1
|
Yang PC, Belardinelli L, Clancy CE. Mechanisms of Chemical Atrial Defibrillation by Flecainide and Ibutilide. JACC Clin Electrophysiol 2024:S2405-500X(24)00751-5. [PMID: 39387743 DOI: 10.1016/j.jacep.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/16/2024] [Accepted: 08/13/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Effective and safe pharmacological approaches for atrial defibrillation offer several potential advantages over techniques like ablation. Pharmacological therapy is noninvasive, involving no risk associated with the procedure or resulting complications. Moreover, acute drug intervention with existing drugs is likely to be low cost and broadly accessible, thereby addressing a central tenet of health equity. OBJECTIVES This study aims to investigate ibutilide-mediated action potential prolongation to promote use-dependent effects of flecainide on Na+ channels by reducing the diastolic interval and, consequently, drug unbinding to reduce action potential excitability in atrial tissue and terminate re-entrant arrhythmia. METHODS Here we utilize a modeling and simulation approach to predict the specific combinations of sodium- and potassium-channel blocking drugs to chemically terminate atrial re-entry. RESULTS Computational modeling and simulation show that acute application of flecainide and ibutilide is a promising example of drug repurposing that may constitute a promising combination for chemical atrial defibrillation. CONCLUSIONS We predict the drug concentrations that promote efficacy of flecainide and ibutilide used in combination for atrial chemical defibrillation. We also predict the potential safety pharmacology impact of this drug combination on ventricular electrophysiology.
Collapse
Affiliation(s)
- Pei-Chi Yang
- Department of Physiology and Membrane Biology, Center for Precision Medicine and Data Science, School of Medicine, University of California, Davis, California, USA.
| | | | - Colleen E Clancy
- Department of Physiology and Membrane Biology, Center for Precision Medicine and Data Science, School of Medicine, University of California, Davis, California, USA.
| |
Collapse
|
2
|
Li E, van der Heyden MAG. The network of cardiac K IR2.1: its function, cellular regulation, electrical signaling, diseases and new drug avenues. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6369-6389. [PMID: 38683369 PMCID: PMC11422472 DOI: 10.1007/s00210-024-03116-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
The functioning of the human heart relies on complex electrical and communication systems that coordinate cardiac contractions and sustain rhythmicity. One of the key players contributing to this intricate system is the KIR2.1 potassium ion channel, which is encoded by the KCNJ2 gene. KIR2.1 channels exhibit abundant expression in both ventricular myocytes and Purkinje fibers, exerting an important role in maintaining the balance of intracellular potassium ion levels within the heart. And by stabilizing the resting membrane potential and contributing to action potential repolarization, these channels have an important role in cardiac excitability also. Either gain- or loss-of-function mutations, but also acquired impairments of their function, are implicated in the pathogenesis of diverse types of cardiac arrhythmias. In this review, we aim to elucidate the system functions of KIR2.1 channels related to cellular electrical signaling, communication, and their contributions to cardiovascular disease. Based on this knowledge, we will discuss existing and new pharmacological avenues to modulate their function.
Collapse
Affiliation(s)
- Encan Li
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM, Utrecht, Netherlands
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM, Utrecht, Netherlands.
| |
Collapse
|
3
|
Ebrahim MA, Pham TD, Niu MC, Etheridge SP, Tristani-Firouzi M, Miyake CY. Pediatric and Familial Genetic Arrhythmia Syndromes: Evaluation of Bidirectional Ventricular Tachycardia-Differential Diagnosis. Card Electrophysiol Clin 2024; 16:203-210. [PMID: 38749642 DOI: 10.1016/j.ccep.2023.10.002] [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] [Indexed: 05/26/2024]
Abstract
Bidirectional ventricular tachycardia is a unique arrhythmia that can herald lethal arrhythmia syndromes. Using cases based on real patient stories, this article examines 3 different presentations to help clinicians learn the differential diagnosis associated with this condition. Each associated genetic disorder will be briefly discussed, and valuable tips for distinguishing them from each other will be provided.
Collapse
Affiliation(s)
- Mohammad A Ebrahim
- Department of Pediatrics, Chest Diseases Hospital, Kuwait University, Jabriya, Block 4, Street 102, Kuwait City, 46300, Kuwait
| | - Tam Dan Pham
- Department of Pediatrics, Texas Children's Hospital, 6651 Main Street, Houston, TX 77003, USA
| | - Mary C Niu
- Department of Pediatrics, Division of Cardiology, University of Utah and Primary Children's Hospital, 100 Mario Capecchi Drive, Salt Lake City, UT 84113, USA
| | - Susan P Etheridge
- Department of Pediatrics, Division of Cardiology, University of Utah and Primary Children's Hospital, 100 Mario Capecchi Drive, Salt Lake City, UT 84113, USA
| | - Martin Tristani-Firouzi
- Department of Pediatrics, Division of Cardiology, University of Utah and Primary Children's Hospital, 100 Mario Capecchi Drive, Salt Lake City, UT 84113, USA
| | - Christina Y Miyake
- Department of Pediatrics, Texas Children's Hospital, 6651 Main Street, Houston, TX 77003, USA.
