Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Circ Genom Precis Med ; 16(4): 328-336, 2023 08.
Article in English | MEDLINE | ID: mdl-37199186

ABSTRACT

BACKGROUND: Genetic variants in TNNI3K (troponin-I interacting kinase) have previously been associated with dilated cardiomyopathy (DCM), cardiac conduction disease, and supraventricular tachycardias. However, the link between TNNI3K variants and these cardiac phenotypes shows a lack of consensus concerning phenotype and protein function. METHODS: We describe a systematic retrospective study of a cohort of patients undergoing genetic testing for cardiac arrhythmias and cardiomyopathy including TNNI3K. We further performed burden testing of TNNI3K in the UK Biobank. For 2 novel TNNI3K variants, we tested cosegregation. TNNI3K kinase function was estimated by TNNI3K autophosphorylation assays. RESULTS: We demonstrate enrichment of rare coding TNNI3K variants in DCM patients in the Amsterdam cohort. In the UK Biobank, we observed an association between TNNI3K missense (but not loss-of-function) variants and DCM and atrial fibrillation. Furthermore, we demonstrate genetic segregation for 2 rare variants, TNNI3K-p.Ile512Thr and TNNI3K-p.His592Tyr, with phenotypes consisting of DCM, cardiac conduction disease, and supraventricular tachycardia, together with increased autophosphorylation. In contrast, TNNI3K-p.Arg556_Asn590del, a likely benign variant, demonstrated depleted autophosphorylation. CONCLUSIONS: Our findings demonstrate an increased burden of rare coding TNNI3K variants in cardiac patients with DCM. Furthermore, we present 2 novel likely pathogenic TNNI3K variants with increased autophosphorylation, suggesting that enhanced autophosphorylation is likely to drive pathogenicity.


Subject(s)
Cardiomyopathy, Dilated , Humans , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/genetics , Retrospective Studies , Arrhythmias, Cardiac/genetics , Genetic Testing , Cardiac Conduction System Disease/genetics , Protein Serine-Threonine Kinases/genetics
2.
Cardiovasc Res ; 118(7): 1742-1757, 2022 06 22.
Article in English | MEDLINE | ID: mdl-34142125

ABSTRACT

AIMS: Cardiac arrhythmias comprise a major health and economic burden and are associated with significant morbidity and mortality, including cardiac failure, stroke, and sudden cardiac death (SCD). Development of efficient preventive and therapeutic strategies is hampered by incomplete knowledge of disease mechanisms and pathways. Our aim is to identify novel mechanisms underlying cardiac arrhythmia and SCD using an unbiased approach. METHODS AND RESULTS: We employed a phenotype-driven N-ethyl-N-nitrosourea mutagenesis screen and identified a mouse line with a high incidence of sudden death at young age (6-9 weeks) in the absence of prior symptoms. Affected mice were found to be homozygous for the nonsense mutation Bcat2p.Q300*/p.Q300* in the Bcat2 gene encoding branched chain amino acid transaminase 2. At the age of 4-5 weeks, Bcat2p.Q300*/p.Q300* mice displayed drastic increase of plasma levels of branch chain amino acids (BCAAs-leucine, isoleucine, valine) due to the incomplete catabolism of BCAAs, in addition to inducible arrhythmias ex vivo as well as cardiac conduction and repolarization disturbances. In line with these findings, plasma BCAA levels were positively correlated to electrocardiogram indices of conduction and repolarization in the German community-based KORA F4 Study. Isolated cardiomyocytes from Bcat2p.Q300*/p.Q300* mice revealed action potential (AP) prolongation, pro-arrhythmic events (early and late afterdepolarizations, triggered APs), and dysregulated calcium homeostasis. Incubation of human pluripotent stem cell-derived cardiomyocytes with elevated concentration of BCAAs induced similar calcium dysregulation and pro-arrhythmic events which were prevented by rapamycin, demonstrating the crucial involvement of mTOR pathway activation. CONCLUSIONS: Our findings identify for the first time a causative link between elevated BCAAs and arrhythmia, which has implications for arrhythmogenesis in conditions associated with BCAA metabolism dysregulation such as diabetes, metabolic syndrome, and heart failure.


