1
|
Bussmann B, Ayagama T, Liu K, Li D, Herring N. Bayliss Starling Prize Lecture 2023: Neuropeptide-Y being 'unsympathetic' to the broken hearted. J Physiol 2025; 603:1841-1864. [PMID: 38847435 PMCID: PMC11955873 DOI: 10.1113/jp285370] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/01/2024] [Indexed: 04/01/2025] Open
Abstract
William Bayliss and Ernest Starling are not only famous as pioneers in cardiovascular physiology, but also responsible for the discovery of the first hormone (from the Greek 'excite or arouse'), the intestinal signalling molecule and neuropeptide secretin in 1902. Our research group focuses on neuropeptides and neuromodulators that influence cardiovascular autonomic control as potential biomarkers in disease and tractable targets for therapeutic intervention. Acute myocardial infarction (AMI) and chronic heart failure (CHF) result in high levels of cardiac sympathetic stimulation, which is a poor prognostic indicator. Although beta-blockers improve mortality in these conditions by preventing the action of the neurotransmitter noradrenaline, a substantial residual risk remains. Recently, we have identified the sympathetic co-transmitter neuropeptide-Y (NPY) as being released during AMI, leading to larger infarcts and life-threatening arrhythmia in both animal models and patients. Here, we discuss recently published data demonstrating that peripheral venous NPY levels are associated with heart failure hospitalisation and mortality after AMI, and all cause cardiovascular mortality in CHF, even when adjusting for known risk factors (including brain natriuretic peptide). We have investigated the mechanistic basis for these observations in human and rat stellate ganglia and cardiac tissue, manipulating NPY neurochemistry at the same time as using state-of-the-art imaging techniques, to establish the receptor pathways responsible for NPY signalling. We propose NPY as a new mechanistic biomarker in AMI and CHF patients and aim to determine whether specific NPY receptor blockers can prevent arrhythmia and attenuate the development of heart failure.
Collapse
Affiliation(s)
- Benjamin Bussmann
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Thamali Ayagama
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Kun Liu
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Blair GA, Depman M, Adams WP, Maisonneuve RO, Hoeker GS, Weinberg SH, Poelzing S. Sequence-Dependent Repolarization Is Modulated by Endogenous Action Potential Duration Gradients Rather Than Electrical Coupling in Ventricular Myocardium. J Am Heart Assoc 2025; 14:e030433. [PMID: 39719415 PMCID: PMC12054505 DOI: 10.1161/jaha.123.030433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/12/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Previous studies suggest the relationship between activation time (AT) and action potential duration (APD) in the heart is dependent on electrotonic coupling, but this has not been directly tested. This study assessed whether acute changes in electrical coupling, or other determinants of conduction or repolarization, modulate APD heterogeneity. METHODS AND RESULTS Langendorff-perfused guinea pig hearts were epicardially paced and optically mapped after treatment with the gap junction uncoupler carbenoxolone, ephaptic uncoupler mannitol, ephaptic enhancer dextran 2MDa, sodium channel inhibitor flecainide, or rapid component of the delayed rectifier potassium channel inhibitor E4031. SD of APD and the AT-APD slope and coefficient of determination were quantified as metrics of APD heterogeneity. SD of APD increased with carbenoxolone, mannitol, and altered activation sequence. The AT-APD slope was insensitive to carbenoxolone, mannitol, dextran, flecainide, or E4031 but changed in response to activation sequence. The coefficient of determination did not change with carbenoxolone; decreased with mannitol, E4031, and activation sequence; but increased with dextran and flecainide. APD heterogeneity changes were dependent on whether the estimation used SD of APD or the AT-APD relationship. The pacing stimulus increased APD at the site of stimulation, revealing a confounding stimulus effect on APD within the measurement area. Simulations predict that the stimulus artifact and endogenous APD gradients are stronger determinants of APD heterogeneity than AT. CONCLUSIONS APD dependence on conduction is relatively small. Furthermore, APD heterogeneity within a mapping field of view is dependent on endogenous gradients, the stimulus artifact, and the experimental approach, rather than electrical coupling.
Collapse
Affiliation(s)
- Grace A. Blair
- Graduate Program in Translational BiologyMedicine and Health, Virginia TechRoanokeVAUSA
- Center for Vascular and Heart ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
| | - Madeline Depman
- Graduate Program in Translational BiologyMedicine and Health, Virginia TechRoanokeVAUSA
- Center for Vascular and Heart ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
| | - William P. Adams
- Center for Vascular and Heart ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
| | - Rowan O. Maisonneuve
- Center for Vascular and Heart ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
| | - Gregory S. Hoeker
- Center for Vascular and Heart ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
| | - Seth H. Weinberg
- Department of Biomedical EngineeringDavis Heart and Lung Research Institute, The Ohio State UniversityColumbusOHUSA
| | - Steve Poelzing
- Graduate Program in Translational BiologyMedicine and Health, Virginia TechRoanokeVAUSA
- Center for Vascular and Heart ResearchFralin Biomedical Research Institute at Virginia Tech CarilionRoanokeVAUSA
- Department of Biomedical Engineering and MechanicsVirginia Polytechnic Institute and State UniversityBlacksburgVAUSA
| |
Collapse
|
3
|
De la Cruz A, Wu X, Rainer QC, Hiniesto-Iñigo I, Perez ME, Edler I, Liin SI, Larsson HP. Pharmacological Screening of Kv7.1 and Kv7.1/KCNE1 Activators as Potential Antiarrhythmic Drugs in the Zebrafish Heart. Int J Mol Sci 2023; 24:12092. [PMID: 37569465 PMCID: PMC10418701 DOI: 10.3390/ijms241512092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Long QT syndrome (LQTS) can lead to ventricular arrhythmia and sudden cardiac death. The most common congenital cause of LQTS is mutations in the channel subunits generating the cardiac potassium current IKs. Zebrafish (Danio rerio) have been proposed as a powerful system to model human cardiac diseases due to the similar electrical properties of the zebrafish heart and the human heart. We used high-resolution all-optical electrophysiology on ex vivo zebrafish hearts to assess the effects of IKs analogues on the cardiac action potential. We found that chromanol 293B (an IKs inhibitor) prolonged the action potential duration (APD) in the presence of E4031 (an IKr inhibitor applied to drug-induced LQT2), and to a lesser extent, in the absence of E4031. Moreover, we showed that PUFA analogues slightly shortened the APD of the zebrafish heart. However, PUFA analogues failed to reverse the APD prolongation in drug-induced LQT2. However, a more potent IKs activator, ML-277, partially reversed the APD prolongation in drug-induced LQT2 zebrafish hearts. Our results suggest that IKs plays a limited role in ventricular repolarizations in the zebrafish heart under resting conditions, although it plays a more important role when the IKr is compromised, as if the IKs in zebrafish serves as a repolarization reserve as in human hearts. This study shows that potent IKs activators can restore the action potential duration in drug-induced LQT2 in the zebrafish heart.
Collapse
Affiliation(s)
- Alicia De la Cruz
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| | - Xiaoan Wu
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| | - Quinn C. Rainer
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| | - Irene Hiniesto-Iñigo
- Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85 Linköping, Sweden
| | - Marta E. Perez
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| | - Isak Edler
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
- Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85 Linköping, Sweden
| | - Sara I. Liin
- Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85 Linköping, Sweden
| | - H. Peter Larsson
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
4
|
Verkerk AO, Wilders R. Human Sinoatrial Node Pacemaker Activity: Role of the Slow Component of the Delayed Rectifier K + Current, I Ks. Int J Mol Sci 2023; 24:7264. [PMID: 37108427 PMCID: PMC10138838 DOI: 10.3390/ijms24087264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The pacemaker activity of the sinoatrial node (SAN) has been studied extensively in animal species but is virtually unexplored in humans. Here we assess the role of the slowly activating component of the delayed rectifier K+ current (IKs) in human SAN pacemaker activity and its dependence on heart rate and β-adrenergic stimulation. HEK-293 cells were transiently transfected with wild-type KCNQ1 and KCNE1 cDNA, encoding the α- and β-subunits of the IKs channel, respectively. KCNQ1/KCNE1 currents were recorded both during a traditional voltage clamp and during an action potential (AP) clamp with human SAN-like APs. Forskolin (10 µmol/L) was used to increase the intracellular cAMP level, thus mimicking β-adrenergic stimulation. The experimentally observed effects were evaluated in the Fabbri-Severi computer model of an isolated human SAN cell. Transfected HEK-293 cells displayed large IKs-like outward currents in response to depolarizing voltage clamp steps. Forskolin significantly increased the current density and significantly shifted the half-maximal activation voltage towards more negative potentials. Furthermore, forskolin significantly accelerated activation without affecting the rate of deactivation. During an AP clamp, the KCNQ1/KCNE1 current was substantial during the AP phase, but relatively small during diastolic depolarization. In the presence of forskolin, the KCNQ1/KCNE1 current during both the AP phase and diastolic depolarization increased, resulting in a clearly active KCNQ1/KCNE1 current during diastolic depolarization, particularly at shorter cycle lengths. Computer simulations demonstrated that IKs reduces the intrinsic beating rate through its slowing effect on diastolic depolarization at all levels of autonomic tone and that gain-of-function mutations in KCNQ1 may exert a marked bradycardic effect during vagal tone. In conclusion, IKs is active during human SAN pacemaker activity and has a strong dependence on heart rate and cAMP level, with a prominent role at all levels of autonomic tone.
