Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Bioorg Med Chem Lett ; 46: 128162, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34062251

ABSTRACT

In the United States, approximately one million individuals are hospitalized every year for arrhythmias, making arrhythmias one of the top causes of healthcare expenditures. Mexiletine is currently used as an antiarrhythmic drug but has limitations. The purpose of this work was to use normal and Long QT syndrome Type 3 (LQTS3) patient-derived human induced pluripotent stem cell (iPSC)-derived cardiomyocytes to identify an analog of mexiletine with superior drug-like properties. Compared to racemic mexiletine, medicinal chemistry optimization of substituted racemic pyridyl phenyl mexiletine analogs resulted in a more potent sodium channel inhibitor with greater selectivity for the sodium over the potassium channel and for late over peak sodium current.


Subject(s)
Cardiac Conduction System Disease/pathology , Induced Pluripotent Stem Cells/chemistry , Long QT Syndrome/pathology , Mexiletine/pharmacology , Myocytes, Cardiac/pathology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Pyridines/pharmacology , Dose-Response Relationship, Drug , Humans , Mexiletine/chemistry , Molecular Structure , Pyridines/chemistry , Structure-Activity Relationship
2.
Circ Res ; 112(10): 1310-22, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23532596

ABSTRACT

RATIONALE: A chromosomal haplotype producing cardiac overexpression of dipeptidyl peptidase-like protein-6 (DPP6) causes familial idiopathic ventricular fibrillation. The molecular basis of transient outward current (I(to)) in Purkinje fibers (PFs) is poorly understood. We hypothesized that DPP6 contributes to PF I(to) and that its overexpression might specifically alter PF I(to) properties and repolarization. OBJECTIVE: To assess the potential role of DPP6 in PF I(to). METHODS AND RESULTS: Clinical data in 5 idiopathic ventricular fibrillation patients suggested arrhythmia origin in the PF-conducting system. PF and ventricular muscle I(to) had similar density, but PF I(to) differed from ventricular muscle in having tetraethylammonium sensitivity and slower recovery. DPP6 overexpression significantly increased, whereas DPP6 knockdown reduced, I(to) density and tetraethylammonium sensitivity in canine PF but not in ventricular muscle cells. The K(+)-channel interacting ß-subunit K(+)-channel interacting protein type-2, essential for normal expression of I(to) in ventricular muscle, was weakly expressed in human PFs, whereas DPP6 and frequenin (neuronal calcium sensor-1) were enriched. Heterologous expression of Kv4.3 in Chinese hamster ovary cells produced small I(to); I(to) amplitude was greatly enhanced by coexpression with K(+)-channel interacting protein type-2 or DPP6. Coexpression of DPP6 with Kv4.3 and K(+)-channel interacting protein type-2 failed to alter I(to) compared with Kv4.3/K(+)-channel interacting protein type-2 alone, but DPP6 expression with Kv4.3 and neuronal calcium sensor-1 (to mimic PF I(to) composition) greatly enhanced I(to) compared with Kv4.3/neuronal calcium sensor-1 and recapitulated characteristic PF kinetic/pharmacological properties. A mathematical model of cardiac PF action potentials showed that I(to) enhancement can greatly accelerate PF repolarization. CONCLUSIONS: These results point to a previously unknown central role of DPP6 in PF I(to), with DPP6 gain of function selectively enhancing PF current, and suggest that a DPP6-mediated PF early-repolarization syndrome might be a novel molecular paradigm for some forms of idiopathic ventricular fibrillation.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/physiology , Kv Channel-Interacting Proteins/physiology , Nerve Tissue Proteins/physiology , Potassium Channels/physiology , Purkinje Fibers/physiology , Shal Potassium Channels/physiology , Ventricular Fibrillation/physiopathology , Adult , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Disease Models, Animal , Dogs , Female , Gene Knockdown Techniques , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , In Vitro Techniques , Kv Channel-Interacting Proteins/drug effects , Kv Channel-Interacting Proteins/genetics , Male , Middle Aged , Models, Theoretical , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/genetics , Purkinje Fibers/pathology , Shal Potassium Channels/drug effects , Shal Potassium Channels/genetics , Tetraethylammonium/pharmacology , Transfection
3.
Proc Natl Acad Sci U S A ; 109(18): 7103-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22509038

