Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nat Med ; 30(1): 106-111, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38092897

ABSTRACT

Existing antiarrhythmic drugs to treat atrial fibrillation (AF) have incomplete efficacy, contraindications and adverse effects, including proarrhythmia. AP30663, an inhibitor of the KCa2 channel, has demonstrated AF efficacy in animals; however, its efficacy in humans with AF is unknown. Here we conducted a phase 2 trial in which patients with a current episode of AF lasting for 7 days or less were randomized to receive an intravenous infusion of 3 or 5 mg kg-1 AP30663 or placebo. The trial was prematurely discontinued because of slow enrollment during the coronavirus disease 2019 pandemic. The primary endpoint of the trial was cardioversion from AF to sinus rhythm within 90 min from the start of the infusion, analyzed with Bayesian statistics. Among 59 patients randomized and included in the efficacy analyses, the primary endpoint occurred in 42% (5 of 12), 55% (12 of 22) and 0% (0 of 25) of patients treated with 3 mg kg-1 AP30663, 5 mg kg-1 AP30663 or placebo, respectively. Both doses demonstrated more than 99.9% probability of superiority over placebo, surpassing the prespecified 95% threshold. The mean time to cardioversion, a secondary endpoint, was 47 (s.d. = 23) and 41 (s.d. = 24) minutes for 3 mg kg-1 and 5 mg kg-1 AP30663, respectively. AP30663 caused a transient increase in the QTcF interval, with a maximum mean effect of 37.7 ms for the 5 mg kg-1 dose. For both dose groups, no ventricular arrhythmias occurred and adverse event rates were comparable to the placebo group. AP30663 demonstrated AF cardioversion efficacy in patients with recent-onset AF episodes. KCa2 channel inhibition may be an attractive mechanism for rhythm control of AF that should be studied further in randomized trials. ClinicalTrials.gov registration: NCT04571385 .


Subject(s)
Atrial Fibrillation , Humans , Atrial Fibrillation/drug therapy , Bayes Theorem , Treatment Outcome , Anti-Arrhythmia Agents/adverse effects , Infusions, Intravenous
2.
Biophys J ; 122(7): 1143-1157, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36760125

ABSTRACT

Small-conductance (SK) calcium-activated potassium channels are a promising treatment target in atrial fibrillation. However, the functional properties that differentiate SK inhibitors remain poorly understood. The objective of this study was to determine how two unrelated SK channel inhibitors, apamin and AP14145, impact SK channel function in excised inside-out single-channel recordings. Surprisingly, both apamin and AP14145 exert much of their inhibition by inducing a class of very-long-lived channel closures (apamin: τc,vl = 11.8 ± 7.1 s, and AP14145: τc,vl = 10.3 ± 7.2 s), which were never observed under control conditions. Both inhibitors also induced changes to the three closed and two open durations typical of normal SK channel gating. AP14145 shifted the open duration distribution to favor longer open durations, whereas apamin did not alter open-state kinetics. AP14145 also prolonged the two shortest channel closed durations (AP14145: τc,s = 3.50 ± 0.81 ms, and τc,i = 32.0 ± 6.76 ms versus control: τc,s = 1.59 ± 0.19 ms, and τc,i = 13.5 ± 1.17 ms), thus slowing overall gating kinetics within bursts of channel activity. In contrast, apamin accelerated intraburst gating kinetics by shortening the two shortest closed durations (τc,s = 0.75 ± 0.10 ms and τc,i = 5.08 ± 0.49 ms) and inducing periods of flickery activity. Finally, AP14145 introduced a unique form of inhibition by decreasing unitary current amplitude. SK channels exhibited two clearly distinguishable amplitudes (control: Ahigh = 0.76 ± 0.03 pA, and Alow = 0.54 ± 0.03 pA). AP14145 both reduced the fraction of patches exhibiting the higher amplitude (AP14145: 4/9 patches versus control: 16/16 patches) and reduced the mean low amplitude (0.38 ± 0.03 pA). Here, we have demonstrated that both inhibitors introduce very long channel closures but that each also exhibits unique effects on other components of SK gating kinetics and unitary current. The combination of these effects is likely to be critical for understanding the functional differences of each inhibitor in the context of cyclical Ca2+-dependent channel activation in vivo.