| |
Collapse
|
4
|
Cámara-Checa A, Rubio-Alarcón M, Dago M, Crespo-García T, Rapún J, Marín M, Tamargo J, Gómez R, Caballero R, Delpón E. Reply to Benndorff and DiFrancesco: Reliable human HCN4 single-channel recordings using the cell-attached configuration in expression systems. Proc Natl Acad Sci U S A 2024; 121:e2402992121. [PMID: 38588416 PMCID: PMC11032473 DOI: 10.1073/pnas.2402992121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Affiliation(s)
- Anabel Cámara-Checa
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Marcos Rubio-Alarcón
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Dago
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Teresa Crespo-García
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Josu Rapún
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Juan Tamargo
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
| | - Ricardo Gómez
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Ricardo Caballero
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Eva Delpón
- Cardiac Cellular Electrophysiology Group at the Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| |
Collapse
|
5
|
Cruz FM, Macías Á, Moreno-Manuel AI, Gutiérrez LK, Vera-Pedrosa ML, Martínez-Carrascoso I, Pérez PS, Robles JMR, Bermúdez-Jiménez FJ, Díaz-Agustín A, de Benito FM, Arias-Santiago S, Braza-Boils A, Martín-Martínez M, Gutierrez-Rodríguez M, Bernal JA, Zorio E, Jiménez-Jaimez J, Jalife J. Extracellular Kir2.1 C122Y Mutant Upsets Kir2.1-PIP 2 Bonds and Is Arrhythmogenic in Andersen-Tawil Syndrome. Circ Res 2024; 134:e52-e71. [PMID: 38497220 PMCID: PMC11009053 DOI: 10.1161/circresaha.123.323895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Andersen-Tawil syndrome type 1 is a rare heritable disease caused by mutations in the gene coding the strong inwardly rectifying K+ channel Kir2.1. The extracellular Cys (cysteine)122-to-Cys154 disulfide bond in the channel structure is crucial for proper folding but has not been associated with correct channel function at the membrane. We evaluated whether a human mutation at the Cys122-to-Cys154 disulfide bridge leads to Kir2.1 channel dysfunction and arrhythmias by reorganizing the overall Kir2.1 channel structure and destabilizing its open state. METHODS We identified a Kir2.1 loss-of-function mutation (c.366 A>T; p.Cys122Tyr) in an ATS1 family. To investigate its pathophysiological implications, we generated an AAV9-mediated cardiac-specific mouse model expressing the Kir2.1C122Y variant. We employed a multidisciplinary approach, integrating patch clamping and intracardiac stimulation, molecular biology techniques, molecular dynamics, and bioluminescence resonance energy transfer experiments. RESULTS Kir2.1C122Y mice recapitulated the ECG features of ATS1 independently of sex, including corrected QT prolongation, conduction defects, and increased arrhythmia susceptibility. Isolated Kir2.1C122Y cardiomyocytes showed significantly reduced inwardly rectifier K+ (IK1) and inward Na+ (INa) current densities independently of normal trafficking. Molecular dynamics predicted that the C122Y mutation provoked a conformational change over the 2000-ns simulation, characterized by a greater loss of hydrogen bonds between Kir2.1 and phosphatidylinositol 4,5-bisphosphate than wild type (WT). Therefore, the phosphatidylinositol 4,5-bisphosphate-binding pocket was destabilized, resulting in a lower conductance state compared with WT. Accordingly, on inside-out patch clamping, the C122Y mutation significantly blunted Kir2.1 sensitivity to increasing phosphatidylinositol 4,5-bisphosphate concentrations. In addition, the Kir2.1C122Y mutation resulted in channelosome degradation, demonstrating temporal instability of both Kir2.1 and NaV1.5 proteins. CONCLUSIONS The extracellular Cys122-to-Cys154 disulfide bond in the tridimensional Kir2.1 channel structure is essential for the channel function. We demonstrate that breaking disulfide bonds in the extracellular domain disrupts phosphatidylinositol 4,5-bisphosphate-dependent regulation, leading to channel dysfunction and defects in Kir2.1 energetic stability. The mutation also alters functional expression of the NaV1.5 channel and ultimately leads to conduction disturbances and life-threatening arrhythmia characteristic of Andersen-Tawil syndrome type 1.
Collapse
Affiliation(s)
- Francisco M. Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | | | - Lilian K. Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | | | | | | | | | - Francisco J Bermúdez-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - Aitor Díaz-Agustín
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Fernando Martínez de Benito
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Salvador Arias-Santiago
- Servicio de Dermatología Hospital Universitario Virgen de las Nieves
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - Aitana Braza-Boils
- Unit of Inherited Cardiomyopathies and Sudden Death (CAFAMUSME), Health Research Institute La Fe, La Fe Hospital, Valencia, Spain
- Cardiology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Mercedes Martín-Martínez
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Marta Gutierrez-Rodríguez
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Juan A. Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Esther Zorio
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Unit of Inherited Cardiomyopathies and Sudden Death (CAFAMUSME), Health Research Institute La Fe, La Fe Hospital, Valencia, Spain
- Cardiology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Juan Jiménez-Jaimez
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departments of Medicine and Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
6
|
Cámara-Checa A, Perin F, Rubio-Alarcón M, Dago M, Crespo-García T, Rapún J, Marín M, Cebrián J, Gómez R, Bermúdez-Jiménez F, Monserrat L, Tamargo J, Caballero R, Jiménez-Jáimez J, Delpón E. A gain-of-function HCN4 mutant in the HCN domain is responsible for inappropriate sinus tachycardia in a Spanish family. Proc Natl Acad Sci U S A 2023; 120:e2305135120. [PMID: 38032931 PMCID: PMC10710060 DOI: 10.1073/pnas.2305135120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/12/2023] [Indexed: 12/02/2023] Open
Abstract
In a family with inappropriate sinus tachycardia (IST), we identified a mutation (p.V240M) of the hyperpolarization-activated cyclic nucleotide-gated type 4 (HCN4) channel, which contributes to the pacemaker current (If) in human sinoatrial node cells. Here, we clinically study fifteen family members and functionally analyze the p.V240M variant. Macroscopic (IHCN4) and single-channel currents were recorded using patch-clamp in cells expressing human native (WT) and/or p.V240M HCN4 channels. All p.V240M mutation carriers exhibited IST that was accompanied by cardiomyopathy in adults. IHCN4 generated by p.V240M channels either alone or in combination with WT was significantly greater than that generated by WT channels alone. The variant, which lies in the N-terminal HCN domain, increased the single-channel conductance and opening frequency and probability of HCN4 channels. Conversely, it did not modify the channel sensitivity for cAMP and ivabradine or the level of expression at the membrane. Treatment with ivabradine based on functional data reversed the IST and the cardiomyopathy of the carriers. In computer simulations, the p.V240M gain-of-function variant increases If and beating rate and thus explains the IST of the carriers. The results demonstrate the importance of the unique HCN domain in HCN4, which stabilizes the channels in the closed state.