Collapse
Affiliation(s)
- Arie O. Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| |
Collapse
|
5
|
Ovechkin AO, Vaykshnorayte MA, Sedova KA, Shmakov DN, Shumikhin KV, Medvedeva SY, Danilova IG, Azarov JE. Beta-Receptor Blockade Reproduces Electrophysiological Effects of Early Diabetes Mellitus in Ventricular Myocardium. J EVOL BIOCHEM PHYS+ 2022. [PMCID: PMC9910273 DOI: 10.1134/s0022093022070080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- A. O. Ovechkin
- Department of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, Syktyvkar, Russia
- Institute of Medicine, Pitirim Sorokin Syktyvkar State University, Syktyvkar, Russia
| | - M. A. Vaykshnorayte
- Department of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, Syktyvkar, Russia
| | - K. A. Sedova
- Faculty of Biomedical Engineering, Czech Technical University, Kladno, Czech Republic
| | - D. N. Shmakov
- Department of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, Syktyvkar, Russia
| | - K. V. Shumikhin
- Institute of Medicine, Pitirim Sorokin Syktyvkar State University, Syktyvkar, Russia
| | - S. Yu. Medvedeva
- Department of morphology and biochemistry, Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg, Russia
| | - I. G. Danilova
- Department of morphology and biochemistry, Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg, Russia
| | - J. E. Azarov
- Department of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, Syktyvkar, Russia
- Institute of Medicine, Pitirim Sorokin Syktyvkar State University, Syktyvkar, Russia
| |
Collapse
|
6
|
Yuasa M, Kojima A, Mi X, Ding WG, Omatsu-Kanbe M, Kitagawa H, Matsuura H. Characterization and functional role of rapid- and slow-activating delayed rectifier K + currents in atrioventricular node cells of guinea pigs. Pflugers Arch 2021; 473:1885-1898. [PMID: 34704178 DOI: 10.1007/s00424-021-02617-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 01/28/2023]
Abstract
The atrioventricular (AV) node is the only conduction pathway where electrical impulse can pass from atria to ventricles and exhibits spontaneous automaticity. This study examined the function of the rapid- and slow-activating delayed rectifier K+ currents (IKr and IKs) in the regulation of AV node automaticity. Isolated AV node cells from guinea pigs were current- and voltage-clamped to record the action potentials and the IKr and IKs current. The expression of IKr or IKs was confirmed in the AV node cells by immunocytochemistry, and the positive signals of both channels were localized mainly on the cell membrane. The basal spontaneous automaticity was equally reduced by E4031 and HMR-1556, selective blockers of IKr and IKs, respectively. The nonselective β-adrenoceptor agonist isoproterenol markedly increased the firing rate of action potentials. In the presence of isoproterenol, the firing rate of action potentials was more effectively reduced by the IKs inhibitor HMR-1556 than by the IKr inhibitor E4031. Both E4031 and HMR-1556 prolonged the action potential duration and depolarized the maximum diastolic potential under basal and β-adrenoceptor-stimulated conditions. IKr was not significantly influenced by β-adrenoceptor stimulation, but IKs was concentration-dependently enhanced by isoproterenol (EC50: 15 nM), with a significant negative voltage shift in the channel activation. These findings suggest that both the IKr and IKs channels might exert similar effects on regulating the repolarization process of AV node action potentials under basal conditions; however, when the β-adrenoceptor is activated, IKs modulation may become more important.
Collapse
Affiliation(s)
- Mayumi Yuasa
- Department of Anesthesiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Akiko Kojima
- Department of Anesthesiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Xinya Mi
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Wei-Guang Ding
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| | - Mariko Omatsu-Kanbe
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Hirotoshi Kitagawa
- Department of Anesthesiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Hiroshi Matsuura
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
7
|
Kasak L, Rull K, Yang T, Roden DM, Laan M. Recurrent Pregnancy Loss and Concealed Long-QT Syndrome. J Am Heart Assoc 2021; 10:e021236. [PMID: 34398675 PMCID: PMC8649249 DOI: 10.1161/jaha.121.021236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Recurrent pregnancy loss affects 1% to 2% of couples attempting childbirth. A large fraction of all cases remains idiopathic, which warrants research into monogenic causes of this distressing disorder. Methods and Results We investigated a nonconsanguineous Estonian family who had experienced 5 live births, intersected by 3 early pregnancy losses, and 6 fetal deaths, 3 of which occurred during the second trimester. No fetal malformations were described at the autopsies performed in 3 of 6 cases of fetal death. Parental and fetal chromosomal abnormalities (including submicroscopic) and maternal risk factors were excluded. Material for genetic testing was available from 4 miscarried cases (gestational weeks 11, 14, 17, and 18). Exome sequencing in 3 pregnancy losses and the mother identified no rare variants explicitly shared by the miscarried conceptuses. However, the mother and 2 pregnancy losses carried a heterozygous nonsynonymous variant, resulting in p.Val173Asp (rs199472695) in the ion channel gene KCNQ1. It is expressed not only in heart, where mutations cause type 1 long‐QT syndrome, but also in other tissues, including uterus. The p.Val173Asp variant has been previously identified in a patient with type 1 long‐QT syndrome, but not reported in the Genome Aggregation Database. With heterologous expression in CHO cells, our in vitro electrophysiologic studies indicated that the mutant slowly activating voltage‐gated K+ channel (IKs) is dysfunctional. It showed reduced total activating and deactivating currents (P<0.01), with dramatically positive shift of voltage dependence of activation by ≈10 mV (P<0.05). Conclusions The current study uncovered concealed maternal type 1 long‐QT syndrome as a potential novel cause behind recurrent fetal loss.
Collapse
Affiliation(s)
- Laura Kasak
- Institute of Biomedicine and Translational Medicine University of Tartu Estonia
| | - Kristiina Rull
- Institute of Biomedicine and Translational Medicine University of Tartu Estonia.,Women's Clinic Tartu University Hospital Tartu Estonia.,Institute of Clinical Medicine University of Tartu Estonia
| | - Tao Yang
- Departments of Medicine, Pharmacology and Biomedical Informatics Vanderbilt University Medical Center Nashville TN
| | - Dan M Roden
- Departments of Medicine, Pharmacology and Biomedical Informatics Vanderbilt University Medical Center Nashville TN
| | - Maris Laan
- Institute of Biomedicine and Translational Medicine University of Tartu Estonia
| |
Collapse
|
8
|
Filatova TS, Abramochkin DV, Pavlova NS, Pustovit KB, Konovalova OP, Kuzmin VS, Dobrzynski H. Repolarizing potassium currents in working myocardium of Japanese quail: a novel translational model for cardiac electrophysiology. Comp Biochem Physiol A Mol Integr Physiol 2021; 255:110919. [PMID: 33582263 DOI: 10.1016/j.cbpa.2021.110919] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/06/2021] [Accepted: 02/06/2021] [Indexed: 12/14/2022]
|
9
|
Hoogendijk MG, Géczy T, Yap SC, Szili-Torok T. Pathophysiological Mechanisms of Premature Ventricular Complexes. Front Physiol 2020; 11:406. [PMID: 32528299 PMCID: PMC7247859 DOI: 10.3389/fphys.2020.00406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/06/2020] [Indexed: 11/29/2022] Open
Abstract
Premature ventricular complexes (PVCs) are the most common ventricular arrhythmia. Despite the high prevalence, the cause of PVCs remains elusive in most patients. A better understanding of the underlying pathophysiological mechanism may help to steer future research. This review aims to provide an overview of the potential pathophysiological mechanisms of PVCs and their differentiation.
Collapse
Affiliation(s)
- Mark G Hoogendijk
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Tamás Géczy
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sing-Chien Yap
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Tamas Szili-Torok
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
10
|
Shroff SN, Das SL, Tseng HA, Noueihed J, Fernandez F, White JA, Chen CS, Han X. Voltage Imaging of Cardiac Cells and Tissue Using the Genetically Encoded Voltage Sensor Archon1. iScience 2020; 23:100974. [PMID: 32299055 PMCID: PMC7160579 DOI: 10.1016/j.isci.2020.100974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 02/21/2020] [Accepted: 03/05/2020] [Indexed: 01/19/2023] Open
Abstract
Precise measurement of action potentials (APs) is needed to observe electrical activity and cellular communication within cardiac tissue. Voltage-sensitive dyes (VSDs) are traditionally used to measure cardiac APs; however, they require acute chemical addition that prevents chronic imaging. Genetically encoded voltage indicators (GEVIs) enable long-term studies of APs without the need of chemical additions, but current GEVIs used in cardiac tissue exhibit poor kinetics and/or low signal to noise (SNR). Here, we demonstrate the use of Archon1, a recently developed GEVI, in hiPSC-derived cardiomyocytes (CMs). When expressed in CMs, Archon1 demonstrated fast kinetics comparable with patch-clamp electrophysiology and high SNR significantly greater than the VSD Di-8-ANEPPS. Additionally, Archon1 enabled monitoring of APs across multiple cells simultaneously in 3D cardiac tissues. These results highlight Archon1's capability to investigate the electrical activity of CMs in a variety of applications and its potential to probe functionally complex in vitro models, as well as in vivo systems.
Collapse
Affiliation(s)
- Sanaya N Shroff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Shoshana L Das
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Hua-An Tseng
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Jad Noueihed
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Fernando Fernandez
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - John A White
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
11
|
Shults NV, Rybka V, Suzuki YJ, Brelidze TI. Increased Smooth Muscle Kv11.1 Channel Expression in Pulmonary Hypertension and Protective Role of Kv11.1 Channel Blocker Dofetilide. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:48-56. [PMID: 31839145 PMCID: PMC6943378 DOI: 10.1016/j.ajpath.2019.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 01/08/2023]
Abstract
Kv11.1 potassium channels are essential for heart repolarization. Prescription medication that blocks Kv11.1 channels lengthens the ventricular action potential and causes cardiac arrhythmias. Surprisingly little is known about the Kv11.1 channel expression and function in the lung tissue. Here we report that Kv11.1 channels were abundantly expressed in the large pulmonary arteries (PAs) of healthy lung tissues from humans and rats. Kv11.1 channel expression was increased in the lungs of humans affected by chronic obstructive pulmonary disease-associated pulmonary hypertension and in the lungs of rats with pulmonary arterial hypertension (PAH). In healthy lung tissues from humans and rats, Kv11.1 channels were confined to the large PAs. In humans with chronic obstructive pulmonary disease-associated pulmonary hypertension and in rats with PAH, Kv11.1 channels were expressed in both the large and small PAs. The increase in Kv11.1 channel expression closely followed the time-course of the development of pulmonary vascular remodeling in PAH rats. Treatment of PAH rats with dofetilide, an Kv11.1 channel blocker approved by the US Food and Drug Administration for use in the treatment of arrythmia, inhibited PAH-associated pulmonary vascular remodeling. Taken together, the findings from this study uncovered a novel role of Kv11.1 channels in lung function and their potential as new drug targets in the treatment of pulmonary hypertension. The protective effect of dofetilide raises the possibility of repurposing this antiarrhythmic drug for the treatment of patients with pulmonary hypertension.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/prevention & control
- Case-Control Studies
- ERG1 Potassium Channel/antagonists & inhibitors
- ERG1 Potassium Channel/metabolism
- Female
- Follow-Up Studies
- Humans
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Phenethylamines/pharmacology
- Potassium Channel Blockers/pharmacology
- Prognosis
- Pulmonary Arterial Hypertension/complications
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Rats, Sprague-Dawley
- Sulfonamides/pharmacology
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Nataliia V Shults
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| | - Vladyslava Rybka
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| | - Yuichiro J Suzuki
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| | - Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia.
| |
Collapse
|
12
|
Winbo A, Paterson DJ. The Brain-Heart Connection in Sympathetically Triggered Inherited Arrhythmia Syndromes. Heart Lung Circ 2019; 29:529-537. [PMID: 31959550 DOI: 10.1016/j.hlc.2019.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/25/2019] [Accepted: 11/11/2019] [Indexed: 12/31/2022]
Abstract
Sympathetically triggered inherited arrhythmia syndromes, including the long QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT), can cause sudden cardiac death in young individuals with structurally normal hearts. With cardiac events typically triggered by physical or emotional stress, not surprisingly, two of the most common treatments are neuromodulators, including mainstay beta blocker pharmacotherapy, and surgical sympathetic cardiac denervation. This review updates the clinician on the relevant anatomy and physiology of the cardiac autonomic nervous system, outlines neurocardiac arrhythmia mechanisms, and discusses the latest rationale for a neurocardiac therapeutic approach to manage sympathetic-induced arrhythmia in patients with inherited cardiac disease.