Subject(s)
Potassium Channels , Small-Conductance Calcium-Activated Potassium Channels , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Apamin/pharmacology , Acetamides , Kinetics , Calcium/metabolism
3.
J Cardiovasc Electrophysiol ; 34(1): 126-134, 2023 01.
Article in English | MEDLINE | ID: mdl-36482155

ABSTRACT

BACKGROUND: Obstructive sleep apnea (OSA) creates a complex substrate for atrial fibrillation (AF), which is refractory to many clinically available pharmacological interventions. We investigated atrial antiarrhythmogenic properties and ventricular electrophysiological safety of small-conductance Ca2+ -activated K+ (SK)-channel inhibition in a porcine model for obstructive respiratory events. METHODS: In spontaneously breathing pigs, obstructive respiratory events were simulated by intermittent negative upper airway pressure (INAP) applied via a pressure device connected to the intubation tube. INAP was applied for 75 s, every 10 min, three times before and three times during infusion of the SK-channel inhibitor AP14145. Atrial effective refractory periods (AERP) were acquired before (pre-INAP), during (INAP) and after (post-) INAP. AF-inducibility was determined by a S1S2 atrial pacing protocol. Ventricular arrhythmicity was evaluated by heart rate adjusted QT-interval duration (QT-paced) and electromechanical window (EMW) shortening. RESULTS: During vehicle infusion, INAP transiently shortened AERP (pre-INAP: 135 ± 10 ms vs. post-INAP 101 ± 11 ms; p = .008) and increased AF-inducibility. QT-paced prolonged during INAP (pre-INAP 270 ± 7 ms vs. INAP 275 ± 7 ms; p = .04) and EMW shortened progressively throughout INAP and post-INAP (pre-INAP 80 ± 4 ms; INAP 59 ± 6 ms, post-INAP 46 ± 10 ms). AP14145 prolonged baseline AERP, partially prevented INAP-induced AERP-shortening and reduced AF-susceptibility. AP14145 did not alter QT-paced at baseline (pre-AP14145 270 ± 7 ms vs. AP14145 268 ± 6 ms, p = .83) or QT-paced and EMW-shortening during INAP. CONCLUSION: In a pig model for obstructive respiratory events, the SK-channel-inhibitor AP14145 prevented INAP-associated AERP-shortening and AF-susceptibility without impairing ventricular electrophysiology. Whether SK-channels represent a target for OSA-related AF in humans warrants further study.


Subject(s)
Atrial Fibrillation , Sleep Apnea, Obstructive , Humans , Swine , Animals , Atrial Fibrillation/prevention & control , Acetamides
4.
Ugeskr Laeger ; 184(24)2022 06 13.
Article in Danish | MEDLINE | ID: mdl-35703076

ABSTRACT

Viroporins are ion channels found in many viruses, where they contribute to virus life cycle and thereby pathogenesis. Viroporin targeting is a known, yet largely unexplored, therapeutic strategy so far only used in Influenza A with the drugs amantadine and rimantadine. In this review, we seek to utilize the inhibition by amantadine of the viroporin Protein E in SARS-CoV-2 in an attempt to treat COVID-19 in its early stages. We are executing a double-blinded placebo-controlled trial based on promising in vivo and in vitro work as a stepping-stone for establishing a therapeutic antiviral regime: blocking of viroporins.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Amantadine , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Humans , SARS-CoV-2 , Viroporin Proteins
5.
ACS Chem Biol ; 16(9): 1654-1662, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34423964

ABSTRACT

Marine tunicates produce defensive amino-acid-derived metabolites, including 2-(3,5-diiodo-4-methoxyphenyl)ethan-1-amine (DIMTA), but their mechanisms of action are rarely known. Using an assay-guided approach, we found that out of the many different sensory cells in the mouse dorsal root ganglion (DRG), DIMTA selectively affected low-threshold cold thermosensors. Whole-cell electrophysiology experiments using DRG cells, channels expressed in Xenopus oocytes, and human cell lines revealed that DIMTA blocks several potassium channels, reducing the magnitude of the afterhyperpolarization and increasing the baseline intracellular calcium concentration [Ca2+]i of low-threshold cold thermosensors. When injected into mice, DIMTA increased the threshold of cold sensation by >3 °C. DIMTA may thus serve as a lead in the further design of compounds that inhibit problems in the cold-sensory system, such as cold allodynia and other neuropathic pain conditions.