Subject(s)
Calcium , Heart Failure , Amino Acids, Branched-Chain/metabolism , Animals , Humans , Mice , Myocytes, Cardiac/metabolism , Sirolimus
3.
Circ Res ; 130(2): 166-180, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34886679

ABSTRACT

RATIONALE: Dextro-transposition of the great arteries (D-TGA) is a severe congenital heart defect which affects approximately 1 in 4,000 live births. While there are several reports of D-TGA patients with rare variants in individual genes, the majority of D-TGA cases remain genetically elusive. Familial recurrence patterns and the observation that most cases with D-TGA are sporadic suggest a polygenic inheritance for the disorder, yet this remains unexplored. OBJECTIVE: We sought to study the role of common single nucleotide polymorphisms (SNPs) in risk for D-TGA. METHODS AND RESULTS: We conducted a genome-wide association study in an international set of 1,237 patients with D-TGA and identified a genome-wide significant susceptibility locus on chromosome 3p14.3, which was subsequently replicated in an independent case-control set (rs56219800, meta-analysis P=8.6x10-10, OR=0.69 per C allele). SNP-based heritability analysis showed that 25% of variance in susceptibility to D-TGA may be explained by common variants. A genome-wide polygenic risk score derived from the discovery set was significantly associated to D-TGA in the replication set (P=4x10-5). The genome-wide significant locus (3p14.3) co-localizes with a putative regulatory element that interacts with the promoter of WNT5A, which encodes the Wnt Family Member 5A protein known for its role in cardiac development in mice. We show that this element drives reporter gene activity in the developing heart of mice and zebrafish and is bound by the developmental transcription factor TBX20. We further demonstrate that TBX20 attenuates Wnt5a expression levels in the developing mouse heart. CONCLUSIONS: This work provides support for a polygenic architecture in D-TGA and identifies a susceptibility locus on chromosome 3p14.3 near WNT5A. Genomic and functional data support a causal role of WNT5A at the locus.


Subject(s)
Polymorphism, Single Nucleotide , Transposition of Great Vessels/genetics , Animals , Cells, Cultured , Humans , Mice , Multifactorial Inheritance , Myocytes, Cardiac/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transposition of Great Vessels/metabolism , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism , Zebrafish
5.
Genet Med ; 23(10): 1952-1960, 2021 10.
Article in English | MEDLINE | ID: mdl-34113005

ABSTRACT

PURPOSE: Rare genetic variants in KDR, encoding the vascular endothelial growth factor receptor 2 (VEGFR2), have been reported in patients with tetralogy of Fallot (TOF). However, their role in disease causality and pathogenesis remains unclear. METHODS: We conducted exome sequencing in a familial case of TOF and large-scale genetic studies, including burden testing, in >1,500 patients with TOF. We studied gene-targeted mice and conducted cell-based assays to explore the role of KDR genetic variation in the etiology of TOF. RESULTS: Exome sequencing in a family with two siblings affected by TOF revealed biallelic missense variants in KDR. Studies in knock-in mice and in HEK 293T cells identified embryonic lethality for one variant when occurring in the homozygous state, and a significantly reduced VEGFR2 phosphorylation for both variants. Rare variant burden analysis conducted in a set of 1,569 patients of European descent with TOF identified a 46-fold enrichment of protein-truncating variants (PTVs) in TOF cases compared to controls (P = 7 × 10-11). CONCLUSION: Rare KDR variants, in particular PTVs, strongly associate with TOF, likely in the setting of different inheritance patterns. Supported by genetic and in vivo and in vitro functional analysis, we propose loss-of-function of VEGFR2 as one of the mechanisms involved in the pathogenesis of TOF.


Subject(s)
Tetralogy of Fallot , Vascular Endothelial Growth Factor Receptor-2 , Animals , Genetic Predisposition to Disease , HEK293 Cells , Humans , Mice , Tetralogy of Fallot/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Exome Sequencing
6.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33645542

ABSTRACT

Congenital heart disease is the most common type of birth defect, accounting for one-third of all congenital anomalies. Using whole-exome sequencing of 2718 patients with congenital heart disease and a search in GeneMatcher, we identified 30 patients from 21 unrelated families of different ancestries with biallelic phospholipase D1 (PLD1) variants who presented predominantly with congenital cardiac valve defects. We also associated recessive PLD1 variants with isolated neonatal cardiomyopathy. Furthermore, we established that p.I668F is a founder variant among Ashkenazi Jews (allele frequency of ~2%) and describe the phenotypic spectrum of PLD1-associated congenital heart defects. PLD1 missense variants were overrepresented in regions of the protein critical for catalytic activity, and, correspondingly, we observed a strong reduction in enzymatic activity for most of the mutant proteins in an enzymatic assay. Finally, we demonstrate that PLD1 inhibition decreased endothelial-mesenchymal transition, an established pivotal early step in valvulogenesis. In conclusion, our study provides a more detailed understanding of disease mechanisms and phenotypic expression associated with PLD1 loss of function.


