51
|
Takahashi M, Yokoshiki H, Mitsuyama H, Watanabe M, Temma T, Kamada R, Hagiwara H, Takahashi Y, Anzai T. SK channel blockade prevents hypoxia-induced ventricular arrhythmias through inhibition of Ca 2+/voltage uncoupling in hypertrophied hearts. Am J Physiol Heart Circ Physiol 2021; 320:H1456-H1469. [PMID: 33635168 DOI: 10.1152/ajpheart.00777.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/17/2021] [Indexed: 11/22/2022]
Abstract
Ventricular arrhythmia (VA) is the major cause of death in patients with left ventricular (LV) hypertrophy and/or acute ischemia. We hypothesized that apamin, a blocker of small-conductance Ca2+-activated K+ (SK) channels, alters Ca2+ handling and exhibits anti-arrhythmic effects in ventricular myocardium. Spontaneous hypertensive rats were used as a model of LV hypertrophy. A dual optical mapping of membrane potential (Vm) and intracellular calcium (Cai) was performed during global hypoxia (GH) on the Langendorff perfusion system. The majority of pacing-induced VAs during GH were initiated by triggered activities. Pretreatment of apamin (100 nmol/L) significantly inhibited the VA inducibility. Compared with SK channel blockers (apamin and NS8593), non-SK channel blockers (glibenclamide and 4-AP) did not exhibit anti-arrhythmic effects. Apamin prevented not only action potential duration (APD80) shortening (-18.7 [95% confidence interval, -35.2 to -6.05] ms vs. -2.75 [95% CI, -10.45 to 12.65] ms, P = 0.04) but also calcium transient duration (CaTD80) prolongation (14.52 [95% CI, 8.8-20.35] ms vs. 3.85 [95% CI, -3.3 to 12.1] ms, P < 0.01), thereby reducing CaTD80 - APD80, which denotes "Cai/Vm uncoupling" (33.22 [95% CI, 22-48.4] ms vs. 6.6 [95% CI, 0-14.85] ms, P < 0.01). The reduction of Cai/Vm uncoupling was attributable to less prolonged Ca2+ decay constant and suppression of diastolic Cai increase by apamin. The inhibition of VA inducibility and changes in APs/CaTs parameters caused by apamin was negated by the addition of ouabain, an inhibitor of Na+/K+ pump. Apamin attenuates APD shortening, Ca2+ handling abnormalities, and Cai/Vm uncoupling, leading to inhibition of VA occurrence in hypoxic hypertrophied hearts.NEW & NOTEWORTHY We demonstrated that hypoxia-induced ventricular arrhythmias were mainly initiated by Ca2+-loaded triggered activities in hypertrophied hearts. The blockades of small-conductance Ca2+-activated K+ channels, especially "apamin," showed anti-arrhythmic effects by alleviation of not only action potential duration shortening but also Ca2+ handling abnormalities, most notably the "Ca2+/voltage uncoupling."
Collapse
Affiliation(s)
- Masayuki Takahashi
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Cardiovascular Medicine, National Hospital Organization Hokkaido Medical Center, Sapporo, Japan
| | - Hisashi Yokoshiki
- Department of Cardiovascular Medicine, Sapporo City General Hospital, Sapporo, Japan
| | - Hirofumi Mitsuyama
- Department of Cardiovascular Medicine, Hokkaido Ohno Memorial Hospital, Sapporo, Japan
| | - Masaya Watanabe
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Taro Temma
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Rui Kamada
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hikaru Hagiwara
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yumi Takahashi
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
52
|
Rebecchi M, Panattoni G, Edoardo B, de Ruvo E, Sciarra L, Politano A, Sgueglia M, Ricagni C, Verbena S, Crescenzi C, Sangiorgi C, Borrelli A, De Luca L, Scarà A, Grieco D, Jacomelli I, Martino A, Calò L. Atrial fibrillation and autonomic nervous system: A translational approach to guide therapeutic goals. J Arrhythm 2021; 37:320-330. [PMID: 33850573 PMCID: PMC8022002 DOI: 10.1002/joa3.12512] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/21/2020] [Accepted: 01/15/2021] [Indexed: 12/19/2022] Open
Abstract
The autonomic nervous system (ANS) is known to play an important role in the genesis and maintenance of atrial fibrillation (AF). Biomolecular and genetic mechanisms, anatomical knowledges with recent diagnostic techniques acquisitions, both invasive and non-invasive, have enabled greater therapeutic goals in patients affected by AF related to ANS imbalance. Catheter ablation of ganglionated plexi (GP) in the left and right atrium has been proposed in varied clinical conditions. Moreover interesting results arise from renal sympathetic denervation and vagal nerve stimulation. Despite all this, in the scenario of ANS modulation translational strategies we necessary must consider the treatment or correction of dynamic factors such as obesity, obstructive sleep apnea, lifestyle, food, and stress. Finally, new antiarrhythmic drugs, gene therapy and "ablatogenomic" could be represent exciting future therapeutic perspectives.
Collapse
Affiliation(s)
| | | | | | | | - Luigi Sciarra
- Department of CardiologyPoliclinico CasilinoRomeItaly
| | | | | | | | - Sara Verbena
- Department of CardiologyPoliclinico CasilinoRomeItaly
| | | | | | | | - Lucia De Luca
- Department of CardiologyPoliclinico CasilinoRomeItaly
| | - Antonio Scarà
- Department of CardiologyPoliclinico CasilinoRomeItaly
| | | | | | | | - Leonardo Calò
- Department of CardiologyPoliclinico CasilinoRomeItaly
| |
Collapse
|
53
|
Darkow E, Nguyen TT, Stolina M, Kari FA, Schmidt C, Wiedmann F, Baczkó I, Kohl P, Rajamani S, Ravens U, Peyronnet R. Small Conductance Ca 2 +-Activated K + (SK) Channel mRNA Expression in Human Atrial and Ventricular Tissue: Comparison Between Donor, Atrial Fibrillation and Heart Failure Tissue. Front Physiol 2021; 12:650964. [PMID: 33868017 PMCID: PMC8047327 DOI: 10.3389/fphys.2021.650964] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/01/2021] [Indexed: 12/25/2022] Open
Abstract
In search of more efficacious and safe pharmacological treatments for atrial fibrillation (AF), atria-selective antiarrhythmic agents have been promoted that target ion channels principally expressed in the atria. This concept allows one to engage antiarrhythmic effects in atria, but spares the ventricles from potentially proarrhythmic side effects. It has been suggested that cardiac small conductance Ca2+-activated K+ (SK) channels may represent an atria-selective target in mammals including humans. However, there are conflicting data concerning the expression of SK channels in different stages of AF, and recent findings suggest that SK channels are upregulated in ventricular myocardium when patients develop heart failure. To address this issue, RNA-sequencing was performed to compare expression levels of three SK channels (KCNN1, KCNN2, and KCNN3) in human atrial and ventricular tissue samples from transplant donor hearts (no cardiac disease), and patients with cardiac disease in sinus rhythm or with AF. In addition, for control purposes expression levels of several genes known to be either chamber-selective or differentially expressed in AF and heart failure were determined. In atria, as compared to ventricle from transplant donor hearts, we confirmed higher expression of KCNN1 and KCNA5, and lower expression of KCNJ2, whereas KCNN2 and KCNN3 were statistically not differentially expressed. Overall expression of KCNN1 was low compared to KCNN2 and KCNN3. Comparing atrial tissue from patients with AF to sinus rhythm samples we saw downregulation of KCNN2 in AF, as previously reported. When comparing ventricular tissue from heart failure patients to non-diseased samples, we found significantly increased ventricular expression of KCNN3 in heart failure, as previously published. The other channels showed no significant difference in expression in either disease. Our results add weight to the view that SK channels are not likely to be an atria-selective target, especially in failing human hearts, and modulators of these channels may prove to have less utility in treating AF than hoped. Whether targeting SK1 holds potential remains to be elucidated.
Collapse
Affiliation(s)
- Elisa Darkow
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Freiburg im Breisgau, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Thong T Nguyen
- Genome Analysis Unit, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Marina Stolina
- Department of Cardiometabolic Disorders, Amgen Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Fabian A Kari
- Medical Center and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Department of Cardiovascular Surgery, University Heart Center Freiburg-Bad Krozingen, Freiburg im Breisgau, Germany
| | - Constanze Schmidt
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
| | - Felix Wiedmann
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Heidelberg University, Heidelberg, Germany
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Freiburg im Breisgau, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg im Breisgau, Germany
| | - Sridharan Rajamani
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Freiburg im Breisgau, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Freiburg im Breisgau, Germany.,Medical Center and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
54
|
Atrial resting membrane potential confers sodium current sensitivity to propafenone, flecainide and dronedarone. Heart Rhythm 2021; 18:1212-1220. [PMID: 33737232 PMCID: PMC8259123 DOI: 10.1016/j.hrthm.2021.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Although atrial fibrillation ablation is increasingly used for rhythm control therapy, antiarrhythmic drugs (AADs) are commonly used, either alone or in combination with ablation. The effectiveness of AADs is highly variable. Previous work from our group suggests that alterations in atrial resting membrane potential (RMP) induced by low Pitx2 expression could explain the variable effect of flecainide. OBJECTIVE The purpose of this study was to assess whether alterations in atrial/cardiac RMP modify the effectiveness of multiple clinically used AADs. METHODS The sodium channel blocking effects of propafenone (300 nM, 1 μM), flecainide (1 μM), and dronedarone (5 μM, 10 μM) were measured in human stem cell-derived cardiac myocytes, HEK293 expressing human NaV1.5, primary murine atrial cardiac myocytes, and murine hearts with reduced Pitx2c. RESULTS A more positive atrial RMP delayed INa recovery, slowed channel inactivation, and decreased peak action potential (AP) upstroke velocity. All 3 AADs displayed enhanced sodium channel block at more positive atrial RMPs. Dronedarone was the most sensitive to changes in atrial RMP. Dronedarone caused greater reductions in AP amplitude and peak AP upstroke velocity at more positive RMPs. Dronedarone evoked greater prolongation of the atrial effective refractory period and postrepolarization refractoriness in murine Langendorff-perfused Pitx2c+/- hearts, which have a more positive RMP compared to wild type. CONCLUSION Atrial RMP modifies the effectiveness of several clinically used AADs. Dronedarone is more sensitive to changes in atrial RMP than flecainide or propafenone. Identifying and modifying atrial RMP may offer a novel approach to enhancing the effectiveness of AADs or personalizing AAD selection.
Collapse
|
55
|
Kraft M, Büscher A, Wiedmann F, L’hoste Y, Haefeli WE, Frey N, Katus HA, Schmidt C. Current Drug Treatment Strategies for Atrial Fibrillation and TASK-1 Inhibition as an Emerging Novel Therapy Option. Front Pharmacol 2021; 12:638445. [PMID: 33897427 PMCID: PMC8058608 DOI: 10.3389/fphar.2021.638445] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia with a prevalence of up to 4% and an upwards trend due to demographic changes. It is associated with an increase in mortality and stroke incidences. While stroke risk can be significantly reduced through anticoagulant therapy, adequate treatment of other AF related symptoms remains an unmet medical need in many cases. Two main treatment strategies are available: rate control that modulates ventricular heart rate and prevents tachymyopathy as well as rhythm control that aims to restore and sustain sinus rhythm. Rate control can be achieved through drugs or ablation of the atrioventricular node, rendering the patient pacemaker-dependent. For rhythm control electrical cardioversion and pharmacological cardioversion can be used. While electrical cardioversion requires fasting and sedation of the patient, antiarrhythmic drugs have other limitations. Most antiarrhythmic drugs carry a risk for pro-arrhythmic effects and are contraindicated in patients with structural heart diseases. Furthermore, catheter ablation of pulmonary veins can be performed with its risk of intraprocedural complications and varying success. In recent years TASK-1 has been introduced as a new target for AF therapy. Upregulation of TASK-1 in AF patients contributes to prolongation of the action potential duration. In a porcine model of AF, TASK-1 inhibition by gene therapy or pharmacological compounds induced cardioversion to sinus rhythm. The DOxapram Conversion TO Sinus rhythm (DOCTOS)-Trial will reveal whether doxapram, a potent TASK-1 inhibitor, can be used for acute cardioversion of persistent and paroxysmal AF in patients, potentially leading to a new treatment option for AF.
Collapse
Affiliation(s)
- Manuel Kraft
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Antonius Büscher
- Clinic for Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Felix Wiedmann
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Yannick L’hoste
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Walter E. Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Hugo A. Katus
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| | - Constanze Schmidt
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
- HCR, Heidelberg Center for Heart Rhythm Disorders, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
56
|
Qi MM, Qian LL, Wang RX. Modulation of SK Channels: Insight Into Therapeutics of Atrial Fibrillation. Heart Lung Circ 2021; 30:1130-1139. [PMID: 33642173 DOI: 10.1016/j.hlc.2021.01.009] [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: 05/27/2020] [Revised: 01/12/2021] [Accepted: 01/31/2021] [Indexed: 11/19/2022]
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia in the world. Although much technological progress in the treatment of AF has been made, there is an urgent need for better treatment of AF due to its high rates of morbidity and mortality. The anti-arrhythmic drugs currently approved for marketing have significant limitations and side effects such as life-threatening ventricular arrhythmias and hypotension. The small conductance Ca2+-activated K+ channels (SK channels) are dependent on intracellular Ca2+ concentrations, which tightly integrate with membrane potential. Given the predominant expression in the atria of many species, including humans, they are now emerging as a therapeutic target for treating AF. This review aimed to illustrate the characteristics and function of SK channels. Moreover, it discussed the regulation of SK channels and their potential as a therapeutic target of AF.