ABSTRACT

KCNQ1 (Kv7.1) is a unique member of the superfamily of voltage-gated K(+) channels in that it displays a remarkable range of gating behaviors tuned by coassembly with different ß subunits of the KCNE family of proteins. To better understand the basis for the biophysical diversity of KCNQ1 channels, we here investigate the basis of KCNQ1 gating in the absence of ß subunits using voltage-clamp fluorometry (VCF). In our previous study, we found the kinetics and voltage dependence of voltage-sensor movements are very similar to those of the channel gate, as if multiple voltage-sensor movements are not required to precede gate opening. Here, we have tested two different hypotheses to explain KCNQ1 gating: (i) KCNQ1 voltage sensors undergo a single concerted movement that leads to channel opening, or (ii) individual voltage-sensor movements lead to channel opening before all voltage sensors have moved. Here, we find that KCNQ1 voltage sensors move relatively independently, but that the channel can conduct before all voltage sensors have activated. We explore a KCNQ1 point mutation that causes some channels to transition to the open state even in the absence of voltage-sensor movement. To interpret these results, we adopt an allosteric gating scheme wherein KCNQ1 is able to transition to the open state after zero to four voltage-sensor movements. This model allows for widely varying gating behavior, depending on the relative strength of the opening transition, and suggests how KCNQ1 could be controlled by coassembly with different KCNE family members.


Subject(s)
Ion Channel Gating , KCNQ1 Potassium Channel/metabolism , Allosteric Site , Amino Acid Substitution , Animals , Female , Humans , In Vitro Techniques , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/genetics , Models, Biological , Mutagenesis, Site-Directed , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Xenopus laevis
4.
Proc Natl Acad Sci U S A ; 107(52): 22710-5, 2010 Dec 28.
Article in English | MEDLINE | ID: mdl-21149716

ABSTRACT

The delayed rectifier I(Ks) potassium channel, formed by coassembly of α- (KCNQ1) and ß- (KCNE1) subunits, is essential for cardiac function. Although KCNE1 is necessary to reproduce the functional properties of the native I(Ks) channel, the mechanism(s) through which KCNE1 modulates KCNQ1 is unknown. Here we report measurements of voltage sensor movements in KCNQ1 and KCNQ1/KCNE1 channels using voltage clamp fluorometry. KCNQ1 channels exhibit indistinguishable voltage dependence of fluorescence and current signals, suggesting a one-to-one relationship between voltage sensor movement and channel opening. KCNE1 coexpression dramatically separates the voltage dependence of KCNQ1/KCNE1 current and fluorescence, suggesting an imposed requirement for movements of multiple voltage sensors before KCNQ1/KCNE1 channel opening. This work provides insight into the mechanism by which KCNE1 modulates the I(Ks) channel and presents a mechanism for distinct ß-subunit regulation of ion channel proteins.


Subject(s)
Ion Channel Gating/physiology , KCNQ1 Potassium Channel/physiology , Potassium Channels, Voltage-Gated/physiology , Signal Transduction/physiology , Algorithms , Animals , Female , Fluorometry/methods , Humans , Ion Channel Gating/genetics , KCNQ1 Potassium Channel/genetics , Membrane Potentials , Microinjections , Models, Biological , Mutation , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/genetics , RNA, Complementary/administration & dosage , RNA, Complementary/genetics , Signal Transduction/genetics , Xenopus laevis
5.
Elife ; 122023 08 31.
Article in English | MEDLINE | ID: mdl-37650513

ABSTRACT

The slow delayed rectifier potassium current, IKs, conducted through pore-forming Q1 and auxiliary E1 ion channel complexes is important for human cardiac action potential repolarization. During exercise or fright, IKs is up-regulated by protein kinase A (PKA)-mediated Q1 phosphorylation to maintain heart rhythm and optimum cardiac performance. Sympathetic up-regulation of IKs requires recruitment of PKA holoenzyme (two regulatory - RI or RII - and two catalytic Cα subunits) to Q1 C-terminus by an A kinase anchoring protein (AKAP9). Mutations in Q1 or AKAP9 that abolish their functional interaction result in long QT syndrome type 1 and 11, respectively, which increases the risk of sudden cardiac death during exercise. Here, we investigated the utility of a targeted protein phosphorylation (TPP) approach to reconstitute PKA regulation of IKs in the absence of AKAP9. Targeted recruitment of endogenous Cα to E1-YFP using a GFP/YFP nanobody (nano) fused to RIIα enabled acute cAMP-mediated enhancement of IKs, reconstituting physiological regulation of the channel complex. By contrast, nano-mediated tethering of RIIα or Cα to Q1-YFP constitutively inhibited IKs by retaining the channel intracellularly in the endoplasmic reticulum and Golgi. Proteomic analysis revealed that distinct phosphorylation sites are modified by Cα targeted to Q1-YFP compared to free Cα. Thus, functional outcomes of synthetically recruited PKA on IKs regulation is critically dependent on the site of recruitment within the channel complex. The results reveal insights into divergent regulation of IKs by phosphorylation across different spatial and time scales, and suggest a TPP approach to develop new drugs to prevent exercise-induced sudden cardiac death.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , KCNQ1 Potassium Channel , Humans , Proteomics , Action Potentials , Death, Sudden, Cardiac
7.
Front Physiol ; 13: 902224, 2022.
Article in English | MEDLINE | ID: mdl-36505078