Collapse
Affiliation(s)
- Annika Winbo
- Department of Physiology, University of Auckland, Auckland, New Zealand; Department of Paediatric and Congenital Cardiac Services, Starship Children's Hospital, Auckland, New Zealand.
| | - David J Paterson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Martinez-Mateu L, Saiz J, Aromolaran AS. Differential Modulation of IK and ICa,L Channels in High-Fat Diet-Induced Obese Guinea Pig Atria. Front Physiol 2019; 10:1212. [PMID: 31607952 PMCID: PMC6773813 DOI: 10.3389/fphys.2019.01212] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022] Open
Abstract
Obesity mechanisms that make atrial tissue vulnerable to arrhythmia are poorly understood. Voltage-dependent potassium (IK, IKur, and IK1) and L-type calcium currents (ICa,L) are electrically relevant and represent key substrates for modulation in obesity. We investigated whether electrical remodeling produced by high-fat diet (HFD) alone or in concert with acute atrial stimulation were different. Electrophysiology was used to assess atrial electrical function after short-term HFD-feeding in guinea pigs. HFD atria displayed spontaneous beats, increased IK (IKr + IKs) and decreased ICa,L densities. Only with pacing did a reduction in IKur and increased IK1 phenotype emerge, leading to a further shortening of action potential duration. Computer modeling studies further indicate that the measured changes in potassium and calcium current densities contribute prominently to shortened atrial action potential duration in human heart. Our data are the first to show that multiple mechanisms (shortened action potential duration, early afterdepolarizations and increased incidence of spontaneous beats) may underlie initiation of supraventricular arrhythmias in obese guinea pig hearts. These results offer different mechanistic insights with implications for obese patients harboring supraventricular arrhythmias.
Collapse
Affiliation(s)
- Laura Martinez-Mateu
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain
| | - Javier Saiz
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain
| | - Ademuyiwa S Aromolaran
- Cardiac Electrophysiology and Metabolism Research Group, VA New York Harbor Healthcare System, Brooklyn, NY, United States.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Physiology & Cellular Biophysics, Columbia University, New York, NY, United States
| |
Collapse
|
14
|
The autonomic nervous system and cardiac arrhythmias: current concepts and emerging therapies. Nat Rev Cardiol 2019; 16:707-726. [DOI: 10.1038/s41569-019-0221-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/07/2019] [Indexed: 12/19/2022]
|
15
|
Tinker A, Aziz Q, Li Y, Specterman M. ATP‐Sensitive Potassium Channels and Their Physiological and Pathophysiological Roles. Compr Physiol 2018; 8:1463-1511. [DOI: 10.1002/cphy.c170048] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
16
|
Abramochkin DV, Hassinen M, Vornanen M. Transcripts of Kv7.1 and MinK channels and slow delayed rectifier K + current (I Ks) are expressed in zebrafish (Danio rerio) heart. Pflugers Arch 2018; 470:1753-1764. [PMID: 30116893 DOI: 10.1007/s00424-018-2193-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/24/2018] [Accepted: 08/06/2018] [Indexed: 12/18/2022]
Abstract
Zebrafish are increasingly used as a model for human cardiac electrophysiology, arrhythmias, and drug screening. However, K+ ion channels of the zebrafish heart, which determine the rate of repolarization and duration of cardiac action potential (AP) are still incompletely known and characterized. Here, we provide the first evidence for the presence of the slow component of the delayed rectifier K+channels in the zebrafish heart and characterize electrophysiological properties of the slow component of the delayed rectifier K+current, IKs. Zebrafish atrium and ventricle showed strong transcript expression of the kcnq1 gene, which encodes the Kv7.1 α-subunit of the slow delayed rectifier K+ channel. In contrast, the kcne1 gene, encoding the MinK β-subunit of the delayed rectifier, was expressed at 21 and 17 times lower level in ventricle and atrium, respectively, in comparison to the kcnq1. IKs was observed in 62% of ventricular myocytes with mean (± SEM) density of 1.23 ± 0.37 pA/pF at + 30 mV. Activation rate of IKs was 38% faster (τ50 = 1248 ± 215 ms) than kcnq1:kcne1 channels (1725 ± 792 ms) expressed in 3:1 ratio in Chinese hamster ovary cells. Microelectrode experiments demonstrated the functional relevance of IKs in the zebrafish heart, since 100 μM chromanol 293B produced a significant prolongation of AP in zebrafish ventricle. We conclude that AP repolarization in zebrafish ventricle is contributed by IKs, which is mainly generated by homotetrameric Kv7.1 channels not coupled to MinK ancillary β-subunits. This is a clear difference to the human heart, where MinK is an essential component of the slow delayed rectifier K+channel.
Collapse
Affiliation(s)
- Denis V Abramochkin
- Department of Human and Animal Physiology, Biological faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia.
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russia.
- Laboratory of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, Syktyvkar, Russia.
| | - Minna Hassinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| |
Collapse
|
17
|
Gou X, Wang W, Zou S, Qi Y, Xu Y. Protein kinase C epsilon mediates the inhibition of angiotensin II on the slowly activating delayed-rectifier potassium current through channel phosphorylation. J Mol Cell Cardiol 2018; 116:165-174. [PMID: 29452158 DOI: 10.1016/j.yjmcc.2018.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 01/25/2018] [Accepted: 02/12/2018] [Indexed: 01/14/2023]
Abstract
The slowly activating delayed rectifier K+ current (IKs) is one of the main repolarizing currents in the human heart. Evidence has shown that angiotensin II (Ang II) regulates IKs through the protein kinase C (PKC) pathway, but the related results are controversial. This study was designed to identify PKC isoenzymes involved in the regulation of IKs by Ang II and the underlying molecular mechanism. The whole-cell patch-clamp technique was used to record IKs in isolated guinea pig ventricular cardiomyocytes and in human embryonic kidney (HEK) 293 cells co-transfected with human KCNQ1/KCNE1 genes and Ang II type 1 receptor genes. Ang II inhibited IKs in a concentration-dependent manner in native cardiomyocytes. A broad PKC inhibitor Gö6983 (not inhibiting PKCε) and a selective cPKC inhibitor Gö6976 did not affect the inhibitory action of Ang II. In contrast, the inhibition was significantly attenuated by PKCε-selective peptide inhibitor εV1-2. However, direct activation of PKC by phorbol 12-myristate 13-acetate (PMA) increased the cloned human IKs in HEK293 cells. Similarly, the cPKC peptide activator significantly enhanced the current. In contrast, the PKCε peptide activator inhibited the current. Further evidence showed that PKCε knockdown by siRNA antagonized the Ang II-induced inhibition on KCNQ1/KCNE1 current, whereas knockdown of cPKCs (PKCα and PKCβ) attenuated the potentiation of the current by PMA. Moreover, deletion of four putative phosphorylation sites in the C-terminus of KCNQ1 abolished the action of PMA. Mutation of two putative phosphorylation sites in the N-terminus of KCNQ1 and one site in KCNE1 (S102) blocked the inhibition of Ang II. Our results demonstrate that PKCε isoenzyme mediates the inhibitory action of Ang II on IKs and by phosphorylating distinct sites in KCNQ1/KCNE1, cPKC and PKCε isoenzymes produce the contrary regulatory effects on the channel. These findings have provided new insight into the molecular mechanism underlying the modulation of the KCNQ1/KCNE1 channel.
Collapse
Affiliation(s)
- Xiangbo Gou
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China; Department of Pharmacology, North China University of Science and Technology, Tangshan 063210, China
| | - Wenying Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Sihao Zou
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Yajuan Qi
- Department of Pharmacology, North China University of Science and Technology, Tangshan 063210, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China.
| |
Collapse
|
18
|
Lin Z, Xing W, Gao C, Wang X, Qi D, Dai G, Zhao W, Yan G. Inhibitory Effect of Vascular Endothelial Growth Factor on the Slowly Activating Delayed Rectifier Potassium Current in Guinea Pig Ventricular Myocytes. J Am Heart Assoc 2018; 7:JAHA.117.007730. [PMID: 29374044 PMCID: PMC5850256 DOI: 10.1161/jaha.117.007730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Vascular endothelial growth factor (VEGF) exerts a number of beneficial effects on ischemic myocardium via its angiogenic properties. However, little is known about whether VEGF has a direct effect on the electrical properties of cardiomyocytes. In the present study, we investigated the effects of different concentrations of VEGF on delayed rectifier potassium currents (IK) in guinea pig ventricular myocytes and their effects on action potential (AP) parameters. Methods and Results IK and AP were recorded by the whole‐cell patch clamp method in ventricular myocytes. Cells were superfused with control solution or solution containing VEGF at different concentrations for 10 minutes before recording. Some ventricular myocytes were pretreated with a phosphatidylinositol 3‐kinase inhibitor for 1 hour before the addition of VEGF. We found that VEGF inhibited the slowly activating delayed rectifier potassium current (IKs) in a concentration‐dependent manner (18.13±1.04 versus 12.73±0.34, n=5, P=0.001; 12.73±0.34 versus 9.05±1.20, n=5, P=0.036) and prolonged AP duration (894.5±36.92 versus 746.3±33.71, n=5, P=0.021). Wortmannin, a phosphatidylinositol 3‐kinase inhibitor, eliminated these VEGF‐induced effects. VEGF had no significant effect on the rapidly activating delayed rectifier potassium current (IKr), resting membrane potential, AP amplitude, or maximal velocity of depolarization. Conclusions VEGF inhibited IKs in a concentration‐dependent manner through a phosphatidylinositol 3‐kinase–mediated signaling pathway, leading to AP prolongation. The results indicate a promising therapeutic potential of VEGF in prevention of ventricular tachyarrhythmias under conditions of high sympathetic activity and ischemia.
Collapse
Affiliation(s)
- Zhenhao Lin
- Department of Cardiology, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Wenlu Xing
- Department of Cardiology, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Chuanyu Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Xianpei Wang
- Department of Cardiology, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Datun Qi
- Department of Cardiology, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Guoyou Dai
- Department of Cardiology, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Wen Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ganxin Yan
- Main Line Health Heart Center, Lankenau Institute for Medical Research, Wynnewood, PA
| |
Collapse
|
19
|
Aziz Q, Li Y, Tinker A. Potassium channels in the sinoatrial node and their role in heart rate control. Channels (Austin) 2018; 12:356-366. [PMID: 30301404 PMCID: PMC6207292 DOI: 10.1080/19336950.2018.1532255] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/25/2018] [Accepted: 09/28/2018] [Indexed: 10/28/2022] Open
Abstract
Potassium currents determine the resting membrane potential and govern repolarisation in cardiac myocytes. Here, we review the various currents in the sinoatrial node focussing on their molecular and cellular properties and their role in pacemaking and heart rate control. We also describe how our recent finding of a novel ATP-sensitive potassium channel population in these cells fits into this picture.