Subject(s)
Amines/metabolism , Calcium Channels/metabolism , Sensory Receptor Cells/metabolism , Amines/administration & dosage , Animals , Calcium/metabolism , Ganglia, Spinal/metabolism , Male , Mice , Patch-Clamp Techniques , Signal Transduction , Thermosensing/physiology , Urochordata , Vertebrates
6.
PLoS Biol ; 19(4): e3001144, 2021 04.
Article in English | MEDLINE | ID: mdl-33872299

ABSTRACT

Delineating human cardiac pathologies and their basic molecular mechanisms relies on research conducted in model organisms. Yet translating findings from preclinical models to humans present a significant challenge, in part due to differences in cardiac protein expression between humans and model organisms. Proteins immediately determine cellular function, yet their large-scale investigation in hearts has lagged behind those of genes and transcripts. Here, we set out to bridge this knowledge gap: By analyzing protein profiles in humans and commonly used model organisms across cardiac chambers, we determine their commonalities and regional differences. We analyzed cardiac tissue from each chamber of human, pig, horse, rat, mouse, and zebrafish in biological replicates. Using mass spectrometry-based proteomics workflows, we measured and evaluated the abundance of approximately 7,000 proteins in each species. The resulting knowledgebase of cardiac protein signatures is accessible through an online database: atlas.cardiacproteomics.com. Our combined analysis allows for quantitative evaluation of protein abundances across cardiac chambers, as well as comparisons of cardiac protein profiles across model organisms. Up to a quarter of proteins with differential abundances between atria and ventricles showed opposite chamber-specific enrichment between species; these included numerous proteins implicated in cardiac disease. The generated proteomics resource facilitates translational prospects of cardiac studies from model organisms to humans by comparisons of disease-linked protein networks across species.


Subject(s)
Myocardium/metabolism , Proteome/metabolism , Animals , Heart/physiology , Heart Ventricles/chemistry , Heart Ventricles/metabolism , Horses , Humans , Mice , Models, Animal , Myocardium/chemistry , Organ Specificity , Protein Processing, Post-Translational , Proteome/analysis , Proteomics/methods , Rats , Species Specificity , Swine , Zebrafish
7.
Clin Transl Sci ; 13(6): 1336-1344, 2020 11.
Article in English | MEDLINE | ID: mdl-32725783

ABSTRACT

Pharmacological cardioversion of atrial fibrillation (AF) is frequently inefficacious. AP30663, a small conductance Ca2+ activated K+ (KCa 2) channel blocker, prolonged the atrial effective refractory period in preclinical studies and subsequently converted AF into normal sinus rhythm. This first-in-human study evaluated the safety and tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) effects were explored. Forty-seven healthy male volunteers (23.7 ± 3.0 years) received AP30663 intravenously in ascending doses. Due to infusion site reactions, changes to the formulation and administration were implemented in the latter 24 volunteers. Extractions from a 24-hour continuous electrocardiogram were used to evaluate the PD effect of AP30663. Data were analyzed with a repeated measure analysis of covariance, noncompartmental analysis, and concentration-effect analysis. In total, 33 of 34 adverse events considered related to AP30663 exposure were related to the infusion site, mild in severity, and temporary in nature, although full recovery took up to 110 days. After formulation and administration changes, the local infusion site reaction remained, but the median duration was shorter despite higher dose levels. AP30663 displayed a less than dose proportional increase in peak plasma concentration (Cmax ) and a terminal half-life of around 5 hours. In healthy volunteers, no effect of AP30663 was observed on electrocardiographic parameters, other than a concentration-dependent effect on the corrected QT Fridericia's formula interval (+18.8 ± 4.3 ms for the highest dose level compared with time matched placebo). In conclusion, administration of AP30663, a novel KCa 2 channel inhibitor, was safe and well-tolerated systemically in humans, supporting further development in patients with AF undergoing cardioversion.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Electrocardiography/drug effects , Injection Site Reaction/diagnosis , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Adolescent , Adult , Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/pharmacokinetics , Atrial Fibrillation/drug therapy , Dose-Response Relationship, Drug , Double-Blind Method , Half-Life , Healthy Volunteers , Heart Rate/drug effects , Humans , Infusions, Intravenous , Injection Site Reaction/etiology , Male , Middle Aged , Severity of Illness Index , Young Adult
8.
Acta Physiol (Oxf) ; 229(4): e13471, 2020 08.
Article in English | MEDLINE | ID: mdl-32223014