Collapse
Affiliation(s)
- Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Francesca Perin
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
| | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Josu Rapún
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Jorge Cebrián
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Ricardo Gómez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Francisco Bermúdez-Jiménez
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
- Centro Nacional de Investigaciones Cardiovasculares, Madrid28029, Spain
| | - Lorenzo Monserrat
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
- Health in Code Sociedad Limitada, A Coruña15008, Spain
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| | - Juan Jiménez-Jáimez
- Department of Pediatric Cardiology, Virgen de las Nieves University Hospital, Granada18014, Spain
- Instituto de Investigación Biosanitaria de Granada, Granada18014, Spain
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, 28040Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid28029, Spain
| |
Collapse
|
7
|
Kawabata Y, Takai S, Sanematsu K, Yoshida R, Kawabata F, Shigemura N. The Antiarrhythmic Drug Flecainide Enhances Aversion to HCl in Mice. eNeuro 2023; 10:ENEURO.0048-23.2023. [PMID: 37696662 PMCID: PMC10515741 DOI: 10.1523/eneuro.0048-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
Drug-induced taste disorders reduce quality of life, but little is known about the molecular mechanisms by which drugs induce taste disturbances. In this study, we investigated the short-term and long-term effects of the antiarrhythmic drug flecainide, which is known to cause taste dysfunction. Analyses of behavioral responses (licking tests) revealed that mice given a single intraperitoneal injection of flecainide exhibited a significant reduction in preference for a sour tastant (HCl) but not for other taste solutions (NaCl, quinine, sucrose, KCl and monopotassium glutamate) when compared with controls. Mice administered a single dose of flecainide also had significantly higher taste nerve responses to HCl but not to other taste solutions. Compared with controls, mice administered flecainide once-daily for 30 d showed a reduced preference for HCl without any changes in the behavioral responses to other taste solutions. The electrophysiological experiments using HEK293T cells transiently expressing otopetrin-1 (Otop1; the mouse sour taste receptor) showed that flecainide did not alter the responses to HCl. Taken together, our results suggest that flecainide specifically enhances the response to HCl in mice during short-term and long-term administration. Although further studies will be needed to elucidate the molecular mechanisms, these findings provide new insights into the pathophysiology of drug-induced taste disorders.
Collapse
Affiliation(s)
- Yuko Kawabata
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Shingo Takai
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Keisuke Sanematsu
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
- Oral Health/Brain Health/Total Health Research Center, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan
| | - Fuminori Kawabata
- Physiology of Domestic Animals, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki 036-8561, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
8
|
Cruz FM, Macías Á, Moreno-Manuel AI, Gutiérrez LK, Vera-Pedrosa ML, Martínez-Carrascoso I, Pérez PS, Robles JMR, Bermúdez-Jiménez FJ, Díaz-Agustín A, de Benito FM, Santiago SA, Braza-Boils A, Martín-Martínez M, Gutierrez-Rodríguez M, Bernal JA, Zorio E, Jiménez-Jaimez J, Jalife J. Extracellular cysteine disulfide bond break at Cys122 disrupts PIP 2-dependent Kir2.1 channel function and leads to arrhythmias in Andersen-Tawil Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544151. [PMID: 37333254 PMCID: PMC10274791 DOI: 10.1101/2023.06.07.544151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Andersen-Tawil Syndrome Type 1 (ATS1) is a rare heritable disease caused by mutations in the strong inwardly rectifying K+ channel Kir2.1. The extracellular Cys122-to-Cys154 disulfide bond in the Kir2.1 channel structure is crucial for proper folding, but has not been associated with correct channel function at the membrane. We tested whether a human mutation at the Cys122-to-Cys154 disulfide bridge leads to Kir2.1 channel dysfunction and arrhythmias by reorganizing the overall Kir2.1 channel structure and destabilizing the open state of the channel. Methods and Results We identified a Kir2.1 loss-of-function mutation in Cys122 (c.366 A>T; p.Cys122Tyr) in a family with ATS1. To study the consequences of this mutation on Kir2.1 function we generated a cardiac specific mouse model expressing the Kir2.1C122Y mutation. Kir2.1C122Y animals recapitulated the abnormal ECG features of ATS1, like QT prolongation, conduction defects, and increased arrhythmia susceptibility. Kir2.1C122Y mouse cardiomyocytes showed significantly reduced inward rectifier K+ (IK1) and inward Na+ (INa) current densities independently of normal trafficking ability and localization at the sarcolemma and the sarcoplasmic reticulum. Kir2.1C122Y formed heterotetramers with wildtype (WT) subunits. However, molecular dynamic modeling predicted that the Cys122-to-Cys154 disulfide-bond break induced by the C122Y mutation provoked a conformational change over the 2000 ns simulation, characterized by larger loss of the hydrogen bonds between Kir2.1 and phosphatidylinositol-4,5-bisphosphate (PIP2) than WT. Therefore, consistent with the inability of Kir2.1C122Y channels to bind directly to PIP2 in bioluminescence resonance energy transfer experiments, the PIP2 binding pocket was destabilized, resulting in a lower conductance state compared with WT. Accordingly, on inside-out patch-clamping the C122Y mutation significantly blunted Kir2.1 sensitivity to increasing PIP2 concentrations. Conclusion The extracellular Cys122-to-Cys154 disulfide bond in the tridimensional Kir2.1 channel structure is essential to channel function. We demonstrated that ATS1 mutations that break disulfide bonds in the extracellular domain disrupt PIP2-dependent regulation, leading to channel dysfunction and life-threatening arrhythmias.