Collapse
Affiliation(s)
- Qadeer Aziz
- William Harvey Heart Centre, Barts & The London School of Medicine & Dentistry, Queen Mary, University of London, London, UK
| | - Yiwen Li
- William Harvey Heart Centre, Barts & The London School of Medicine & Dentistry, Queen Mary, University of London, London, UK
| | - Andrew Tinker
- William Harvey Heart Centre, Barts & The London School of Medicine & Dentistry, Queen Mary, University of London, London, UK
| |
Collapse
|
20
|
β-adrenergic stimulation augments transmural dispersion of repolarization via modulation of delayed rectifier currents I Ks and I Kr in the human ventricle. Sci Rep 2017; 7:15922. [PMID: 29162896 PMCID: PMC5698468 DOI: 10.1038/s41598-017-16218-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/09/2017] [Indexed: 12/23/2022] Open
Abstract
Long QT syndrome (LQTS) is an inherited or drug induced condition associated with delayed repolarization and sudden cardiac death. The cardiac potassium channel, IKr, and the adrenergic-sensitive cardiac potassium current, IKs, are two primary contributors to cardiac repolarization. This study aimed to elucidate the role of β-adrenergic (β-AR) stimulation in mediating the contributions of IKr and IKs to repolarizing the human left ventricle (n = 18). Optical mapping was used to measure action potential durations (APDs) in the presence of the IKs blocker JNJ-303 and the IKr blocker E-4031. We found that JNJ-303 alone did not increase APD. However, under isoprenaline (ISO), both the application of JNJ-303 and additional E-4031 significantly increased APD. With JNJ-303, ISO decreased APD significantly more in the epicardium as compared to the endocardium, with subsequent application E-4031 increasing mid- and endocardial APD80 more significantly than in the epicardium. We found that β-AR stimulation significantly augmented the effect of IKs blocker JNJ-303, in contrast to the reduced effect of IKr blocker E-4031. We also observed synergistic augmentation of transmural repolarization gradient by the combination of ISO and E-4031. Our results suggest β-AR-mediated increase of transmural dispersion of repolarization, which could pose arrhythmogenic risk in LQTS patients.
Collapse
|
21
|
Lee W, Windley MJ, Vandenberg JI, Hill AP. In Vitro and In Silico Risk Assessment in Acquired Long QT Syndrome: The Devil Is in the Details. Front Physiol 2017; 8:934. [PMID: 29201009 PMCID: PMC5696636 DOI: 10.3389/fphys.2017.00934] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/03/2017] [Indexed: 12/16/2022] Open
Abstract
Acquired long QT syndrome, mostly as a result of drug block of the Kv11. 1 potassium channel in the heart, is characterized by delayed cardiac myocyte repolarization, prolongation of the T interval on the ECG, syncope and sudden cardiac death due to the polymorphic ventricular arrhythmia Torsade de Pointes (TdP). In recent years, efforts are underway through the Comprehensive in vitro proarrhythmic assay (CiPA) initiative, to develop better tests for this drug induced arrhythmia based in part on in silico simulations of pharmacological disruption of repolarization. However, drug binding to Kv11.1 is more complex than a simple binary molecular reaction, meaning simple steady state measures of potency are poor surrogates for risk. As a result, there is a plethora of mechanistic detail describing the drug/Kv11.1 interaction—such as drug binding kinetics, state preference, temperature dependence and trapping—that needs to be considered when developing in silico models for risk prediction. In addition to this, other factors, such as multichannel pharmacological profile and the nature of the ventricular cell models used in simulations also need to be considered in the search for the optimum in silico approach. Here we consider how much of mechanistic detail needs to be included for in silico models to accurately predict risk and further, how much of this detail can be retrieved from protocols that are practical to implement in high throughout screens as part of next generation of preclinical in silico drug screening approaches?
Collapse
Affiliation(s)
- William Lee
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Monique J Windley
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Jamie I Vandenberg
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Adam P Hill
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
22
|
Jiménez MAV, Nascimento JHM, Monnerat G, Maciel L, Paiva CN, Pedrosa RC, Campos de Carvalho AC, Medei E. Autoantibodies with beta-adrenergic activity from chronic chagasic patients induce cardiac arrhythmias and early afterdepolarization in a drug-induced LQT2 rabbit hearts. Int J Cardiol 2017; 240:354-359. [PMID: 28320606 DOI: 10.1016/j.ijcard.2017.02.066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cardiac arrhythmias are one of the main causes of death in ChCP and other dilated cardiomyopathies. Previous studies demonstrated that ventricular arrhythmias are associated with the presence of autoantibodies with beta-adrenergic activity, Ab-β. OBJECTIVES The aim of this study was to investigate whether Ab-β, present in chronic chagasic patients (ChCP), induce cardiac arrhythmias in the pharmacological type-2 long QT syndrome model (LQTS-2). METHODS/RESULTS The LQTS2 was established by perfusion of Tyrode saline solution with a potassium channel blocker E-4031 (5μM) in isolated rabbit hearts or in rabbit cardiac strips, in order to record ECG or action potential, respectively. Autoantibodies from ChCP activating (Ab-β) or not (Ab-NR) cardiac beta 1-adrenergic receptors were used. Ab-β, but not Ab-NR, were able to significantly shorten QT, QTc and increase Tpeak-Tend interval in the LQTS-2. A positive correlation between higher QTc and Tpeak-Tend was found after Ab-β perfusion in the same model. In addition, in the LQTS-2 model, in almost 75% (11/15) of the hearts perfused with Ab-β, ventricular and atrio-ventricular electrical disturbances were observed. Atenolol abolished all Ab-β-induced arrhythmias. Ab-β, when perfused in a cellular LQTS-2, drastically reduced the action potential duration and evoked early afterdepolarization (EAD's), while Ab-NR did not modulate the AP properties in the LQTS-2. CONCLUSION The results indicate that Ab-β were able to induce cardiac arrhythmias and EAD's. This phenomenon can explain, at least in part, the cellular mechanism of Ab-β-induced arrhythmias. Furthermore, atenolol is effective for the treatment of Ab-β-induced arrhythmias.
Collapse
Affiliation(s)
| | - José H M Nascimento
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Monnerat
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Maciel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia N Paiva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Roberto Coury Pedrosa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Emiliano Medei
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
23
|
Osadchii OE. Role of abnormal repolarization in the mechanism of cardiac arrhythmia. Acta Physiol (Oxf) 2017; 220 Suppl 712:1-71. [PMID: 28707396 DOI: 10.1111/apha.12902] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In cardiac patients, life-threatening tachyarrhythmia is often precipitated by abnormal changes in ventricular repolarization and refractoriness. Repolarization abnormalities typically evolve as a consequence of impaired function of outward K+ currents in cardiac myocytes, which may be caused by genetic defects or result from various acquired pathophysiological conditions, including electrical remodelling in cardiac disease, ion channel modulation by clinically used pharmacological agents, and systemic electrolyte disorders seen in heart failure, such as hypokalaemia. Cardiac electrical instability attributed to abnormal repolarization relies on the complex interplay between a provocative arrhythmic trigger and vulnerable arrhythmic substrate, with a central role played by the excessive prolongation of ventricular action potential duration, impaired intracellular Ca2+ handling, and slowed impulse conduction. This review outlines the electrical activity of ventricular myocytes in normal conditions and cardiac disease, describes classical electrophysiological mechanisms of cardiac arrhythmia, and provides an update on repolarization-related surrogates currently used to assess arrhythmic propensity, including spatial dispersion of repolarization, activation-repolarization coupling, electrical restitution, TRIaD (triangulation, reverse use dependence, instability, and dispersion), and the electromechanical window. This is followed by a discussion of the mechanisms that account for the dependence of arrhythmic vulnerability on the location of the ventricular pacing site. Finally, the review clarifies the electrophysiological basis for cardiac arrhythmia produced by hypokalaemia, and gives insight into the clinical importance and pathophysiology of drug-induced arrhythmia, with particular focus on class Ia (quinidine, procainamide) and Ic (flecainide) Na+ channel blockers, and class III antiarrhythmic agents that block the delayed rectifier K+ channel (dofetilide).
Collapse
Affiliation(s)
- O. E. Osadchii
- Department of Health Science and Technology; University of Aalborg; Aalborg Denmark
| |
Collapse
|
24
|
Yokoyama H, Nakamura Y, Saito H, Nagayama Y, Hoshiai K, Wada T, Izumi-Nakaseko H, Ando K, Akie Y, Sugiyama A. Pharmacological characterization of microminipig as a model to assess the drug-induced cardiovascular responses for non-clinical toxicity and/or safety pharmacology studies. J Toxicol Sci 2017; 42:93-101. [PMID: 28070113 DOI: 10.2131/jts.42.93] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
We tried to establish the halothane-anesthetized microminipigs as an alternative animal model for non-clinical toxicity and/or safety pharmacology studies. In order to characterize the halothane-anesthetized microminipigs, we firstly clarified the effects of halothane anesthesia on their cardiovascular system (n = 5). Then, we examined the cardiovascular effects of dl-sotalol in doses of 0.1, 0.3 and 1 mg/kg, i.v. on the halothane-anesthetized microminipigs (n = 6). Induction of the halothane anesthesia by itself prolonged the QT interval as well as QTcF, suggesting that the halothane anesthesia can reduce the cardiac repolarization reserve in microminipigs like in dogs. dl-Sotalol showed more potent negative chronotropic, dromotropic and hypotensive effects together with repolarization delay in microminipigs than in dogs, although each cardiovascular response to dl-sotalol was directionally similar between them, suggesting greater basal sympathetic tone and/or smaller volume of distribution of the drug in microminipigs than in dogs. Analyses of proarrhythmic surrogate markers indicate that Tpeak-Tend and short-term variability of QT interval may be more sensitive to detect the dl-sotalol-induced direct electrophysiological changes in microminipigs than in dogs, but its reverse will be true for J-Tpeakc. Thus, these results may help better understand the drug-induced cardiovascular responses in microminipigs.
Collapse
|
25
|
Finlay M, Harmer SC, Tinker A. The control of cardiac ventricular excitability by autonomic pathways. Pharmacol Ther 2017; 174:97-111. [PMID: 28223225 DOI: 10.1016/j.pharmthera.2017.02.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Central to the genesis of ventricular cardiac arrhythmia are variations in determinants of excitability. These involve individual ionic channels and transporters in cardiac myocytes but also tissue factors such as variable conduction of the excitation wave, fibrosis and source-sink mismatch. It is also known that in certain diseases and particularly the channelopathies critical events occur with specific stressors. For example, in hereditary long QT syndrome due to mutations in KCNQ1 arrhythmic episodes are provoked by exercise and in particular swimming. Thus not only is the static substrate important but also how this is modified by dynamic signalling events associated with common physiological responses. In this review, we examine the regulation of ventricular excitability by signalling pathways from a cellular and tissue perspective in an effort to identify key processes, effectors and potential therapeutic approaches. We specifically focus on the autonomic nervous system and related signalling pathways.