Collapse
Affiliation(s)
- Miao-Miao Qi
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Ling-Ling Qian
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Ru-Xing Wang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China.
| |
Collapse
|
57
|
Chen H, Xia F, Chen X, Cai Y, Jin Z. Ablation of small conductance calcium-activated potassium type-2 channel (SK 2) delays occurrence of bupivacaine-induced cardiotoxicity in isolated mouse hearts. Hum Exp Toxicol 2021; 40:464-471. [PMID: 32909839 DOI: 10.1177/0960327120958102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bupivacaine is frequently used for conducting regional anesthesia. When accidentally injected or excessively absorbed into circulation, bupivacaine can induce severe arrhythmia and potentially lead to cardiac arrest. The specific mechanisms underlying this cardiotoxicity, however, remain to be clarified. We transfected HEK-293 cells to express the small conductance calcium-activated potassium type-2 channel (SK2), and used a whole-cell patch clamp method in order to explore how bupivacaine affected these channels. We subsequently used SK2 knockout mice to explore the relevance of SK2 channels in bupivacaine-induced cardiotoxicity in isolating mouse hearts, mounting them on a Langendorff apparatus, and perfusing them with bupivacaine. Using this system, arrhythmia, asystole, and cardiac functions were monitored. We observed dose-dependent inhibition of SK2 channels by bupivacaine: half-maximal inhibitory concentration (IC50) value = 18.6 μM (95% CI 10.8-32.1). When SK2 knockout (SK2 -/-) or wild-type (WT) mice were perfused with Krebs-Henseleit buffer (KHB), we did not observe any instances of arrhythmia. When SK2 -/- mice or WT were perfused with KHB containing bupivacaine (40 μM), the time to arrhythmia (Tarrhythmia) and time to asystole (Tasystole) were both significantly longer in SK2 -/- mice relative to WT mice (P < 0.001). Similarly, SK2 -/- mice exhibited a significantly longer time to 25%, 50%, and 75% reductions in heart rate (HR) and rate-pressure product (RPP) relative to WT mice following bupivacaine perfusion (P < 0.001). These results reveal that bupivacaine was able to mediate a dose-dependent inhibition of SK2 channels in HEK-293 cells, and deletion of SK2 channels can delay bupivacaine-induced cardiotoxicity in isolated mouse hearts.
Collapse
Affiliation(s)
- H Chen
- Department of Anesthesiology, the 89657First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - F Xia
- Department of Anesthesiology, the 89657First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - X Chen
- Department of Anesthesiology, the 89657First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Y Cai
- Department of Anesthesiology, the 89657First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Z Jin
- Department of Anesthesiology, the 89657First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
58
|
Zhang XD, Thai PN, Lieu DK, Chiamvimonvat N. Cardiac small-conductance calcium-activated potassium channels in health and disease. Pflugers Arch 2021; 473:477-489. [PMID: 33624131 PMCID: PMC7940285 DOI: 10.1007/s00424-021-02535-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/22/2022]
Abstract
Small-conductance Ca2+-activated K+ (SK, KCa2) channels are encoded by KCNN genes, including KCNN1, 2, and 3. The channels play critical roles in the regulation of cardiac excitability and are gated solely by beat-to-beat changes in intracellular Ca2+. The family of SK channels consists of three members with differential sensitivity to apamin. All three isoforms are expressed in human hearts. Studies over the past two decades have provided evidence to substantiate the pivotal roles of SK channels, not only in healthy heart but also with diseases including atrial fibrillation (AF), ventricular arrhythmia, and heart failure (HF). SK channels are prominently expressed in atrial myocytes and pacemaking cells, compared to ventricular cells. However, the channels are significantly upregulated in ventricular myocytes in HF and pulmonary veins in AF models. Interests in cardiac SK channels are further fueled by recent studies suggesting the possible roles of SK channels in human AF. Therefore, SK channel may represent a novel therapeutic target for atrial arrhythmias. Furthermore, SK channel function is significantly altered by human calmodulin (CaM) mutations, linked to life-threatening arrhythmia syndromes. The current review will summarize recent progress in our understanding of cardiac SK channels and the roles of SK channels in the heart in health and disease.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA.
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA.
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA
| | - Deborah K Lieu
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA.
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA.
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
59
|
Saljic A, Jespersen T, Buhl R. Anti-arrhythmic investigations in large animal models of atrial fibrillation. Br J Pharmacol 2021; 179:838-858. [PMID: 33624840 DOI: 10.1111/bph.15417] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/13/2022] Open
Abstract
Atrial fibrillation (AF) constitutes an increasing health problem in the aging population. Animal models reflecting human phenotypes are needed to understand the mechanisms of AF, as well as to test new pharmacological interventions. In recent years, a number of large animal models, primarily pigs, goats, dog and horses have been used in AF research. These animals can to a certain extent recapitulate the human pathophysiological characteristics and serve as valuable tools in investigating new pharmacological interventions for treating AF. This review focuses on anti-arrhythmic investigations in large animals. Initially, spontaneous AF in small and large mammals is discussed. This is followed by a short presentation of frequently used methods for inducing short- and long-term AF. The major focus of the review is on anti-arrhythmic compounds either frequently used in the human clinic (ranolazine, flecainide, vernakalant and amiodarone) or being promising new AF medicine candidates (IK,Ach , ISK,Ca and IK2P blockers).
Collapse
Affiliation(s)
- Arnela Saljic
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Jespersen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
60
|
Fenner MF, Gatta G, Sattler S, Kuiper M, Hesselkilde EM, Adler DMT, Smerup M, Schotten U, Sørensen U, Diness JG, Jespersen T, Verheule S, Van Hunnik A, Buhl R. Inhibition of Small-Conductance Calcium-Activated Potassium Current ( I K,Ca) Leads to Differential Atrial Electrophysiological Effects in a Horse Model of Persistent Atrial Fibrillation. Front Physiol 2021; 12:614483. [PMID: 33633584 PMCID: PMC7900437 DOI: 10.3389/fphys.2021.614483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Background Small-conductance Ca2+-activated K+ (KCa2) channels have been proposed as a possible atrial-selective target to pharmacologically terminate atrial fibrillation (AF) and to maintain sinus rhythm. However, it has been hypothesized that the importance of the KCa2 current—and thereby the efficacy of small-conductance Ca2+-activated K+ current (IK,Ca) inhibition—might be negatively related to AF duration and the extent of AF-induced remodeling. Experimental Approach and Methods To address the hypothesis of the efficacy of IK,Ca inhibition being dependent on AF duration, the anti-arrhythmic properties of the IK,Ca inhibitor NS8593 (5 mg/kg) and its influence on atrial conduction were studied using epicardial high-density contact mapping in horses with persistent AF. Eleven Standardbred mares with tachypacing-induced persistent AF (42 ± 5 days of AF) were studied in an open-chest experiment. Unipolar AF electrograms were recorded and isochronal high-density maps analyzed to allow for the reconstruction of wave patterns and changes in electrophysiological parameters, such as atrial conduction velocity and AF cycle length. Atrial anti-arrhythmic properties and adverse effects of NS8593 on ventricular electrophysiology were evaluated by continuous surface ECG monitoring. Results IK,Ca inhibition by NS8593 administered intravenously had divergent effects on right and left AF complexity and propagation properties in this equine model of persistent AF. Despite global prolongation of AF cycle length, a slowing of conduction in the right atrium led to increased anisotropy and electrical dissociation, thus increasing AF complexity. In contrast, there was no significant change in AF complexity in the LA, and cardioversion of AF was not achieved. Conclusions Intra-atrial heterogeneity in response to IK,Ca inhibition by NS8593 was observed. The investigated dose of NS8593 increased the AF cycle length but was not sufficient to induce cardioversion. In terms of propagation properties during AF, IK,Ca inhibition by NS8593 led to divergent effects in the right and left atrium. This divergent behavior may have impeded the cardioversion success.
Collapse
Affiliation(s)
- Merle Friederike Fenner
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Giulia Gatta
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Stefan Sattler
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marion Kuiper
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Eva Melis Hesselkilde
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ditte M T Adler
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Morten Smerup
- Department of Cardiothoracic Surgery, The Heart Centre, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ulrich Schotten
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | | | | | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sander Verheule
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Arne Van Hunnik
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Rikke Buhl
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
61
|
Auricchio A, Özkartal T, Salghetti F, Neumann L, Pezzuto S, Gharaviri A, Demarchi A, Caputo ML, Regoli F, De Asmundis C, Chierchia GB, Brugada P, Klersy C, Moccetti T, Schotten U, Conte G. Short P-Wave Duration is a Marker of Higher Rate of Atrial Fibrillation Recurrences after Pulmonary Vein Isolation: New Insights into the Pathophysiological Mechanisms Through Computer Simulations. J Am Heart Assoc 2021; 10:e018572. [PMID: 33410337 PMCID: PMC7955300 DOI: 10.1161/jaha.120.018572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Short ECG P-wave duration has recently been demonstrated to be associated with higher risk of atrial fibrillation (AF). The aim of this study was to assess the rate of AF recurrence after pulmonary vein isolation in patients with a short P wave, and to mechanistically elucidate the observation by computer modeling. Methods and Results A total of 282 consecutive patients undergoing a first single-pulmonary vein isolation procedure for paroxysmal or persistent AF were included. Computational models studied the effect of adenosine and sodium conductance on action potential duration and P-wave duration (PWD). About 16% of the patients had a PWD of 110 ms or shorter (median PWD 126 ms, interquartile range, 115 ms-138 ms; range, 71 ms-180 ms). At Cox regression, PWD was significantly associated with AF recurrence (P=0.012). Patients with a PWD <110 ms (hazard ratio [HR], 2.20; 95% CI, 1.24-3.88; P=0.007) and patients with a PWD ≥140 (HR, 1.87, 95% CI, 1.06-3.30; P=0.031) had a nearly 2-fold increase in risk with respect to the other group. In the computational model, adenosine yielded a significant reduction of action potential duration 90 (52%) and PWD (7%). An increased sodium conductance (up to 200%) was robustly accompanied by an increase in conduction velocity (26%), a reduction in action potential duration 90 (28%), and PWD (22%). Conclusions One out of 5 patients referred for pulmonary vein isolation has a short PWD which was associated with a higher rate of AF after the index procedure. Computer simulations suggest that shortening of atrial action potential duration leading to a faster atrial conduction may be the cause of this clinical observation.
Collapse
Affiliation(s)
- Angelo Auricchio
- Division of Cardiology Cardiocentro Ticino Lugano Switzerland.,Center for Computational Medicine in Cardiology Università della Svizzera Italiana Lugano Switzerland
| | - Tardu Özkartal
- Division of Cardiology Cardiocentro Ticino Lugano Switzerland.,Department of Cardiology Ospedale San Giovanni Bellinzona Switzerland
| | - Francesca Salghetti
- Heart Rhythm Management Centre Postgraduate course in Cardiac Electrophysiology and PacingVrije Universiteit BrusselUniversitair Ziekenhuis Brussel Brussels Belgium
| | - Laura Neumann
- Division of Cardiology Cardiocentro Ticino Lugano Switzerland
| | - Simone Pezzuto
- Center for Computational Medicine in Cardiology Università della Svizzera Italiana Lugano Switzerland
| | - Ali Gharaviri
- Center for Computational Medicine in Cardiology Università della Svizzera Italiana Lugano Switzerland
| | - Andrea Demarchi
- Division of Cardiology Cardiocentro Ticino Lugano Switzerland
| | | | - François Regoli
- Division of Cardiology Cardiocentro Ticino Lugano Switzerland
| | - Carlo De Asmundis
- Heart Rhythm Management Centre Postgraduate course in Cardiac Electrophysiology and PacingVrije Universiteit BrusselUniversitair Ziekenhuis Brussel Brussels Belgium
| | - Gian-Battista Chierchia
- Heart Rhythm Management Centre Postgraduate course in Cardiac Electrophysiology and PacingVrije Universiteit BrusselUniversitair Ziekenhuis Brussel Brussels Belgium
| | - Pedro Brugada
- Heart Rhythm Management Centre Postgraduate course in Cardiac Electrophysiology and PacingVrije Universiteit BrusselUniversitair Ziekenhuis Brussel Brussels Belgium
| | - Catherine Klersy
- Service of Clinical Epidemiology & Biometry Fondazione IRCCS Policlinico San Matteo Pavia Italy
| | | | - Ulrich Schotten
- Department of Physiology Cardiovascular Research Institute MaastrichtMaastricht University Maastricht The Netherlands
| | - Giulio Conte
- Division of Cardiology Cardiocentro Ticino Lugano Switzerland
| |
Collapse
|
62
|
The regulation of the small-conductance calcium-activated potassium current and the mechanisms of sex dimorphism in J wave syndrome. Pflugers Arch 2021; 473:491-506. [PMID: 33411079 DOI: 10.1007/s00424-020-02500-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/20/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022]
Abstract
Apamin-sensitive small-conductance calcium-activated potassium (SK) current (IKAS) plays an important role in cardiac repolarization under a variety of physiological and pathological conditions. The regulation of cardiac IKAS relies on SK channel expression, intracellular Ca2+, and interaction between SK channel and intracellular Ca2+. IKAS activation participates in multiple types of arrhythmias, including atrial fibrillation, ventricular tachyarrhythmias, and automaticity and conduction abnormality. Recently, sex dimorphisms in autonomic control have been noticed in IKAS activation, resulting in sex-differentiated action potential morphology and arrhythmogenesis. This review provides an update on the Ca2+-dependent regulation of cardiac IKAS and the role of IKAS on arrhythmias, with a special focus on sex differences in IKAS activation. We propose that sex dimorphism in autonomic control of IKAS may play a role in J wave syndrome.