ABSTRACT

The congenital Long QT Syndrome (LQTS) is an inherited disorder in which cardiac ventricular repolarization is delayed and predisposes patients to cardiac arrhythmias and sudden cardiac death. LQT1 and LQT5 are LQTS variants caused by mutations in KCNQ1 or KCNE1 genes respectively. KCNQ1 and KCNE1 co-assemble to form critical IKS potassium channels. Beta-blockers are the standard of care for the treatment of LQT1, however, doing so based on mechanisms other than correcting the loss-of-function of K+ channels. ML277 and R-L3 are compounds that enhance IKS channels and slow channel deactivation in a manner that is dependent on the stoichiometry of KCNE1 subunits in the assembled channels. In this paper, we used expression of IKS channels in Chinese hamster ovary (CHO) cells and Xenopus oocytes to study the potential of these two drugs (ML277 and R-L3) for the rescue of LQT1 and LQT5 mutant channels. We focused on the LQT1 mutation KCNQ1-S546L, and two LQT5 mutations, KCNE1-L51H and KCNE1-G52R. We found ML277 and R-L3 potentiated homozygote LQTS mutations in the IKS complexes-KCNE1-G52R and KCNE1-L51H and in heterogeneous IKS channel complexes which mimic heterogeneous expression of mutations in patients. ML277 and R-L3 increased the mutant IKS current amplitude and slowed current deactivation, but not in wild type (WT) IKS. We obtained similar results in the LQT1 mutant (KCNQ1 S546L/KCNE1) with ML277 and R-L3. ML277 and R-L3 had a similar effect on the LQT1 and LQT5 mutants, however, ML277 was more effective than R-L3 in this modulation. Importantly we found that not all LQT5 mutants expressed with KCNQ1 resulted in channels that are potentiated by these drugs as the KCNE1 mutant D76N inhibited drug action when expressed with KCNQ1. Thus, our work shows that by directly studying the treatment of LQT1 and LQT5 mutations with ML277 and R-L3, we will understand the potential utility of these activators as options in specific LQTS therapeutics.

8.
Biomolecules ; 12(10)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36291551

ABSTRACT

Pulmonary arterial hypertension (PAH) is a devastating disease with high morbidity and mortality. Deleterious remodeling in the pulmonary arterial system leads to irreversible arterial constriction and elevated pulmonary arterial pressures, right heart failure, and eventually death. The difficulty in treating PAH stems in part from the complex nature of disease pathogenesis, with several signaling compounds known to be involved (e.g., endothelin-1, prostacyclins) which are indeed targets of PAH therapy. Over the last decade, potassium channelopathies were established as novel causes of PAH. More specifically, loss-of-function mutations in the KCNK3 gene that encodes the two-pore-domain potassium channel KCNK3 (or TASK-1) and loss-of-function mutations in the ABCC8 gene that encodes a key subunit, SUR1, of the ATP-sensitive potassium channel (KATP) were established as the first two potassium channelopathies in human cohorts with pulmonary arterial hypertension. Moreover, voltage-gated potassium channels (Kv) represent a third family of potassium channels with genetic changes observed in association with PAH. While other ion channel genes have since been reported in association with PAH, this review focuses on KCNK3, KATP, and Kv potassium channels as promising therapeutic targets in PAH, with recent experimental pharmacologic discoveries significantly advancing the field.