Collapse
Affiliation(s)
- Malcolm Finlay
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK
| | - Stephen C Harmer
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK
| | - Andrew Tinker
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK.
| |
Collapse
|
26
|
Chiamvimonvat N, Chen-Izu Y, Clancy CE, Deschenes I, Dobrev D, Heijman J, Izu L, Qu Z, Ripplinger CM, Vandenberg JI, Weiss JN, Koren G, Banyasz T, Grandi E, Sanguinetti MC, Bers DM, Nerbonne JM. Potassium currents in the heart: functional roles in repolarization, arrhythmia and therapeutics. J Physiol 2017; 595:2229-2252. [PMID: 27808412 DOI: 10.1113/jp272883] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/11/2016] [Indexed: 12/19/2022] Open
Abstract
This is the second of the two White Papers from the fourth UC Davis Cardiovascular Symposium Systems Approach to Understanding Cardiac Excitation-Contraction Coupling and Arrhythmias (3-4 March 2016), a biennial event that brings together leading experts in different fields of cardiovascular research. The theme of the 2016 symposium was 'K+ channels and regulation', and the objectives of the conference were severalfold: (1) to identify current knowledge gaps; (2) to understand what may go wrong in the diseased heart and why; (3) to identify possible novel therapeutic targets; and (4) to further the development of systems biology approaches to decipher the molecular mechanisms and treatment of cardiac arrhythmias. The sessions of the Symposium focusing on the functional roles of the cardiac K+ channel in health and disease, as well as K+ channels as therapeutic targets, were contributed by Ye Chen-Izu, Gideon Koren, James Weiss, David Paterson, David Christini, Dobromir Dobrev, Jordi Heijman, Thomas O'Hara, Crystal Ripplinger, Zhilin Qu, Jamie Vandenberg, Colleen Clancy, Isabelle Deschenes, Leighton Izu, Tamas Banyasz, Andras Varro, Heike Wulff, Eleonora Grandi, Michael Sanguinetti, Donald Bers, Jeanne Nerbonne and Nipavan Chiamvimonvat as speakers and panel discussants. This article summarizes state-of-the-art knowledge and controversies on the functional roles of cardiac K+ channels in normal and diseased heart. We endeavour to integrate current knowledge at multiple scales, from the single cell to the whole organ levels, and from both experimental and computational studies.
Collapse
Affiliation(s)
- Nipavan Chiamvimonvat
- Department of Internal Medicine, University of California, Davis, Genome and Biomedical Science Facility, Rm 6315, Davis, CA, 95616, USA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA, 95655, USA
| | - Ye Chen-Izu
- Department of Internal Medicine, University of California, Davis, Genome and Biomedical Science Facility, Rm 6315, Davis, CA, 95616, USA.,Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA.,Department of Biomedical Engineering, University of California, Davis, Genome and Biomedical Science Facility, Rm 2303, Davis, CA, 95616, USA
| | - Colleen E Clancy
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Isabelle Deschenes
- Department of Physiology and Biophysics, and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44109, USA.,Heart and Vascular Research Center, MetroHealth Medical Center, Cleveland, OH, 44109, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Leighton Izu
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Zhilin Qu
- Division of Cardiology, Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, 3645 MRL, Los Angeles, CA, 90095, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW, 2010, Australia
| | - James N Weiss
- Division of Cardiology, Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, 3645 MRL, Los Angeles, CA, 90095, USA
| | - Gideon Koren
- Cardiovascular Research Center, Rhode Island Hospital and the Cardiovascular Institute, The Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Tamas Banyasz
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Michael C Sanguinetti
- Department of Internal Medicine, University of Utah, Nora Eccles Harrison Cardiovascular Research & Training Institute, Salt Lake City, UT, 84112, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Science Facility, Rm 3503, Davis, CA, 95616, USA
| | - Jeanne M Nerbonne
- Departments of Developmental Biology and Internal Medicine, Cardiovascular Division, Washington University Medical School, St Louis, MO, 63110, USA
| |
Collapse
|
27
|
Pointer KB, Clark PA, Eliceiri KW, Salamat MS, Robertson GA, Kuo JS. Administration of Non-Torsadogenic human Ether-à-go-go-Related Gene Inhibitors Is Associated with Better Survival for High hERG-Expressing Glioblastoma Patients. Clin Cancer Res 2016; 23:73-80. [PMID: 27635088 DOI: 10.1158/1078-0432.ccr-15-3169] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 08/12/2016] [Accepted: 09/03/2016] [Indexed: 12/29/2022]
Abstract
PURPOSE Glioblastoma is the most malignant primary brain tumor, with a median survival of less than 2 years. More effective therapeutic approaches are needed to improve clinical outcomes. EXPERIMENTAL DESIGN Glioblastoma patient-derived cells (GPDC) were isolated from patient glioblastomas and implanted in mice to form xenografts. IHC was performed for human Ether-à-go-go-Related Gene (hERG) expression and tumor proliferation. Sphere-forming assays with the hERG blocker E-4031 were performed on a high and low hERG-expressing lines. A glioblastoma tissue microarray (TMA; 115 patients) was used to correlate hERG expression with patient survival. Clinical data were analyzed to determine whether patient survival was affected by incidental administration of hERG inhibitory drugs and the correlative effect of patient glioblastoma hERG expression levels. RESULTS hERG expression was upregulated in glioblastoma xenografts with higher proliferative indices. High hERG-expressing GPDCs showed a reduction in sphere formation when treated with hERG inhibitors compared with low hERG-expressing GPDCs. Glioblastoma TMA analysis showed worse survival for glioblastoma patients with high hERG expression versus low expression-43.5 weeks versus 60.9 weeks, respectively (P = 0.022). Furthermore, patients who received at least one hERG blocker had a better survival rate compared with patients who did not (P = 0.0015). Subgroup analysis showed that glioblastoma patients with high hERG expression who received hERG blockers had improved survival (P = 0.0458). There was no difference in survival for low hERG-expressing glioblastoma patients who received hERG blockers (P = 0.4136). CONCLUSIONS Our findings suggest that hERG is a potential glioblastoma survival marker, and that already approved drugs with non-torsadogenic hERG inhibitory activity may potentially be repurposed as adjuvant glioblastoma therapy in high hERG-expressing glioblastoma patients. Clin Cancer Res; 23(1); 73-80. ©2016 AACRSee related commentary by Arcangeli and Becchetti, p. 3.
Collapse
Affiliation(s)
- Kelli B Pointer
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin.,Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin-Madison, Madison, Wisconsin
| | - Paul A Clark
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kevin W Eliceiri
- Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin-Madison, Madison, Wisconsin.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - M Shahriar Salamat
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Gail A Robertson
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin. .,Cardiovascular Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - John S Kuo
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin. .,Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Surgery, National University of Singapore, Singapore
| |
Collapse
|
28
|
Rothenberg I, Piccini I, Wrobel E, Stallmeyer B, Müller J, Greber B, Strutz-Seebohm N, Schulze-Bahr E, Schmitt N, Seebohm G. Structural interplay of K V7.1 and KCNE1 is essential for normal repolarization and is compromised in short QT syndrome 2 (K V7.1-A287T). HeartRhythm Case Rep 2016; 2:521-529. [PMID: 28491751 PMCID: PMC5420010 DOI: 10.1016/j.hrcr.2016.08.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Ina Rothenberg
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Ilaria Piccini
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Eva Wrobel
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Birgit Stallmeyer
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Jovanca Müller
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Boris Greber
- Human Stem Cell Pluripotency Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany
| | - Nathalie Strutz-Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
- Interdisziplinäres Zentrum für Klinische Forschung Münster (IZKF Münster) and Innovative Medizinische Forschung (IMF Münster), Faculty of Medicine, University of Münster, Münster, Germany
| | - Nicole Schmitt
- Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
- Interdisziplinäres Zentrum für Klinische Forschung Münster (IZKF Münster) and Innovative Medizinische Forschung (IMF Münster), Faculty of Medicine, University of Münster, Münster, Germany
- Address reprint requests and correspondence: Dr Guiscard Seebohm, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D48149 Münster, Germany.Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D48149MünsterGermany
| |
Collapse
|
29
|
Abstract
Any disturbance of electrical impulse formation in the heart and of impulse conduction or action potential (AP) repolarization can lead to rhythm disorders. Potassium (K(+)) channels play a prominent role in the AP repolarization process. In this review we describe the causes and mechanisms of proarrhythmic effects that arise as a response to blockers of cardiac K(+) channels. The largest and chemically most diverse groups of compound targets are Kv11.1 (hERG) and Kv7.1 (KvLQT1) channels. Finally, the proarrhythmic propensity of atrial-selective K(+) blockers inhibiting Kv1.5, Kir3.1/3.4, SK, and K2P channels is discussed.
Collapse
Affiliation(s)
- Lasse Skibsbye
- Danish Arrhythmia Research Centre, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, 3 Copenhagen N DK-2200, Denmark
| | - Ursula Ravens
- Department of Pharmacology and Toxicology, Medical Faculty Carl Gustav Carus, Institut für Pharmakologie und Toxikologie, TU Dresden, Fetscherstrasse 74, Dresden D-01307, Germany.
| |
Collapse
|
30
|
Liu J, Laksman Z, Backx PH. The electrophysiological development of cardiomyocytes. Adv Drug Deliv Rev 2016; 96:253-73. [PMID: 26788696 DOI: 10.1016/j.addr.2015.12.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/23/2015] [Accepted: 12/31/2015] [Indexed: 02/07/2023]
Abstract
The generation of human cardiomyocytes (CMs) from human pluripotent stem cells (hPSCs) has become an important resource for modeling human cardiac disease and for drug screening, and also holds significant potential for cardiac regeneration. Many challenges remain to be overcome however, before innovation in this field can translate into a change in the morbidity and mortality associated with heart disease. Of particular importance for the future application of this technology is an improved understanding of the electrophysiologic characteristics of CMs, so that better protocols can be developed and optimized for generating hPSC-CMs. Many different cell culture protocols are currently utilized to generate CMs from hPSCs and all appear to yield relatively “developmentally” immature CMs with highly heterogeneous electrical properties. These hPSC-CMs are characterized by spontaneous beating at highly variable rates with a broad range of depolarization-repolarization patterns, suggestive of mixed populations containing atrial, ventricular and nodal cells. Many recent studies have attempted to introduce approaches to promote maturation and to create cells with specific functional properties. In this review, we summarize the studies in which the electrical properties of CMs derived from stem cells have been examined. In order to place this information in a useful context, we also review the electrical properties of CMs as they transition from the developing embryo to the adult human heart. The signal pathways involved in the regulation of ion channel expression during development are also briefly considered.