Collapse
|
63
|
Luo T, Li L, Peng Y, Xie R, Yan N, Fan H, Zhang Q. The MORN domain of Junctophilin2 regulates functional interactions with small-conductance Ca 2+ -activated potassium channel subtype2 (SK2). Biofactors 2021; 47:69-79. [PMID: 31904168 DOI: 10.1002/biof.1608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/17/2019] [Indexed: 01/15/2023]
Abstract
Small-conductance Ca2+ -activated K+ channel subtype2 (SK2) are stable macromolecular complexes that regulate myocardial excitability and Ca2+ homeostasis. Junctophilin-2 (JP2) is a membrane-binding protein, which provides functional crosstalk by physically linking with the cell-surface and intracellular ion channels. We previously demonstrated that the MORN domain of JP2 interacts with SK2 channels. However, the roles of the JP2 MORN domain in regulating the precise subcellular localization and molecular modulation of SK2 have not yet been incompletely understood. In the present study, in vitro and in vivo assays were used to confirm the physical interactions between the SK2 channel and JP2 in H9c2 and HEK293 cells, with a concentration on the association between the C-terminus of SK2 channels and the MORN domain of JP2. Furthermore, the membrane expression of SK2 were found to be significantly impaired by the mutation or knockdown of JP2. Using immunofluorescence staining along with Golgi/early endosome markers, we studied the mechanisms of JP2-regulated SK2 membrane trafficking, which indicates that the JP2 MORN domain is probably necessary for the retrograde trafficking of SK2 channels. The functional study demonstrates that whole cell SK2 current densities recorded from the HEK293 cells co-expressing the JP2-MORN domain with SK2 were significantly augmented, compared with cells expressing SK2 alone. Our findings suggest that the MORN domain of JP2 directly modulates SK2 channel current amplitude and trafficking, through its interaction with an overlapping region of the JP2 MORN domain on the SK2 C-terminus.
Collapse
Affiliation(s)
- Tianxia Luo
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Liren Li
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanghao Peng
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Rongrong Xie
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ningning Yan
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hongkun Fan
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qian Zhang
- Department of Physiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
64
|
Tiffner A, Derler I. Molecular Choreography and Structure of Ca 2+ Release-Activated Ca 2+ (CRAC) and K Ca2+ Channels and Their Relevance in Disease with Special Focus on Cancer. MEMBRANES 2020; 10:E425. [PMID: 33333945 PMCID: PMC7765462 DOI: 10.3390/membranes10120425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Ca2+ ions play a variety of roles in the human body as well as within a single cell. Cellular Ca2+ signal transduction processes are governed by Ca2+ sensing and Ca2+ transporting proteins. In this review, we discuss the Ca2+ and the Ca2+-sensing ion channels with particular focus on the structure-function relationship of the Ca2+ release-activated Ca2+ (CRAC) ion channel, the Ca2+-activated K+ (KCa2+) ion channels, and their modulation via other cellular components. Moreover, we highlight their roles in healthy signaling processes as well as in disease with a special focus on cancer. As KCa2+ channels are activated via elevations of intracellular Ca2+ levels, we summarize the current knowledge on the action mechanisms of the interplay of CRAC and KCa2+ ion channels and their role in cancer cell development.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
65
|
Rahm AK, Wieder T, Gramlich D, Müller ME, Wunsch MN, El Tahry FA, Heimberger T, Weis T, Most P, Katus HA, Thomas D, Lugenbiel P. HDAC2-dependent remodeling of K Ca2.2 (KCNN2) and K Ca2.3 (KCNN3) K + channels in atrial fibrillation with concomitant heart failure. Life Sci 2020; 266:118892. [PMID: 33310041 DOI: 10.1016/j.lfs.2020.118892] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
AIMS Atrial fibrillation (AF) with concomitant heart failure (HF) is associated with prolonged atrial refractoriness. Small-conductance, calcium-activated K+ (KCa, KCNN) channels promote action potential (AP) repolarization. KCNN2 and KCNN3 variants are associated with AF risk. In addition, histone deacetylase (HDAC)-related epigenetic mechanisms have been implicated in AP regulation. We hypothesized that HDAC2-dependent remodeling of KCNN2 and KCNN3 expression contributes to atrial arrhythmogenesis in AF complicated by HF. The objectives were to assess HDAC2 and KCNN2/3 transcript levels in AF/HF patients and in a pig model, and to investigate cellular epigenetic effects of HDAC2 inactivation on KCNN expression. MATERIALS AND METHODS HDAC2 and KCNN2/3 transcript levels were quantified in patients with AF and HF, and in a porcine model of atrial tachypacing-induced AF and reduced left ventricular function. Tachypacing and anti-Hdac2 siRNA treatment were employed in HL-1 atrial myocytes to study effects on KCNN2/3 mRNA and KCa protein abundance. KEY FINDINGS Atrial KCNN2 and KCNN3 expression was reduced in AF/HF patients and in a corresponding pig model. HDAC2 displayed significant downregulation in humans and a tendency towards reduced expression in right atrial tissue of pigs. Tachypacing recapitulated downregulation of Kcnn2/KCa2.2, Kcnn3/KCa2.3 and Hdac2/HDAC2, indicating that high atrial rates trigger epigenetic remodeling mechanisms. Finally, knock-down of Hdac2 in vitro reduced Kcnn3/KCa2.3 expression. SIGNIFICANCE KCNN2/3 and HDAC2 expression is suppressed in AF complicated by HF. Hdac2 directly regulates Kcnn3 mRNA levels in atrial cells. The mechanistic and therapeutic significance of epigenetic electrophysiological effects in AF requires further validation.
Collapse
Affiliation(s)
- Ann-Kathrin Rahm
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Teresa Wieder
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Dominik Gramlich
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Mara Elena Müller
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Maximilian N Wunsch
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Fadwa A El Tahry
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Tanja Heimberger
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Tanja Weis
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Patrick Most
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Patrick Lugenbiel
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; HCR (Heidelberg Center for Heart Rhythm Disorders), University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
66
|
Weisbrod D. Small and Intermediate Calcium Activated Potassium Channels in the Heart: Role and Strategies in the Treatment of Cardiovascular Diseases. Front Physiol 2020; 11:590534. [PMID: 33329039 PMCID: PMC7719780 DOI: 10.3389/fphys.2020.590534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium-activated potassium channels are a heterogeneous family of channels that, despite their different biophysical characteristics, structures, and pharmacological signatures, play a role of transducer between the ubiquitous intracellular calcium signaling and the electric variations of the membrane. Although this family of channels was extensively described in various excitable and non-excitable tissues, an increasing amount of evidences shows their functional role in the heart. This review aims to focus on the physiological role and the contribution of the small and intermediate calcium-activated potassium channels in cardiac pathologies.
Collapse
|
67
|
Varró A, Tomek J, Nagy N, Virág L, Passini E, Rodriguez B, Baczkó I. Cardiac transmembrane ion channels and action potentials: cellular physiology and arrhythmogenic behavior. Physiol Rev 2020; 101:1083-1176. [PMID: 33118864 DOI: 10.1152/physrev.00024.2019] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrhythmias are among the leading causes of mortality. They often arise from alterations in the electrophysiological properties of cardiac cells and their underlying ionic mechanisms. It is therefore critical to further unravel the pathophysiology of the ionic basis of human cardiac electrophysiology in health and disease. In the first part of this review, current knowledge on the differences in ion channel expression and properties of the ionic processes that determine the morphology and properties of cardiac action potentials and calcium dynamics from cardiomyocytes in different regions of the heart are described. Then the cellular mechanisms promoting arrhythmias in congenital or acquired conditions of ion channel function (electrical remodeling) are discussed. The focus is on human-relevant findings obtained with clinical, experimental, and computational studies, given that interspecies differences make the extrapolation from animal experiments to human clinical settings difficult. Deepening the understanding of the diverse pathophysiology of human cellular electrophysiology will help in developing novel and effective antiarrhythmic strategies for specific subpopulations and disease conditions.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
68
|
Lan H, Xu Q, El-Battrawy I, Zhong R, Li X, Lang S, Cyganek L, Borggrefe M, Zhou X, Akin I. Ionic Mechanisms of Disopyramide Prolonging Action Potential Duration in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes From a Patient With Short QT Syndrome Type 1. Front Pharmacol 2020; 11:554422. [PMID: 33154722 PMCID: PMC7586889 DOI: 10.3389/fphar.2020.554422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/16/2020] [Indexed: 11/29/2022] Open
Abstract
Short QT syndrome (SQTS) is associated with tachyarrhythmias and sudden cardiac death. So far, only quinidine has been demonstrated to be effective in patients with SQTS type 1(SQTS1). The aim of this study was to investigate the mechanisms of disopyramide underlying its antiarrhythmic effects in SQTS1 with the N588K mutation in HERG channel. Human-induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) from a patient with SQTS1 and a healthy donor, patch clamp, and calcium imaging measurements were employed to assess the drug effects. Disopyramide prolonged the action potential duration (APD) in hiPSC-CMs from a SQTS1-patient (SQTS1-hiPSC-CMs). In spontaneously beating SQTS1-hiPSC-CMs challenged by carbachol plus epinephrine, disopyramide reduced the arrhythmic events. Disopyramide enhanced the inward L-type calcium channel current (ICa-L), the late sodium channel current (late INa) and the Na/Ca exchanger current (INCX), but it reduced the outward small-conductance calcium-activated potassium channel current (ISK), leading to APD-prolongation. Disopyramide displayed no effects on the rapidly and slowly activating delayed rectifier and ATP-sensitive potassium channel currents. In hiPSC-CMs from the healthy donor, disopyramide reduced peak INa, ICa-L, IKr, and ISK but enhanced late INa and INCX. The results demonstrated that disopyramide may be effective for preventing tachyarrhythmias in SQTS1-patients carrying the N588K mutation in HERG channel by APD-prolongation via enhancing ICa-L, late INa, INCX, and reducing ISK.
Collapse
Affiliation(s)
- Huan Lan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Qiang Xu
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,Department of Histology and Embryology, Southwest Medical University, Luzhou, China
| | - Ibrahim El-Battrawy
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Mannheim, Germany
| | - Rujia Zhong
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Xin Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Siegfried Lang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Mannheim, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.,Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Martin Borggrefe
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Mannheim, Germany
| | - Xiaobo Zhou
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.,First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Mannheim, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Mannheim, Germany
| |
Collapse
|
69
|
Celotto C, Sanchez C, Mountris KA, Laguna P, Pueyo E. SK Channel Block and Adrenergic Stimulation Counteract Acetylcholine-Induced Arrhythmogenic Effects in Human Atria. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:2303-2306. [PMID: 33018468 DOI: 10.1109/embc44109.2020.9175886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
There is increasing evidence on the role of the autonomic nervous system in the pathogenesis of atrial fibrillation. Interventions targeting autonomic modulation of atrial electrical activity have been shown to reduce the incidence of atrial arrhythmias. Additionally, recent investigations have proved that pharmacological therapies inhibiting small-conductance calcium-activated potassium (SK) channels are able to lessen cholinergic effects in the atria.In this study we use computational modeling and simulation to test individual and combined effects of SK channel block and adrenergic stimulation in counteracting detrimental effects induced by the parasympathetic neurotransmitter acetylcholine (ACh) on human atrial electrophysiology. Cell and tissue models are built that incorporate descriptions of SK channels as well as of isoproterenol (Iso)- and ACh-mediated regulation of the atrial action potential (AP). Three different cellular AP models, representing a range of physiological AP shapes, are considered and both homogeneous and heterogeneous ACh distributions in atrial tissue are simulated.At the cellular level, SK channel block is demonstrated to partially revert shortening of AP duration (APD) mediated by ACh at various doses, whereas 1 µM Iso has a variable response depending on the AP shape. The combination of SK block and Iso is in all cases able to take APD back to baseline levels, recovering between 82% and 120% of the APD shortening induced by 0.1 µM ACh. At the tissue level, SK block and Iso alone or in combination do not exert remarkable effects on conduction velocity, but the combination of the two is able to notably prolong the ACh-mediated APD shortening, thus increasing the wavelength for reentry.In conclusion, the results from this study support the combination of SK channel block and adrenergic stimulation as a potential option to counteract parasympathetically-mediated proarrhythmic effects in the human atria.
Collapse
|
70
|
Mapping TRPM7 Function by NS8593. Int J Mol Sci 2020; 21:ijms21197017. [PMID: 32977698 PMCID: PMC7582524 DOI: 10.3390/ijms21197017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
The transient receptor potential cation channel, subfamily M, member 7 (TRPM7) is a ubiquitously expressed membrane protein, which forms a channel linked to a cytosolic protein kinase. Genetic inactivation of TRPM7 in animal models uncovered the critical role of TRPM7 in early embryonic development, immune responses, and the organismal balance of Zn2+, Mg2+, and Ca2+. TRPM7 emerged as a new therapeutic target because malfunctions of TRPM7 have been associated with anoxic neuronal death, tissue fibrosis, tumour progression, and giant platelet disorder. Recently, several laboratories have identified pharmacological compounds allowing to modulate either channel or kinase activity of TRPM7. Among other small molecules, NS8593 has been defined as a potent negative gating regulator of the TRPM7 channel. Consequently, several groups applied NS8593 to investigate cellular pathways regulated by TRPM7. Here, we summarize the progress in this research area. In particular, two notable milestones have been reached in the assessment of TRPM7 druggability. Firstly, several laboratories demonstrated that NS8593 treatment reliably mirrors prominent phenotypes of cells manipulated by genetic inactivation of TRPM7. Secondly, it has been shown that NS8593 allows us to probe the therapeutic potential of TRPM7 in animal models of human diseases. Collectively, these studies employing NS8593 may serve as a blueprint for the preclinical assessment of TRPM7-targeting drugs.