Subject(s)
Alleles , Heart Defects, Congenital , Heart Valve Diseases , Loss of Function Mutation , Phospholipase D , Female , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/genetics , Heart Valve Diseases/enzymology , Heart Valve Diseases/genetics , Humans , Male , Phospholipase D/genetics , Phospholipase D/metabolism
7.
Genet Med ; 23(5): 856-864, 2021 05.
Article in English | MEDLINE | ID: mdl-33500567

ABSTRACT

PURPOSE: To characterize the genetic architecture of left ventricular noncompaction (LVNC) and investigate the extent to which it may represent a distinct pathology or a secondary phenotype associated with other cardiac diseases. METHODS: We performed rare variant association analysis with 840 LVNC cases and 125,748 gnomAD population controls, and compared results to similar analyses on dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM). RESULTS: We observed substantial genetic overlap indicating that LVNC often represents a phenotypic variation of DCM or HCM. In contrast, truncating variants in MYH7, ACTN2, and PRDM16 were uniquely associated with LVNC and may reflect a distinct LVNC etiology. In particular, MYH7 truncating variants (MYH7tv), generally considered nonpathogenic for cardiomyopathies, were 20-fold enriched in LVNC cases over controls. MYH7tv heterozygotes identified in the UK Biobank and healthy volunteer cohorts also displayed significantly greater noncompaction compared with matched controls. RYR2 exon deletions and HCN4 transmembrane variants were also enriched in LVNC, supporting prior reports of association with arrhythmogenic LVNC phenotypes. CONCLUSION: LVNC is characterized by substantial genetic overlap with DCM/HCM but is also associated with distinct noncompaction and arrhythmia etiologies. These results will enable enhanced application of LVNC genetic testing and help to distinguish pathological from physiological noncompaction.


Subject(s)
Cardiomyopathies , Cardiomyopathy, Dilated , Cardiomyopathy, Hypertrophic , Heart Defects, Congenital , Cardiomyopathies/genetics , Cardiomyopathy, Dilated/genetics , Genetic Testing , Humans
8.
Nat Genet ; 53(2): 128-134, 2021 02.
Article in English | MEDLINE | ID: mdl-33495596

ABSTRACT

The heart muscle diseases hypertrophic (HCM) and dilated (DCM) cardiomyopathies are leading causes of sudden death and heart failure in young, otherwise healthy, individuals. We conducted genome-wide association studies and multi-trait analyses in HCM (1,733 cases), DCM (5,521 cases) and nine left ventricular (LV) traits (19,260 UK Biobank participants with structurally normal hearts). We identified 16 loci associated with HCM, 13 with DCM and 23 with LV traits. We show strong genetic correlations between LV traits and cardiomyopathies, with opposing effects in HCM and DCM. Two-sample Mendelian randomization supports a causal association linking increased LV contractility with HCM risk. A polygenic risk score explains a significant portion of phenotypic variability in carriers of HCM-causing rare variants. Our findings thus provide evidence that polygenic risk score may account for variability in Mendelian diseases. More broadly, we provide insights into how genetic pathways may lead to distinct disorders through opposing genetic effects.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Dilated/mortality , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Hypertrophic/mortality , Cardiomyopathy, Hypertrophic/physiopathology , Case-Control Studies , Gene Frequency , Genetic Predisposition to Disease , Genome-Wide Association Study , Heart Ventricles/physiopathology , Humans , Kaplan-Meier Estimate , Linkage Disequilibrium , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Ventricular Function, Left/genetics
9.
Europace ; 22(10): 1579-1589, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32778883

ABSTRACT

AIMS: SCN5A mutations are associated with arrhythmia syndromes, including Brugada syndrome, long QT syndrome type 3 (LQT3), and cardiac conduction disease. Long QT syndrome type 3 patients display atrio-ventricular (AV) conduction slowing which may contribute to arrhythmogenesis. We here investigated the as yet unknown underlying mechanisms. METHODS AND RESULTS: We assessed electrophysiological and molecular alterations underlying AV-conduction abnormalities in mice carrying the Scn5a1798insD/+ mutation. Langendorff-perfused Scn5a1798insD/+ hearts showed prolonged AV-conduction compared to wild type (WT) without changes in atrial and His-ventricular (HV) conduction. The late sodium current (INa,L) inhibitor ranolazine (RAN) normalized AV-conduction in Scn5a1798insD/+ mice, likely by preventing the mutation-induced increase in intracellular sodium ([Na+]i) and calcium ([Ca2+]i) concentrations. Indeed, further enhancement of [Na+]i and [Ca2+]i by the Na+/K+-ATPase inhibitor ouabain caused excessive increase in AV-conduction time in Scn5a1798insD/+ hearts. Scn5a1798insD/+ mice from the 129P2 strain displayed more severe AV-conduction abnormalities than FVB/N-Scn5a1798insD/+ mice, in line with their larger mutation-induced INa,L. Transverse aortic constriction (TAC) caused excessive prolongation of AV-conduction in FVB/N-Scn5a1798insD/+ mice (while HV-intervals remained unchanged), which was prevented by chronic RAN treatment. Scn5a1798insD/+-TAC hearts showed decreased mRNA levels of conduction genes in the AV-nodal region, but no structural changes in the AV-node or His bundle. In Scn5a1798insD/+-TAC mice deficient for the transcription factor Nfatc2 (effector of the calcium-calcineurin pathway), AV-conduction and conduction gene expression were restored to WT levels. CONCLUSIONS: Our findings indicate a detrimental role for enhanced INa,L and consequent calcium dysregulation on AV-conduction in Scn5a1798insD/+ mice, providing evidence for a functional mechanism underlying AV-conduction disturbances secondary to gain-of-function SCN5A mutations.