Collapse
|
31
|
Lee W, Mann SA, Windley MJ, Imtiaz MS, Vandenberg JI, Hill AP. In silico assessment of kinetics and state dependent binding properties of drugs causing acquired LQTS. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 120:89-99. [PMID: 26713558 DOI: 10.1016/j.pbiomolbio.2015.12.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/06/2015] [Accepted: 12/16/2015] [Indexed: 11/24/2022]
Abstract
The Kv11.1 or hERG potassium channel is responsible for one of the major repolarising currents (IKr) in cardiac myocytes. Drug binding to hERG can result in reduction in IKr, action potential prolongation, acquired long QT syndrome and fatal cardiac arrhythmias. The current guidelines for pre-clinical assessment of drugs in development is based on the measurement of the drug concentration that causes 50% current block, i.e., IC50. However, drugs with the same apparent IC50 may have very different kinetics of binding and unbinding, as well as different affinities for the open and inactivated states of Kv11.1. Therefore, IC50 measurements may not reflect the true risk of drug induced arrhythmias. Here we have used an in silico approach to test the hypothesis that drug binding kinetics and differences in state-dependent affinity will influence the extent of cardiac action potential prolongation independent of apparent IC50 values. We found, in general that drugs with faster overall kinetics and drugs with higher affinity for the open state relative to the inactivated state cause more action potential prolongation. These characteristics of drug-hERG interaction are likely to be more arrhythmogenic but cannot be predicted by IC50 measurement alone. Our results suggest that the pre-clinical assessment of Kv11.1-drug interactions should include descriptions of the kinetics and state dependence of drug binding. Further, incorporation of this information into sophisticated in silico models should be able to better predict arrhythmia risk and therefore more accurately assess safety of new drugs in development.
Collapse
Affiliation(s)
- William Lee
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia
| | - Stefan A Mann
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia
| | - Monique J Windley
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia
| | - Mohammad S Imtiaz
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia.
| |
Collapse
|
32
|
Zhao Y, Huang H, Du Y, Li X, Lv T, Zhang S, Wei H, Shang J, Liu P, Liu H. β1-Adrenoceptor autoantibodies affect action potential duration and delayed rectifier potassium currents in guinea pigs. Cardiovasc Toxicol 2015; 15:1-9. [PMID: 24894912 DOI: 10.1007/s12012-014-9261-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
β1-Adrenoceptor autoantibodies (β1-AAs) affect the action potential duration (APD) in cardiomyocytes and are related to ventricular arrhythmias. The delayed rectifier potassium current (I K) plays a crucial role in APD, but the effects of β1-AAs on I K have not been completely illuminated. This work aimed to observe the effects of β1-AAs on I K and APD and further explore the mechanisms of β1-AA-mediated ventricular arrhythmias. β1-AAs were obtained from sera of patients with coronary heart disease (CHD) and nonsustained ventricular tachycardia. With whole-cell patch clamp technique, action potentials and I K were recorded. The results illustrated 0.1 μmol/L β1-AAs shortened APD at 50 % (APD50) and 90 % (APD90) of the repolarization. However, at 0.01 μmol/L, β1-AAs had no effects on either APD90 or APD50 (P > 0.05). At 0.001 μmol/L, β1-AAs significantly prolonged APD90 and APD50. Moreover, β1-AAs (0.001, 0.01, 0.1 μmol/L) dose-dependently increased the rapidly activating delayed rectifier potassium current (I Kr), but similarly decreased the slowly activating delayed rectifier potassium current (I Ks) and increased L-type calcium currents at the different concentrations. Taken together, the IKr increase induced by high β1-AA concentrations is responsible for a significant APD reduction which would contribute to repolarization changes and trigger the malignant ventricular arrhythmias in CHD patients.
Collapse
Affiliation(s)
- Yuhui Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Negroni JA, Morotti S, Lascano EC, Gomes AV, Grandi E, Puglisi JL, Bers DM. β-adrenergic effects on cardiac myofilaments and contraction in an integrated rabbit ventricular myocyte model. J Mol Cell Cardiol 2015; 81:162-75. [PMID: 25724724 DOI: 10.1016/j.yjmcc.2015.02.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/10/2015] [Accepted: 02/17/2015] [Indexed: 12/21/2022]
Abstract
A five-state model of myofilament contraction was integrated into a well-established rabbit ventricular myocyte model of ion channels, Ca(2+) transporters and kinase signaling to analyze the relative contribution of different phosphorylation targets to the overall mechanical response driven by β-adrenergic stimulation (β-AS). β-AS effect on sarcoplasmic reticulum Ca(2+) handling, Ca(2+), K(+) and Cl(-) currents, and Na(+)/K(+)-ATPase properties was included based on experimental data. The inotropic effect on the myofilaments was represented as reduced myofilament Ca(2+) sensitivity (XBCa) and titin stiffness, and increased cross-bridge (XB) cycling rate (XBcy). Assuming independent roles of XBCa and XBcy, the model reproduced experimental β-AS responses on action potentials and Ca(2+) transient amplitude and kinetics. It also replicated the behavior of force-Ca(2+), release-restretch, length-step, stiffness-frequency and force-velocity relationships, and increased force and shortening in isometric and isotonic twitch contractions. The β-AS effect was then switched off from individual targets to analyze their relative impact on contractility. Preventing β-AS effects on L-type Ca(2+) channels or phospholamban limited Ca(2+) transients and contractile responses in parallel, while blocking phospholemman and K(+) channel (IKs) effects enhanced Ca(2+) and inotropy. Removal of β-AS effects from XBCa enhanced contractile force while decreasing peak Ca(2+) (due to greater Ca(2+) buffering), but had less effect on shortening. Conversely, preventing β-AS effects on XBcy preserved Ca(2+) transient effects, but blunted inotropy (both isometric force and especially shortening). Removal of titin effects had little impact on contraction. Finally, exclusion of β-AS from XBCa and XBcy while preserving effects on other targets resulted in preserved peak isometric force response (with slower kinetics) but nearly abolished enhanced shortening. β-AS effects on XBCa and XBcy have greater impact on isometric and isotonic contraction, respectively.
Collapse
Affiliation(s)
- Jorge A Negroni
- Department of Comparative, Cellular and Molecular Biology, Universidad Favaloro, Buenos Aires, Argentina.
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis, CA, USA
| | - Elena C Lascano
- Department of Comparative, Cellular and Molecular Biology, Universidad Favaloro, Buenos Aires, Argentina
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology and Behavior, University of California Davis, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, CA, USA
| | - José L Puglisi
- Department of Pharmacology, University of California Davis, CA, USA
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, CA, USA.
| |
Collapse
|
34
|
The potassium current carried by TREK-1 channels in rat cardiac ventricular muscle. Pflugers Arch 2014; 467:1069-79. [PMID: 25539776 DOI: 10.1007/s00424-014-1678-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 01/13/2023]
Abstract
We studied the potassium current flowing through TREK-1 channels in rat cardiac ventricular myocytes. We separated the TREK-1 current from other current components by blocking most other channels with a blocker cocktail. We tried to inhibit the TREK-1 current by activating protein kinase A (PKA) with a mixture of forskolin and isobutyl-methylxanthine (IBMX). Activation of PKA blocked an outwardly rectifying current component at membrane potentials positive to -40 mV. At 37 °C, application of forskolin plus IBMX reduced the steady-state outward current measured at positive voltages by about 52 %. Application of the potassium channel blockers quinidine or tetrahexylammonium also reduced the steady-state outward current by about 50 %. Taken together, our results suggest that the increase in temperature from 22 to 37 °C increased the TREK-1 current by a factor of at least 5 and that the average density of the TREK-1 current in rat cardiomyocytes at 37 °C is about 1.5 pA/pF at +30 mV. The contribution of TREK-1 to the action potential was assessed by using a dynamic patch clamp technique. After subtraction of simulated TREK-1 currents, action potential duration at 50 or 90 % repolarisation was increased by about 12 %, indicating that TREK-1 may be functionally important in rat ventricular muscle. During sympathetic stimulation, inhibition of TREK-1 channels via PKA is expected to prolong the action potential primarily in subendocardial myocytes; this may decrease the transmural dispersion of repolarisation and thus may serve to prevent the occurrence of arrhythmias.
Collapse
|
35
|
Coppini R, Ferrantini C, Aiazzi A, Mazzoni L, Sartiani L, Mugelli A, Poggesi C, Cerbai E. Isolation and functional characterization of human ventricular cardiomyocytes from fresh surgical samples. J Vis Exp 2014. [PMID: 24798397 DOI: 10.3791/51116] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cardiomyocytes from diseased hearts are subjected to complex remodeling processes involving changes in cell structure, excitation contraction coupling and membrane ion currents. Those changes are likely to be responsible for the increased arrhythmogenic risk and the contractile alterations leading to systolic and diastolic dysfunction in cardiac patients. However, most information on the alterations of myocyte function in cardiac diseases has come from animal models. Here we describe and validate a protocol to isolate viable myocytes from small surgical samples of ventricular myocardium from patients undergoing cardiac surgery operations. The protocol is described in detail. Electrophysiological and intracellular calcium measurements are reported to demonstrate the feasibility of a number of single cell measurements in human ventricular cardiomyocytes obtained with this method. The protocol reported here can be useful for future investigations of the cellular and molecular basis of functional alterations of the human heart in the presence of different cardiac diseases. Further, this method can be used to identify novel therapeutic targets at cellular level and to test the effectiveness of new compounds on human cardiomyocytes, with direct translational value.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department NeuroFarBa, Division of Pharmacology, University of Florence;
| | - Cecila Ferrantini
- Department of Clinical and Experimental Medicine, Division of Physiology, University of Florence
| | - Alessandro Aiazzi
- Department of Clinical and Experimental Medicine, Division of Physiology, University of Florence
| | - Luca Mazzoni
- Department NeuroFarBa, Division of Pharmacology, University of Florence
| | - Laura Sartiani
- Department NeuroFarBa, Division of Pharmacology, University of Florence
| | | | - Corrado Poggesi
- Department of Clinical and Experimental Medicine, Division of Physiology, University of Florence
| | - Elisabetta Cerbai
- Department NeuroFarBa, Division of Pharmacology, University of Florence
| |
Collapse
|
36
|
Beta-adrenergic stimulation reverses the I Kr-I Ks dominant pattern during cardiac action potential. Pflugers Arch 2014; 466:2067-76. [PMID: 24535581 DOI: 10.1007/s00424-014-1465-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 01/06/2014] [Accepted: 01/28/2014] [Indexed: 12/21/2022]
Abstract
β-Adrenergic stimulation differentially modulates different K(+) channels and thus fine-tunes cardiac action potential (AP) repolarization. However, it remains unclear how the proportion of I Ks, I Kr, and I K1 currents in the same cell would be altered by β-adrenergic stimulation, which would change the relative contribution of individual K(+) current to the total repolarization reserve. In this study, we used an innovative AP-clamp sequential dissection technique to directly record the dynamic I Ks, I Kr, and I K1 currents during the AP in guinea pig ventricular myocytes under physiologically relevant conditions. Our data provide quantitative measures of the magnitude and time course of I Ks, I Kr, and I K1 currents in the same cell under its own steady-state AP, in a physiological milieu, and with preserved Ca(2+) homeostasis. We found that isoproterenol treatment significantly enhanced I Ks, moderately increased I K1, but slightly decreased I Kr in a dose-dependent manner. The dominance pattern of the K(+) currents was I Kr > I K1 > I Ks at the control condition, but reversed to I Kr < I K1 < I Ks following β-adrenergic stimulation. We systematically determined the changes in the relative contribution of I Ks, I Kr, and I K1 to cardiac repolarization during AP at different adrenergic states. In conclusion, the β-adrenergic stimulation fine-tunes the cardiac AP morphology by shifting the power of different K(+) currents in a dose-dependent manner. This knowledge is important for designing antiarrhythmic drug strategies to treat hearts exposed to various sympathetic tones.