ABSTRACT

AIM: We aimed to assess the ability of natural and modified polyunsaturated fatty acids (PUFAs) to shorten QT interval in ex-vivo and in-vivo guinea pig hearts. METHODS: The effect of one natural (docosahexaenoic acid [DHA]) and three modified (linoleoyl glycine [Lin-GLY], docosahexaenoyl glycine [DHA-GLY], N-arachidonoyl taurine [N-AT]) PUFAs on ventricular action potential duration (APD) and QT interval was studied in a E4031 drug-induced long QT2 model of ex-vivo guinea pig hearts. The effect of DHA-GLY on QT interval was also studied in in-vivo guinea pig hearts upon intravenous administration. The effect of modified PUFAs on IKs was studied using Xenopus laevis oocytes expressing human KCNQ1 and KCNE1. RESULTS: All tested PUFAs shortened ADP and QT interval in ex-vivo guinea pig hearts, however, with different ability in restoring baseline APD/QT interval with specific modified PUFAs being most efficacious. Despite comparable ability in activating the human KCNQ1/KCNE1 channel, Lin-GLY was not as effective in shortening APD/QT interval as DHA-GLY in ex-vivo hearts. By constructing a guinea pig-like KCNE1, we found Lin-GLY to induce less activating effect compared with DHA-GLY on human KCNQ1 co-expressed with guinea pig-like KCNE1. Docosahexaenoyl glycine was studied in more detail and was found to shorten QT interval in in-vivo guinea pig hearts. CONCLUSION: Our results show that specific PUFAs shorten QT interval in guinea pig hearts. The tendency of modified PUFAs with pronounced IKs channel activating effect to better restore QT interval suggests that modifying PUFAs to target the IKs channel is a means to improve the QT-shortening effect.


Subject(s)
Fatty Acids, Unsaturated/pharmacology , Heart/drug effects , KCNQ1 Potassium Channel/agonists , Action Potentials , Animals , Guinea Pigs , Heart Ventricles , In Vitro Techniques , Long QT Syndrome , Oocytes , Potassium Channels, Voltage-Gated/agonists , Xenopus laevis
9.
Front Pharmacol ; 9: 1409, 2018.
Article in English | MEDLINE | ID: mdl-30559671

ABSTRACT

A variety of polycyclic pyridines have been proposed as inhibitors of the small conductance calcium-activated potassium (SK) channel. To this group belongs 2,6-bis(2-benzimidazolyl)pyridine (BBP), a commercially and readily available small organic compound which has earlier been described in a broad range of chemical and biological uses. Here, we show how BBP can also be used as a potent and specific SK channel blocker in vitro. The potency of BBP was measured using automatic patch clamp on all three SK channel subtypes, resulting in similar IC50 of 0.4 µM. We also assessed the selectivity of BBP on a panel of calcium-activated and voltage-activated potassium channels using two-electrode voltage clamp, automatic and manual patch clamp. BBP did not have any effect on IK, Kir2.1, Kir3.1+Kir3.4, Kv1.5, Kv4.3/KCHIP2 and Kv7.1/KCNE1 currents and was 4.8-fold and 46-fold more potent on all SK channel subtypes vs. BK and hERG channels, respectively. Moreover, we were able to identify H491 as a critical amino acid for the pharmacological effect of BBP on the SK channel. From a medicinal chemistry perspective, BBP could be used as a starting point for the design of new and improved SK inhibitors.

10.
Br J Pharmacol ; 175(3): 501-516, 2018 02.
Article in English | MEDLINE | ID: mdl-29156097

ABSTRACT

BACKGROUND AND PURPOSE: Kv 7.4 and Kv 7.5 channels are regulators of vascular tone. 4-Aminopyridine (4-AP) is considered a broad inhibitor of voltage-gated potassium (KV ) channels, with little inhibitory effect on Kv 7 family members at mmol concentrations. However, the effect of 4-AP on Kv 7 channels has not been systematically studied. The aim of this study was to investigate the pharmacological activity of 4-AP on Kv 7.4 and Kv 7.5 channels and characterize the effect of 4-AP on rat resistance arteries. EXPERIMENTAL APPROACH: Voltage clamp experiments were performed on Xenopus laevis oocytes injected with cRNA encoding KCNQ4 or KCNQ5, HEK cells expressing Kv 7.4 channels and on rat, freshly isolated mesenteric artery smooth muscle cells. The effect of 4-AP on tension, membrane potential, intracellular calcium and pH was assessed in rat mesenteric artery segments. KEY RESULTS: 4-AP increased the Kv 7.4-mediated current in oocytes and HEK cells but did not affect Kv 7.5 current. 4-AP also enhanced native mesenteric artery myocyte K+ current at sub-mmol concentrations. When applied to NA-preconstricted mesenteric artery segments, 4-AP hyperpolarized the membrane, decreased [Ca2+ ]i and caused concentration-dependent relaxations that were independent of 4-AP-mediated changes in intracellular pH. Application of the Kv 7 channel blocker XE991 and BKCa channel blocker iberiotoxin attenuated 4-AP-mediated relaxation. 4-AP also inhibited the NA-mediated signal transduction to elicit a relaxation. CONCLUSIONS AND IMPLICATIONS: These data show that 4-AP is able to relax NA-preconstricted rat mesenteric arteries by enhancing the activity of Kv 7.4 and BKCa channels and attenuating NA-mediated signalling.