Collapse
|
71
|
Saljic A, Muthukumarasamy KM, la Cour JM, Boddum K, Grunnet M, Berchtold MW, Jespersen T. Impact of arrhythmogenic calmodulin variants on small conductance Ca 2+ -activated K + (SK3) channels. Physiol Rep 2020; 7:e14210. [PMID: 31587513 PMCID: PMC6778599 DOI: 10.14814/phy2.14210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Calmodulin (CaM) is a ubiquitous Ca2+‐sensing protein regulating many important cellular processes. Several CaM‐associated variants have been identified in a small group of patients with cardiac arrhythmias. The mechanism remains largely unknown, even though a number of ion channels, including the ryanodine receptors and the L‐type calcium channels have been shown to be functionally affected by the presence of mutant CaM. CaM is constitutively bound to the SK channel, which underlies the calcium‐gated ISK contributing to cardiac repolarization. The CaM binding to SK channels is essential for gating, correct assembly, and membrane expression. To elucidate the effect of nine different arrhythmogenic CaM variants on SK3 channel function, HEK293 cells stably expressing SK3 were transiently co‐transfected with CaMWT or variant and whole‐cell patch‐clamp recordings were performed with a calculated free Ca2+ concentration of 400 nmol/L. MDCK cells were transiently transfected with SK3 and/or CaMWT or variant to address SK3 and CaM localization by immunocytochemistry. The LQTS‐associated variants CaMD96V, CaMD130G, and CaMF142L reduced ISK,Ca compared with CaMWT (P < 0.01, P < 0.001, and P < 0.05, respectively). The CPVT associated variant CaMN54I also reduced the ISK,Ca (P < 0.05), which was linked to an accumulation of SK3/CaMN54I channel complexes in intracellular compartments (P < 0.05). The CPVT associated variants, CaMA103V and CaMD132E only revealed a tendency toward reduced current, while the variants CaMF90L and CaMN98S, causing LQTS syndrome, did not have any impact on ISK,Ca. In conclusion, we found that the arrhythmogenic CaM variants CaMN54I, CaMD96V, CaMD130G, and CaMF142L significantly down‐regulate the SK3 channel current, but with distinct mechanism.
Collapse
Affiliation(s)
- Arnela Saljic
- Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kalai Mangai Muthukumarasamy
- Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Marstrand la Cour
- Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Boddum
- Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Martin Werner Berchtold
- Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Jespersen
- Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
72
|
Ni H, Fogli Iseppe A, Giles WR, Narayan SM, Zhang H, Edwards AG, Morotti S, Grandi E. Populations of in silico myocytes and tissues reveal synergy of multiatrial-predominant K + -current block in atrial fibrillation. Br J Pharmacol 2020; 177:4497-4515. [PMID: 32667679 DOI: 10.1111/bph.15198] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 06/22/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Pharmacotherapy of atrial fibrillation (AF), the most common cardiac arrhythmia, remains unsatisfactory due to low efficacy and safety concerns. New therapeutic strategies target atrial-predominant ion-channels and involve multichannel block (poly)therapy. As AF is characterized by rapid and irregular atrial activations, compounds displaying potent antiarrhythmic effects at fast and minimal effects at slow rates are desirable. We present a novel systems pharmacology framework to quantitatively evaluate synergistic anti-AF effects of combined block of multiple atrial-predominant K+ currents (ultra-rapid delayed rectifier K+ current, IKur , small conductance Ca2+ -activated K+ current, IKCa , K2P 3.1 2-pore-domain K+ current, IK2P ) in AF. EXPERIMENTAL APPROACH We constructed experimentally calibrated populations of virtual atrial myocyte models in normal sinus rhythm and AF-remodelled conditions using two distinct, well-established atrial models. Sensitivity analyses on our atrial populations was used to investigate the rate dependence of action potential duration (APD) changes due to blocking IKur , IK2P or IKCa and interactions caused by blocking of these currents in modulating APD. Block was simulated in both single myocytes and one-dimensional tissue strands to confirm insights from the sensitivity analyses and examine anti-arrhythmic effects of multi-atrial-predominant K+ current block in single cells and coupled tissue. KEY RESULTS In both virtual atrial myocytes and tissues, multiple atrial-predominant K+ -current block promoted favourable positive rate-dependent APD prolongation and displayed positive rate-dependent synergy, that is, increasing synergistic antiarrhythmic effects at fast pacing versus slow rates. CONCLUSION AND IMPLICATIONS Simultaneous block of multiple atrial-predominant K+ currents may be a valuable antiarrhythmic pharmacotherapeutic strategy for AF.
Collapse
Affiliation(s)
- Haibo Ni
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Alex Fogli Iseppe
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Wayne R Giles
- Faculties of Kinesiology and Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sanjiv M Narayan
- Division of Cardiology, Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, UK
| | - Andrew G Edwards
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Stefano Morotti
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
73
|
Saljic A, Soattin L, Trachsel DS, Boddum K, Jespersen T. In vivo knockdown of SK3 channels using antisense oligonucleotides protects against atrial fibrillation in rats. J Mol Cell Cardiol 2020; 147:18-26. [PMID: 32768409 DOI: 10.1016/j.yjmcc.2020.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 07/06/2020] [Accepted: 07/28/2020] [Indexed: 02/02/2023]
Abstract
INTRODUCTION GapmeRs are oligonucleotides that bind to a specific RNA sequence and thereby affecting posttranscriptional gene regulation. They therefore hold the potential to manipulate targets where current pharmacological modulators are inefficient or exhibit adverse side effects. Here, we show that a treatment with a GapmeR, mediating knockdown of small conductance Ca2+-activated K+ channels (SK3), has an in vivo protective effect against atrial fibrillation (AF) in rats. MATERIAL AND METHODS A unique SK3-GapmeR design was selected after thorough in vitro evaluation. 22 rats were randomly assigned to receive either 50 mg/kg SK3-GapmeR or vehicle subcutaneously once a week for two weeks. Langendorff experiments were performed seven days after the last injection, where action potential duration (APD90), effective refractory period (ERP) and AF propensity were investigated. SK3 channel activity was evaluated using the SK channel blocker, ICA (N-(pyridin-2-yl)-4-(pyridine-2-yl)thiazol-2-amine). SK3 protein expression was assessed by Western Blot. RESULTS The designed GapmeR effectively down-regulate the SK3 protein expression in the heart (48% downregulation, p = 0.0095) and did indeed protect against AF. Duration of AF episodes elicited by burst pacing in the rats treated with SK3-GapmeR was reduced 78% compared to controls (3.7 s vs. 16.8 s, p = 0.0353). The number of spontaneous AF episodes were decreased by 68% in the SK3-GapmeR group (39 episodes versus 123 in the control group, respectively) and were also significantly shorter in duration (7.2 s versus 29.7 s in the control group, p = 0.0327). Refractoriness was not altered at sinus rhythm, but ERP prolongation following ICA application was blunted in the SK3-GapmeR group. CONCLUSION The selected GapmeR silenced the cardiac SK3 channels, thereby preventing AF in rats. Thus, GapmeR technology can be applied as an experimental tool of downregulation of cardiac proteins and could potentially offer a novel modality for treatment of cardiac diseases.
Collapse
Affiliation(s)
- Arnela Saljic
- Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Luca Soattin
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom; Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Dagmar S Trachsel
- Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Kim Boddum
- Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Thomas Jespersen
- Laboratory of Cardiac Physiology, Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
74
|
Impact of I SK Voltage and Ca 2+/Mg 2+-Dependent Rectification on Cardiac Repolarization. Biophys J 2020; 119:690-704. [PMID: 32668235 DOI: 10.1016/j.bpj.2020.06.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 10/24/2022] Open
Abstract
Cardiac small conductance Ca2+-activated K+ (SK) channels are activated solely by Ca2+, but the SK current (ISK) is inwardly rectified. However, the impact of inward rectification in shaping action potentials (APs) in ventricular cardiomyocytes under β-adrenergic stimulation or in disease states remains undefined. Two processes underlie this inward rectification: an intrinsic rectification caused by an electrostatic energy barrier from positively charged amino acids at the inner pore and a voltage-dependent Ca2+/Mg2+ block. Thus, Ca2+ has a biphasic effect on ISK, activating at low [Ca2+] yet inhibiting ISK at high [Ca2+]. We examined the effect of ISK rectification on APs in rat cardiomyocytes by simultaneously recording whole-cell apamin-sensitive currents and Ca2+ transients during an AP waveform and developed a computer model of SK channels with rectification features. The typical profile of ISK during AP clamp included an initial peak (mean 1.6 pA/pF) followed by decay to the point that submembrane [Ca2+] reached ∼10 μM. During the rest of the AP stimulus, ISK either plateaued or gradually increased as the cell repolarized and submembrane [Ca2+] decreased further. We used a six-state gating model combined with intrinsic and Ca2+/Mg2+-dependent rectification to simulate ISK and investigated the relative contributions of each type of rectification to AP shape. This SK channel model replicates key features of ISK recording during AP clamp showing that intrinsic rectification limits ISK at high Vm during the early and plateau phase of APs. Furthermore, the initial rise of Ca2+ transients activates, but higher [Ca2+] blocks SK channels, yielding a transient outward-like ISK trajectory. During the decay phase of Ca2+, the Ca2+-dependent block is released, causing ISK to rise again and contribute to repolarization. Therefore, ISK is an important repolarizing current, and the rectification characteristics of an SK channel determine its impact on early, plateau, and repolarization phases of APs.
Collapse
|
75
|
Gal P, Klaassen ES, Bergmann KR, Saghari M, Burggraaf J, Kemme MJB, Sylvest C, Sørensen U, Bentzen BH, Grunnet M, Diness JG, Edvardsson N. First Clinical Study with AP30663 - a K Ca 2 Channel Inhibitor in Development for Conversion of Atrial Fibrillation. Clin Transl Sci 2020; 13:1336-1344. [PMID: 32725783 PMCID: PMC7719388 DOI: 10.1111/cts.12835] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/22/2020] [Indexed: 11/30/2022] Open
Abstract
Pharmacological cardioversion of atrial fibrillation (AF) is frequently inefficacious. AP30663, a small conductance Ca2+ activated K+ (KCa2) channel blocker, prolonged the atrial effective refractory period in preclinical studies and subsequently converted AF into normal sinus rhythm. This first‐in‐human study evaluated the safety and tolerability, and pharmacokinetic (PK) and pharmacodynamic (PD) effects were explored. Forty‐seven healthy male volunteers (23.7 ± 3.0 years) received AP30663 intravenously in ascending doses. Due to infusion site reactions, changes to the formulation and administration were implemented in the latter 24 volunteers. Extractions from a 24‐hour continuous electrocardiogram were used to evaluate the PD effect of AP30663. Data were analyzed with a repeated measure analysis of covariance, noncompartmental analysis, and concentration‐effect analysis. In total, 33 of 34 adverse events considered related to AP30663 exposure were related to the infusion site, mild in severity, and temporary in nature, although full recovery took up to 110 days. After formulation and administration changes, the local infusion site reaction remained, but the median duration was shorter despite higher dose levels. AP30663 displayed a less than dose proportional increase in peak plasma concentration (Cmax) and a terminal half‐life of around 5 hours. In healthy volunteers, no effect of AP30663 was observed on electrocardiographic parameters, other than a concentration‐dependent effect on the corrected QT Fridericia’s formula interval (+18.8 ± 4.3 ms for the highest dose level compared with time matched placebo). In conclusion, administration of AP30663, a novel KCa2 channel inhibitor, was safe and well‐tolerated systemically in humans, supporting further development in patients with AF undergoing cardioversion.
Collapse
Affiliation(s)
- Pim Gal
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | - Mahdi Saghari
- Centre for Human Drug Research, Leiden, The Netherlands
| | - Jacobus Burggraaf
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Centre, Leiden, The Netherlands.,Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | | | | | | | | | | | | | - Nils Edvardsson
- Acesion Pharma ApS, Copenhagen, Denmark.,Department of Molecular and Clinical Medicine/Cardiology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
76
|
Fontaine D, Figiel S, Félix R, Kouba S, Fromont G, Mahéo K, Potier-Cartereau M, Chantôme A, Vandier C. Roles of endogenous ether lipids and associated PUFAs in the regulation of ion channels and their relevance for disease. J Lipid Res 2020; 61:840-858. [PMID: 32265321 PMCID: PMC7269763 DOI: 10.1194/jlr.ra120000634] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Indexed: 12/16/2022] Open
Abstract
Ether lipids (ELs) are lipids characterized by the presence of either an ether linkage (alkyl lipids) or a vinyl ether linkage [i.e., plasmalogens (Pls)] at the sn1 position of the glycerol backbone, and they are enriched in PUFAs at the sn2 position. In this review, we highlight that ELs have various biological functions, act as a reservoir for second messengers (such as PUFAs) and have roles in many diseases. Some of the biological effects of ELs may be associated with their ability to regulate ion channels that control excitation-contraction/secretion/mobility coupling and therefore cell physiology. These channels are embedded in lipid membranes, and lipids can regulate their activities directly or indirectly as second messengers or by incorporating into membranes. Interestingly, ELs and EL-derived PUFAs have been reported to play a key role in several pathologies, including neurological disorders, cardiovascular diseases, and cancers. Investigations leading to a better understanding of their mechanisms of action in pathologies have opened a new field in cancer research. In summary, newly identified lipid regulators of ion channels, such as ELs and PUFAs, may represent valuable targets to improve disease diagnosis and advance the development of new therapeutic strategies for managing a range of diseases and conditions.