Collapse
|
37
|
Membrane channels as integrators of G-protein-mediated signaling. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:521-31. [PMID: 24028827 DOI: 10.1016/j.bbamem.2013.08.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 08/14/2013] [Accepted: 08/21/2013] [Indexed: 01/03/2023]
Abstract
A variety of extracellular stimuli regulate cellular responses via membrane receptors. A well-known group of seven-transmembrane domain-containing proteins referred to as G protein-coupled receptors, directly couple with the intracellular GTP-binding proteins (G proteins) across cell membranes and trigger various cellular responses by regulating the activity of several enzymes as well as ion channels. Many specific populations of ion channels are directly controlled by G proteins; however, indirect modulation of some channels by G protein-dependent phosphorylation events and lipid metabolism is also observed. G protein-mediated diverse modifications affect the ion channel activities and spatio-temporally regulate membrane potentials as well as of intracellular Ca(2+) concentrations in both excitatory and non-excitatory cells. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.
Collapse
|
38
|
Structure of the C-terminal region of an ERG channel and functional implications. Proc Natl Acad Sci U S A 2013; 110:11648-53. [PMID: 23801759 DOI: 10.1073/pnas.1306887110] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The human ether-à-go-go-related gene (hERG) encodes a K(+) channel crucial for repolarization of the cardiac action potential. EAG-related gene (ERG) channels contain a C-terminal cyclic nucleotide-binding homology domain coupled to the pore of the channel by a C-linker. Here, we report the structure of the C-linker/cyclic nucleotide-binding homology domain of a mosquito ERG channel at 2.5-Å resolution. The structure reveals that the region expected to form the cyclic nucleotide-binding pocket is negatively charged and is occupied by a short β-strand, referred to as the intrinsic ligand, explaining the lack of direct regulation of ERG channels by cyclic nucleotides. In hERG channels, the intrinsic ligand harbors hereditary mutations associated with long-QT syndrome (LQTS), a potentially lethal cardiac arrhythmia. Mutations in the intrinsic ligand affected hERG channel gating and LQTS mutations abolished hERG currents and altered trafficking of hERG channels, which explains the LQT phenotype. The structure also reveals a dramatically different conformation of the C-linker compared with the structures of the related ether-à-go-go-like K(+) and hyperpolarization-activated cyclic nucleotide-modulated channels, suggesting that the C-linker region may be highly dynamic in the KCNH, hyperpolarization-activated cyclic nucleotide-modulated, and cyclic nucleotide-gated channels.
Collapse
|
39
|
Lan YF, Zhang JC, Gao JL, Wang XP, Fang Z, Fu YC, Chen MY, Lin M, Xue Q, Li Y. Effects of nerve growth factor on the action potential duration and repolarizing currents in a rabbit model of myocardial infarction. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2013; 10:39-51. [PMID: 23610573 PMCID: PMC3627714 DOI: 10.3969/j.issn.1671-5411.2013.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 09/11/2012] [Accepted: 01/18/2013] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To investigate the effect of nerve growth factor (NGF) on the action potential and potassium currents of non-infarcted myocardium in the myocardial infarcted rabbit model. METHODS Rabbits with occlusion of the left anterior descending coronary artery were prepared and allowed to recover for eight weeks (healed myocardial infarction, HMI). During ligation surgery of the left coronary artery, a polyethylene tube was placed near the left stellate ganglion in the subcutis of the neck for the purpose of administering NGF 400 U/d for eight weeks (HMI + NGF group). Cardiomyocytes were isolated from regions of the non-infarcted left ventricular wall and the action potentials and ion currents in these cells were recorded using whole-cell patch clamps. RESULTS Compared with HMI and control cardiomyocytes, significant prolongation of APD50 or APD90 (Action potential duration (APD) measured at 50% and 90% of repolarization) in HMI + NGF cardiomyocytes was found. The results showed that the 4-aminopyridine sensitive transient outward potassium current (I to), the rapidly activated omponent of delayed rectifier potassium current (I Kr), the slowly activated component of delayed rectifier potassium current (I Ks), and the L-type calcium current (I CaL) were significantly altered in NGF + HMI cardiomyocytes compared with HMI and control cells. CONCLUSIONS Our results suggest that NGF treatment significantly prolongs APD in HMI cardiomyocytes and that a decrease in outward potassium currents and an increase of inward Ca(2+) current are likely the underlying mechanism of action.
Collapse
Affiliation(s)
- Yun-Feng Lan
- Institute of Geriatric Cardiology of Chinese PLA General Hospital, 28 Fuxing Road, 100853 Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sex differences in repolarization and slow delayed rectifier potassium current and their regulation by sympathetic stimulation in rabbits. Pflugers Arch 2012; 465:805-18. [PMID: 23242028 DOI: 10.1007/s00424-012-1193-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 11/27/2012] [Accepted: 11/28/2012] [Indexed: 12/25/2022]
Abstract
Slow delayed rectifier potassium current (IKs) is important in action potential (AP) repolarization and repolarization reserve. We tested the hypothesis that there are sex-specific differences in IKs, AP, and their regulation by β-adrenergic receptors (β-AR's) using whole-cell patch-clamp. AP duration (APD90) was significantly longer in control female (F) than in control male (M) myocytes. Isoproterenol (ISO, 500 nM) shortened APD90 comparably in M and F, and was largely reversed by β1-AR blocker CGP 20712A (CGP, 300 nM). Inhibition of IKs with chromanol 293B (10 μM) resulted in less APD prolongation in F at baseline (3.0 vs 8.9 %, p < 0.05 vs M) and even in the presence of ISO (5.4 vs 20.9 %, p < 0.05). This suggests that much of the ISO-induced APD abbreviation in F is independent of IKs. In F, baseline IKs was 42 % less and was more weakly activated by ISO (19 vs 68 % in M, p < 0.01). ISO enhancement of IKs was comparably attenuated by CGP in M and F. After ovariectomy, IKs in F had greater enhancement by ISO (72 %), now comparable to control M. After orchiectomy, IKs in M was only slightly enhanced by ISO (23 %), comparable to control F. Pretreatment with thapsigargin (to block SR Ca release) had bigger impact on ISO-induced APD shortening in F than that in M (p < 0.01). In conclusion, we found that there are sex differences in IKs, AP, and their regulation by β-AR's that are modulated by sex hormones, suggesting the potential for sex-specific antiarrhythmic therapy.
Collapse
|
41
|
Vandenberg JI, Perry MD, Perrin MJ, Mann SA, Ke Y, Hill AP. hERG K+ Channels: Structure, Function, and Clinical Significance. Physiol Rev 2012; 92:1393-478. [DOI: 10.1152/physrev.00036.2011] [Citation(s) in RCA: 463] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The human ether-a-go-go related gene (hERG) encodes the pore-forming subunit of the rapid component of the delayed rectifier K+ channel, Kv11.1, which are expressed in the heart, various brain regions, smooth muscle cells, endocrine cells, and a wide range of tumor cell lines. However, it is the role that Kv11.1 channels play in the heart that has been best characterized, for two main reasons. First, it is the gene product involved in chromosome 7-associated long QT syndrome (LQTS), an inherited disorder associated with a markedly increased risk of ventricular arrhythmias and sudden cardiac death. Second, blockade of Kv11.1, by a wide range of prescription medications, causes drug-induced QT prolongation with an increase in risk of sudden cardiac arrest. In the first part of this review, the properties of Kv11.1 channels, including biogenesis, trafficking, gating, and pharmacology are discussed, while the second part focuses on the pathophysiology of Kv11.1 channels.
Collapse
Affiliation(s)
- Jamie I. Vandenberg
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Matthew D. Perry
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Mark J. Perrin
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Stefan A. Mann
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Ying Ke
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| | - Adam P. Hill
- Mark Cowley Lidwill Research Programme in Cardiac Electrophysiology, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, New South Wales, Australia; and University of Ottawa Heart Institute, Ottawa, Canada
| |
Collapse
|
42
|
Kallergis EM, Goudis CA, Simantirakis EN, Kochiadakis GE, Vardas PE. Mechanisms, risk factors, and management of acquired long QT syndrome: a comprehensive review. ScientificWorldJournal 2012; 2012:212178. [PMID: 22593664 PMCID: PMC3347892 DOI: 10.1100/2012/212178] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 12/22/2011] [Indexed: 12/19/2022] Open
Abstract
Long QT syndrome is characterized by prolongation of the corrected QT (QTc) interval on the surface electrocardiogram and is associated with precipitation of torsade de pointes (TdP), a polymorphic ventricular tachycardia that may cause sudden death. Acquired long QT syndrome describes pathologic excessive prolongation of the QT interval, upon exposure to an environmental stressor, with reversion back to normal following removal of the stressor. The most common environmental stressor in acquired long QT syndrome is drug therapy. Acquired long QT syndrome is an important issue for clinicians and a significant public health problem concerning the large number of drugs with this adverse effect with a potentially fatal outcome, the large number of patients exposed to these drugs, and our inability to predict the risk for a given individual. In this paper, we focus on mechanisms underlying QT prolongation, risk factors for torsades de pointes and describe the short- and long-term treatment of acquired long QT syndrome.