Subject(s)
4-Aminopyridine/pharmacology , KCNQ Potassium Channels/physiology , Mesenteric Arteries/physiology , Norepinephrine/pharmacology , Potassium Channel Blockers/pharmacology , Vasoconstriction/physiology , Animals , Dose-Response Relationship, Drug , Female , HEK293 Cells , Humans , KCNQ Potassium Channels/antagonists & inhibitors , Male , Mesenteric Arteries/drug effects , Norepinephrine/antagonists & inhibitors , Organ Culture Techniques , Rats , Rats, Wistar , Vasoconstriction/drug effects , Xenopus laevis
11.
Br J Pharmacol ; 174(23): 4396-4408, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28925012

ABSTRACT

BACKGROUND AND PURPOSE: Small conductance calcium-activated potassium (KCa 2) channels represent a promising atrial-selective target for treatment of atrial fibrillation. Here, we establish the mechanism of KCa 2 channel inhibition by the new compound AP14145. EXPERIMENTAL APPROACH: Using site-directed mutagenesis, binding determinants for AP14145 inhibition were explored. AP14145 selectivity and mechanism of action were investigated by patch-clamp recordings of heterologously expressed KCa 2 channels. The biological efficacy of AP14145 was assessed by measuring atrial effective refractory period (AERP) prolongation in anaesthetized rats, and a beam walk test was performed in mice to determine acute CNS-related effects of the drug. KEY RESULTS: AP14145 was found to be an equipotent negative allosteric modulator of KCa 2.2 and KCa 2.3 channels (IC50  = 1.1 ± 0.3 µM). The presence of AP14145 (10 µM) increased the EC50 of Ca2+ on KCa 2.3 channels from 0.36 ± 0.02 to 1.2 ± 0.1 µM. The inhibitory effect strongly depended on two amino acids, S508 and A533 in the channel. AP14145 concentration-dependently prolonged AERP in rats. Moreover, AP14145 (10 mg·kg-1 ) did not trigger any apparent CNS effects in mice. CONCLUSIONS AND IMPLICATIONS: AP14145 is a negative allosteric modulator of KCa 2.2 and KCa 2.3 channels that shifted the calcium dependence of channel activation, an effect strongly dependent on two identified amino acids. AP14145 prolonged AERP in rats and did not trigger any acute CNS effects in mice. The understanding of how KCa 2 channels are inhibited, at the molecular level, will help further development of drugs targeting KCa 2 channels.


Subject(s)
Acetamides/pharmacology , Allosteric Regulation/drug effects , Potassium Channel Blockers/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Acetamides/administration & dosage , Animals , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Potassium Channel Blockers/administration & dosage , Rats , Rats, Sprague-Dawley , Small-Conductance Calcium-Activated Potassium Channels/metabolism
12.
Heart Rhythm ; 14(10): 1531-1538, 2017 10.
Article in English | MEDLINE | ID: mdl-28549997

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is the most common cardiac arrhythmia. Valvular heart disease is a strong predictor, yet the underlying molecular mechanisms are unknown. OBJECTIVE: The purpose of this study was to investigate the prevalence of somatic variants in AF candidate genes in an AF patient population undergoing surgery for mitral valve regurgitation (MVR) to determine whether these patients are genetically predisposed to AF. METHODS: DNA was extracted from blood and left atrial tissue from 44 AF patients with MVR. Using next-generation sequencing, we investigated 110 genes using the HaloPlex Target Enrichment System. MuTect software was used for identification of somatic point variants. We functionally characterized selected variants using electrophysiologic techniques. RESULTS: No somatic variants were identified in the cardiac tissue. Thirty-three patients (75%) had a rare germline variation in ≥1 candidate genes. Fourteen variants were novel. Fifteen variants were predicted damaging or likely damaging in ≥6 in silico predictions. We identified rare variants in genes never directly associated with AF: KCNE4, SCN4B, NEURL1, and CAND2. Interestingly, 7 patients (16%) had variants in genes involved in cellular potassium handling. The variants KCNQ1 (p.G272S) and KCNH2 (p.A913V) resulted in gain of function due to faster activation (KCNQ1) and slowed deactivation kinetics (KCNQ1, KCNH2). CONCLUSION: We did not find any somatic variants in patients with AF and MVR. Surprisingly, we found that our cohort of non-lone AF patients might, like lone AF patients, be predisposed to AF by rare germline variants. Our findings emphasize the extent of still unknown factors in the pathogenesis of AF.