Subject(s)
Channelopathies , Hypertension, Pulmonary , Potassium Channels, Tandem Pore Domain , Potassium Channels, Voltage-Gated , Pulmonary Arterial Hypertension , Humans , Potassium Channels, Tandem Pore Domain/genetics , Channelopathies/drug therapy , Channelopathies/genetics , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Endothelin-1 , Nerve Tissue Proteins/metabolism , Familial Primary Pulmonary Hypertension/genetics , Prostaglandins I , Potassium , KATP Channels/genetics
9.
Front Physiol ; 13: 903050, 2022.
Article in English | MEDLINE | ID: mdl-35957984

ABSTRACT

ML277 and R-L3 are two small-molecule activators of KCNQ1, the pore-forming subunit of the slowly activating potassium channel IKs. KCNQ1 loss-of-function mutations prolong cardiac action potential duration and are associated with long QT syndrome, which predispose patients to lethal ventricular arrhythmia. ML277 and R-L3 enhance KCNQ1 current amplitude and slow deactivation. However, the presence of KCNE1, an auxiliary subunit of IKs channels, renders the channel insensitive to both activators. We found that ML277 effects are dependent on several residues in the KCNQ1 pore domain. Some of these residues are also necessary for R-L3 effects. These residues form a putative hydrophobic pocket located between two adjacent KCNQ1 subunits, where KCNE1 subunits are thought to dwell, thus providing an explanation for how KCNE1 renders the IKs channel insensitive to these activators. Our experiments showed that the effect of R-L3 on voltage sensor movement during channel deactivation was much more prominent than that of ML277. Simulations using a KCNQ1 kinetic model showed that the effects of ML277 and R-L3 could be reproduced through two different effects on channel gating: ML277 enhances KCNQ1 channel function through a pore-dependent and voltage sensor-independent mechanism, while R-L3 affects both channel pore and voltage sensor.

10.
Channels (Austin) ; 16(1): 173-184, 2022 12.
Article in English | MEDLINE | ID: mdl-35949058

ABSTRACT

The congenital long QT syndrome (LQTS), one of the most common cardiac channelopathies, is characterized by delayed ventricular repolarization underlying prolongation of the QT interval of the surface electrocardiogram. LQTS is caused by mutations in genes coding for cardiac ion channels or ion channel-associated proteins. The major therapeutic approach to LQTS management is beta blocker therapy which has been shown to be effective in treatment of LQTS variants caused by mutations in K+ channels. However, this approach has been questioned in the treatment of patients identified as LQTS variant 3(LQT3) patients who carry mutations in SCN5A, the gene coding for the principal cardiac Na+ channel. LQT3 mutations are gain of function mutations that disrupt spontaneous Na+ channel inactivation and promote persistent or late Na+ channel current (INaL) that delays repolarization and underlies QT prolongation. Clinical investigation of patients with the two most common LQT3 mutations, the ΔKPQ and the E1784K mutations, found beta blocker treatment a useful therapeutic approach for managing arrhythmias in this patient population. However, there is little experimental data that reveals the mechanisms underlying these antiarrhythmic actions. Here, we have investigated the effects of the beta blocker propranolol on INaL expressed by ΔKPQ and E1784K channels in induced pluripotent stem cells derived from patients carrying these mutations. Our results indicate that propranolol preferentially inhibits INaL expressed by these channels suggesting that the protective effects of propranolol in treating LQT3 patients is due in part to modulation of INaL.


Subject(s)
Long QT Syndrome , Pluripotent Stem Cells , Arrhythmias, Cardiac/genetics , Humans , Long QT Syndrome/drug therapy , Long QT Syndrome/genetics , Muscle Cells/metabolism , Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , Pluripotent Stem Cells/metabolism , Propranolol/pharmacology , Propranolol/therapeutic use , Sodium Channels
11.
J Physiol ; 589(Pt 24): 6093-104, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22025662