Collapse
Affiliation(s)
- Eleftherios M Kallergis
- Department of Cardiology, University Hospital of Heraklion, 711 10 Heraklion, Crete, Greece.
| | | | | | | | | |
Collapse
|
43
|
Kato S, Honjo H, Takemoto Y, Takanari H, Suzuki T, Okuno Y, Opthof T, Sakuma I, Inada S, Nakazawa K, Ashihara T, Kodama I, Kamiya K. Pharmacological Blockade of IKs Destabilizes Spiral-Wave Reentry Under β-Adrenergic Stimulation in Favor of Its Early Termination. J Pharmacol Sci 2012; 119:52-63. [DOI: 10.1254/jphs.12008fp] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
44
|
Liu GX, Choi BR, Ziv O, Li W, de Lange E, Qu Z, Koren G. Differential conditions for early after-depolarizations and triggered activity in cardiomyocytes derived from transgenic LQT1 and LQT2 rabbits. J Physiol 2011; 590:1171-80. [PMID: 22183728 DOI: 10.1113/jphysiol.2011.218164] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Early after-depolarization (EAD), or abnormal depolarization during the plateau phase of action potentials, is a hallmark of long-QT syndrome (LQTS). More than 13 genes have been identified as responsible for LQTS, and elevated risks for EADs may depend on genotypes, such as exercise in LQT1 vs. sudden arousal in LQT2 patients. We investigated mechanisms underlying different high-risk conditions that trigger EADs using transgenic rabbit models of LQT1 and LQT2, which lack I(Ks) and I(Kr) (slow and fast components of delayed rectifying K(+) current), respectively. Single-cell patch-clamp studies show that prolongation of action potential duration (APD) can be further enhanced by lowering extracellular potassium concentration ([K(+)](o)) from 5.4 to 3.6 mm. However, only LQT2 myocytes developed spontaneous EADs following perfusion with lower [K(+)](o), while there was no EAD formation in littermate control (LMC) or LQT1 myocytes, although APDs were also prolonged in LMC myocytes and LQT1 myocytes. Isoprenaline (ISO) prolonged APDs and triggered EADs in LQT1 myocytes in the presence of lower [K(+)](o). In contrast, continuous ISO perfusion diminished APD prolongation and reduced the incidence of EADs in LQT2 myocytes. These different effects of ISO on LQT1 and LQT2 were verified by optical mapping of the whole heart, suggesting that ISO-induced EADs are genotype specific. Further voltage-clamp studies revealed that ISO increases L-type calcium current (I(Ca)) faster than I(Ks) (time constant 9.2 s for I(Ca) and 43.6 s for I(Ks)), and computer simulation demonstrated a high-risk window of EADs in LQT2 during ISO perfusion owing to mismatch in the time courses of I(Ca) and I(Ks), which may explain why a sympathetic surge rather than high sympathetic tone can be an effective trigger of EADs in LQT2 perfused hearts. In summary, EAD formation is genotype specific, such that EADs can be elicited in LQT2 myocytes simply by lowering [K(+)](o), while LQT1 myocytes require sympathetic stimulation. Slower activation of I(Ks) than of I(Ca) by ISO may explain why different sympathetic modes, i.e. sympathetic surge vs. high sympathetic tone, are associated with polymorphic ventricular tachycardia in LQTS patients.
Collapse
Affiliation(s)
- Gong-Xin Liu
- Cardiovascular Research Centre, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Sedova KA, Goshka SL, Vityazev VA, Shmakov DN, Azarov JE. Load-induced changes in ventricular repolarization: evidence of autonomic modulation. Can J Physiol Pharmacol 2011; 89:935-44. [DOI: 10.1139/y11-098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Augmented hemodynamic load increases the risk of arrhythmogenesis by modulating cardiac repolarization duration. We hypothesized that the intervention on the autonomic tone may affect the load-dependent changes in ventricular repolarization. Activation–recovery intervals were measured in unipolar electrograms simultaneously recorded from 64 ventricular epicardial leads, in a total of 26 chinchilla rabbits in resting conditions, and after 1 and 10 min of aortic stenosis. Eleven animals were given atropine and propranolol before the loading. The short-term stenosis decreased the activation–recovery intervals in the right ventricle, whereas the prolonged overload increased the repolarization duration in both ventricles. The treatment with the β-adrenergic and M-cholinergic blockers prolonged the activation–recovery intervals, especially at the left ventricle, attenuating the apicobasal and interventricular gradients of repolarization duration seen in the baseline state. Further ventricular loading shortened the repolarization duration in both ventricles in animals with autonomic blockade. Thus, the autonomic tone was shown to be essential for the development of repolarization heterogeneity across the ventricles. The autonomic blockade transformed the biphasic changes of activation–recovery intervals into their monophasic shortening at aortic stenosis.
Collapse
Affiliation(s)
- Ksenia A. Sedova
- Laboratory of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 50 Pervomayskaya Street, Syktyvkar, 167982 Russia
| | - Sergey L. Goshka
- Laboratory of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 50 Pervomayskaya Street, Syktyvkar, 167982 Russia
| | - Vladimir A. Vityazev
- Laboratory of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 50 Pervomayskaya Street, Syktyvkar, 167982 Russia
| | - Dmitriy N. Shmakov
- Laboratory of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 50 Pervomayskaya Street, Syktyvkar, 167982 Russia
| | - Jan E. Azarov
- Laboratory of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 50 Pervomayskaya Street, Syktyvkar, 167982 Russia
- Department of Physiology, Komi Branch of Kirov State Medical Academy, 11 Babushkin Street, Syktyvkar, 167000 Russia
| |
Collapse
|
46
|
Sroubek J, McDonald TV. Protein kinase A activity at the endoplasmic reticulum surface is responsible for augmentation of human ether-a-go-go-related gene product (HERG). J Biol Chem 2011; 286:21927-36. [PMID: 21536683 DOI: 10.1074/jbc.m110.201699] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human ether-a-go-go-related gene product (HERG) is a cardiac potassium channel commonly implicated in the pathogenesis of the long QT syndrome, type 2 (LQT2). LQT2 mutations typically have incomplete penetrance and affect individuals at various stages of their lives; this may mirror variations in intracellular signaling and HERG regulation. Previous work showed that sustained protein kinase A (PKA) activity augments HERG protein abundance by a mechanism that includes enhanced protein translation. To investigate the subcellular site of this regulation, we generated site-specific probes to the cytoplasmic surface of the endoplasmic reticulum (ER), the presumed locale of channel synthesis. Real-time FRET-based indicators demonstrated both cAMP and PKA activity at the ER. A PKA inhibitor targeted to the ER surface (termed p4PKIg) completely abolished PKA-mediated augmentation of HERG in HEK293 cells as well as rat neonatal cardiomyocytes. Immunofluorescence co-localization, targeted FRET-based PKA biosensors, phospho-specific antibodies, and in vivo phosphorylation experiments confirmed that p4PKIg is preferentially active at the ER surface rather than the plasma membrane. Rerouting this inhibitor to the outer mitochondrial membrane diminishes its ability to block cAMP-dependent HERG induction. Our results support a model where PKA-dependent regulation of HERG synthesis occurs at the ER surface. Furthermore, reagents generated for this study provide novel experimental tools to probe compartmentalized cAMP/PKA signaling within cells.
Collapse
Affiliation(s)
- Jakub Sroubek
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
47
|
Abstract
Pharmacological and molecular approaches have shown that an atypical β-adrenoceptor (AR), called β(3)-AR, that is distinct from β(1)-ARs and β(2)-ARs, exists in some tissues in heterogeneous populations such as β(3a)-ARs and β(3b)-ARs. β(3)-ARs belong to a superfamily of receptors linked to guanine nucleotide binding proteins (G proteins). The β(3)-AR gene contains two introns whereas the β(1)-AR and β(2)-AR genes are intronless, leading to splice variants. β(3)-ARs can couple to G(i) and G(s) and they are reported to be present in brown adipose tissue, vasculature, the heart, among other tissues. β(3)-ARs cause vasodilation of microvessels in the islets of Langerhans and may participate in the pathogenesis of cardiac failure, during which modification of β(1)-AR and β(2)-AR expression occurs. The development of β(3)-AR agonists has led to the elaboration of promising new drugs, including antiobesity and antidiabetic drugs. This article reviews the various pharmacological actions of β(3)-ARs and their clinical implications for diabetes and cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Anita A. Mehta
- Department of Pharmacology, LM College of Pharmacy, Ahmedabad, Gujarat, India
| | | |
Collapse
|
48
|
Laursen M, Olesen SP, Grunnet M, Mow T, Jespersen T. Characterization of cardiac repolarization in the Göttingen minipig. J Pharmacol Toxicol Methods 2011; 63:186-95. [DOI: 10.1016/j.vascn.2010.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 10/06/2010] [Accepted: 10/06/2010] [Indexed: 10/18/2022]
|
49
|
Namekata I, Tsuruoka N, Tsuneoka Y, Matsuda T, Takahara A, Tanaka Y, Suzuki T, Takahashi T, Iida-Tanaka N, Tanaka H. Blocking Effect of NIP-142 on the KCNQ1/KCNE1 Channel Current Expressed in HEK293 Cells. Biol Pharm Bull 2011; 34:153-5. [DOI: 10.1248/bpb.34.153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Iyuki Namekata
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Noriko Tsuruoka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Yayoi Tsuneoka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Tomoyuki Matsuda
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
- Biological Research Laboratories, Nissan Chemical Industries, Ltd
| | - Akira Takahara
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Yoshio Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
- Department of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Takeshi Suzuki
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
- School of Material Science, Japan Advanced Institute of Science and Technology (JAIST)
| | - Tetsuo Takahashi
- Department of Biophysical Chemistry, Faculty of Pharmaceutical Sciences, Toho University
| | - Naoko Iida-Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
- Department of Food Science, Otsuma Women's University
| | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| |
Collapse
|
50
|
Schwoerer AP, Kebernik J, Ehmke H, Friederich P. The subtype-specific effects of droperidol on action potential duration in cellular and computational models of long QT syndrome. Anesth Analg 2010; 111:638-46. [PMID: 20601449 DOI: 10.1213/ane.0b013e3181e41996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Droperidol is a highly potent butyrophenone used for the therapy of postoperative nausea and vomiting. Its cardiac safety in cardiovascular-healthy patients and those with long QT (LQT) syndrome is a matter of debate. In this study, we investigated whether droperidol has subtype-specific effects in cellular and computational models of LQT syndrome. METHODS Left ventricular cardiac myocytes were isolated from adult guinea pig hearts. LQT1-like behavior was pharmacologically induced by chromanol 293B (10 micromol/L) and LQT2-like states by E4031 (10 micromol/L). Computational analysis was performed using the Luo-Rudy dynamic model. Data are given as mean + or - SEM. RESULTS In control myocytes, droperidol lengthened action potentials in a concentration-dependent manner with a maximal prolongation of 37% + or - 13% (n = 4) at a concentration of 0.6 micromol/L. In LQT1-like myocytes, droperidol (0.6 micromol/L) further prolonged action potentials by 31% + or - 6% (n = 6) but shortened action potentials of LQT2-like myocytes by 11% + or - 2% (n = 8). Computational modeling supported the concept that droperidol, in addition to the rapid component of the delayed K(+) current, blocks depolarizing targets, such as the L-type Ca(2+) current, the Na(+)-Ca(2+) exchanger, and the Na(+)-K(+) adenosine triphosphatase. CONCLUSIONS Droperidol has more detrimental effects on cardiac repolarization of LQT1-like than of LQT2-like myocytes suggesting subtype-specific cardiotoxic effects in patients with LQT syndrome. The subtype specificity of droperidol seems to be caused by a complex interaction of droperidol with several different molecular targets. This interaction deserves further investigation to establish the feasibility of a subtype-directed approach in the perioperative management of patients with LQT syndrome.
Collapse
Affiliation(s)
- Alexander P Schwoerer
- Department of Vegetative Physiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | | | | | |
Collapse
|