Subject(s)
Calcium , Long QT Syndrome , Animals , Humans , Long QT Syndrome/genetics , Long QT Syndrome/therapy , Mice , Mice, Transgenic , NAV1.5 Voltage-Gated Sodium Channel/genetics , Sodium/metabolism
10.
Circulation ; 142(4): 324-338, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32429735

ABSTRACT

BACKGROUND: Long QT syndrome (LQTS) is a rare genetic disorder and a major preventable cause of sudden cardiac death in the young. A causal rare genetic variant with large effect size is identified in up to 80% of probands (genotype positive) and cascade family screening shows incomplete penetrance of genetic variants. Furthermore, a proportion of cases meeting diagnostic criteria for LQTS remain genetically elusive despite genetic testing of established genes (genotype negative). These observations raise the possibility that common genetic variants with small effect size contribute to the clinical picture of LQTS. This study aimed to characterize and quantify the contribution of common genetic variation to LQTS disease susceptibility. METHODS: We conducted genome-wide association studies followed by transethnic meta-analysis in 1656 unrelated patients with LQTS of European or Japanese ancestry and 9890 controls to identify susceptibility single nucleotide polymorphisms. We estimated the common variant heritability of LQTS and tested the genetic correlation between LQTS susceptibility and other cardiac traits. Furthermore, we tested the aggregate effect of the 68 single nucleotide polymorphisms previously associated with the QT-interval in the general population using a polygenic risk score. RESULTS: Genome-wide association analysis identified 3 loci associated with LQTS at genome-wide statistical significance (P<5×10-8) near NOS1AP, KCNQ1, and KLF12, and 1 missense variant in KCNE1(p.Asp85Asn) at the suggestive threshold (P<10-6). Heritability analyses showed that ≈15% of variance in overall LQTS susceptibility was attributable to common genetic variation (h2SNP 0.148; standard error 0.019). LQTS susceptibility showed a strong genome-wide genetic correlation with the QT-interval in the general population (rg=0.40; P=3.2×10-3). The polygenic risk score comprising common variants previously associated with the QT-interval in the general population was greater in LQTS cases compared with controls (P<10-13), and it is notable that, among patients with LQTS, this polygenic risk score was greater in patients who were genotype negative compared with those who were genotype positive (P<0.005). CONCLUSIONS: This work establishes an important role for common genetic variation in susceptibility to LQTS. We demonstrate overlap between genetic control of the QT-interval in the general population and genetic factors contributing to LQTS susceptibility. Using polygenic risk score analyses aggregating common genetic variants that modulate the QT-interval in the general population, we provide evidence for a polygenic architecture in genotype negative LQTS.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Long QT Syndrome/genetics , Adolescent , Adult , Age of Onset , Alleles , Case-Control Studies , Electrocardiography , Genetic Association Studies , Genome-Wide Association Study/methods , Genotype , Humans , Long QT Syndrome/diagnosis , Long QT Syndrome/mortality , Long QT Syndrome/therapy , Multifactorial Inheritance , Phenotype , Polymorphism, Single Nucleotide , Prognosis , Severity of Illness Index , Young Adult
11.
Eur Heart J ; 40(37): 3097-3107, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31504448