Collapse
Affiliation(s)
- Delphine Fontaine
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sandy Figiel
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Romain Félix
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sana Kouba
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Gaëlle Fromont
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Department of Pathology, CHRU Bretonneau, F-37044 Tours CEDEX 9, France
| | - Karine Mahéo
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | | | - Aurélie Chantôme
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | - Christophe Vandier
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France. mailto:
| |
Collapse
|
77
|
Diness JG, Abildgaard L, Bomholtz SH, Skarsfeldt MA, Edvardsson N, Sørensen US, Grunnet M, Bentzen BH. Inhibition of K Ca2 Channels Decreased the Risk of Ventricular Arrhythmia in the Guinea Pig Heart During Induced Hypokalemia. Front Pharmacol 2020; 11:749. [PMID: 32508659 PMCID: PMC7251152 DOI: 10.3389/fphar.2020.00749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/06/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Hypokalemia reduces the cardiac repolarization reserve. This prolongs the QT-interval and increases the risk of ventricular arrhythmia; a risk that is exacerbated by administration of classical class 3 anti-arrhythmic agents.Small conductance Ca2+-activated K+-channels (KCa2) are a promising new atrial selective target for treatment of atrial fibrillation. Under physiological conditions KCa2 plays a minor role in ventricular repolarization. However, this might change under hypokalemia because of concomitant increases in ventriculay -60r intracellur Ca2+. PURPOSE To study the effects of pharmacological KCa2 channel inhibition by the compounds AP14145, ICA, or AP30663 under hypokalemic conditions as compared to dofetilide and hypokalemia alone time-matched controls (TMC). METHODS The current at +10 mV was compared in HEK293 cells stably expressing KCa2.3 perfused first with normo- and then hypokalemic solutions (4 mM K+ and 2.5 mM K+, respectively). Guinea pig hearts were isolated and perfused with normokalemic (4 mM K+) Krebs-Henseleit solution, followed by perfusion with drug or vehicle control. The perfusion was then changed to hypokalemic solution (2.5 mM K+) in presence of drug. 30 animals were randomly assigned to 5 groups: ICA, AP14145, AP30663, dofetilide, or TMC. QT-interval, the interval from the peak to the end of the T wave (Tp-Te), ventricular effective refractory period (VERP), arrhythmia score, and ventricular fibrillation (VF) incidence were recorded. RESULTS Hypokalemia slightly increased KCa2.3 current compared to normokalemia. Application of KCa2 channel inhibitors and dofetilide prolonged the QT interval corrected for heart rate. Dofetilide, but none of the KCa2 channel inhibitors increased Tp-Te during hypokalemia. During hypokalemia 4/6 hearts in the TMC group developed VF (two spontaneously, two by S1S2 stimulation) whereas 5/6 hearts developed VF in the dofetilide group (two spontaneously, three by S1S2 stimulation). In comparison, 0/6, 1/6, and 1/6 hearts developed VF when treated with the KCa2 channel inhibitors AP30663, ICA, or AP14145, respectively. CONCLUSION Hypokalemia was associated with an increased incidence of VF, an effect that also seen in the presence of dofetilide. In comparison, the structurally and functionally different KCa2 channel inhibitors, ICA, AP14145, and AP30663 protected the heart from hypokalemia induced VF. These results support that KCa2 inhibition may be associated with a better safety and tolerability profile than dofetilide.
Collapse
Affiliation(s)
| | | | - Sofia Hammami Bomholtz
- Acesion Pharma, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mark Alexander Skarsfeldt
- Acesion Pharma, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nils Edvardsson
- Acesion Pharma, Copenhagen, Denmark
- Department of Molecular and Clinical Medicine/Cardiology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Bo Hjorth Bentzen
- Acesion Pharma, Copenhagen, Denmark
- Department of Molecular and Clinical Medicine/Cardiology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
78
|
Larupa Santos J, Rodríguez I, S. Olesen M, Hjorth Bentzen B, Schmitt N. Investigating gene-microRNA networks in atrial fibrillation patients with mitral valve regurgitation. PLoS One 2020; 15:e0232719. [PMID: 32392228 PMCID: PMC7213724 DOI: 10.1371/journal.pone.0232719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/20/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is predicted to affect around 17.9 million individuals in Europe by 2060. The disease is associated with severe electrical and structural remodelling of the heart, and increased the risk of stroke and heart failure. In order to improve treatment and find new drug targets, the field needs to better comprehend the exact molecular mechanisms in these remodelling processes. OBJECTIVES This study aims to identify gene and miRNA networks involved in the remodelling of AF hearts in AF patients with mitral valve regurgitation (MVR). METHODS Total RNA was extracted from right atrial biopsies from patients undergoing surgery for mitral valve replacement or repair with AF and without history of AF to test for differentially expressed genes and miRNAs using RNA-sequencing and miRNA microarray. In silico predictions were used to construct a mRNA-miRNA network including differentially expressed mRNAs and miRNAs. Gene and chromosome enrichment analysis were used to identify molecular pathways and high-density AF loci. RESULTS We found 644 genes and 43 miRNAs differentially expressed in AF patients compared to controls. From these lists, we identified 905 pairs of putative miRNA-mRNA interactions, including 37 miRNAs and 295 genes. Of particular note, AF-associated miR-130b-3p, miR-338-5p and miR-208a-3p were differentially expressed in our AF tissue samples. These miRNAs are predicted regulators of several differentially expressed genes associated with cardiac conduction and fibrosis. We identified two high-density AF loci in chromosomes 14q11.2 and 6p21.3. CONCLUSIONS AF in MVR patients is associated with down-regulation of ion channel genes and up-regulation of extracellular matrix genes. Other AF related genes are dysregulated and several are predicted to be targeted by miRNAs. Our novel miRNA-mRNA regulatory network provides new insights into the mechanisms of AF.
Collapse
Affiliation(s)
- Joana Larupa Santos
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Ismael Rodríguez
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Morten S. Olesen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Department of Cardiology, Laboratory for Molecular Cardiology, The Heart Centre, Rigshospitalet, University Hospital of Copenhagen, Copenhagen Ø, Denmark
| | - Bo Hjorth Bentzen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Nicole Schmitt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
79
|
Yang M, Wang Y, Xiong X, Xie B, Liu J, Yin J, Zi L, Wang X, Tang Y, Huang C, Zhao Q. SK4 calcium-activated potassium channels activated by sympathetic nerves enhances atrial fibrillation vulnerability in a canine model of acute stroke. Heliyon 2020; 6:e03928. [PMID: 32420493 PMCID: PMC7215192 DOI: 10.1016/j.heliyon.2020.e03928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/24/2020] [Accepted: 05/01/2020] [Indexed: 12/02/2022] Open
Abstract
Background New-onset atrial fibrillation (AF) is common in patients with acute stroke (AS). Studies have shown that intermediate-conductance calcium-activated potassium channel channels (SK4) play an important role in cardiomyocyte automaticity. The aim of this study was to investigate the effects of SK4 on AF vulnerability in dogs with AS. Experimental Eighteen dogs were randomly divided into a control group, AS group and left stellate ganglion ablation (LSGA) group. In the control group, dogs received craniotomy without right middle cerebral artery occlusion (MCAO). AS dogs were established using a cerebral ischemic model with right MCAO. LSGA dogs underwent MCAO, and LSGA was performed. Results Three days later, the dispersion of the effective refractory period (dERP) and AF vulnerability in the AS group were significantly increased compared with those in the control group and LSGA group. However, no significant difference in dERP and AF vulnerability was found between the control group and the LSGA group. The SK4 inhibitor (TRAM-34) completely inhibited the inducibility of AF in AS dogs. SK4 expression and levels of noradrenaline (NE), β1-AR, p38 and c-Fos in the atrium were higher in the AS dogs than in the control group or LSGA group. However, no significant difference in SK4 expression or levels of NE, β1-AR, p38 and c-Fos in the left atrium was observed between the control group and LSGA group. Conclusion SK4 plays a key role in AF vulnerability in a canine model with AS. The effects of LSGA on AF vulnerability were associated with the p38 signaling pathways.
Collapse
Affiliation(s)
- Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Youcheng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, 430060, PR China
| | - Baojun Xie
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan City, 430060, PR China
| | - Jia Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, 430060, PR China
| | - Junkui Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Liuliu Zi
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Corresponding author.
| |
Collapse
|
80
|
Bentzen BH, Bomholtz SH, Simó-Vicens R, Folkersen L, Abildgaard L, Speerschneider T, Muthukumarasamy KM, Edvardsson N, Sørensen US, Grunnet M, Diness JG. Mechanisms of Action of the KCa2-Negative Modulator AP30663, a Novel Compound in Development for Treatment of Atrial Fibrillation in Man. Front Pharmacol 2020; 11:610. [PMID: 32477117 PMCID: PMC7232560 DOI: 10.3389/fphar.2020.00610] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/20/2020] [Indexed: 12/17/2022] Open
Abstract
Aims Small conductance Ca2+-activated K+ channels (SK channels, KCa2) are a new target for treatment of atrial fibrillation (AF). AP30663 is a small molecule inhibitor of KCa2 channels that is currently in clinical development for treatment of AF. The aim of this study is to present the electrophysiological profile and mechanism of action of AP30663 and its efficacy in prolonging atrial refractoriness in rodents, and by bioinformatic analysis investigate if genetic variants in KCNN2 or KCNN3 influence the expression level of these in human heart tissue. Methods and Results Whole-cell and inside-out patch-clamp recordings of heterologously expressed KCa2 channels revealed that AP30663 inhibits KCa2 channels with minor effects on other relevant cardiac ion channels. AP30663 modulates the KCa2.3 channel by right-shifting the Ca2+-activation curve. In isolated guinea pig hearts AP30663 significantly prolonged the atrial effective refractory period (AERP) with minor effects on the QT-interval corrected for heart rate. Similarly, in anaesthetized rats 5 and 10 mg/kg of AP30663 changed the AERP to 130.7±5.4% and 189.9±18.6 of baseline values. The expression quantitative trait loci analyses revealed that the genome wide association studies for AF SNP rs13376333 in KCNN3 is associated with increased mRNA expression of KCNN3 in human atrial appendage tissue. Conclusions AP30663 is a novel negative allosteric modulator of KCa2 channels that concentration-dependently prolonged rodent atrial refractoriness with minor effects on the QT-interval. Moreover, AF associated SNPs in KCNN3 influence KCNN3 mRNA expression in human atrial tissue. These properties support continued development of AP30663 for treatment of AF in man.
Collapse
Affiliation(s)
- Bo Hjorth Bentzen
- Acesion Pharma, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofia Hammami Bomholtz
- Acesion Pharma, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rafel Simó-Vicens
- Acesion Pharma, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Folkersen
- Institute of Biological Psychiatry, Sankt Hans Hospital, Roskilde, Denmark
| | | | - Tobias Speerschneider
- Acesion Pharma, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kalai Mangai Muthukumarasamy
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nils Edvardsson
- Acesion Pharma, Copenhagen, Denmark.,Department of Molecular and Clinical Medicine/Cardiology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | | |
Collapse
|
81
|
Citerni C, Kirchhoff J, Olsen LH, Sattler SM, Grunnet M, Edvardsson N, Bentzen BH, Diness JG. Inhibition of K Ca2 and K v11.1 Channels in Pigs With Left Ventricular Dysfunction. Front Pharmacol 2020; 11:556. [PMID: 32435191 PMCID: PMC7219273 DOI: 10.3389/fphar.2020.00556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/14/2020] [Indexed: 12/17/2022] Open
Abstract
Background Inhibition of KCa2 channels, conducting IKCa, can convert atrial fibrillation (AF) to sinus rhythm and protect against its induction. IKCa inhibition has been shown to possess functional atrial selectivity with minor effects on ventricles. Under pathophysiological conditions with ventricular remodeling, however, inhibiting IKCa can exhibit both proarrhythmic and antiarrhythmic ventricular effects. The aim of this study was to evaluate the effects of the IKCa inhibitor AP14145, when given before or after the IKr blocker dofetilide, on cardiac function and ventricular proarrhythmia markers in pigs with or without left ventricular dysfunction (LVD). Methods Landrace pigs were randomized into an AF group (n = 6) and two control groups: SHAM1 (n = 8) and SHAM2 (n = 4). AF pigs were atrially tachypaced (A-TP) for 43 ± 4 days until sustained AF and LVD developed. A-TP and SHAM1 pigs received 20 mg/kg AP14145 followed by 100 µg/kg dofetilide whereas SHAM2 pigs received the same drugs in the opposite order. Proarrhythmic markers such as short-term variability of QT (STVQT) and RR (STVRR) intervals, and the number of premature ventricular complexes (PVCs) were measured at baseline and after administration of drugs. The influence on cardiac function was assessed by measuring cardiac output, stroke volume, and relevant echocardiographic parameters. Results IKCa inhibition by AP14145 did not increase STVQT or STVRR in any of the pigs. IKr inhibition by dofetilide markedly increased STVQT in the A-TP pigs, but not in SHAM operated pigs. Upon infusion of AP14145 the number of PVCs decreased or remained unchanged both when AP14145 was infused after baseline and after dofetilide. Conversely, the number of PVCs increased or remained unchanged upon dofetilide infusion. Neither AP14145 nor dofetilide affected relevant echocardiographic parameters, cardiac output, or stroke volume in any of the groups. Conclusion IKCa inhibition with AP14145 was not proarrhythmic in healthy pigs, or in the presence of LVD resulting from A-TP. In pigs already challenged with 100 µg/kg dofetilide there were no signs of proarrhythmia when 20 mg/kg AP14145 were infused. KCa2 channel inhibition did not affect cardiac function, implying that KCa2 inhibitors can be administered safely also in the presence of LV dysfunction.