ABSTRACT

Human embryonic stem cells (hESCs) are an important cellular model for studying ion channel function in the context of a human cardiac cell and will provide a wealth of information about both heritable arrhythmias and acquired electrophysiological disorders. However, detailed electrophysiological characterization of the important cardiac ion channels has been so far overlooked. Because mutations in the gene for the I(Ks) α subunit, KCNQ1, constitute the majority of long QT syndrome (LQT-1) cases, we have carried out a detailed biophysical analysis of this channel expressed in hESCs to establish baseline I(Ks) channel biophysical properties in cardiac myocytes derived from hESCs (hESC-CMs). I(Ks) channels are heteromultimeric proteins consisting of four identical α-subunits (KCNQ1) assembled with auxiliary ß-subunits (KCNE1). We found that the half-maximal I(Ks) activation voltage in hESC-CMs and in myocytes derived from human induced pluripotent stems cells (hiPSC-CMs) falls between that of KCNQ1 channels expressed alone and with full complement of KCNE1, the major KCNE subunit expressed in hESC-CMs as shown by qPCR analysis. Overexpression of KCNE1 by transfection of hESC-CMs markedly shifted and slowed native I(Ks) activation implying assembly of additional KCNE1 subunits with endogenous channels. Our results in hESC-CMs, which indicate an I(Ks) subunit stoichiometry that can be altered by variable KCNE1 expression, suggest the possibility for variable I(Ks) function in the developing heart, in different tissues in the heart, and in disease. This establishes a new baseline for I(Ks) channel properties in myocytes derived from pluripotent stem cells and will guide future studies in patient-specific hiPSCs.


Subject(s)
KCNQ1 Potassium Channel/physiology , Myocytes, Cardiac/physiology , Potassium Channels, Voltage-Gated/physiology , Protein Subunits/physiology , Action Potentials/physiology , Cell Line , Cells, Cultured , Charybdotoxin/pharmacology , Cytokines/pharmacology , Embryonic Stem Cells/cytology , Fibroblasts/physiology , HEK293 Cells , Humans , Neurotoxins/pharmacology
12.
J Med Chem ; 64(9): 5384-5403, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33942619

ABSTRACT

Ventricular cardiac arrhythmia (VA) arises in acquired or congenital heart disease. Long QT syndrome type-3 (LQT3) is a congenital form of VA caused by cardiac sodium channel (INaL) SCN5A mutations that prolongs cardiac action potential (AP) and enhances INaL current. Mexiletine inhibits INaL and shortens the QT interval in LQT3 patients. Above therapeutic doses, mexiletine prolongs the cardiac AP. We explored structure-activity relationships (SAR) for AP shortening and prolongation using dynamic medicinal chemistry and AP kinetics in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using patient-derived LQT3 and healthy hiPSC-CMs, we resolved distinct SAR for AP shortening and prolongation effects in mexiletine analogues and synthesized new analogues with enhanced potency and selectivity for INaL. This resulted in compounds with decreased AP prolongation effects, increased metabolic stability, increased INaL selectivity, and decreased avidity for the potassium channel. This study highlights using hiPSC-CMs to guide medicinal chemistry and "drug development in a dish".


Subject(s)
Anti-Arrhythmia Agents/chemistry , Cardiac Conduction System Disease/pathology , Long QT Syndrome/pathology , Mexiletine/analogs & derivatives , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Behavior, Animal/drug effects , Cardiac Conduction System Disease/metabolism , Cells, Cultured , Drug Design , Drug Stability , Half-Life , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Long QT Syndrome/metabolism , Male , Mexiletine/pharmacology , Mice , Mice, Inbred BALB C , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
13.
Proc Natl Acad Sci U S A ; 104(52): 20990-5, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18093912

ABSTRACT

A-kinase anchoring proteins (AKAPs) recruit signaling molecules and present them to downstream targets to achieve efficient spatial and temporal control of their phosphorylation state. In the heart, sympathetic nervous system (SNS) regulation of cardiac action potential duration (APD), mediated by beta-adrenergic receptor (betaAR) activation, requires assembly of AKAP9 (Yotiao) with the I(Ks) potassium channel alpha subunit (KCNQ1). KCNQ1 mutations that disrupt this complex cause type 1 long-QT syndrome (LQT1), one of the potentially lethal heritable arrhythmia syndromes. Here, we report identification of (i) regions on Yotiao critical to its binding to KCNQ1 and (ii) a single putative LQTS-causing mutation (S1570L) in AKAP9 (Yotiao) localized to the KCNQ1 binding domain in 1/50 (2%) subjects with a clinically robust phenotype for LQTS but absent in 1,320 reference alleles. The inherited S1570L mutation reduces the interaction between KCNQ1 and Yotiao, reduces the cAMP-induced phosphorylation of the channel, eliminates the functional response of the I(Ks) channel to cAMP, and prolongs the action potential in a computational model of the ventricular cardiocyte. These reconstituted cellular consequences of the inherited S1570L-Yotiao mutation are consistent with delayed repolarization of the ventricular action potential observed in the affected siblings. Thus, we have demonstrated a link between genetic perturbations in AKAP and human disease in general and AKAP9 and LQTS in particular.