ABSTRACT

AIMS: Sodium-channel blockers (SCBs) are associated with arrhythmia, but variability of cardiac electrical response remains unexplained. We sought to identify predictors of ajmaline-induced PR and QRS changes and Type I Brugada syndrome (BrS) electrocardiogram (ECG). METHODS AND RESULTS: In 1368 patients that underwent ajmaline infusion for suspected BrS, we performed measurements of 26 721 ECGs, dose-response mixed modelling and genotyping. We calculated polygenic risk scores (PRS) for PR interval (PRSPR), QRS duration (PRSQRS), and Brugada syndrome (PRSBrS) derived from published genome-wide association studies and used regression analysis to identify predictors of ajmaline dose related PR change (slope) and QRS slope. We derived and validated using bootstrapping a predictive model for ajmaline-induced Type I BrS ECG. Higher PRSPR, baseline PR, and female sex are associated with more pronounced PR slope, while PRSQRS and age are positively associated with QRS slope (P < 0.01 for all). PRSBrS, baseline QRS duration, presence of Type II or III BrS ECG at baseline, and family history of BrS are independently associated with the occurrence of a Type I BrS ECG, with good predictive accuracy (optimism-corrected C-statistic 0.74). CONCLUSION: We show for the first time that genetic factors underlie the variability of cardiac electrical response to SCB. PRSBrS, family history, and a baseline ECG can predict the development of a diagnostic drug-induced Type I BrS ECG with clinically relevant accuracy. These findings could lead to the use of PRS in the diagnosis of BrS and, if confirmed in population studies, to identify patients at risk for toxicity when given SCB.


Subject(s)
Ajmaline/adverse effects , Brugada Syndrome/drug therapy , Clinical Decision Rules , Genome-Wide Association Study , Heart Rate/drug effects , Polymorphism, Single Nucleotide , Sodium Channel Blockers/adverse effects , Ajmaline/therapeutic use , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/therapeutic use , Brugada Syndrome/diagnosis , Brugada Syndrome/genetics , Brugada Syndrome/physiopathology , Dose-Response Relationship, Drug , Electrocardiography , Female , Genetic Markers , Genotyping Techniques , Heart Rate/genetics , Humans , Infusions, Intravenous , Male , Risk Assessment , Sodium Channel Blockers/therapeutic use
12.
Am J Med Genet A ; 179(9): 1836-1845, 2019 09.
Article in English | MEDLINE | ID: mdl-31301121

ABSTRACT

The first human mutations in GATA6 were described in a cohort of patients with persistent truncus arteriosus, and the phenotypic spectrum has expanded since then. This study underscores the broad phenotypic spectrum by presenting two patients with de novo GATA6 mutations, both exhibiting complex cardiac defects, pancreatic, and other abnormalities. Furthermore, we provided a detailed overview of all published human genetic variation in/near GATA6 published to date and the associated phenotypes (n = 78). We conclude that the most common phenotypes associated with a mutation in GATA6 were structural cardiac and pancreatic abnormalities, with a penetrance of 87 and 60%, respectively. Other common malformations were gallbladder agenesis, congenital diaphragmatic hernia, and neurocognitive abnormalities, mostly developmental delay. Fifty-eight percent of the mutations were de novo, and these patients more often had an anomaly of intracardiac connections, an anomaly of the great arteries, and hypothyroidism, compared with those with inherited mutations. Functional studies mostly support loss-of-function as the pathophysiological mechanism. In conclusion, GATA6 mutations give a wide range of phenotypic defects, most frequently malformations of the heart and pancreas. This highlights the importance of detailed clinical evaluation of identified carriers to evaluate their full phenotypic spectrum.


Subject(s)
GATA6 Transcription Factor/genetics , Heart Defects, Congenital/genetics , Heart/physiopathology , Truncus Arteriosus, Persistent/genetics , Adult , Child , Gallbladder/physiopathology , Genetic Predisposition to Disease , Genotype , Heart/diagnostic imaging , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/physiopathology , Hernias, Diaphragmatic, Congenital/diagnostic imaging , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/physiopathology , Heterozygote , Humans , Loss of Function Mutation/genetics , Male , Mutation , Pancreas/diagnostic imaging , Pancreas/physiopathology , Phenotype , Truncus Arteriosus, Persistent/diagnostic imaging , Truncus Arteriosus, Persistent/physiopathology , Exome Sequencing
13.
Int J Cardiol ; 263: 54-62, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29754923