Subject(s)
Atrial Fibrillation/genetics , DNA/genetics , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing/methods , Mitral Valve Insufficiency/complications , Mosaicism , Aged , Atrial Fibrillation/etiology , Computational Biology/methods , DNA Mutational Analysis , Female , Follow-Up Studies , Germ-Line Mutation , Humans , Male , Middle Aged , Mitral Valve Insufficiency/genetics , Retrospective Studies
13.
Eur J Pharmacol ; 803: 118-123, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28322838

ABSTRACT

Atrial fibrillation (AF) is the most common type of arrhythmia. Current pharmacological treatment for AF is moderately effective and/or increases the risk of serious ventricular adverse effects. To avoid ventricular adverse effects, a new target has been considered, the small conductance calcium-activated K+ channels (KCa2.X, SK channels). In the heart, KCa2.X channels are functionally more important in atria compared to ventricles, and pharmacological inhibition of the channel confers atrial selective prolongation of the cardiac action potential and converts AF to sinus rhythm in animal models of AF. Whether antiarrhythmic drugs (AADs) recommended for treating AF target KCa2.X channels is unknown. To this end, we tested a large number of AADs on the human KCa2.2 and KCa2.3 channels to assess their effect on this new target using automated whole-cell patch clamp. Of the AADs recommended for treatment of AF only dofetilide and propafenone inhibited hKCa2.X channels, with no subtype selectivity. The calculated IC50 were 90±10µmol/l vs 60±10µmol/l for dofetilide and 42±4µmol/l vs 80±20µmol/l for propafenone (hKCa2.3 vs hKCa2.2). Whether this inhibition has clinical importance for their antiarrhythmic effect is unlikely, as the calculated IC50 values are very high compared to the effective free therapeutic plasma concentration of the drugs when used for AF treatment, 40,000-fold for dofetilide and 140-fold higher for propafenone.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , HEK293 Cells , Humans
14.
Physiol Rep ; 4(8)2016 Apr.
Article in English | MEDLINE | ID: mdl-27117805

ABSTRACT

The inwardly rectifying potassium current (IK 1) conducted through Kir2.X channels contribute to repolarization of the cardiac action potential and to stabilization of the resting membrane potential in cardiomyocytes. Our aim was to investigate the effect of the recently discovered IK 1 inhibitor PA-6 on action potential repolarization and refractoriness in isolated rat hearts. Transiently transfected HEK-293 cells expressing IK 1 were voltage-clamped with ramp protocols. Langendorff-perfused heart experiments were performed on male Sprague-Dawley rats, effective refractory period, Wenckebach cycle length, and ventricular effective refractory period were determined following 200 nmol/L PA-6 perfusion. 200 nmol/L PA-6 resulted in a significant time-latency in drug effect on the IK 1 current expressed in HEK-293 cells, giving rise to a maximal effect at 20 min. In the Langendorff-perfused heart experiments, PA-6 prolonged the ventricular action potential duration at 90% repolarization (from 41.8 ± 6.5 msec to 72.6 ± 21.1 msec, 74% compared to baseline, P < 0.01, n = 6). In parallel, PA-6 significantly prolonged the ventricular effective refractory period compared to baseline (from 34.8 ± 4.6 msec to 58.1 ± 14.7 msec, 67%, P < 0.01, n = 6). PA-6 increased the short-term beat-to-beat variability and ventricular fibrillation was observed in two of six hearts. Neither atrial ERP nor duration of atrial fibrillation was altered following PA-6 application. The results show that pharmacological inhibition of cardiac IK 1 affects ventricular action potential repolarization and refractoriness and increases the risk of ventricular arrhythmia in isolated rat hearts.


Subject(s)
Action Potentials/physiology , Heart Ventricles/metabolism , Myocytes, Cardiac/physiology , Pentamidine/analogs & derivatives , Potassium Channels/metabolism , Action Potentials/drug effects , Animals , HEK293 Cells , Humans , Isolated Heart Preparation , Male , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Transfection
15.
Pflugers Arch ; 468(4): 643-54, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26729267