Collapse
Affiliation(s)
- Carlotta Citerni
- Biomedical Institute, University of Copenhagen, Copenhagen, Denmark.,Acesion Pharma, Copenhagen, Denmark
| | | | - Lisbeth Høier Olsen
- Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark
| | - Stefan Michael Sattler
- Biomedical Institute, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiology, Heart Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Nils Edvardsson
- Acesion Pharma, Copenhagen, Denmark.,Department of Molecular and Clinical Medicine, Sahlgrenska Academy at Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bo Hjorth Bentzen
- Biomedical Institute, University of Copenhagen, Copenhagen, Denmark.,Acesion Pharma, Copenhagen, Denmark
| | | |
Collapse
|
82
|
Diness JG, Kirchhoff JE, Speerschneider T, Abildgaard L, Edvardsson N, Sørensen US, Grunnet M, Bentzen BH. The K Ca2 Channel Inhibitor AP30663 Selectively Increases Atrial Refractoriness, Converts Vernakalant-Resistant Atrial Fibrillation and Prevents Its Reinduction in Conscious Pigs. Front Pharmacol 2020; 11:159. [PMID: 32180722 PMCID: PMC7059611 DOI: 10.3389/fphar.2020.00159] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/07/2020] [Indexed: 12/20/2022] Open
Abstract
AIMS To describe the effects of the KCa2 channel inhibitor AP30663 in pigs regarding tolerability, cardiac electrophysiology, pharmacokinetics, atrial functional selectivity, effectiveness in cardioversion of tachy-pacing induced vernakalant-resistant atrial fibrillation (AF), and prevention of reinduction of AF. METHODS AND RESULTS Six healthy pigs with implanted pacemakers and equipped with a Holter monitor were used to compare the effects of increasing doses (0, 5, 10, 15, 20, and 25 mg/kg) of AP30663 on the right atrial effective refractory period (AERP) and on various ECG parameters, including the QT interval. Ten pigs with implanted neurostimulators were long-term atrially tachypaced (A-TP) until sustained vernakalant-resistant AF was present. 20 mg/kg AP30663 was tested to discover if it could successfully convert vernakalant-resistant AF to sinus rhythm (SR) and protect against reinduction of AF. Seven anesthetized pigs were used for pharmacokinetic experiments. Two pigs received an infusion of 20 mg/kg AP30663 over 60 min while five pigs received 5 mg/kg AP30663 over 30 min. Blood samples were collected before, during, and after infusion on AP30663. AP30663 was well-tolerated and prominently increased the AERP in pigs with little effect on ventricular repolarization. Furthermore, it converted A-TP induced AF that had become unresponsive to vernakalant, and it prevented reinduction of AF in pigs. Both a >30 ms increase of the AERP and conversion of AF occurred in different pigs at a free plasma concentration level of around 1.0-1.4 µM of AP30663, which was achieved at a dose level of 5 mg/kg. CONCLUSION AP30663 has shown properties in animals that would be of clinical interest in man.
Collapse
Affiliation(s)
| | | | - Tobias Speerschneider
- Department of In Vivo Pharmacology, Acesion Pharma, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lea Abildgaard
- Department of In Vivo Pharmacology, Acesion Pharma, Copenhagen, Denmark
| | - Nils Edvardsson
- Department of In Vivo Pharmacology, Acesion Pharma, Copenhagen, Denmark
- Department of Molecular and Clinical Medicine/Cardiology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulrik S. Sørensen
- Department of In Vivo Pharmacology, Acesion Pharma, Copenhagen, Denmark
| | - Morten Grunnet
- Department of In Vivo Pharmacology, Acesion Pharma, Copenhagen, Denmark
| | - Bo Hjorth Bentzen
- Department of In Vivo Pharmacology, Acesion Pharma, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
83
|
Kouba S, Braire J, Félix R, Chantôme A, Jaffrès PA, Lebreton J, Dubreuil D, Pipelier M, Zhang X, Trebak M, Vandier C, Mathé-Allainmat M, Potier-Cartereau M. Lipidic synthetic alkaloids as SK3 channel modulators. Synthesis and biological evaluation of 2-substituted tetrahydropyridine derivatives with potential anti-metastatic activity. Eur J Med Chem 2020; 186:111854. [PMID: 31753515 DOI: 10.1016/j.ejmech.2019.111854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/04/2019] [Indexed: 11/18/2022]
Abstract
Small Conductance Calcium (Ca2+)-activated potassium (K+) channels (SKCa) are now proved to be involved in many cancer cell behaviors such as proliferation or migration. The SK3 channel isoform was particularly described in breast cancer where it can be associated with the Orai1 Ca2+ channel to form a complex that regulates the Ca2+ homeostasis during tumor development and acts as a potent mediator of bone metastases development in vivo. Until now, very few specific blockers of Orai1 and/or SK3 have been developed as potential anti-metastatic compounds. In this study, we illustrated the synthesis of new families of lipophilic pyridine and tetrahydropyridine derivatives designed as potential modulators of SK3 channel. The toxicity of the newly synthesized compounds and their migration effects were evaluated on the breast cancer cell line MDA-MB-435s. Two molecules (7a and 10c) demonstrated a significant decrease in the SK3 channel-dependent migration as well as the SK3/Orai1-related Ca2+ entry. Current measurements showed that these compounds are more likely SK3-selective. Taken all together these results suggest that such molecules could be considered as promising anti-metastatic drugs in breast cancer.
Collapse
Affiliation(s)
- Sana Kouba
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France
| | - Julien Braire
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France
| | - Romain Félix
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France
| | - Aurélie Chantôme
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France
| | - Paul-Alain Jaffrès
- University of Brest, CNRS, CEMCA, UMR 6521, 6 Avenue Victor Le Gorgeu, 29238, Brest, France
| | - Jacques Lebreton
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France
| | - Didier Dubreuil
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France
| | - Muriel Pipelier
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France
| | - Xuexin Zhang
- Pennylvania State University College of Medicine, Department of Cellular and Molecular Physiology, 700 HMC Crescent Road, Hershey, PA, 17033, USA
| | - Mohamed Trebak
- Pennylvania State University College of Medicine, Department of Cellular and Molecular Physiology, 700 HMC Crescent Road, Hershey, PA, 17033, USA
| | - Christophe Vandier
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France
| | - Monique Mathé-Allainmat
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France.
| | - Marie Potier-Cartereau
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France.
| |
Collapse
|
84
|
Abi-Gerges N, Miller PE, Ghetti A. Human Heart Cardiomyocytes in Drug Discovery and Research: New Opportunities in Translational Sciences. Curr Pharm Biotechnol 2019; 21:787-806. [PMID: 31820682 DOI: 10.2174/1389201021666191210142023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
In preclinical drug development, accurate prediction of drug effects on the human heart is critically important, whether in the context of cardiovascular safety or for the purpose of modulating cardiac function to treat heart disease. Current strategies have significant limitations, whereby, cardiotoxic drugs can escape detection or potential life-saving therapies are abandoned due to false positive toxicity signals. Thus, new and more reliable translational approaches are urgently needed to help accelerate the rate of new therapy development. Renewed efforts in the recovery of human donor hearts for research and in cardiomyocyte isolation methods, are providing new opportunities for preclinical studies in adult primary cardiomyocytes. These cells exhibit the native physiological and pharmacological properties, overcoming the limitations presented by artificial cellular models, animal models and have great potential for providing an excellent tool for preclinical drug testing. Adult human primary cardiomyocytes have already shown utility in assessing drug-induced cardiotoxicity risk and helping in the identification of new treatments for cardiac diseases, such as heart failure and atrial fibrillation. Finally, strategies with actionable decision-making trees that rely on data derived from adult human primary cardiomyocytes will provide the holistic insights necessary to accurately predict human heart effects of drugs.
Collapse
Affiliation(s)
- Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Paul E Miller
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Andre Ghetti
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| |
Collapse
|
85
|
Lubberding AF, Sattler SM, Grunnet M, Sørensen US, Tfelt-Hansen J, Jespersen T. Arrhythmia development during inhibition of small-conductance calcium-activated potassium channels in acute myocardial infarction in a porcine model. Europace 2019; 21:1584-1593. [DOI: 10.1093/europace/euz223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/22/2019] [Indexed: 12/19/2022] Open
Abstract
AbstractAims Acute myocardial infarction (AMI) is associated with intracellular Ca2+ build-up. In healthy ventricles, small conductance Ca2+-activated K+ (SK) channels are present but do not participate in repolarization. However, SK current is increased in chronic myocardial infarction and heart failure, and recently, SK channel inhibition was demonstrated to reduce arrhythmias in AMI rats. Hence, we hypothesized that SK channel inhibitors (NS8593 and AP14145) could reduce arrhythmia development during AMI in a porcine model.Methods and results Twenty-seven pigs were randomized 1:1:1 to control, NS8593, or AP14145. Haemodynamic and electrophysiological parameters [electrocardiogram (ECG) and monophasic action potentials (MAP)] were continuously recorded. A balloon was placed in the mid-left anterior descending artery, blinded to treatment. Infusion lasted from 10 min before occlusion until 30 min after. Occlusion was maintained for 1 h, followed by 2 h of reperfusion. Upon occlusion, cardiac output dropped similarly in all groups, while blood pressure remained stable. Heart rate decreased in the NS8593 and AP14145 groups. QRS duration increased upon occlusion in all groups but more prominently in AP14145-treated pigs. Inhibition of SK channels did not affect QT interval. Infarct MAP duration shortened comparably in all groups. Ventricular fibrillation developed in 4/9 control-, 4/9 AP14145-, and 2/9 NS8593-treated pigs. Ventricular tachycardia was rarely observed in either group, whereas ventricular extrasystoles occurred comparably in all groups.Conclusion Inhibition of SK channels was neither beneficial nor detrimental to ventricular arrhythmia development in the setting of AMI in this porcine model.
Collapse
Affiliation(s)
- Anniek F Lubberding
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Norre Alle 14, Copenhagen, Denmark
| | - Stefan M Sattler
- Department of Cardiology, Heart Centre, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
- Medical Department I, University Hospital Grosshadern, LMU, Marchioninistraße 15, Munich, Germany
| | | | | | - Jacob Tfelt-Hansen
- Department of Cardiology, Heart Centre, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
- Department of Forensic Medicine, Faculty of Medical Sciences, University of Copenhagen, Frederik V's Vej, Copenhagen, Denmark
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Norre Alle 14, Copenhagen, Denmark
| |
Collapse
|
86
|
Brown BM, Shim H, Christophersen P, Wulff H. Pharmacology of Small- and Intermediate-Conductance Calcium-Activated Potassium Channels. Annu Rev Pharmacol Toxicol 2019; 60:219-240. [PMID: 31337271 DOI: 10.1146/annurev-pharmtox-010919-023420] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The three small-conductance calcium-activated potassium (KCa2) channels and the related intermediate-conductance KCa3.1 channel are voltage-independent K+ channels that mediate calcium-induced membrane hyperpolarization. When intracellular calcium increases in the channel vicinity, it calcifies the flexible N lobe of the channel-bound calmodulin, which then swings over to the S4-S5 linker and opens the channel. KCa2 and KCa3.1 channels are highly druggable and offer multiple binding sites for venom peptides and small-molecule blockers as well as for positive- and negative-gating modulators. In this review, we briefly summarize the physiological role of KCa channels and then discuss the pharmacophores and the mechanism of action of the most commonly used peptidic and small-molecule KCa2 and KCa3.1 modulators. Finally, we describe the progress that has been made in advancing KCa3.1 blockers and KCa2.2 negative- and positive-gating modulators toward the clinic for neurological and cardiovascular diseases and discuss the remaining challenges.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Heesung Shim
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | | | - Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| |
Collapse
|
87
|
Kirchhoff JE, Skarsfeldt MA, Muthukumarasamy KM, Simó-Vicens R, Bomholtz SH, Abildgaard L, Jespersen T, Sørensen US, Grunnet M, Bentzen BH, Diness JG. The K Ca2 Channel Inhibitor AP14145, But Not Dofetilide or Ondansetron, Provides Functional Atrial Selectivity in Guinea Pig Hearts. Front Pharmacol 2019; 10:668. [PMID: 31275147 PMCID: PMC6593233 DOI: 10.3389/fphar.2019.00668] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/23/2019] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: Prolongation of cardiac action potentials is considered antiarrhythmic in the atria but can be proarrhythmic in ventricles if the current carried by Kv11.1-channels (IKr) is inhibited. The current mediated by KCa2-channels, IKCa, is considered a promising new target for treatment of atrial fibrillation (AF). Selective inhibitors of IKr (dofetilide) and IKCa (AP14145) were used to compare the effects on ventricular and atrial repolarization. Ondansetron, which has been reported to be a potent blocker of both IKr and IKCa, was included to examine its potential atrial antiarrhythmic properties. Experimental Approach: The expression of KCa2- and Kv11.1-channels in the guinea pig heart was investigated using quantitative polymerase chain reaction (qPCR). Whole-cell patch clamp technique was used to investigate the effects of dofetilide, AP14145, and ondansetron on IKCa and/or IKr. The effect of dofetilide, AP14145, and ondansetron on atrial and ventricular repolarization was investigated in isolated hearts. A novel atrial paced in vivo guinea pig model was further validated using AP14145 and dofetilide. Key Results: AP14145 increased the atrial effective refractory period (AERP) without prolonging the QT interval with Bazett's correction for heart rate (QTcB) both ex vivo and in vivo. In contrast, dofetilide increased QTcB and, to a lesser extent, AERP in isolated hearts and prolonged QTcB with no effects on AERP in the in vivo guinea pig model. Ondansetron did not inhibit IKCa, but did inhibit IKr in vitro. Ondansetron prolonged ventricular, but not atrial repolarization ex vivo. Conclusion and Implications: IKCa inhibition by AP14145 selectively increases atrial repolarization, whereas IKr inhibition by dofetilide and ondansetron increases ventricular repolarization to a larger extent than atrial repolarization.