ABSTRACT

BACKGROUND: Long QT syndrome mutations in the SCN5A gene are associated with an enhanced late sodium current (INa,L) which may lead to pro-arrhythmic action potential prolongation and intracellular calcium dysregulation. We here investigated the dynamic relation between INa,L, intracellular sodium ([Na+]i) and calcium ([Ca2+]i) homeostasis and pro-arrhythmic events in the setting of a SCN5A mutation. METHODS AND RESULTS: Wild-type (WT) and Scn5a1798insD/+ (MUT) mice (age 3-5 months) carrying the murine homolog of the SCN5A-1795insD mutation on two distinct genetic backgrounds (FVB/N and 129P2) were studied. [Na+]i, [Ca2+]i and Ca2+ transient amplitude were significantly increased in 129P2-MUT myocytes as compared to WT, but not in FVB/N-MUT. Accordingly, INa,L wassignificantly more enhanced in 129P2-MUT than in FVB/N-MUT myocytes, consistent with a dose-dependent correlation. Quantitative RT-PCR analysis revealed intrinsic differences in mRNA expression levels of the sodium/potassium pump, the sodium/hydrogen exchanger, and sodium­calcium exchanger between the two mouse strains. The rate of increase in [Na+]i, [Ca2+]i and Ca2+ transient amplitude following the application of the Na+/K+-ATPase inhibitor ouabain was significantly greater in 129P2-MUT than in 129P2-WT myocytes and was normalized by the INa,L inhibitor ranolazine. Furthermore, ranolazine decreased the incidence of pro-arrhythmic calcium after-transients elicited in 129P2-MUT myocytes. CONCLUSIONS: In this study we established a causal link between the magnitude of INa,L, extent of Na+ and Ca2+ dysregulation, and incidence of pro-arrhythmic events in murine Scn5a1798insD/+ myocytes. Furthermore, our findings provide mechanistic insight into the anti-arrhythmic potential of pharmacological inhibition of INa,L in patients with LQT3 syndrome.


Subject(s)
Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Calcium/physiology , Intracellular Fluid/physiology , NAV1.5 Voltage-Gated Sodium Channel/physiology , Sodium/physiology , Animals , Arrhythmias, Cardiac/etiology , Cells, Cultured , Intracellular Fluid/drug effects , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Sodium-Calcium Exchanger/drug effects , Sodium-Calcium Exchanger/physiology
14.
Circ Res ; 121(5): 537-548, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28637782

ABSTRACT

RATIONALE: Genome-wide association studies previously identified an association of rs9388451 at chromosome 6q22.3 (near HEY2) with Brugada syndrome. The causal gene and underlying mechanism remain unresolved. OBJECTIVE: We used an integrative approach entailing transcriptomic studies in human hearts and electrophysiological studies in Hey2+/- (Hey2 heterozygous knockout) mice to dissect the underpinnings of the 6q22.31 association with Brugada syndrome. METHODS AND RESULTS: We queried expression quantitative trait locus data acquired in 190 human left ventricular samples from the genotype-tissue expression consortium for cis-expression quantitative trait locus effects of rs9388451, which revealed an association between Brugada syndrome risk allele dosage and HEY2 expression (ß=+0.159; P=0.0036). In the same transcriptomic data, we conducted genome-wide coexpression analysis for HEY2, which uncovered KCNIP2, encoding the ß-subunit of the channel underlying the transient outward current (Ito), as the transcript most robustly correlating with HEY2 expression (ß=+1.47; P=2×10-34). Transcript abundance of Hey2 and the Ito subunits Kcnip2 and Kcnd2, assessed by quantitative reverse transcription-polymerase chain reaction, was higher in subepicardium versus subendocardium in both left and right ventricles, with lower levels in Hey2+/- mice compared with wild type. Surface ECG measurements showed less prominent J waves in Hey2+/- mice compared with wild-type. In wild-type mice, patch-clamp electrophysiological studies on cardiomyocytes from right ventricle demonstrated a shorter action potential duration and a lower Vmax in subepicardium compared with subendocardium cardiomyocytes, which was paralleled by a higher Ito and a lower sodium current (INa) density in subepicardium versus subendocardium. These transmural differences were diminished in Hey2+/- mice because of changes in subepicardial cardiomyocytes. CONCLUSIONS: This study uncovers a role of HEY2 in the normal transmural electrophysiological gradient in the ventricle and provides compelling evidence that genetic variation at 6q22.31 (rs9388451) is associated with Brugada syndrome through a HEY2-dependent alteration of ion channel expression across the cardiac ventricular wall.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Genetic Predisposition to Disease/genetics , Heart Ventricles/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Animals , Brugada Syndrome/physiopathology , Electrocardiography/methods , Female , Genome-Wide Association Study/methods , Heart Ventricles/physiopathology , Humans , Ion Channels/biosynthesis , Ion Channels/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic
15.
Int J Cardiol ; 236: 187-193, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28262340