ABSTRACT

In isolated human atrial cardiomyocytes, inhibition of K2P3.1 K(+) channels results in action potential (action potential duration (APD)) prolongation. It has therefore been postulated that K2P3.1 (KCNK3), together with K2P9.1 (KCNK9), could represent novel drug targets for the treatment of atrial fibrillation (AF). However, it is unknown whether these findings in isolated cells translate to the whole heart. The purposes of this study were to investigate the expression levels of KCNK3 and KCNK9 in human hearts and two relevant rodent models and determine the antiarrhythmic potential of K2P3.1 inhibition in isolated whole-heart preparations. By quantitative PCR, we found that KCNK3 is predominantly expressed in human atria whereas KCNK9 was not detectable in heart human tissue. No differences were found between patients in AF or sinus rhythm. The expression in guinea pig heart resembled humans whereas rats displayed a more uniform expression of KCNK3 between atria and ventricle. In voltage-clamp experiments, ML365 and A293 were found to be potent and selective inhibitors of K2P3.1, but at pH 7.4, they failed to prolong atrial APD and refractory period (effective refractory period (ERP)) in isolated perfused rat and guinea pig hearts. At pH 7.8, which augments K2P3.1 currents, pharmacological channel inhibition produced a significant prolongation of atrial ERP (11.6 %, p = 0.004) without prolonging ventricular APD but did not display a significant antiarrhythmic effect in our guinea pig AF model (3/8 hearts converted on A293 vs 0/7 hearts in time-matched controls). These results suggest that when K2P3.1 current is augmented, K2P3.1 inhibition leads to atrial-specific prolongation of ERP; however, this ERP prolongation did not translate into significant antiarrhythmic effects in our AF model.


Subject(s)
Action Potentials , Arrhythmias, Cardiac/metabolism , Atrial Function , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Protons , Refractory Period, Electrophysiological , Adolescent , Adult , Animals , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Female , Guinea Pigs , Heart Atria/cytology , Heart Atria/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Rats , Rats, Wistar , Species Specificity , Ventricular Function
16.
Gen Physiol Biophys ; 34(4): 383-92, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26001288

ABSTRACT

Although several physiological, pathophysiological and regulatory properties of classical inward rectifier K+ current I(K1), G-protein coupled inwardly-rectifying K+ current I(K,ACh) and the small-conductance Ca2+ activated K+ current I(K,Ca) have been identified, quantitative biophysical details remain unclear. Both I(K1) and I(K,ACh) are implicated in atrial fibrillation (AF), and recently also I(K,Ca) has been speculated to be linked with the genesis and sustainability of AF. All these three currents have been shown to be involved in the electrical remodeling in the atria of patients suffering from AF, and it is therefore important to characterize their biophysical properties and compare their relative current contribution in atrial electrophysiology in both sinus rhythm (SR) and AF. The aim of this study is to investigate the contribution of the three potassium currents when subjected to voltage protocols adapted from atrial action potentials recorded in human tissue at 1 and 3 Hz. The current recordings were performed in the HEK-293 heterologous cell system expressing either I(K1), I(K,ACh) or I(K,Ca) to establish the individual contribution of each of these currents during the voltage changes of atrial action potential waveforms. I(K1) primarily contributes to the atrial electrophysiology at the latter part of repolarization and during the diastolic phase, while both I(K,Ca) under high [Ca2+]i and I(K,ACh) contribute relatively most during repolarization.


Subject(s)
Action Potentials , Atrial Fibrillation/physiopathology , Calcium/metabolism , Membrane Potentials , Potassium Channels, Inwardly Rectifying/metabolism , Potassium/metabolism , HEK293 Cells , Heart Rate , Humans , Ion Channel Gating
17.
J Cardiovasc Pharmacol ; 66(5): 441-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25830485

ABSTRACT

Small-conductance Ca(2+)-activated potassium (SK) channels are relative newcomers within the field of cardiac electrophysiology. In recent years, an increased focus has been given to these channels because they might constitute a relatively atrial-selective target. This review will give a general introduction to SK channels followed by their proposed function in the heart under normal and pathophysiological conditions. It is revealed how antiarrhythmic effects can be obtained by SK channel inhibition in a number of species in situations of atrial fibrillation. On the contrary, the beneficial effects of SK channel inhibition in situations of heart failure are questionable and still needs investigation. The understanding of cardiac SK channels is rapidly increasing these years, and it is hoped that this will clarify whether SK channel inhibition has potential as a new anti-atrial fibrillation principle.