Collapse
Affiliation(s)
- Francisco M. Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | | | - Lilian K. Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | | | | | | | | | - Francisco J Bermúdez-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - Aitor Díaz-Agustín
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Fernando Martínez de Benito
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Salvador Arias Santiago
- Servicio de Dermatología Hospital Universitario Virgen de las Nieves
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - Aitana Braza-Boils
- Unit of Inherited Cardiomyopathies and Sudden Death (CAFAMUSME), Health Research Institute La Fe, La Fe Hospital, Valencia, Spain
- Cardiology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Mercedes Martín-Martínez
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Marta Gutierrez-Rodríguez
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Juan A. Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Esther Zorio
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Unit of Inherited Cardiomyopathies and Sudden Death (CAFAMUSME), Health Research Institute La Fe, La Fe Hospital, Valencia, Spain
- Cardiology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Juan Jiménez-Jaimez
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada IBS, Granada, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departments of Medicine and Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Moreno-Manuel AI, Gutiérrez LK, Vera-Pedrosa ML, Cruz FM, Bermúdez-Jiménez FJ, Martínez-Carrascoso I, Sánchez-Pérez P, Macías Á, Jalife J. Molecular stratification of arrhythmogenic mechanisms in the Andersen Tawil syndrome. Cardiovasc Res 2023; 119:919-932. [PMID: 35892314 PMCID: PMC10153646 DOI: 10.1093/cvr/cvac118] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 11/12/2022] Open
Abstract
Andersen-Tawil syndrome (ATS) is a rare inheritable disease associated with loss-of-function mutations in KCNJ2, the gene coding the strong inward rectifier potassium channel Kir2.1, which forms an essential membrane protein controlling cardiac excitability. ATS is usually marked by a triad of periodic paralysis, life-threatening cardiac arrhythmias and dysmorphic features, but its expression is variable and not all patients with a phenotype linked to ATS have a known genetic alteration. The mechanisms underlying this arrhythmogenic syndrome are poorly understood. Knowing such mechanisms would be essential to distinguish ATS from other channelopathies with overlapping phenotypes and to develop individualized therapies. For example, the recently suggested role of Kir2.1 as a countercurrent to sarcoplasmic calcium reuptake might explain the arrhythmogenic mechanisms of ATS and its overlap with catecholaminergic polymorphic ventricular tachycardia. Here we summarize current knowledge on the mechanisms of arrhythmias leading to sudden cardiac death in ATS. We first provide an overview of the syndrome and its pathophysiology, from the patient's bedside to the protein and discuss the role of essential regulators and interactors that could play a role in cases of ATS. The review highlights novel ideas related to some post-translational channel interactions with partner proteins that might help define the molecular bases of the arrhythmia phenotype. We then propose a new all-embracing classification of the currently known ATS loss-of-function mutations according to their position in the Kir2.1 channel structure and their functional implications. We also discuss specific ATS pathogenic variants, their clinical manifestations, and treatment stratification. The goal is to provide a deeper mechanistic understanding of the syndrome toward the development of novel targets and personalized treatment strategies.
Collapse
Affiliation(s)
| | - Lilian K Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | | | - Francisco Miguel Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | - Francisco José Bermúdez-Jiménez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
- Departamento de Cardiología, Hospital Virgen de las Nieves, GranadaSpain
| | | | - Patricia Sánchez-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departments of Medicine and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Li E, Kool W, Woolschot L, van der Heyden MAG. Chronic Propafenone Application Increases Functional K IR2.1 Expression In Vitro. Pharmaceuticals (Basel) 2023; 16:ph16030404. [PMID: 36986503 PMCID: PMC10056987 DOI: 10.3390/ph16030404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Expression and activity of inwardly rectifying potassium (KIR) channels within the heart are strictly regulated. KIR channels have an important role in shaping cardiac action potentials, having a limited conductance at depolarized potentials but contributing to the final stage of repolarization and resting membrane stability. Impaired KIR2.1 function causes Andersen-Tawil Syndrome (ATS) and is associated with heart failure. Restoring KIR2.1 function by agonists of KIR2.1 (AgoKirs) would be beneficial. The class 1c antiarrhythmic drug propafenone is identified as an AgoKir; however, its long-term effects on KIR2.1 protein expression, subcellular localization, and function are unknown. Propafenone's long-term effect on KIR2.1 expression and its underlying mechanisms in vitro were investigated. KIR2.1-carried currents were measured by single-cell patch-clamp electrophysiology. KIR2.1 protein expression levels were determined by Western blot analysis, whereas conventional immunofluorescence and advanced live-imaging microscopy were used to assess the subcellular localization of KIR2.1 proteins. Acute propafenone treatment at low concentrations supports the ability of propafenone to function as an AgoKir without disturbing KIR2.1 protein handling. Chronic propafenone treatment (at 25-100 times higher concentrations than in the acute treatment) increases KIR2.1 protein expression and KIR2.1 current densities in vitro, which are potentially associated with pre-lysosomal trafficking inhibition.