ABSTRACT

BACKGROUND: Mutations in SCN5A, the gene encoding the α-subunit of the cardiac sodium channel (NaV1.5), are associated with a broad spectrum of inherited cardiac arrhythmia disorders. The purpose of this study was to identify the genetic and functional determinants underlying a Dutch family that presented with a combined phenotype of ventricular arrhythmias with a likely adrenergic component, either in isolation or in combination with a mildly decreased heart function and early onset (<55years) atrial fibrillation. METHODS AND RESULTS: We performed next generation sequencing in the proband of a two-generation Dutch family and demonstrated a novel missense mutation in SCN5A-(p.M1851V) which co-segregated with the clinical phenotype in the family. We functionally evaluated the putative genetic defect by patch clamp electrophysiological studies in human embryonic kidney cells transfected with mutant or wild-type Nav1.5. The current inactivation was slower and recovery from inactivation was faster in SCN5A-M1851V channels. The voltage dependence of inactivation was shifted towards more positive potentials and consequently, a larger TTX-sensitive window current was observed in SCN5A-M1851V channels. Furthermore, a higher upstroke velocity was observed for the SCN5A-M1851V channels, while the depolarization voltage was more negative, both indicating increased excitability. CONCLUSIONS: This mutation leads to a gain-of-function mechanism based on increased channel availability and increased window current, fitting the observed clinical phenotype of (likely adrenergic-induced) ventricular arrhythmias and atrial fibrillation. These findings further expand the range of cardiac arrhythmias associated with mutations in SCN5A.


Subject(s)
Atrial Fibrillation/genetics , Mutation, Missense/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Ventricular Fibrillation/genetics , Adolescent , Adult , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Female , Humans , Male , Middle Aged , Pedigree , Ventricular Fibrillation/diagnosis , Ventricular Fibrillation/physiopathology , Young Adult
16.
Heart Rhythm ; 14(2): 265-272, 2017 02.
Article in English | MEDLINE | ID: mdl-27737802

ABSTRACT

BACKGROUND: The repolarization pattern of the human heart is unknown. OBJECTIVE: The purpose of this study was to perform a multisite analysis of the activation-repolarization patterns and mRNA expression patterns of ion channel subunits in isolated human hearts. METHODS: Hearts from 3 donors without reported cardiac disease were Langendorff perfused with the patient's own blood. A standard ECG was obtained before explantation. Up to 92 unipolar electrograms from 24 transmural needles were obtained during right atrial pacing. Local activation and repolarization times and activation-recovery intervals (ARI) were measured. The mRNA levels of subunits of the channels carrying the transient outward current and slow and rapid components of the delayed rectifier current were determined by quantitative reverse transcriptase polymerase chain reaction at up to 63 sites. RESULTS: The repolarization gradients in the 3 hearts were different and occurred along all axes without midmural late repolarization. A negative activation-repolarization relationship occurred along the epicardium, but this relationship was positive in the whole hearts. Coefficients of variation of mRNA levels (40%-80%) and of the Kv7.1 protein (alpha-subunit slow delayed rectifier channel) were larger than those of ARIs (7%-17%). The regional mRNA expression patterns were similar in the 3 hearts, unlike the ARI profiles. The expression level of individual mRNAs and of Kv7.1 did not correlate with local ARIs at the same sites. CONCLUSION: In the normal human heart, repolarization gradients encompass all axes, without late midmural repolarization. Last activated areas do not repolarize first as previously assumed. Gradients of mRNAs of single ion channel subunits and of ARIs do not correlate.


Subject(s)
Heart , KCNQ1 Potassium Channel/metabolism , Action Potentials/physiology , Electrophysiologic Techniques, Cardiac/methods , Heart/physiology , Heart/physiopathology , Humans , Refractory Period, Electrophysiological/physiology , Research Design
18.
Am J Hum Genet ; 99(3): 704-710, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27523599

ABSTRACT

GNB5 encodes the G protein ß subunit 5 and is involved in inhibitory G protein signaling. Here, we report mutations in GNB5 that are associated with heart-rate disturbance, eye disease, intellectual disability, gastric problems, hypotonia, and seizures in nine individuals from six families. We observed an association between the nature of the variants and clinical severity; individuals with loss-of-function alleles had more severe symptoms, including substantial developmental delay, speech defects, severe hypotonia, pathological gastro-esophageal reflux, retinal disease, and sinus-node dysfunction, whereas related heterozygotes harboring missense variants presented with a clinically milder phenotype. Zebrafish gnb5 knockouts recapitulated the phenotypic spectrum of affected individuals, including cardiac, neurological, and ophthalmological abnormalities, supporting a direct role of GNB5 in the control of heart rate, hypotonia, and vision.