Subject(s)
A Kinase Anchor Proteins/genetics , Cytoskeletal Proteins/genetics , KCNQ1 Potassium Channel/genetics , Long QT Syndrome/genetics , Mutation , A Kinase Anchor Proteins/physiology , Adolescent , Adult , Aged , Binding Sites , Child , Child, Preschool , Cytoskeletal Proteins/physiology , DNA Mutational Analysis , Female , Humans , Infant , KCNQ1 Potassium Channel/physiology , Male , Middle Aged , Potassium Channels/metabolism , Protein Structure, Tertiary , Receptors, Adrenergic, beta/metabolism
14.
Cell Stem Cell ; 27(5): 813-821.e6, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32931730

ABSTRACT

Modeling cardiac disorders with human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes is a new paradigm for preclinical testing of candidate therapeutics. However, disease-relevant physiological assays can be complex, and the use of hiPSC-cardiomyocyte models of congenital disease phenotypes for guiding large-scale screening and medicinal chemistry have not been shown. We report chemical refinement of the antiarrhythmic drug mexiletine via high-throughput screening of hiPSC-CMs derived from patients with the cardiac rhythm disorder long QT syndrome 3 (LQT3) carrying SCN5A sodium channel variants. Using iterative cycles of medicinal chemistry synthesis and testing, we identified drug analogs with increased potency and selectivity for inhibiting late sodium current across a panel of 7 LQT3 sodium channel variants and suppressing arrhythmic activity across multiple genetic and pharmacological hiPSC-CM models of LQT3 with diverse backgrounds. These mexiletine analogs can be exploited as mechanistic probes and for clinical development.


Subject(s)
Induced Pluripotent Stem Cells , Action Potentials , Anti-Arrhythmia Agents/pharmacology , Humans , Myocytes, Cardiac , Patch-Clamp Techniques
15.
Circ Res ; 99(11): 1225-32, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-17082480

ABSTRACT

Long QT syndrome (LQTS) type 3 (LQT3), typified by the DeltaKPQ mutation (LQT3 mutation in which amino acid residues 1505 to 1507 [KPQ] are deleted), is caused by increased sodium entry during the action potential plateau resulting from mutation-altered inactivation of the Na(v)1.5 channel. Although rare, LQT3 is the most lethal of common LQTS variants. Here we tested the hypothesis that cellular electrical dysfunction, caused not only by action potential prolongation but also by mutation-altered Na(+) entry, distinguishes LQT3 from other LQTS variants and may contribute to its distinct lethality. We compared cellular electrical activity in myocytes isolated from mice heterozygous for the DeltaKPQ mutation (DeltaKPQ) and myocytes from wild-type littermates. Current-clamp pause protocols induced rate-dependent spontaneous diastolic activity (delayed after depolarizations) in 6 of 7 DeltaKPQ, but no wild-type, myocytes (n=11) tested. Voltage-clamp pause protocols that independently control depolarization duration and interpulse interval identified a distinct contribution of both depolarization duration and mutant Na(+) channel activity to the generation of Ca(i)(2+)-dependent diastolic transient inward current. This was found at rates and depolarization durations relevant both to the mouse model and to LQT3 patients. Flecainide, which preferentially inhibits mutation-altered late Na(+) current and is used to treat LQT3 patients, suppresses transient inward current formation in voltage-clamped DeltaKPQ myocytes. Our results demonstrate a marked contribution of mutation-altered Na(+) entry to the incidence of pause-dependent spontaneous diastolic activity in DeltaKPQ myocytes and suggest that altered Na(+) entry may contribute to the elevated lethality of LQT3 versus other LQTS variants.