Collapse
Affiliation(s)
- Encan Li
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Willy Kool
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Liset Woolschot
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | - Marcel A G van der Heyden
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
11
|
Abstract
Long QT syndrome (LQTS) is a detrimental arrhythmia syndrome mainly caused by dysregulated expression or aberrant function of ion channels. The major clinical symptoms of ventricular arrhythmia, palpitations and syncope vary among LQTS subtypes. Susceptibility to malignant arrhythmia is a result of delayed repolarisation of the cardiomyocyte action potential (AP). There are 17 distinct subtypes of LQTS linked to 15 autosomal dominant genes with monogenic mutations. However, due to the presence of modifier genes, the identical mutation may result in completely different clinical manifestations in different carriers. In this review, we describe the roles of various ion channels in orchestrating APs and discuss molecular aetiologies of various types of LQTS. We highlight the usage of patient-specific induced pluripotent stem cell (iPSC) models in characterising fundamental mechanisms associated with LQTS. To mitigate the outcomes of LQTS, treatment strategies are initially focused on small molecules targeting ion channel activities. Next-generation treatments will reap the benefits from development of LQTS patient-specific iPSC platform, which is bolstered by the state-of-the-art technologies including whole-genome sequencing, CRISPR genome editing and machine learning. Deep phenotyping and high-throughput drug testing using LQTS patient-specific cardiomyocytes herald the upcoming precision medicine in LQTS.
Collapse
|
12
|
Dago M, Crespo-García T, Cámara-Checa A, Rapún J, Rubio-Alarcón M, Marín M, Tamargo J, Caballero R, Delpón E. Empagliflozin and Dapagliflozin Increase Na + and Inward Rectifier K + Current Densities in Human Cardiomyocytes Derived from Induced Pluripotent Stem Cells (hiPSC-CMs). Cells 2022; 11:3707. [PMID: 36496967 PMCID: PMC9738206 DOI: 10.3390/cells11233707] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022] Open
Abstract
Dapagliflozin (dapa) and empagliflozin (empa) are sodium-glucose cotransporter-2 inhibitors (SGLT2is) that reduce morbidity and mortality in heart failure (HF) patients. Sodium and inward rectifier K+ currents (INa and IK1), carried by Nav1.5 and Kir2.1 channels, respectively, are responsible for cardiac excitability, conduction velocity, and refractoriness. In HF patients, Nav1.5 and Kir2.1 expression are reduced, enhancing risk of arrhythmia. Incubation with dapa or empa (24-h,1 µM) significantly increased INa and IK1 densities recorded in human-induced pluripotent stem cell-cardiomyocytes (hiPSC-CMs) using patch-clamp techniques. Dapa and empa, respectively, shifted to more hyperpolarized potentials the INa activation and inactivation curves. Identical effects were observed in Chinese hamster ovary (CHO) cells that were incubated with dapa or empa and transiently expressed human Nav1.5 channels. Conversely, empa but not dapa significantly increased human Kir2.1 currents in CHO cells. Dapa and empa effects on INa and IK1 were also apparent in Ca-calmodulin kinase II-silenced CHO cells. Cariporide, a Na+/H+ exchanger type 1 (NHE1) inhibitor, did not increase INa or IK1 in hiPSC-CMs. Dapa and empa at therapeutic concentrations increased INa and IK1 in healthy human cardiomyocytes. These SGLT2is could represent a new class of drugs with a novel and long-pursued antiarrhythmic mechanism of action.
Collapse
Affiliation(s)
- María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Josu Rapún
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
13
|
Crespo-García T, Rubio-Alarcón M, Cámara-Checa A, Dago M, Rapún J, Nieto-Marín P, Marín M, Cebrián J, Tamargo J, Delpón E, Caballero R. A Cantú syndrome mutation produces dual effects on KATP channels by disrupting ankyrin B regulation. J Gen Physiol 2022; 155:213613. [PMID: 36287534 PMCID: PMC9614705 DOI: 10.1085/jgp.202112995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/09/2022] [Accepted: 10/04/2022] [Indexed: 02/01/2023] Open
Abstract
ATP-sensitive potassium (KATP) channels composed of Kir6.x and sulfonylurea receptor (SURs) subunits couple cellular metabolism to electrical activity. Cantú syndrome (CS) is a rare disease caused by mutations in the genes encoding Kir6.1 (KCNJ8) and SUR2A (ABCC9) that produce KATP channel hyperactivity due to a reduced channel block by physiological ATP concentrations. We functionally characterized the p.S1054Y SUR2A mutation identified in two CS carriers, who exhibited a mild phenotype although the mutation was predicted as highly pathogenic. We recorded macroscopic and single-channel currents in CHO and HEK-293 cells and measured the membrane expression of the channel subunits by biotinylation assays in HEK-293 cells. The mutation increased basal whole-cell current density and at the single-channel level, it augmented opening frequency, slope conductance, and open probability (Po), and promoted the appearance of multiple conductance levels. p.S1054Y also reduced Kir6.2 and SUR2A expression specifically at the membrane. Overexpression of ankyrin B (AnkB) prevented these gain- and loss-of-function effects, as well as the p.S1054Y-induced reduction of ATP inhibition of currents measured in inside-out macropatches. Yeast two-hybrid assays suggested that SUR2A WT and AnkB interact, while p.S1054Y interaction with AnkB is decreased. The p.E322K Kir6.2 mutation, which prevents AnkB binding to Kir6.2, produced similar biophysical alterations than p.S1054Y. Our results are the first demonstration of a CS mutation whose functional consequences involve the disruption of AnkB effects on KATP channels providing a novel mechanism by which CS mutations can reduce ATP block. Furthermore, they may help explain the mild phenotype associated with this mutation.