Subject(s)
Bradycardia/genetics , Bradycardia/physiopathology , Developmental Disabilities/genetics , GTP-Binding Protein beta Subunits/genetics , Genes, Recessive/genetics , Mutation/genetics , Sinoatrial Node/physiopathology , Adolescent , Animals , Child , Developmental Disabilities/physiopathology , Female , GTP-Binding Protein beta Subunits/deficiency , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/physiopathology , Gene Deletion , Heart Rate/genetics , Heterozygote , Humans , Male , Muscle Hypotonia/genetics , Mutation, Missense/genetics , Pedigree , Phenotype , Retinal Diseases/genetics , Retinal Diseases/physiopathology , Seizures/genetics , Syndrome , Young Adult , Zebrafish/genetics , Zebrafish/physiology , Zebrafish Proteins
19.
Circ Cardiovasc Genet ; 9(2): 147-53, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26800703

ABSTRACT

BACKGROUND: Sudden cardiac arrest (SCA) ranks among the most common causes of death worldwide. Because SCA is most often lethal, yet mostly occurs in individuals without previously known cardiac disease, the identification of patients at risk for SCA could save many lives. In unselected SCA victims from the community, common genetic variants (which are not disease-causing per se, but may increase susceptibility to ventricular fibrillation) are found to be associated with increased SCA risk. However, whether rare genetic variants contribute to SCA risk in the community is largely unexplored. METHODS AND RESULTS: We here investigated the involvement of rare genetic variants in SCA risk at the population level by studying the prevalence of 6 founder genetic variants present in the Dutch population (PLN-p.Arg14del, MYBPC3-p.Trp792fsX17, MYBPC3-p.Arg943X, MYBPC3-p.Pro955fsX95, PKP2-p.Arg79X, and the Chr7q36 idiopathic ventricular fibrillation risk haplotype) in a cohort of 1440 unselected Dutch SCA victims included in the Amsterdam Resuscitation Study (ARREST). The six studied founder mutations were found to be more prevalent (1.1%) in the ARREST SCA cohort compared with an ethnically and geographically matched set of controls (0.4%, n=1379; P<0.05) or a set of Dutch individuals drawn from the Genome of the Netherlands (GoNL) study (0%, n=500; P<0.02). CONCLUSIONS: This finding provides proof-of-concept for the notion that rare genetic variants contribute to some extent to SCA risk in the community.


Subject(s)
Death, Sudden, Cardiac/etiology , Genetic Variation , Residence Characteristics , Case-Control Studies , Death, Sudden, Cardiac/epidemiology , Female , Founder Effect , Geography , Humans , Male , Middle Aged , Mutation/genetics , Netherlands/epidemiology , Prevalence
20.
Circ Cardiovasc Genet ; 7(6): 790-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25217174

ABSTRACT

BACKGROUND: With aging and in cardiac disease, fibrosis caused by collagen deposition is increased, impairing contractility and providing a substrate for arrhythmia. In this study, we set out to identify genetic modifiers of collagen deposition in the heart by exploiting the genetic variability among F2 progeny of 129P2 and FVBN/J mice carrying the Scn5a(tm1Care/+) mutation. METHODS AND RESULTS: Relative amounts of collagen were determined in left ventricular myocardium of 65 F2-mice and combined with genome-wide genotypic and cardiac expression data to identify collagen quantitative trait loci (QTLs) and overlapping expression QTLs (eQTLs). A significant collagen QTL was identified on mouse Chr8; an additional collagen QTL was identified on mouse Chr2 after correction for a genetic covariate uncovered on Chr18 using multiple QTL mapping. Of the 24 eQTLs colocalizing with the Chr8-collagen QTL, 6 transcripts were correlated with relative collagen amount. Similarly, of the 7 eQTLs colocalizing with the Chr2-collagen QTL, 1 transcript, Gpr158, correlated with relative collagen. Of the 12 transcripts with an eQTLs in the Chr18-covariate region, only Fgf1 correlated with relative collagen amount. Furthermore, 2 of the transcripts, Pdlim3 (Chr8) and Itga6 (Chr2), with eQTLs overlapping with collagen QTLs, had a genetic covariate on Chr18 that coincided with the Chr18 collagen covariate locus. Application of recombinant human FGF1 to isolated cardiac fibroblasts in culture affected the level of expression of Pdlim3, Itga6, and Gpr158. CONCLUSIONS: We here mapped a possible novel genetic network modulating collagen deposition in mouse left ventricular myocardium.


Subject(s)
Collagen/metabolism , Genome , Animals , Cells, Cultured , Chromosome Mapping , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 1/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression/drug effects , Gene Regulatory Networks , Genotype , Heart Ventricles/metabolism , Integrin alpha6/genetics , Integrin alpha6/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Quantitative Trait Loci , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...