Subject(s)
Genetic Variation , Heart/physiopathology , Long QT Syndrome/physiopathology , Myocytes, Cardiac/metabolism , Sodium Channels/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Diastole , Electrophysiology , Flecainide/pharmacology , Gene Deletion , Glutamine , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Lysine , Mice , Mice, Transgenic , Patch-Clamp Techniques , Proline , Sodium Channels/drug effects
17.
Circ Genom Precis Med ; 11(10): e002087, 2018 10.
Article in English | MEDLINE | ID: mdl-30354297

ABSTRACT

BACKGROUND: In pulmonary arterial hypertension (PAH), pathological changes in pulmonary arterioles progressively raise pulmonary artery pressure and increase pulmonary vascular resistance, leading to right heart failure and high mortality rates. Recently, the first potassium channelopathy in PAH, because of mutations in KCNK3, was identified as a genetic cause and pharmacological target. METHODS: Exome sequencing was performed to identify novel genes in a cohort of 99 pediatric and 134 adult-onset group I PAH patients. Novel rare variants in the gene identified were independently identified in a cohort of 680 adult-onset patients. Variants were expressed in COS cells and function assessed by patch-clamp and rubidium flux analysis. RESULTS: We identified a de novo novel heterozygous predicted deleterious missense variant c.G2873A (p.R958H) in ABCC8 in a child with idiopathic PAH. We then evaluated all individuals in the original and a second cohort for rare or novel variants in ABCC8 and identified 11 additional heterozygous predicted damaging ABCC8 variants. ABCC8 encodes SUR1 (sulfonylurea receptor 1)-a regulatory subunit of the ATP-sensitive potassium channel. We observed loss of ATP-sensitive potassium channel function for all ABCC8 variants evaluated and pharmacological rescue of all channel currents in vitro by the SUR1 activator, diazoxide. CONCLUSIONS: Novel and rare missense variants in ABCC8 are associated with PAH. Identified ABCC8 mutations decreased ATP-sensitive potassium channel function, which was pharmacologically recovered.


Subject(s)
Exome , Familial Primary Pulmonary Hypertension/genetics , Mutation, Missense , Sulfonylurea Receptors/genetics , Adult , Amino Acid Substitution , Child , DNA Mutational Analysis , Familial Primary Pulmonary Hypertension/drug therapy , Female , Humans , Male
18.
Circ Res ; 96(5): e25-34, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15731462

ABSTRACT

I(Ks), the slowly activating component of the delayed rectifier current, plays a major role in repolarization of the cardiac action potential (AP). Genetic mutations in the alpha- (KCNQ1) and beta- (KCNE1) subunits of I(Ks) underlie Long QT Syndrome type 1 and 5 (LQT-1 and LQT-5), respectively, and predispose carriers to the development of polymorphic ventricular arrhythmias and sudden cardiac death. beta-adrenergic stimulation increases I(Ks) and results in rate dependent AP shortening, a control system that can be disrupted by some mutations linked to LQT-1 and LQT-5. The mechanisms by which I(Ks) regulates action potential duration (APD) during beta-adrenergic stimulation at different heart rates are not known, nor are the consequences of mutation induced disruption of this regulation. Here we develop a complementary experimental and theoretical approach to address these questions. We reconstituted I(Ks) in CHO cells (ie, KCNQ1 coexpressed with KCNE1 and the adaptator protein Yotiao) and quantitatively examined the effects of beta-adrenergic stimulation on channel kinetics. We then developed theoretical models of I(Ks) in the absence and presence of beta-adrenergic stimulation. We simulated the effects of sympathetic stimulation on channel activation (speeding) and deactivation (slowing) kinetics on the whole cell action potential under different pacing conditions. The model suggests these kinetic effects are critically important in rate-dependent control of action potential duration. We also investigate the effects of two LQT-5 mutations that alter kinetics and impair sympathetic stimulation of I(Ks) and show the likely mechanism by which they lead to tachyarrhythmias and indicate a distinct role of I(KS) kinetics in this electrical dysfunction. The full text of this article is available online at http://circres.ahajournals.org.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cytoskeletal Proteins/physiology , Myocytes, Cardiac/physiology , Potassium Channels, Voltage-Gated/physiology , Sympathetic Nervous System/physiology , A Kinase Anchor Proteins , Action Potentials/physiology , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Substitution , Animals , CHO Cells , Computer Simulation , Cricetinae , Cricetulus , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeletal Proteins/genetics , Delayed Rectifier Potassium Channels , Humans , Ion Channel Gating/physiology , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Kinetics , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Models, Cardiovascular , Mutation, Missense , Patch-Clamp Techniques , Phosphorylation , Point Mutation , Potassium/metabolism , Potassium Channels, Voltage-Gated/genetics , Protein Processing, Post-Translational , Receptors, Adrenergic, beta/physiology , Recombinant Fusion Proteins/physiology , Second Messenger Systems/physiology , Tachycardia/physiopathology , Transfection
19.
Sci Rep ; 7: 45911, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28383569