Collapse
Affiliation(s)
- Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - Josu Rapún
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - Paloma Nieto-Marín
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - Jorge Cebrián
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain,Correspondence to Eva Delpón:
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, Madrid, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Madrid, Spain
| |
Collapse
|
14
|
Macías Á, González-Guerra A, Moreno-Manuel AI, Cruz FM, Gutiérrez LK, García-Quintáns N, Roche-Molina M, Bermúdez-Jiménez F, Andrés V, Vera-Pedrosa ML, Martínez-Carrascoso I, Bernal JA, Jalife J. Kir2.1 dysfunction at the sarcolemma and the sarcoplasmic reticulum causes arrhythmias in a mouse model of Andersen-Tawil syndrome type 1. NATURE CARDIOVASCULAR RESEARCH 2022; 1:900-917. [PMID: 39195979 PMCID: PMC11358039 DOI: 10.1038/s44161-022-00145-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/02/2022] [Indexed: 08/29/2024]
Abstract
Andersen-Tawil syndrome type 1 (ATS1) is associated with life-threatening arrhythmias of unknown mechanism. In this study, we generated and characterized a mouse model of ATS1 carrying the trafficking-deficient mutant Kir2.1Δ314-315 channel. The mutant mouse recapitulates the electrophysiological phenotype of ATS1, with QT prolongation exacerbated by flecainide or isoproterenol, drug-induced QRS prolongation, increased vulnerability to reentrant arrhythmias and multifocal discharges resembling catecholaminergic polymorphic ventricular tachycardia (CPVT). Kir2.1Δ314-315 cardiomyocytes display significantly reduced inward rectifier K+ and Na+ currents, depolarized resting membrane potential and prolonged action potentials. We show that, in wild-type mouse cardiomyocytes and skeletal muscle cells, Kir2.1 channels localize to sarcoplasmic reticulum (SR) microdomains, contributing to intracellular Ca2+ homeostasis. Kir2.1Δ314-315 cardiomyocytes exhibit defective SR Kir2.1 localization and function, as intact and permeabilized Kir2.1Δ314-315 cardiomyocytes display abnormal spontaneous Ca2+ release events. Overall, defective Kir2.1 channel function at the sarcolemma and the SR explain the life-threatening arrhythmias in ATS1 and its overlap with CPVT.
Collapse
Affiliation(s)
- Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | | | - Francisco M Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Lilian K Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Marta Roche-Molina
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | | | - Juan A Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Departments of Medicine and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Crespo-García T, Cámara-Checa A, Dago M, Rubio-Alarcón M, Rapún J, Tamargo J, Delpón E, Caballero R. Regulation of cardiac ion channels by transcription factors: Looking for new opportunities of druggable targets for the treatment of arrhythmias. Biochem Pharmacol 2022; 204:115206. [PMID: 35963339 DOI: 10.1016/j.bcp.2022.115206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022]
Abstract
Cardiac electrical activity is governed by different ion channels that generate action potentials. Acquired or inherited abnormalities in the expression and/or function of ion channels usually result in electrophysiological changes that can cause cardiac arrhythmias. Transcription factors (TFs) control gene transcription by binding to specific DNA sequences adjacent to target genes. Linkage analysis, candidate-gene screening within families, and genome-wide association studies have linked rare and common genetic variants in the genes encoding TFs with genetically-determined cardiac arrhythmias. Besides its critical role in cardiac development, recent data demonstrated that they control cardiac electrical activity through the direct regulation of the expression and function of cardiac ion channels in adult hearts. This narrative review summarizes some studies showing functional data on regulation of the main human atrial and ventricular Na+, Ca2+, and K+ channels by cardiac TFs such as Pitx2c, Tbx20, Tbx5, Zfhx3, among others. The results have improved our understanding of the mechanisms regulating cardiac electrical activity and may open new avenues for therapeutic interventions in cardiac acquired or inherited arrhythmias through the identification of TFs as potential drug targets. Even though TFs have for a long time been considered as 'undruggable' targets, advances in structural biology have led to the identification of unique pockets in TFs amenable to be targeted with small-molecule drugs or peptides that are emerging as novel therapeutic drugs.
Collapse
Affiliation(s)
- T Crespo-García
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - A Cámara-Checa
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - M Dago
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - M Rubio-Alarcón
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - J Rapún
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - J Tamargo
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | - E Delpón
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain.
| | - R Caballero
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| | -
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, 28040 Madrid, Spain
| |
Collapse
|
16
|
Liu Q, Sun J, Dong Y, Li P, Wang J, Wang Y, Xu Y, Tian X, Wu B, He P, Yu Q, Lu X, Cao J. Tetramisole is a new I K1 channel agonist and exerts I K1 -dependent cardioprotective effects in rats. Pharmacol Res Perspect 2022; 10:e00992. [PMID: 35880674 PMCID: PMC9316008 DOI: 10.1002/prp2.992] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/22/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiac ischemia, hypoxia, arrhythmias, and heart failure share the common electrophysiological changes featured by the elevation of intracellular Ca2+ (Ca2+ overload) and inhibition of the inward rectifier potassium (IK1 ) channel. IK1 channel agonists have been considered a new type of anti-arrhythmia and cardioprotective agents. We predicted using a drug repurposing strategy that tetramisole (Tet), a known anthelminthic agent, was a new IK1 channel agonist. The present study aimed to experimentally identify the above prediction and further demonstrate that Tet has cardioprotective effects. Results of the whole-cell patch clamp technique showed that Tet at 1-100 μmol/L enhanced IK1 current, hyperpolarized resting potential (RP), and shortened action potential duration (APD) in isolated rat cardiomyocytes, while without effects on other ion channels or transporters. In adult Sprague-Dawley (SD) rats in vivo, Tet showed anti-arrhythmia and anticardiac remodeling effects, respectively, in the coronary ligation-induced myocardial infarction model and isoproterenol (Iso, i.p., 3 mg/kg/day, 10 days) infusion-induced cardiac remodeling model. Tet also showed anticardiomyocyte remodeling effect in Iso (1 μmol/L) infused adult rat ventricular myocytes or cultured H9c2 (2-1) cardiomyocytes. Tet at 0.54 mg/kg in vivo or 30 μmol/L in vitro showed promising protections on acute ischemic arrhythmias, myocardial hypertrophy, and fibrosis. Molecular docking was performed and identified the selective binding of Tet with Kir2.1. The cardioprotection of Tet was associated with the facilitation of IK1 channel forward trafficking, deactivation of PKA signaling, and inhibition of intracellular calcium overload. Enhancing IK1 may play dual roles in anti-arrhythmia and antiventricular remodeling mediated by restoration of Ca2+ homeostasis.
Collapse
Affiliation(s)
- Qinghua Liu
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Jiaxing Sun
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Yangdou Dong
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Pan Li
- Department of PathophysiologyShanxi Medical UniversityTaiyuanChina
| | - Jin Wang
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Yulan Wang
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Yanwu Xu
- Department of BiochemistryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xinrui Tian
- Department of Respiratory and Critical Care MedicineSecond Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Bowei Wu
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Peifeng He
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, School of ManagementShanxi Medical UniversityTaiyuanChina
| | - Qi Yu
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, School of ManagementShanxi Medical UniversityTaiyuanChina
| | - Xuechun Lu
- Department of Hematology, The Second Medical Center, Chinese PLA General HospitalNational clinical research center for geriatric diseaseBeijingChina
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Ministry of EducationShanxi Medical UniversityTaiyuanChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| |
Collapse
|
17
|
Polyamines and Their Metabolism: From the Maintenance of Physiological Homeostasis to the Mediation of Disease. MEDICAL SCIENCES (BASEL, SWITZERLAND) 2022; 10:medsci10030038. [PMID: 35893120 PMCID: PMC9326668 DOI: 10.3390/medsci10030038] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 12/13/2022]
Abstract
The polyamines spermidine and spermine are positively charged aliphatic molecules. They are critical in the regulation of nucleic acid and protein structures, protein synthesis, protein and nucleic acid interactions, oxidative balance, and cell proliferation. Cellular polyamine levels are tightly controlled through their import, export, de novo synthesis, and catabolism. Enzymes and enzymatic cascades involved in polyamine metabolism have been well characterized. This knowledge has been used for the development of novel compounds for research and medical applications. Furthermore, studies have shown that disturbances in polyamine levels and their metabolic pathways, as a result of spontaneous mutations in patients, genetic engineering in mice or experimentally induced injuries in rodents, are associated with multiple maladaptive changes. The adverse effects of altered polyamine metabolism have also been demonstrated in in vitro models. These observations highlight the important role these molecules and their metabolism play in the maintenance of physiological normalcy and the mediation of injury. This review will attempt to cover the extensive and diverse knowledge of the biological role of polyamines and their metabolism in the maintenance of physiological homeostasis and the mediation of tissue injury.
Collapse
|
18
|
Ramalho NJD, Švecová O, Kula R, Šimurdová M, Šimurda J, Bébarová M. Aminophylline at clinically relevant concentrations affects inward rectifier potassium current in a dual way. Pflugers Arch 2022; 474:303-313. [PMID: 35084562 DOI: 10.1007/s00424-021-02646-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/26/2021] [Accepted: 11/22/2021] [Indexed: 11/24/2022]
Abstract
Bronchodilator aminophylline may induce atrial or less often ventricular arrhythmias. The mechanism of this proarrhythmic side effect has not been fully explained. Modifications of inward rectifier potassium (Kir) currents including IK1 are known to play an important role in arrhythmogenesis; however, no data on the aminophylline effect on these currents have been published. Hence, we tested the effect of aminophylline (3-100 µM) on IK1 in enzymatically isolated rat ventricular myocytes using the whole-cell patch-clamp technique. A dual steady-state effect of aminophylline was observed; either inhibition or activation was apparent in individual cells during the application of aminophylline at a given concentration. The smaller the magnitude of the control IK1, the more likely the activation of the current by aminophylline and vice versa. The effect was reversible; the relative changes at -50 and -110 mV did not differ. Using IK1 channel population model, the dual effect was explained by the interaction of aminophylline with two different channel populations, the first one being inhibited and the second one being activated. Considering various fractions of these two channel populations in individual cells, varying effects observed in the measured cells could be simulated. We propose that the dual aminophylline effect may be related to the direct and indirect effect of the drug on various Kir2.x subunits forming the homo- and heterotetrameric IK1 channels in a single cell. The observed IK1 changes induced by clinically relevant concentrations of aminophylline might contribute to arrhythmogenesis related to the use of this bronchodilator in clinical medicine.