Subject(s)
Atrial Fibrillation/metabolism , Heart Conduction System/metabolism , Heart Rate , Potassium Channels, Calcium-Activated/metabolism , Animals , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Drugs, Investigational/therapeutic use , Heart Conduction System/drug effects , Heart Conduction System/physiopathology , Heart Rate/drug effects , Humans , Molecular Targeted Therapy , Potassium Channel Blockers/therapeutic use , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Signal Transduction
18.
Front Physiol ; 5: 389, 2014.
Article in English | MEDLINE | ID: mdl-25346695

ABSTRACT

The large conductance calcium- and voltage-activated K(+) channel (KCa1.1, BK, MaxiK) is ubiquitously expressed in the body, and holds the ability to integrate changes in intracellular calcium and membrane potential. This makes the BK channel an important negative feedback system linking increases in intracellular calcium to outward hyperpolarizing potassium currents. Consequently, the channel has many important physiological roles including regulation of smooth muscle tone, neurotransmitter release and neuronal excitability. Additionally, cardioprotective roles have been revealed in recent years. After a short introduction to the structure, function and regulation of BK channels, we review the small organic molecules activating BK channels and how these tool compounds have helped delineate the roles of BK channels in health and disease.

19.
Hypertension ; 62(6): 1090-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24082059

ABSTRACT

The goal of the present study was to determine the role of KCNQ-encoded Kv channels (Kv7 channels) in the passive and active regulation of coronary flow in normotensive and hypertensive rats. In left anterior descending coronary arteries from normotensive rats, structurally different Kv7.2 to 7.5 activators produced relaxations, which were considerably less in arteries from hypertensive rats and were not mimicked by the Kv7.1-specific activator R-L3. In isolated, perfused heart preparations, coronary flow rate increased in response to the Kv7.2 to 7.5 activator (S)-1 and was diminished in the presence of a Kv7 inhibitor. The expression levels of KCNQ1-5 and their known accessory KCNE1-5 subunits in coronary arteries were similar in normotensive and hypertensive rats as measured by quantitative polymerase chain reaction. However, Kv7.4 protein expression was reduced in hypertensive rats. Application of adenosine or A2A receptor agonist CGS-21680 produced concentration-dependent relaxations of coronary arteries from normotensive rats, which were attenuated by application of Kv7 inhibitors. Kv7 blockers also attenuated the ischemia-induced increase in coronary perfusion in Langendorff studies. Overall, these data establish Kv7 channels as crucial regulators of coronary flow at resting and after hypoxic insult.


Subject(s)
Blood Pressure/physiology , Coronary Vessels/metabolism , Hypertension/metabolism , KCNQ Potassium Channels/metabolism , Regional Blood Flow/physiology , Adenosine/pharmacology , Animals , Blood Pressure/drug effects , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Dose-Response Relationship, Drug , Hypertension/physiopathology , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , Male , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
20.
BMC Med Genet ; 13: 24, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22471742

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is the most common arrhythmia. The potassium current IKs is essential for cardiac repolarization. Gain-of-function mutations in KV7.1, the pore-forming α-subunit of the IKs channel, have been associated with AF. We hypothesized that early-onset lone AF is associated with mutations in the IKs channel regulatory subunit KCNE1. METHODS: In 209 unrelated early-onset lone AF patients (< 40 years) the entire coding sequence of KCNE1 was bidirectionally sequenced. We analyzed the identified KCNE1 mutants electrophysiologically in heterologous expression systems. RESULTS: Two non-synonymous mutations G25V and G60D were found in KCNE1 that were not present in the control group (n = 432 alleles) and that have not previously been reported in any publicly available databases or in the exom variant server holding exom data from more than 10.000 alleles. Proband 1 (female, age 45, G25V) had onset of paroxysmal AF at the age of 39 years. Proband 2 (G60D) was diagnosed with lone AF at the age of 33 years. The patient has inherited the mutation from his mother, who also has AF. Both probands had no mutations in genes previously associated with AF. In heterologous expression systems, both mutants showed significant gain-of-function for IKs both with respect to steady-state current levels, kinetic parameters, and heart rate-dependent modulation. CONCLUSIONS: Mutations in KV7.1 leading to gain-of-function of IKs current have previously been described in lone AF, yet this is the first time a mutation in the beta-subunit KCNE1 is associated with the disease. This finding further supports the hypothesis that increased potassium current enhances AF susceptibility.


Subject(s)
Atrial Fibrillation/epidemiology , Atrial Fibrillation/genetics , Genetic Predisposition to Disease , Mutation/genetics , Potassium Channels, Voltage-Gated/genetics , Protein Subunits/genetics , Adult , Age of Onset , Atrial Fibrillation/physiopathology , Base Sequence , Cohort Studies , DNA Mutational Analysis , Denmark/epidemiology , Electric Conductivity , Female , Humans , Ion Channel Gating , Kinetics , Male , Molecular Sequence Data , Mutant Proteins/metabolism , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...