ABSTRACT

KCNQ1 is a voltage-gated potassium channel that is modulated by the beta-subunit KCNE1 to generate IKs, the slow delayed rectifier current, which plays a critical role in repolarizing the cardiac action potential. Two KCNQ1 gain-of-function mutations that cause a genetic form of atrial fibrillation, S140G and V141M, drastically slow IKs deactivation. However, the underlying gating alterations remain unknown. Voltage clamp fluorometry (VCF) allows simultaneous measurement of voltage sensor movement and current through the channel pore. Here, we use VCF and kinetic modeling to determine the effects of mutations on channel voltage-dependent gating. We show that in the absence of KCNE1, S140G, but not V141M, directly slows voltage sensor movement, which indirectly slows current deactivation. In the presence of KCNE1, both S140G and V141M slow pore closing and alter voltage sensor-pore coupling, thereby slowing current deactivation. Our results suggest that KCNE1 can mediate changes in pore movement and voltage sensor-pore coupling to slow IKs deactivation and provide a key step toward developing mechanism-based therapies.


Subject(s)
Atrial Fibrillation/genetics , Genetic Predisposition to Disease/genetics , Ion Channel Gating/genetics , KCNQ1 Potassium Channel/genetics , Mutation, Missense , Action Potentials/genetics , Animals , Female , Fluorometry/methods , Humans , Kinetics , Oocytes/metabolism , Oocytes/physiology , Patch-Clamp Techniques/methods , Potassium Channels, Voltage-Gated/genetics , Xenopus laevis
20.
J Am Heart Assoc ; 6(9)2017 Sep 09.
Article in English | MEDLINE | ID: mdl-28889099

ABSTRACT

BACKGROUND: Heterozygous loss of function mutations in the KCNK3 gene cause hereditary pulmonary arterial hypertension (PAH). KCNK3 encodes an acid-sensitive potassium channel, which contributes to the resting potential of human pulmonary artery smooth muscle cells. KCNK3 is widely expressed in the body, and dimerizes with other KCNK3 subunits, or the closely related, acid-sensitive KCNK9 channel. METHODS AND RESULTS: We engineered homomeric and heterodimeric mutant and nonmutant KCNK3 channels associated with PAH. Using whole-cell patch-clamp electrophysiology in human pulmonary artery smooth muscle and COS7 cell lines, we determined that homomeric and heterodimeric mutant channels in heterozygous KCNK3 conditions lead to mutation-specific severity of channel dysfunction. Both wildtype and mutant KCNK3 channels were activated by ONO-RS-082 (10 µmol/L), causing cell hyperpolarization. We observed robust gene expression of KCNK3 in healthy and familial PAH patient lungs, but no quantifiable expression of KCNK9, and demonstrated in functional studies that KCNK9 minimizes the impact of select KCNK3 mutations when the 2 channel subunits co-assemble. CONCLUSIONS: Heterozygous KCNK3 mutations in PAH lead to variable loss of channel function via distinct mechanisms. Homomeric and heterodimeric mutant KCNK3 channels represent novel therapeutic substrates in PAH. Pharmacological and pH-dependent activation of wildtype and mutant KCNK3 channels in pulmonary artery smooth muscle cells leads to membrane hyperpolarization. Co-assembly of KCNK3 with KCNK9 subunits may provide protection against KCNK3 loss of function in tissues where both KCNK9 and KCNK3 are expressed, contributing to the lung-specific phenotype observed clinically in patients with PAH because of KCNK3 mutations.


Subject(s)
Familial Primary Pulmonary Hypertension/genetics , Heterozygote , Loss of Function Mutation , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Animals , Arterial Pressure/genetics , COS Cells , Case-Control Studies , Chlorobenzoates/pharmacology , Chlorocebus aethiops , Cinnamates/pharmacology , Familial Primary Pulmonary Hypertension/metabolism , Familial Primary Pulmonary Hypertension/physiopathology , Genetic Predisposition to Disease , Humans , Hydrogen-Ion Concentration , Membrane Potentials , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Phenotype , Potassium Channels, Tandem Pore Domain/agonists , Potassium Channels, Tandem Pore Domain/metabolism , Protein Multimerization , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Transfection , ortho-Aminobenzoates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL