variants and genes. We profiled accessible chromatin in single cells obtained from human hearts and leveraged the data to study genetics of Atrial Fibrillation (AF), the most common cardiac arrhythmia. Enrichment analysis of AF risk variants using cell-type-resolved open chromatin regions (OCRs) implicated cardiomyocytes as the main mediator of AF risk. We then performed statistical fine-mapping, leveraging the information in OCRs, and identified putative causal variants in 122 AF-associated loci. Taking advantage of the fine-mapping results, our novel statistical procedure for gene discovery prioritized 46 high-confidence risk genes, highlighting transcription factors and signal transduction pathways important for heart development. In summary, our analysis provides a comprehensive map of AF risk variants and genes, and a general framework to integrate single-cell genomics with genetic studies of complex traits.
Collapse
Affiliation(s)
- Alan Selewa
- Biophysical Sciences Graduate Program, The University of Chicago, Chicago, IL, 60637, USA
| | - Kaixuan Luo
- Department of Human Genetics, The University of Chicago, Chicago, IL, 60637, USA
| | - Michael Wasney
- Department of Medicine, Section of Genetic Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Linsin Smith
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL, 60637, USA
| | - Xiaotong Sun
- Department of Human Genetics, The University of Chicago, Chicago, IL, 60637, USA
| | - Chenwei Tang
- The College, The University of Chicago, Chicago, IL, 60637, USA
| | - Heather Eckart
- Department of Medicine, Section of Genetic Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Ivan P Moskowitz
- Department of Human Genetics, The University of Chicago, Chicago, IL, 60637, USA
- Department of Pediatrics, The University of Chicago, Chicago, IL, 60637, USA
| | - Anindita Basu
- Department of Medicine, Section of Genetic Medicine, The University of Chicago, Chicago, IL, 60637, USA.
| | - Xin He
- Department of Human Genetics, The University of Chicago, Chicago, IL, 60637, USA.
| | - Sebastian Pott
- Department of Medicine, Section of Genetic Medicine, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
36
|
Christiansen MK, Kjær-Sørensen K, Clavsen NC, Dittmann S, Jensen MF, Guldbrandsen HØ, Pedersen LN, Sørensen RH, Lildballe DL, Müller K, Müller P, Vogel K, Rudic B, Borggrefe M, Oxvig C, Aalkjær C, Schulze-Bahr E, Matchkov V, Bundgaard H, Jensen HK. Genetic analysis identifies the SLC4A3 anion exchanger as a major gene for short QT syndrome. Heart Rhythm 2023; 20:1136-1143. [PMID: 36806574 DOI: 10.1016/j.hrthm.2023.02.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 02/07/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND A variant in the SLC4A3 anion exchanger has been identified as a novel cause of short QT syndrome (SQTS), but the clinical importance of SLC4A3 as a cause of SQTS or sudden cardiac death remains unknown. OBJECTIVE The purpose of this study was to investigate the prevalence of potential disease-causing variants in SQTS patients using gene panels including SLC4A3. METHODS In this multicenter study, genetic testing was performed in 34 index patients with SQTS. The pathogenicity of novel SLC4A3variants was validated in a zebrafish embryo heart model. RESULTS Potentially disease-causing variants were identified in 9 (26%) patients and were mainly (15%) located in SLC4A3: 4 patients heterozygous for novel nonsynonymous SLC4A3 variants-p.Arg600Cys, p.Arg621Trp, p.Glu852Asp, and p.Arg952His-and 1 patient with the known p.Arg370His variant. In other SQTS genes, potentially disease-causing variants were less frequent (2× in KCNQ1, 1× in KCNJ2, and CACNA1C each). SLC4A3 variant carriers (n = 5) had a similar heart rate but shorter QT and J point to T wave peak intervals than did noncarriers (n = 29). Knockdown of slc4a3 in zebrafish resulted in shortened heart rate-corrected QT intervals (calculated using the Bazett formula) that could be rescued by overexpression of the native human SLC4A3-encoded protein (AE3), but neither by the mutated AE3 variants p.Arg600Cys, p.Arg621Trp, p.Glu852Asp nor by p.Arg952His, suggesting pathogenicity of these variants. Dysfunction in slc4a3/AE3 was associated with alkaline cytosol and shortened action potential of cardiomyocytes. CONCLUSION In about a quarter of patients with SQTS, a potentially disease-causing variant can be identified. Nonsynonymous variants in SLC4A3 represent the most common cause of SQTS, underscoring the importance of including SLC4A3 in the genetic screening of patients with SQTS or sudden cardiac death.
Collapse
Affiliation(s)
| | - Kasper Kjær-Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Natacha C Clavsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Sven Dittmann
- Institut für Genetik von Herzerkrankungen (IfGH), Universitätsklinikum Münster, Münster, Germany
| | - Maja Fuhlendorff Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark; Department of Biomedicine, Aarhus University, Aarhus C, Denmark; Department of Clinical Medicine, Health, Aarhus University, Aarhus N, Denmark
| | | | | | | | | | - Klara Müller
- Institut für Genetik von Herzerkrankungen (IfGH), Universitätsklinikum Münster, Münster, Germany
| | - Patrick Müller
- Institut für Genetik von Herzerkrankungen (IfGH), Universitätsklinikum Münster, Münster, Germany
| | - Kira Vogel
- Institut für Genetik von Herzerkrankungen (IfGH), Universitätsklinikum Münster, Münster, Germany
| | - Boris Rudic
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine Mannheim, University of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, University Medical Centre Mannheim (UMM), Faculty of Medicine Mannheim, University of Heidelberg, European Center for AngioScience (ECAS), and DZHK (German Center for Cardiovascular Research) partner site Heidelberg/Mannheim, Mannheim, Germany
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | | | - Eric Schulze-Bahr
- Institut für Genetik von Herzerkrankungen (IfGH), Universitätsklinikum Münster, Münster, Germany; ERN Reference Center GUARD-Heart, Münster, Germany
| | | | - Henning Bundgaard
- Unit for Inherited Cardiovascular Diseases, The Heart Centre, National University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Kjærulf Jensen
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark; Department of Clinical Medicine, Health, Aarhus University, Aarhus N, Denmark; ERN Reference Center GUARD-Heart, Aarhus, Denmark
| |
Collapse
|
37
|
Skakkebæk A, Kjær-Sørensen K, Matchkov VV, Christensen LL, Just J, Cömert C, Andersen NH, Oxvig C, Gravholt CH. Dosage of the pseudoautosomal gene SLC25A6 is implicated in QTc interval duration. Sci Rep 2023; 13:12089. [PMID: 37495650 PMCID: PMC10372092 DOI: 10.1038/s41598-023-38867-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/16/2023] [Indexed: 07/28/2023] Open
Abstract
The genetic architecture of the QT interval, defined as the period from onset of depolarisation to completion of repolarisation of the ventricular myocardium, is incompletely understood. Only a minor part of the QT interval variation in the general population has been linked to autosomal variant loci. Altered X chromosome dosage in humans, as seen in sex chromosome aneuploidies such as Turner syndrome (TS) and Klinefelter syndrome (KS), is associated with altered QTc interval (heart rate corrected QT), indicating that genes, located in the pseudoautosomal region 1 of the X and Y chromosomes may contribute to QT interval variation. We investigate the dosage effect of the pseudoautosomal gene SLC25A6, encoding the membrane ADP/ATP translocase 3 in the inner mitochondrial membrane, on QTc interval duration. To this end we used human participants and in vivo zebrafish models. Analyses in humans, based on 44 patients with KS, 44 patients with TS, 59 male and 22 females, revealed a significant negative correlation between SLC25A6 expression level and QTc interval duration. Similarly, downregulation of slc25a6 in zebrafish increased QTc interval duration with pharmacological inhibition of KATP channels restoring the systolic duration, whereas overexpression of SLC25A6 shortened QTc, which was normalized by pharmacological activation of KATP channels. Our study demonstrate an inverse relationship between SLC25A6 dosage and QTc interval indicating that SLC25A6 contributes to QT interval variation.
Collapse
Affiliation(s)
- Anne Skakkebæk
- Department of Clinical Genetics, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | - Kasper Kjær-Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Lise-Lotte Christensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper Just
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Cagla Cömert
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | | | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Claus Højbjerg Gravholt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine and Medical Research Laboratories, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
38
|
Sveinbjornsson G, Benediktsdottir BD, Sigfusson G, Norland K, Davidsson OB, Thorolfsdottir RB, Tragante V, Arnadottir GA, Jensson BO, Katrinardottir H, Fridriksdottir R, Gudmundsdottir H, Aegisdottir HM, Fridriksson B, Thorgeirsson G, Magnusson V, Oddsson A, Sulem P, Gudbjartsson DF, Holm H, Arnar DO, Stefansson K. Screening for Rare Coding Variants That Associate With the QTc Interval in Iceland. J Am Heart Assoc 2023:e029845. [PMID: 37449562 PMCID: PMC10382112 DOI: 10.1161/jaha.123.029845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/17/2023] [Indexed: 07/18/2023]
Abstract
Background Long-QT syndrome (LQTS) is a cardiac repolarization abnormality that can lead to sudden cardiac death. The most common causes are rare coding variants in the genes KCNQ1, KCNH2, and SCN5A. The data on LQTS epidemiology are limited, and information on expressivity and penetrance of pathogenic variants is sparse. Methods and Results We screened for rare coding variants associated with the corrected QT (QTc) interval in Iceland. We explored the frequency of the identified variants, their penetrance, and their association with severe events. Twelve variants were associated with the QTc interval. Five in KCNQ1, 3 in KCNH2, 2 in cardiomyopathy genes MYBPC3 and PKP2, and 2 in genes where coding variants have not been associated with the QTc interval, ISOC1 and MYOM2. The combined carrier frequency of the 8 variants in the previously known LQTS genes was 530 per 100 000 individuals (1:190). p.Tyr315Cys and p.Leu273Phe in KCNQ1 were associated with having a mean QTc interval longer than 500 ms (P=4.2×10-7; odds ratio [OR], 38.6; P=8.4×10-10, OR, 26.5; respectively), and p.Leu273Phe was associated with sudden cardiac death (P=0.0034; OR, 2.99). p.Val215Met in KCNQ1 was carried by 1 in 280 Icelanders, had a smaller effect on the QTc interval (P=1.8×10-44; effect, 22.8 ms), and did not associate with severe clinical events. Conclusions The carrier frequency of associating variants in LQTS genes was higher than previous estimates of the prevalence of LQTS. The variants have variable effects on the QTc interval, and carriers of p.Tyr315Cys and p.Leu273Phe have a more severe disease than carriers of p.Val215Met. These data could lead to improved identification, risk stratification, and a more precise clinical approach to those with QTc prolongation.