Collapse
Affiliation(s)
| | - Mark Alexander Skarsfeldt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kalai Mangai Muthukumarasamy
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rafel Simó-Vicens
- Acesion Pharma, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofia Hammami Bomholtz
- Acesion Pharma, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Bo Hjorth Bentzen
- Acesion Pharma, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
88
|
Metformin Regulates the Expression of SK2 and SK3 in the Atria of Rats With Type 2 Diabetes Mellitus Through the NOX4/p38MAPK Signaling Pathway. J Cardiovasc Pharmacol 2019; 72:205-213. [PMID: 30188871 DOI: 10.1097/fjc.0000000000000615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We previously found that metformin regulates the ion current conducted by the small conductance calcium-activated potassium channels (SK channels) in the atria of rats with type 2 diabetes mellitus (T2DM) as well as the mRNA and protein expression of the SK2 and SK3 subtypes of SK channels. In this study, we hypothesized that the nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4)/p38 mitogen-activated protein kinase (p38MAPK) signaling pathway was involved in the metformin-mediated regulation of SK2 and SK3 expression in the atria of rats with T2DM. We randomly divided Wistar rats into the control group, the untreated T2DM group, the metformin-treated group, the group receiving subcutaneous injections of the nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitor diphenyleneiodonium (DPI), and the group receiving tail vein injections of the p38MAPK agonist anisomycin. Real-time polymerase chain reaction, Western blot, and immunohistochemistry were applied to examine the expression levels of SK2, SK3, NOX4, and phospho-p38MAPK (p-p38MAPK) mRNAs and proteins in the atrial tissue of relevant groups. We observed that the expression levels of NOX4 mRNA and protein and p-p38MAPK protein were significantly elevated in the atria of rats with T2DM compared with the control group. In addition, SK2 protein expression was reduced, whereas SK3 protein expression was increased. The 8-week treatment with metformin markedly reduced the expression levels of NOX4 mRNA and protein and p-p38MAPK protein, upregulated the SK2 expression, and downregulated the SK3 expression. Tail vein injection with anisomycin significantly increased the p-p38MAPK expression while further inhibiting the expression of SK2 and enhancing the expression of SK3. Subcutaneous injection with DPI considerably inhibited the expression of NOX4, further enhanced the expression of SK2 and suppressed the expression of SK3. In addition, subcutaneous injection with DPI significantly suppressed the phosphorylation of p38MAPK. In conclusion, the NOX4/p38MAPK signaling pathway mediates the downregulation of SK2 and the upregulation of SK3 in the atria of rats with T2DM. Long-term metformin treatment upregulates SK2 protein expression and downregulates SK3 protein expression by inhibiting the NOX4/p38MAPK signaling pathway.
Collapse
|
89
|
Heijman J, Dobrev D. Inhibition of Small-Conductance Ca 2+-Activated K + Channels: The Long-Awaited Breakthrough for Antiarrhythmic Drug Therapy of Atrial Fibrillation? Circ Arrhythm Electrophysiol 2019; 10:CIRCEP.117.005776. [PMID: 29018167 DOI: 10.1161/circep.117.005776] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jordi Heijman
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); and Institute of Pharmacology, West German Heart and Vascular Center, School of Medicine, University Duisburg-Essen, Germany (D.D.)
| | - Dobromir Dobrev
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); and Institute of Pharmacology, West German Heart and Vascular Center, School of Medicine, University Duisburg-Essen, Germany (D.D.).
| |
Collapse
|
90
|
Peyronnet R, Ravens U. Atria-selective antiarrhythmic drugs in need of alliance partners. Pharmacol Res 2019; 145:104262. [PMID: 31059791 DOI: 10.1016/j.phrs.2019.104262] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022]
Abstract
Atria-selective antiarrhythmic drugs in need of alliance partners. Guideline-based treatment of atrial fibrillation (AF) comprises prevention of thromboembolism and stroke, as well as antiarrhythmic therapy by drugs, electrical rhythm conversion, ablation and surgical procedures. Conventional antiarrhythmic drugs are burdened with unwanted side effects including a propensity of triggering life-threatening ventricular fibrillation. In order to solve this therapeutic dilemma, 'atria-selective' antiarrhythmic drugs have been developed for the treatment of supraventricular arrhythmias. These drugs are designed to aim at atrial targets, taking advantage of differences in atrial and ventricular ion channel expression and function. However it is not clear, whether such drugs are sufficiently antiarrhythmic or whether they are in need of an alliance partner for clinical efficacy. Atria-selective Na+ channel blockers display fast dissociation kinetics and high binding affinity to inactivated channels. Compounds targeting atria-selective K+ channels include blockers of ultra rapid delayed rectifier (Kv1.5) or acetylcholine-activated inward rectifier K+ channels (Kir3.x), inward rectifying K+ channels (Kir2.x), Ca2+-activated K+ channels of small conductance (SK), weakly rectifying two-pore domain K+ channels (K2P), and transient receptor potential channels (TRP). Despite good antiarrhythmic data from in-vitro and animal model experiments, clinical efficacy of atria-selective antiarrhythmic drugs remains to be demonstrated. In the present review we will briefly summarize the novel compounds and their proposed antiarrhythmic action. In addition, we will discuss the evidence for putative improvement of antiarrhythmic efficacy and potency by addressing multiple pathophysiologically relevant targets as possible alliance partners.
Collapse
Affiliation(s)
- Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg Bad Krozingen, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg Bad Krozingen, Medical Center, University of Freiburg, Freiburg, Germany; Institute of Physiology, Medical Faculty TU Dresden, Dresden, Germany.
| |
Collapse
|
91
|
Shamsaldeen YA, Culliford L, Clout M, James AF, Ascione R, Hancox JC, Marrion NV. Role of SK channel activation in determining the action potential configuration in freshly isolated human atrial myocytes from the SKArF study. Biochem Biophys Res Commun 2019; 512:684-690. [PMID: 30922569 DOI: 10.1016/j.bbrc.2019.03.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/13/2019] [Indexed: 10/27/2022]
Abstract
Inhibition of SK channel function is being pursued in animal models as a possible therapeutic approach to treat atrial fibrillation (AF). However, the pharmacology of SK channels in human atria is unclear. SK channel function is inhibited by both apamin and UCL1684, with the former discriminating between SK channel subtypes. In this proof-of-principle study, the effects of apamin and UCL1684 on right atrial myocytes freshly isolated from patients in sinus rhythm undergoing elective cardiac surgery were investigated. Outward current evoked from voltage clamped human atrial myocytes was reduced by these two inhibitors of SK channel function. In contrast, membrane current underlying the atrial action potential was affected significantly only by UCL1684 and not by apamin. This pharmacology mirrors that observed in mouse atria, suggesting that mammalian atria possess two populations of SK channels, with only one population contributing to the action potential waveform. Immuno-visualization of the subcellular localization of SK2 and SK3 subunits showed a high degree of colocalization, consistent with the formation of heteromeric SK2/SK3 channels. These data reveal that human atrial myocytes express two SK channel subtypes, one exhibiting an unusual pharmacology. These channels contribute to the atrial action potential waveform and might be a target for novel therapeutic approaches to treat supraventricular arrhythmic conditions such as atrial fibrillation.
Collapse
Affiliation(s)
- Yousif A Shamsaldeen
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Lucy Culliford
- Clinical Trials and Evaluation Unit, Bristol Trials Centre, Bristol Medical School, University of Bristol, Bristol, UK
| | - Madeleine Clout
- Clinical Trials and Evaluation Unit, Bristol Trials Centre, Bristol Medical School, University of Bristol, Bristol, UK
| | - Andrew F James
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Raimondo Ascione
- Translational Biomedical Research Centre, Faculty of Health Sciences, University of Bristol, UK
| | - Jules C Hancox
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Neil V Marrion
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
92
|
Hamilton S, Polina I, Terentyeva R, Bronk P, Kim TY, Roder K, Clements RT, Koren G, Choi BR, Terentyev D. PKA phosphorylation underlies functional recruitment of sarcolemmal SK2 channels in ventricular myocytes from hypertrophic hearts. J Physiol 2019; 598:2847-2873. [PMID: 30771223 PMCID: PMC7496687 DOI: 10.1113/jp277618] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 02/08/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Small-conductance Ca2+ -activated K+ (SK) channels expressed in ventricular myocytes are dormant in health, yet become functional in cardiac disease. SK channels are voltage independent and their gating is controlled by intracellular [Ca2+ ] in a biphasic manner. Submicromolar [Ca2+ ] activates the channel via constitutively-bound calmodulin, whereas higher [Ca2+ ] exerts inhibitory effect during depolarization. Using a rat model of cardiac hypertrophy induced by thoracic aortic banding, we found that functional upregulation of SK2 channels in hypertrophic rat ventricular cardiomyocytes is driven by protein kinase A (PKA) phosphorylation. Using site-directed mutagenesis, we identified serine-465 as the site conferring PKA-dependent effects on SK2 channel function. PKA phosphorylation attenuates ISK rectification by reducing the Ca2+ /voltage-dependent inhibition of SK channels without changing their sensitivity to activating submicromolar [Ca2+ ]i . This mechanism underlies the functional recruitment of SK channels not only in cardiac disease, but also in normal physiology, contributing to repolarization under conditions of enhanced adrenergic drive. ABSTRACT Small-conductance Ca2+ -activated K+ (SK) channels expressed in ventricular myocytes (VMs) are dormant in health, yet become functional in cardiac disease. We aimed to test the hypothesis that post-translational modification of SK channels under conditions accompanied by enhanced adrenergic drive plays a central role in disease-related activation of the channels. We investigated this phenomenon using a rat model of hypertrophy induced by thoracic aortic banding (TAB). Western blot analysis using anti-pan-serine/threonine antibodies demonstrated enhanced phosphorylation of immunoprecipitated SK2 channels in VMs from TAB rats vs. Shams, which was reversible by incubation of the VMs with PKA inhibitor H89 (1 μmol L-1 ). Patch clamped VMs under basal conditions from TABs but not Shams exhibited outward current sensitive to the specific SK inhibitor apamin (100 nmol L-1 ), which was eliminated by inhibition of PKA (1 μmol L-1 ). Beta-adrenergic stimulation (isoproterenol, 100 nmol L-1 ) evoked ISK in VMs from Shams, resulting in shortening of action potentials in VMs and ex vivo optically mapped Sham hearts. Using adenoviral gene transfer, wild-type and mutant SK2 channels were overexpressed in adult rat VMs, revealing serine-465 as the site that elicits PKA-dependent phosphorylation effects on SK2 channel function. Concurrent confocal Ca2+ imaging experiments established that PKA phosphorylation lessens rectification of ISK via reduction Ca2+ /voltage-dependent inhibition of the channels at high [Ca2+ ] without affecting their sensitivity to activation by Ca2+ in the submicromolar range. In conclusion, upregulation of SK channels in diseased VMs is mediated by hyperadrenergic drive in cardiac hypertrophy, with functional effects on the channel conferred by PKA-dependent phosphorylation at serine-465.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Iuliia Polina
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Medical University of South Carolina, Department of Medicine, Division of Nephrology, Charleston, SC, USA
| | - Radmila Terentyeva
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Peter Bronk
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Tae Yun Kim
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Karim Roder
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Richard T Clements
- Department of Surgery, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Gideon Koren
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Bum-Rak Choi
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Cardiovascular Research Center, Providence, RI, USA.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
93
|
Bohne LJ, Johnson D, Rose RA, Wilton SB, Gillis AM. The Association Between Diabetes Mellitus and Atrial Fibrillation: Clinical and Mechanistic Insights. Front Physiol 2019; 10:135. [PMID: 30863315 PMCID: PMC6399657 DOI: 10.3389/fphys.2019.00135] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 02/04/2019] [Indexed: 01/16/2023] Open
Abstract
A number of clinical studies have reported that diabetes mellitus (DM) is an independent risk factor for Atrial fibrillation (AF). After adjustment for other known risk factors including age, sex, and cardiovascular risk factors, DM remains a significant if modest risk factor for development of AF. The mechanisms underlying the increased susceptibility to AF in DM are incompletely understood, but are thought to involve electrical, structural, and autonomic remodeling in the atria. Electrical remodeling in DM may involve alterations in gap junction function that affect atrial conduction velocity due to changes in expression or localization of connexins. Electrical remodeling can also occur due to changes in atrial action potential morphology in association with changes in ionic currents, such as sodium or potassium currents, that can affect conduction velocity or susceptibility to triggered activity. Structural remodeling in DM results in atrial fibrosis, which can alter conduction patterns and susceptibility to re-entry in the atria. In addition, increases in atrial adipose tissue, especially in Type II DM, can lead to disruptions in atrial conduction velocity or conduction patterns that may affect arrhythmogenesis. Whether the insulin resistance in type II DM activates unique intracellular signaling pathways independent of obesity requires further investigation. In addition, the relationship between incident AF and glycemic control requires further study.
Collapse
Affiliation(s)
- Loryn J Bohne
- Department of Cardiac Sciences and Department of Physiology and Pharmacology, University of Calgary and Libin Cardiovascular Institute of Alberta, Calgary, AB, Canada
| | - Dustin Johnson
- Department of Cardiac Sciences and Department of Physiology and Pharmacology, University of Calgary and Libin Cardiovascular Institute of Alberta, Calgary, AB, Canada
| | - Robert A Rose
- Department of Cardiac Sciences and Department of Physiology and Pharmacology, University of Calgary and Libin Cardiovascular Institute of Alberta, Calgary, AB, Canada
| | - Stephen B Wilton
- Department of Cardiac Sciences and Department of Physiology and Pharmacology, University of Calgary and Libin Cardiovascular Institute of Alberta, Calgary, AB, Canada
| | - Anne M Gillis
- Department of Cardiac Sciences and Department of Physiology and Pharmacology, University of Calgary and Libin Cardiovascular Institute of Alberta, Calgary, AB, Canada
| |
Collapse
|
94
|
Simó-Vicens R, Bomholtz SH, Sørensen US, Bentzen BH. 2,6-Bis(2-Benzimidazolyl)Pyridine (BBP) Is a Potent and Selective Inhibitor of Small Conductance Calcium-Activated Potassium (SK) Channels. Front Pharmacol 2018; 9:1409. [PMID: 30559671 PMCID: PMC6287599 DOI: 10.3389/fphar.2018.01409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/16/2018] [Indexed: 11/13/2022] Open
Abstract
A variety of polycyclic pyridines have been proposed as inhibitors of the small conductance calcium-activated potassium (SK) channel. To this group belongs 2,6-bis(2-benzimidazolyl)pyridine (BBP), a commercially and readily available small organic compound which has earlier been described in a broad range of chemical and biological uses. Here, we show how BBP can also be used as a potent and specific SK channel blocker in vitro. The potency of BBP was measured using automatic patch clamp on all three SK channel subtypes, resulting in similar IC50 of 0.4 μM. We also assessed the selectivity of BBP on a panel of calcium-activated and voltage-activated potassium channels using two-electrode voltage clamp, automatic and manual patch clamp. BBP did not have any effect on IK, Kir2.1, Kir3.1+Kir3.4, Kv1.5, Kv4.3/KCHIP2 and Kv7.1/KCNE1 currents and was 4.8-fold and 46-fold more potent on all SK channel subtypes vs. BK and hERG channels, respectively. Moreover, we were able to identify H491 as a critical amino acid for the pharmacological effect of BBP on the SK channel. From a medicinal chemistry perspective, BBP could be used as a starting point for the design of new and improved SK inhibitors.
Collapse
Affiliation(s)
- Rafel Simó-Vicens
- Cardiovascular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Acesion Pharma, Copenhagen, Denmark
| | - Sofia H Bomholtz
- Cardiovascular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Acesion Pharma, Copenhagen, Denmark
| | | | - Bo H Bentzen
- Cardiovascular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Acesion Pharma, Copenhagen, Denmark
| |
Collapse
|
95
|
Liu CH, Hua N, Fu X, Pan YL, Li B, Li XD. Metformin regulates atrial SK2 and SK3 expression through inhibiting the PKC/ERK signaling pathway in type 2 diabetic rats. BMC Cardiovasc Disord 2018; 18:236. [PMID: 30545309 PMCID: PMC6293565 DOI: 10.1186/s12872-018-0950-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/09/2018] [Indexed: 01/01/2023] Open
Abstract
Background Our previous study showed that metformin regulates the mRNA and protein levels of type 2 small conductance calcium-activated potassium channel (SK2) and type 3 small conductance calcium-activated potassium channels (SK3) in atrial tissue as well as the ion current of atrial myocytes in rats with type 2 diabetes mellitus (T2DM), but the underlying signaling mechanism is unknown. This study aimed to investigate whether metformin regulates atrial SK2 and SK3 protein expression in T2DM rats though the protein kinase C (PKC)/extracellular signal-regulated kinase (ERK) signaling pathway. Methods A T2DM rat model was established using a high-fat and high-sugar diet combined with a low-dose intraperitoneal injection of streptozotocin (STZ). The rats were randomly divided into the following five groups: the control group, the untreated T2DM group, the metformin-treated only group, the phorbol 12-myristate 13-acetate (PMA; a PKC agonist administered by intraperitoneal injection) treatment group, and the recombinant human epidermal growth factor (rh-EGF; an ERK agonist administered by tail vein injection) treatment group. The activity of PKC in atrial tissues was assayed by a PKC kinase activity assay kit. The protein expression of SK2, SK3, and phosphorylated ERK (pERK) were determined by western blotting and immunohistochemistry. Results Compared with the Control group, atrial PKC activity and pERK and SK3 protein expression were increased, while SK2 protein expression was decreased in atrial tissues of T2DM rats. Eight weeks of metformin treatment inhibited the PKC activity and pERK and SK3 expression, and elevated SK2 expression compared with the T2DM group. Compared with the metformin-treated only group, the injection of rh-EGF increased pERK and SK3 expression, and decreased SK2 expression; the injection of PMA increased PKC activity and SK3 expression, and decreased SK2 expression. In addition, the injection with PMA significantly elevated the expression of pERK. Conclusions The PKC/ERK signaling pathway is involved in the downregulation of SK2 expression and the upregulation of SK3 expression in the atrium of T2DM rats. Long-term metformin treatment prevents the SK2 downregulation and the SK3 upregulation through inhibiting the PKC/ERK signaling pathway. Electronic supplementary material The online version of this article (10.1186/s12872-018-0950-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chang-He Liu
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Na Hua
- Department of Otolaryngology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Xi Fu
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Yi-Long Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Bin Li
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China
| | - Xiao-Dong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
96
|
Dan GA, Dobrev D. Antiarrhythmic drugs for atrial fibrillation: Imminent impulses are emerging. IJC HEART & VASCULATURE 2018; 21:11-15. [PMID: 30225340 PMCID: PMC6138883 DOI: 10.1016/j.ijcha.2018.08.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/15/2018] [Accepted: 08/30/2018] [Indexed: 12/19/2022]
Abstract
Rhythm and rate strategies are considered equivalent for the management of atrial fibrillation (AF). Moreover, both strategies are intended for improving symptoms and quality of life. Despite the clinical availability of several antiarrhythmic drugs (AAD) the alternatives for the patient with comorbidities are significantly fewer because of the concern regarding many adverse effects, including proarrhythmias. The impetuous development of AF ablation gave rise to a false impression that AAD are a second line therapy. All these statements reflect, in fact, the weakness of the classical paradigm and classification regarding AAD and the gap between the current knowledge of AF mechanism and determinants and the "classical" AAD non-discriminatory action. A new paradigm in development of effective and safe AAD is based on modern knowledge of vulnerable parameters involved in the genesis and perpetuation of AF. New AAD will target specific triggers of AF and ion currents which are expressed preferentially in fibrillatory atrium. Such targets will include repolarizing currents and channels, as ultrarapid potassium current, two pore potassium current, the acetylcholine-gated potassium current, small-conductance calcium-dependent potassium channels, but, also, molecular targets involved in intracellular calcium kinetics, as Ca2+-calmodulin-dependent protein kinase, ryanodine receptors and non-coding miRNA. New mechanistic discoveries link AF to inflammation and modern anti-cytokine drugs. There is still a long way to win between basic research and clinical practice, but, without any doubt, antiarrhythmic drug therapy will remain and develop as a cornerstone therapy for AF not in conflict, but complementary and alternative to interventional therapy.
Collapse
Affiliation(s)
- Gheorghe-Andrei Dan
- Carol Davila Medicine University, Bucharest, Romania
- Colentina University Hospital, Bucharest, Romania
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
97
|
Abstract
Women have a longer QT interval than men, which appears to evolve after puberty suggesting that sex hormones have an influence on cardiac electrophysiology. Sex hormones do in fact regulate cardiac ion channels via genomic and nongenomic pathways. Women are at greater risk for life-threatening arrhythmias under conditions that prolong the QT interval. In addition, women exhibit greater sensitivity to QT interval–prolonging drugs. Female sex has also an impact on propensity to cardiovascular disease, including atrial fibrillation. However, ex vivo recorded atrial action potentials (APs) from female and male patients in atrial fibrillation did not exhibit significant differences in shape, except that APs from women had slower upstroke velocity. It is concluded that sex-related differences should be taken into account not only in the clinics, but also in basic research.
Collapse
Affiliation(s)
- Ursula Ravens
- Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg • Bad Krozingen, Medical Faculty, University of Freiburg, Germany; Institute of Physiology, Medical Faculty Carl Gustav Carus, TU Dresden, Germany
- Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg • Bad Krozingen, Medical Faculty, University of Freiburg, Germany; Institute of Physiology, Medical Faculty Carl Gustav Carus, TU Dresden, Germany
| |
Collapse
|
98
|
Colman MA, Saxena P, Kettlewell S, Workman AJ. Description of the Human Atrial Action Potential Derived From a Single, Congruent Data Source: Novel Computational Models for Integrated Experimental-Numerical Study of Atrial Arrhythmia Mechanisms. Front Physiol 2018; 9:1211. [PMID: 30245635 PMCID: PMC6137999 DOI: 10.3389/fphys.2018.01211] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/13/2018] [Indexed: 11/13/2022] Open
Abstract
Introduction: The development of improved diagnosis, management, and treatment strategies for human atrial fibrillation (AF) is a significant and important challenge in order to improve quality of life for millions and reduce the substantial social-economic costs of the condition. As a complex condition demonstrating high variability and relation to other cardiac conditions, the study of AF requires approaches from multiple disciplines including single-cell experimental electrophysiology and computational modeling. Models of human atrial cells are less well parameterized than those of the human ventricle or other mammal species, largely due to the inherent challenges in patch clamping human atrial cells. Such challenges include, frequently, unphysiologically depolarized resting potentials and thus injection of a compensatory hyperpolarizing current, as well as detecting certain ion currents which may be disrupted by the cell isolation process. The aim of this study was to develop a laboratory specific model of human atrial electrophysiology which reproduces exactly the conditions of isolated-cell experiments, including testing of multiple experimental interventions. Methods: Formulations for the primary ion currents characterized by isolated-cell experiments in the Workman laboratory were fit directly to voltage-clamp data; the fast sodium-current was parameterized based on experiments relating resting membrane potential to maximal action potential upstroke velocity; compensatory hyperpolarizing current was included as a constant applied current. These formulations were integrated with three independent human atrial cell models to provide a family of novel models. Extrapolated intact-cell models were developed through removal of the hyperpolarizing current and introduction of terminal repolarization potassium currents. Results: The isolated-cell models quantitatively reproduced experimentally measured properties of excitation in both control and pharmacological and dynamic-clamp interventions. Comparison of isolated and intact-cell models highlighted the importance of reproducing this cellular environment when comparing experimental and simulation data. Conclusion: We have developed a laboratory specific model of the human atrial cell which directly reproduces the experimental isolated-cell conditions and captures human atrial excitation properties. The model may be particularly useful for directly relating model to experiment, and offers a complementary tool to the available set of human atrial cell models with specific advantages resulting from the congruent input data source.
Collapse
Affiliation(s)
- Michael A Colman
- Leeds Computational Physiology Lab, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Priyanka Saxena
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sarah Kettlewell
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Antony J Workman
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
99
|
Vagos M, van Herck IGM, Sundnes J, Arevalo HJ, Edwards AG, Koivumäki JT. Computational Modeling of Electrophysiology and Pharmacotherapy of Atrial Fibrillation: Recent Advances and Future Challenges. Front Physiol 2018; 9:1221. [PMID: 30233399 PMCID: PMC6131668 DOI: 10.3389/fphys.2018.01221] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022] Open
Abstract
The pathophysiology of atrial fibrillation (AF) is broad, with components related to the unique and diverse cellular electrophysiology of atrial myocytes, structural complexity, and heterogeneity of atrial tissue, and pronounced disease-associated remodeling of both cells and tissue. A major challenge for rational design of AF therapy, particularly pharmacotherapy, is integrating these multiscale characteristics to identify approaches that are both efficacious and independent of ventricular contraindications. Computational modeling has long been touted as a basis for achieving such integration in a rapid, economical, and scalable manner. However, computational pipelines for AF-specific drug screening are in their infancy, and while the field is progressing quite rapidly, major challenges remain before computational approaches can fill the role of workhorse in rational design of AF pharmacotherapies. In this review, we briefly detail the unique aspects of AF pathophysiology that determine requirements for compounds targeting AF rhythm control, with emphasis on delimiting mechanisms that promote AF triggers from those providing substrate or supporting reentry. We then describe modeling approaches that have been used to assess the outcomes of drugs acting on established AF targets, as well as on novel promising targets including the ultra-rapidly activating delayed rectifier potassium current, the acetylcholine-activated potassium current and the small conductance calcium-activated potassium channel. Finally, we describe how heterogeneity and variability are being incorporated into AF-specific models, and how these approaches are yielding novel insights into the basic physiology of disease, as well as aiding identification of the important molecular players in the complex AF etiology.
Collapse
Affiliation(s)
- Márcia Vagos
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Ilsbeth G. M. van Herck
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Joakim Sundnes
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Hermenegild J. Arevalo
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Andrew G. Edwards
- Computational Physiology Department, Simula Research Laboratory, Lysaker, Norway
- Center for Cardiological Innovation, Oslo, Norway
| | - Jussi T. Koivumäki
- BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
100
|
Ivanova AD, Kuzmin VS. Electrophysiological characteristics of the rat azygos vein under electrical pacing and adrenergic stimulation. J Physiol Sci 2018; 68:617-628. [PMID: 28929393 PMCID: PMC10717306 DOI: 10.1007/s12576-017-0569-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/12/2017] [Indexed: 02/04/2023]
Abstract
Rodent thoracic veins are characterized by an extended myocardial coating. In the present study, the electrical activity in the cardiac tissue of the rat azygos vein (AZV) was investigated for the first time. The atrial-like action potentials (AP) and atrial-like conduction of the excitation were observed in the rat AZV under continuous electrical pacing. Termination of electrical pacing resulted in spontaneous positive shift of resting membrane potential (RMP) in AZV. Boradrenaline induced biphasic effects on RMP in all quiescent AZV preparations but only in 25% preparations-bursts of spontaneous AP, which were suppressed by both α- and β-adrenoreceptor antagonists. Phenylephrine induced additional depolarization of RMP in quiescent AZV preparations, while isoproterenol caused hyperpolarization. In conclusion, bioelectrical properties of the rat AZV resemble those of atrial myocardium under continuous electrical pacing; however, depolarized RMP and NA-induced spontaneous AP characterize AZV as a tissue prone to rare automaticity.
Collapse
Affiliation(s)
| | - Vlad S Kuzmin
- Biological Department, Moscow State University, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|