Collapse
Affiliation(s)
| | - Bara D Benediktsdottir
- Internal Medicine, Landspitali-The National University Hospital of Iceland Reykjavik Iceland
| | - Gunnlaugur Sigfusson
- Children's Medical Center Landspítali-The National University Hospital of Iceland Reykjavík Iceland
| | | | | | | | | | | | | | | | | | | | | | | | | | - Vidar Magnusson
- The Capital District Fire and Rescue Service Reykjavik Iceland
- Department of Anesthesia, Landspitali The National University Hospital of Iceland Reykjavik Iceland
| | | | | | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Inc. Reykjavik Iceland
- Faculty of Electrical and Computer Engineering University of Iceland Reykjavik Iceland
| | - Hilma Holm
- deCODE Genetics/Amgen, Inc. Reykjavik Iceland
| | - David O Arnar
- deCODE Genetics/Amgen, Inc. Reykjavik Iceland
- Faculty of Medicine University of Iceland Reykjavik Iceland
- Cardiovascular Center, Landspitali The National University Hospital of Iceland Reykjavik Iceland
| | | |
Collapse
|
39
|
Maurya S, Mills RW, Kahnert K, Chiang DY, Bertoli G, Lundegaard PR, Duran MPH, Zhang M, Rothenberg E, George AL, MacRae CA, Delmar M, Lundby A. Outlining cardiac ion channel protein interactors and their signature in the human electrocardiogram. NATURE CARDIOVASCULAR RESEARCH 2023; 2:673-692. [PMID: 38666184 PMCID: PMC11041666 DOI: 10.1038/s44161-023-00294-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 05/31/2023] [Indexed: 04/28/2024]
Abstract
Protein-protein interactions are essential for normal cellular processes and signaling events. Defining these interaction networks is therefore crucial for understanding complex cellular functions and interpretation of disease-associated gene variants. We need to build a comprehensive picture of the interactions, their affinities and interdependencies in the specific organ to decipher hitherto poorly understood signaling mechanisms through ion channels. Here we report the experimental identification of the ensemble of protein interactors for 13 types of ion channels in murine cardiac tissue. Of these, we validated the functional importance of ten interactors on cardiac electrophysiology through genetic knockouts in zebrafish, gene silencing in mice, super-resolution microscopy and patch clamp experiments. Furthermore, we establish a computational framework to reconstruct human cardiomyocyte ion channel networks from deep proteome mapping of human heart tissue and human heart single-cell gene expression data. Finally, we integrate the ion channel interactome with human population genetics data to identify proteins that influence the electrocardiogram (ECG). We demonstrate that the combined channel network is enriched for proteins influencing the ECG, with 44% of the network proteins significantly associated with an ECG phenotype. Altogether, we define interactomes of 13 major cardiac ion channels, contextualize their relevance to human electrophysiology and validate functional roles of ten interactors, including two regulators of the sodium current (epsin-2 and gelsolin). Overall, our data provide a roadmap for our understanding of the molecular machinery that regulates cardiac electrophysiology.
Collapse
Affiliation(s)
- Svetlana Maurya
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert W. Mills
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Kahnert
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David Y. Chiang
- Cardiovascular Medicine Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Giorgia Bertoli
- Division of Cardiology, NYU School of Medicine, New York, NY USA
| | - Pia R. Lundegaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Mingliang Zhang
- Division of Cardiology, NYU School of Medicine, New York, NY USA
| | - Eli Rothenberg
- Division of Pharmacology, NYU School of Medicine, New York, NY USA
| | - Alfred L. George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Calum A. MacRae
- Cardiovascular Medicine Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Mario Delmar
- Division of Cardiology, NYU School of Medicine, New York, NY USA
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Ciochetti NP, Lugli-Moraes B, da Silva BS, Rovaris DL. Genome-wide association studies: utility and limitations for research in physiology. J Physiol 2023; 601:2771-2799. [PMID: 37208942 PMCID: PMC10527550 DOI: 10.1113/jp284241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/10/2023] [Indexed: 05/21/2023] Open
Abstract
Physiological systems are subject to interindividual variation encoded by genetics. Genome-wide association studies (GWAS) operate by surveying thousands of genetic variants from a substantial number of individuals and assessing their association to a trait of interest, be it a physiological variable, a molecular phenotype (e.g. gene expression), or even a disease or condition. Through a myriad of methods, GWAS downstream analyses then explore the functional consequences of each variant and attempt to ascertain a causal relationship to the phenotype of interest, as well as to delve into its links to other traits. This type of investigation allows mechanistic insights into physiological functions, pathological disturbances and shared biological processes between traits (i.e. pleiotropy). An exciting example is the discovery of a new thyroid hormone transporter (SLC17A4) and hormone metabolising enzyme (AADAT) from a GWAS on free thyroxine levels. Therefore, GWAS have substantially contributed with insights into physiology and have been shown to be useful in unveiling the genetic control underlying complex traits and pathological conditions; they will continue to do so with global collaborations and advances in genotyping technology. Finally, the increasing number of trans-ancestry GWAS and initiatives to include ancestry diversity in genomics will boost the power for discoveries, making them also applicable to non-European populations.
Collapse
Affiliation(s)
- Nicolas Pereira Ciochetti
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
| | - Beatriz Lugli-Moraes
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
| | - Bruna Santos da Silva
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
- Laboratory of Developmental Psychiatry, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Diego Luiz Rovaris
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Padmanabhan S, du Toit C, Dominiczak AF. Cardiovascular precision medicine - A pharmacogenomic perspective. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e28. [PMID: 38550953 PMCID: PMC10953758 DOI: 10.1017/pcm.2023.17] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/24/2023] [Accepted: 06/12/2023] [Indexed: 05/16/2024]
Abstract
Precision medicine envisages the integration of an individual's clinical and biological features obtained from laboratory tests, imaging, high-throughput omics and health records, to drive a personalised approach to diagnosis and treatment with a higher chance of success. As only up to half of patients respond to medication prescribed following the current one-size-fits-all treatment strategy, the need for a more personalised approach is evident. One of the routes to transforming healthcare through precision medicine is pharmacogenomics (PGx). Around 95% of the population is estimated to carry one or more actionable pharmacogenetic variants and over 75% of adults over 50 years old are on a prescription with a known PGx association. Whilst there are compelling examples of pharmacogenomic implementation in clinical practice, the case for cardiovascular PGx is still evolving. In this review, we shall summarise the current status of PGx in cardiovascular diseases and look at the key enablers and barriers to PGx implementation in clinical practice.
Collapse
Affiliation(s)
- Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Clea du Toit
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Anna F. Dominiczak
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
42
|
Miles C, Boukens BJ, Scrocco C, Wilde AA, Nademanee K, Haissaguerre M, Coronel R, Behr ER. Subepicardial Cardiomyopathy: A Disease Underlying J-Wave Syndromes and Idiopathic Ventricular Fibrillation. Circulation 2023; 147:1622-1633. [PMID: 37216437 PMCID: PMC11073566 DOI: 10.1161/circulationaha.122.061924] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 03/27/2023] [Indexed: 05/24/2023]
Abstract
Brugada syndrome (BrS), early repolarization syndrome (ERS), and idiopathic ventricular fibrillation (iVF) have long been considered primary electrical disorders associated with malignant ventricular arrhythmia and sudden cardiac death. However, recent studies have revealed the presence of subtle microstructural abnormalities of the extracellular matrix in some cases of BrS, ERS, and iVF, particularly within right ventricular subepicardial myocardium. Substrate-based ablation within this region has been shown to ameliorate the electrocardiographic phenotype and to reduce arrhythmia frequency in BrS. Patients with ERS and iVF may also exhibit low-voltage and fractionated electrograms in the ventricular subepicardial myocardium, which can be treated with ablation. A significant proportion of patients with BrS and ERS, as well as some iVF survivors, harbor pathogenic variants in the voltage-gated sodium channel gene, SCN5A, but the majority of genetic susceptibility of these disorders is likely to be polygenic. Here, we postulate that BrS, ERS, and iVF may form part of a spectrum of subtle subepicardial cardiomyopathy. We propose that impaired sodium current, along with genetic and environmental susceptibility, precipitates a reduction in epicardial conduction reserve, facilitating current-to-load mismatch at sites of structural discontinuity, giving rise to electrocardiographic changes and the arrhythmogenic substrate.
Collapse
Affiliation(s)
- Chris Miles
- Cardiovascular Clinical Academic Group, St. George’s University Hospitals’ NHS Foundation Trust and Molecular and Clinical Sciences Institute, St. George’s, University of London, UK (C.M., C.S., E.R.B.)
| | - Bastiaan J. Boukens
- Department of Medical Biology, University of Amsterdam, the Netherlands (B.J.B.)
- University of Maastricht, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, the Netherlands (B.J.B.)
| | - Chiara Scrocco
- Cardiovascular Clinical Academic Group, St. George’s University Hospitals’ NHS Foundation Trust and Molecular and Clinical Sciences Institute, St. George’s, University of London, UK (C.M., C.S., E.R.B.)
| | - Arthur A.M. Wilde
- Amsterdam UMC, University of Amsterdam, Department of Cardiology, the Netherlands (A.A.M.W.)
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (A.A.M.W.)
- European Reference Network for rare, low-prevalence, and complex diseases of the heart: ERN GUARD-Heart (A.A.M.W., M.H.)
| | - Koonlawee Nademanee
- Center of Excellence in Arrhythmia Research Chulalongkorn University, Department of Medicine, Chulalongkorn University, Thailand (K.N.)
- Pacific Rim Electrophysiology Research Institute, Bumrungrad Hospital, Bangkok, Thailand (K.N.)
| | - Michel Haissaguerre
- European Reference Network for rare, low-prevalence, and complex diseases of the heart: ERN GUARD-Heart (A.A.M.W., M.H.)
- Institut Hospitalo-Universitaire Liryc, Electrophysiology and Heart Modeling Institute, Pessac, France (M.H.)
- Department of Electrophysiology and Cardiac Stimulation, Centre Hospitalier Universitaire de Bordeaux, France (M.H.)
| | - Ruben Coronel
- Department of Experimental Cardiology, Amsterdam University Medical Centers, Cardiovascular Science, the Netherlands (R.C.)
| | - Elijah R. Behr
- Cardiovascular Clinical Academic Group, St. George’s University Hospitals’ NHS Foundation Trust and Molecular and Clinical Sciences Institute, St. George’s, University of London, UK (C.M., C.S., E.R.B.)
- Mayo Clinic Healthcare, London, UK (E.R.B.)
| |
Collapse
|
43
|
Chen Y, Lou L, Zhang X, Jin L, Chen Y, Chen L, Li Z, Zhang F, Fu T, Hu S, Yang J. Association between circulating leukocytes and arrhythmias: Mendelian randomization analysis in immuno-cardiac electrophysiology. Front Immunol 2023; 14:1041591. [PMID: 37090734 PMCID: PMC10113438 DOI: 10.3389/fimmu.2023.1041591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
BackgroundCardiac arrhythmia is a common disease associated with high mortality and morbidity. Circulating leukocyte counts, which serve as a biomarker for assessing systemic immune status, have been linked to arrhythmias in observational studies. However, observational studies are plagued by confounding factors and reverse causality, whether alterations in circulating leukocyte components are causally associated with arrhythmias remains uncertain. The present study explored this question based on genetic evidence.Methods and findingsWe performed Mendelian randomization (MR) analysis to evaluate whether alterations in leukocyte counts affect aggregated risk of all types of arrhythmia or risk of five specific types of arrhythmia. Single-nucleotide polymorphisms serving as proxies for leukocyte differential counts were retrieved from the Blood Cell Consortium, and statistical data on arrhythmias were obtained from the UK Biobank), FinnGenand a meta-analysis of genome-wide association studies for atrial fibrillation. We applied inverse variance-weighted method as the primary analysis, complemented by a series of sensitivity analyses. Bidirectional analyses were conducted to assess reverse causality. Finally, multivariable MR was performed to study the joint effects of multiple risk factors. We found that genetically predicted differential leukocyte counts were not significantly associated with aggregated occurrence of all types of arrhythmia. In contrast, each 1-standard deviation increase in lymphocyte count was associated with 46% higher risk of atrioventricular block (OR 1.46, 95% CI 1.11–1.93, p=0.0065). A similar effect size was observed across all MR sensitivity analyses, with no evidence of horizontal pleiotropy. Reverse MR analysis suggested that atrioventricular block was unlikely to cause changes in lymphocyte count. Primary MR analysis based on the inverse-variance weighted method suggested that changes in neutrophil count alter risk of right bundle branch block, and changes in basophil count alter risk of atrial fibrillation. However, these causal relationships were not robust in sensitivity analyses. We found no compelling evidence that neutrophil or lymphocyte counts cause atrial fibrillation.ConclusionOur data support higher lymphocyte count as a causal risk factor for atrioventricular block. These results highlight the importance of immune cells in the pathogenesis of specific cardiac conduction disorders.
Collapse
Affiliation(s)
- Yuxiao Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lian Lou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Luyang Jin
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lele Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihang Li
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fen Zhang
- Department of Cardiology, Jinhua People's Hospital, Jinhua, China
| | - Ting Fu
- Department of Cardiology, Yiwu Central Hospital, Jinhua, China
| | - Shenjiang Hu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
44
|
Giudicessi JR. Unraveling the influence of genomic context on pleiotropy in SCN5A-mediated cardiac channelopathies: Insights from the Worm Study. Heart Rhythm 2023; 20:728-729. [PMID: 36858161 DOI: 10.1016/j.hrthm.2023.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023]
Affiliation(s)
- John R Giudicessi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
45
|
Isaacs A, Barysenka A, Ter Bekke RMA, Helderman-van den Enden ATJM, van den Wijngaard A, Volders PGA, Stoll M. Standing genetic variation affects phenotypic heterogeneity in an SCN5A-mutation founder population with excess sudden cardiac death. Heart Rhythm 2023; 20:720-727. [PMID: 36764349 DOI: 10.1016/j.hrthm.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/19/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND The Worm Study, ascertained from a multigeneration pedigree segregating a single amino acid deletion in SCN5A (c.4850_4852delTCT, p.(Phe1617del), rs749697698), is characterized by substantial phenotypic heterogeneity and overlap of sudden cardiac death, long-QT syndrome, cardiac conduction disease, Brugada syndrome, and isorhythmic atrioventricular dissociation. Linkage analysis for a synthetic trait derived from these phenotypes identified a single peak (logarithm of the odds [LOD] = 4.52) at the SCN5A/SCN10A/SCN11A locus on chromosome 3. OBJECTIVE This study explored the role of additional genetic variation in the chromosome 3 locus as a source of phenotypic heterogeneity in the Worm Study population. METHODS Genotypes underlying the linkage peak (n = 70) were characterized using microarrays. Haplotypes were determined using family-aware phasing and a population-specific reference panel. Variants with minor allele frequencies >0.10 were tested for association with cardiac conduction disease and isorhythmic dissociation using LAMP and logistic regression. RESULTS Only 1 haplotype carried the p.Phe1617del/rs749697698 deletion, suggesting relatively recent development (∼18 generations); this haplotype contained 5 other missense variants spanning SCN5A/SCN10A/SCN11A. Noncarrier haplotypes (n = 74) ranged in frequency from 0.5% to 5%. Although no variants were associated with cardiac conduction disease, a homozygous missense variant in SCN10A was associated with isorhythmic dissociation after correction for multiple comparisons (odds ratio 11.23; 95% confidence interval 2.76-23.39; P = 1.2 × 10-4). This variant (rs12632942) was previously associated with PR interval. CONCLUSION Our data suggest that other variants, alongside a pathogenic mutation, are associated with phenotypic heterogeneity. Single-mutation screening may be insufficient to predict electrical heart disease in patients and family members. In the Worm Study population, segregating a pathogenic SCN5A mutation, compound variation in the SCN5A/SCN10A/SCN11A locus determines arrhythmic outcome.
Collapse
Affiliation(s)
- Aaron Isaacs
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands; Department of Physiology, Maastricht University, Maastricht, the Netherlands
| | - Andrei Barysenka
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
| | - Rachel M A Ter Bekke
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | | | - Arthur van den Wijngaard
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Paul G A Volders
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Monika Stoll
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands; Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany; Department of Biochemistry, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
46
|
Krijger Juárez C, Amin AS, Offerhaus JA, Bezzina CR, Boukens BJ. Cardiac Repolarization in Health and Disease. JACC Clin Electrophysiol 2023; 9:124-138. [PMID: 36697193 DOI: 10.1016/j.jacep.2022.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022]
Abstract
Abnormal cardiac repolarization is at the basis of life-threatening arrhythmias in various congenital and acquired cardiac diseases. Dysfunction of ion channels involved in repolarization at the cellular level are often the underlying cause of the repolarization abnormality. The expression pattern of the gene encoding the affected ion channel dictates its impact on the shape of the T-wave and duration of the QT interval, thereby setting the stage for both the occurrence of the trigger and the substrate for maintenance of the arrhythmia. Here we discuss how research into the genetic and electrophysiological basis of repolarization has provided us with insights into cardiac repolarization in health and disease and how this in turn may provide the basis for future improved patient-specific management.
Collapse
Affiliation(s)
- Christian Krijger Juárez
- Department of Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Ahmad S Amin
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Joost A Offerhaus
- Department of Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
47
|
Lachaud Q, Aziz MHN, Burton FL, Macquaide N, Myles RC, Simitev RD, Smith GL. Electrophysiological heterogeneity in large populations of rabbit ventricular cardiomyocytes. Cardiovasc Res 2022; 118:3112-3125. [PMID: 35020837 PMCID: PMC9732512 DOI: 10.1093/cvr/cvab375] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 01/07/2022] [Indexed: 01/01/2023] Open
Abstract
AIMS Cardiac electrophysiological heterogeneity includes: (i) regional differences in action potential (AP) waveform, (ii) AP waveform differences in cells isolated from a single region, (iii) variability of the contribution of individual ion currents in cells with similar AP durations (APDs). The aim of this study is to assess intra-regional AP waveform differences, to quantify the contribution of specific ion channels to the APD via drug responses and to generate a population of mathematical models to investigate the mechanisms underlying heterogeneity in rabbit ventricular cells. METHODS AND RESULTS APD in ∼50 isolated cells from subregions of the LV free wall of rabbit hearts were measured using a voltage-sensitive dye. When stimulated at 2 Hz, average APD90 value in cells from the basal epicardial region was 254 ± 25 ms (mean ± standard deviation) in 17 hearts with a mean interquartile range (IQR) of 53 ± 17 ms. Endo-epicardial and apical-basal APD90 differences accounted for ∼10% of the IQR value. Highly variable changes in APD occurred after IK(r) or ICa(L) block that included a sub-population of cells (HR) with an exaggerated (hyper) response to IK(r) inhibition. A set of 4471 AP models matching the experimental APD90 distribution was generated from a larger population of models created by random variation of the maximum conductances (Gmax) of 8 key ion channels/exchangers/pumps. This set reproduced the pattern of cell-specific responses to ICa(L) and IK(r) block, including the HR sub-population. The models exhibited a wide range of Gmax values with constrained relationships linking ICa(L) with IK(r), ICl, INCX, and INaK. CONCLUSION Modelling the measured range of inter-cell APDs required a larger range of key Gmax values indicating that ventricular tissue has considerable inter-cell variation in channel/pump/exchanger activity. AP morphology is retained by relationships linking specific ionic conductances. These interrelationships are necessary for stable repolarization despite large inter-cell variation of individual conductances and this explains the variable sensitivity to ion channel block.
Collapse
Affiliation(s)
- Quentin Lachaud
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Muhamad Hifzhudin Noor Aziz
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
- Institute of Mathematical Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Francis L Burton
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Niall Macquaide
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Rachel C Myles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Radostin D Simitev
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
48
|
Glazer AM. Genetics of congenital arrhythmia syndromes: the challenge of variant interpretation. Curr Opin Genet Dev 2022; 77:102004. [PMID: 36368182 PMCID: PMC9743411 DOI: 10.1016/j.gde.2022.102004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/11/2022]
Abstract
Congenital arrhythmia syndromes are rare genetic disorders that can cause a high risk of sudden cardiac death. Expert panels have affirmed 15 genes that are linked to congenital arrhythmias. These genes mostly encode cardiac ion channel proteins or associated regulatory proteins that generate the cardiac action potential. Common genetic variation modulates the risk of rare variants and partially explains the incomplete penetrance of these disorders. As genetic testing becomes more prevalent, a major challenge is that most detected variants are annotated as variants of uncertain significance. This review will highlight emerging methods that are refining our understanding of arrhythmia genetics, including phenotype risk scores, large cohorts, in vitro functional assays, structural models, and computational predictions.
Collapse
Affiliation(s)
- Andrew M Glazer
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
49
|
Hoffmann TJ, Lu M, Oni-Orisan A, Lee C, Risch N, Iribarren C. A large genome-wide association study of QT interval length utilizing electronic health records. Genetics 2022; 222:iyac157. [PMID: 36271874 PMCID: PMC9713425 DOI: 10.1093/genetics/iyac157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/22/2022] [Indexed: 12/13/2022] Open
Abstract
QT interval length is an important risk factor for adverse cardiovascular outcomes; however, the genetic architecture of QT interval remains incompletely understood. We conducted a genome-wide association study of 76,995 ancestrally diverse Kaiser Permanente Northern California members enrolled in the Genetic Epidemiology Research on Adult Health and Aging cohort using 448,517 longitudinal QT interval measurements, uncovering 9 novel variants, most replicating in 40,537 individuals in the UK Biobank and Population Architecture using Genomics and Epidemiology studies. A meta-analysis of all 3 cohorts (n = 117,532) uncovered an additional 19 novel variants. Conditional analysis identified 15 additional variants, 3 of which were novel. Little, if any, difference was seen when adjusting for putative QT interval lengthening medications genome-wide. Using multiple measurements in Genetic Epidemiology Research on Adult Health and Aging increased variance explained by 163%, and we show that the ≈6 measurements in Genetic Epidemiology Research on Adult Health and Aging was equivalent to a 2.4× increase in sample size of a design with a single measurement. The array heritability was estimated at ≈17%, approximately half of our estimate of 36% from family correlations. Heritability enrichment was estimated highest and most significant in cardiovascular tissue (enrichment 7.2, 95% CI = 5.7-8.7, P = 2.1e-10), and many of the novel variants included expression quantitative trait loci in heart and other relevant tissues. Comparing our results to other cardiac function traits, it appears that QT interval has a multifactorial genetic etiology.
Collapse
Affiliation(s)
- Thomas J Hoffmann
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Meng Lu
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, USA
| | - Akinyemi Oni-Orisan
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Catherine Lee
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, USA
| | - Neil Risch
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94143, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, USA
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, USA
| |
Collapse
|
50
|
Ramírez J, van Duijvenboden S, Young WJ, Tinker A, Lambiase PD, Orini M, Munroe PB. Prediction of Coronary Artery Disease and Major Adverse Cardiovascular Events Using Clinical and Genetic Risk Scores for Cardiovascular Risk Factors. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2022; 15:e003441. [PMID: 35861959 PMCID: PMC9584057 DOI: 10.1161/circgen.121.003441] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/21/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Coronary artery disease (CAD) and major adverse cardiovascular events (MACE) are the leading causes of death in the general population, but risk stratification remains suboptimal. CAD genetic risk scores (GRSs) predict risk independently from clinical tools, like QRISK3. We assessed the added value of GRSs for a variety of cardiovascular traits (CV GRSs) for predicting CAD and MACE and tested their early-life screening potential by comparing against the CAD GRS only. METHODS We used data from 379 581 participants in the UK Biobank without known cardiovascular conditions (follow-up, 11.3 years; 3.3% CAD cases and 5.2% MACE cases). In a training subset (50%) we built 3 scores: QRISK3; QRISK3 and an established CAD GRS; and QRISK3, the CAD GRS and the CV GRSs. In an independent subset (50%), we evaluated each score's performance using the concordance index, odds ratio and net reclassification index. We then repeated the analyses without considering QRISK3. RESULTS For CAD, the combination of QRISK3 and the CAD GRS had a better performance than QRISK3 alone (concordance index, 0.766 versus 0.753; odds ratio, 5.47 versus 4.82; net reclassification index, 7.7%). Adding the CV GRSs did not significantly improve risk stratification. When only looking at genetic information, the combination of CV GRSs and the CAD GRS had a better performance than the CAD GRS alone (concordance index, 0.637 versus 0.625; odds ratio, 2.17 versus 2.07; net reclassification index, 3.3%). Similar results were obtained for MACE. CONCLUSIONS In individuals without known cardiovascular disease, the inclusion of CV GRSs to a clinical tool and an established CAD GRS does not improve CAD or MACE risk stratification. However, their combination only with the CAD GRS increases prediction performance indicating potential use in early-life screening before the advanced development of conventional cardiovascular risk factors.
Collapse
Affiliation(s)
- Julia Ramírez
- Clinical Pharmacology and Precision Medicine Deparment, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (J.R., S.v.D., W.J.Y., A.T., P.B.M.)
- Electronic Engineering and Communications Department, Aragon Institute of Engineering Research, University of Zaragoza, Spain and CIBER's Bioengineering, Biomaterials and Nanomedicine, Spain. (J.R.)
| | - Stefan van Duijvenboden
- Clinical Pharmacology and Precision Medicine Deparment, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (J.R., S.v.D., W.J.Y., A.T., P.B.M.)
- Institute of Cardiovascular Science, University College London, London, United Kingdom (S.v.D., P.D.L., M.O.)
| | - William J. Young
- Clinical Pharmacology and Precision Medicine Deparment, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (J.R., S.v.D., W.J.Y., A.T., P.B.M.)
- Barts Heart Centre, St Bartholomew’s Hospital, London, United Kingdom (W.J.Y., P.D.L., M.O.)
| | - Andrew Tinker
- Clinical Pharmacology and Precision Medicine Deparment, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (J.R., S.v.D., W.J.Y., A.T., P.B.M.)
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (A.T., P.B.M.)
| | - Pier D. Lambiase
- Institute of Cardiovascular Science, University College London, London, United Kingdom (S.v.D., P.D.L., M.O.)
- Barts Heart Centre, St Bartholomew’s Hospital, London, United Kingdom (W.J.Y., P.D.L., M.O.)
| | - Michele Orini
- Institute of Cardiovascular Science, University College London, London, United Kingdom (S.v.D., P.D.L., M.O.)
- Barts Heart Centre, St Bartholomew’s Hospital, London, United Kingdom (W.J.Y., P.D.L., M.O.)
| | - Patricia B. Munroe
- Clinical Pharmacology and Precision Medicine Deparment, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (J.R., S.v.D., W.J.Y., A.T., P.B.M.)
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (A.T., P.B.M.)
| |
Collapse
|