Collapse
Affiliation(s)
- Nuno Jorge Dourado Ramalho
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Olga Švecová
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Roman Kula
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.,Department of Paediatric Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Brno, Masaryk University, Černopolní 9, 662 63, Brno, Czech Republic
| | - Milena Šimurdová
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Jiří Šimurda
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Markéta Bébarová
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| |
Collapse
|
19
|
Akyuz E, Koklu B, Uner A, Angelopoulou E, Paudel YN. Envisioning the role of inwardly rectifying potassium (Kir) channel in epilepsy. J Neurosci Res 2021; 100:413-443. [PMID: 34713909 DOI: 10.1002/jnr.24985] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/23/2021] [Accepted: 10/01/2021] [Indexed: 01/29/2023]
Abstract
Epilepsy is a devastating neurological disorder characterized by recurrent seizures attributed to the disruption of the dynamic excitatory and inhibitory balance in the brain. Epilepsy has emerged as a global health concern affecting about 70 million people worldwide. Despite recent advances in pre-clinical and clinical research, its etiopathogenesis remains obscure, and there are still no treatment strategies modifying disease progression. Although the precise molecular mechanisms underlying epileptogenesis have not been clarified yet, the role of ion channels as regulators of cellular excitability has increasingly gained attention. In this regard, emerging evidence highlights the potential implication of inwardly rectifying potassium (Kir) channels in epileptogenesis. Kir channels consist of seven different subfamilies (Kir1-Kir7), and they are highly expressed in both neuronal and glial cells in the central nervous system. These channels control the cell volume and excitability. In this review, we discuss preclinical and clinical evidence on the role of the several subfamilies of Kir channels in epileptogenesis, aiming to shed more light on the pathogenesis of this disorder and pave the way for future novel therapeutic approaches.
Collapse
Affiliation(s)
- Enes Akyuz
- Faculty of International Medicine, Department of Biophysics, University of Health Sciences, Istanbul, Turkey
| | - Betul Koklu
- Faculty of Medicine, Namık Kemal University, Tekirdağ, Turkey
| | - Arda Uner
- Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
20
|
Déri S, Borbás J, Hartai T, Hategan L, Csányi B, Visnyovszki Á, Madácsy T, Maléth J, Hegedűs Z, Nagy I, Arora R, Labro AJ, Környei L, Varró A, Sepp R, Ördög B. Impaired cytoplasmic domain interactions cause co-assembly defect and loss of function in the p.Glu293Lys KNCJ2 variant isolated from an Andersen-Tawil syndrome patient. Cardiovasc Res 2021; 117:1923-1934. [PMID: 32810216 DOI: 10.1093/cvr/cvaa249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/16/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
AIMS Subunit interactions at the cytoplasmic domain interface (CD-I) have recently been shown to control gating in inward rectifier potassium channels. Here we report the novel KCNJ2 variant p.Glu293Lys that has been found in a patient with Andersen-Tawil syndrome type 1 (ATS1), causing amino acid substitution at the CD-I of the inward rectifier potassium channel subunit Kir2.1. Neither has the role of Glu293 in gating control been investigated nor has a pathogenic variant been described at this position. This study aimed to assess the involvement of Glu293 in CD-I subunit interactions and to establish the pathogenic role of the p.Glu293Lys variant in ATS1. METHODS AND RESULTS The p.Glu293Lys variant produced no current in homomeric form and showed dominant-negative effect over wild-type (WT) subunits. Immunocytochemical labelling showed the p.Glu293Lys subunits to distribute in the subsarcolemmal space. Salt bridge prediction indicated the presence of an intersubunit salt bridge network at the CD-I of Kir2.1, with the involvement of Glu293. Subunit interactions were studied by the NanoLuc® Binary Technology (NanoBiT) split reporter assay. Reporter constructs carrying NanoBiT tags on the intracellular termini produced no bioluminescent signal above background with the p.Glu293Lys variant in homomeric configuration and significantly reduced signals in cells co-expressing WT and p.Glu293Lys subunits simultaneously. Extracellularly presented reporter tags, however, generated comparable bioluminescent signals with heteromeric WT and p.Glu293Lys subunits and with homomeric WT channels. CONCLUSIONS Loss of function and dominant-negative effect confirm the causative role of p.Glu293Lys in ATS1. Co-assembly of Kir2.1 subunits is impaired in homomeric channels consisting of p.Glu293Lys subunits and is partially rescued in heteromeric complexes of WT and p.Glu293Lys Kir2.1 variants. These data point to an important role of Glu293 in mediating subunit assembly, as well as in gating of Kir2.1 channels.
Collapse
Affiliation(s)
- Szilvia Déri
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, PO Box 427, Szeged 6720, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 12, 6720 Szeged, Hungary
| | - János Borbás
- 2nd Department of Internal Medicine and Cardiology Centre, University of Szeged, Semmelweis u. 8, 6725 Szeged, Hungary
| | - Teodóra Hartai
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, PO Box 427, Szeged 6720, Hungary
| | - Lidia Hategan
- 2nd Department of Internal Medicine and Cardiology Centre, University of Szeged, Semmelweis u. 8, 6725 Szeged, Hungary
| | - Beáta Csányi
- 2nd Department of Internal Medicine and Cardiology Centre, University of Szeged, Semmelweis u. 8, 6725 Szeged, Hungary
| | - Ádám Visnyovszki
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, PO Box 427, Szeged 6720, Hungary
| | - Tamara Madácsy
- 1st Department of Internal Medicine, University of Szeged, Korányi fasor 8-10, 6720 Szeged, Hungary, Hungary
| | - József Maléth
- 1st Department of Internal Medicine, University of Szeged, Korányi fasor 8-10, 6720 Szeged, Hungary, Hungary
| | - Zoltán Hegedűs
- Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pécs, Szigeti út 12, 7624 Pécs, Hungary
| | - István Nagy
- Institute of Biochemistry, Biological Research Centre the Hungarian Academy of Sciences, Temesvári krt. 62, 6726 Szeged, Hungary
- Seqomics Biotechnology Ltd, Vállalkozók útja 7, 6782 Mórahalom, Hungary
| | - Rohit Arora
- Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Alain J Labro
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
- Department of Basic Medical Sciences, University of Ghent, C. Heymanslaan 10, 9000 Ghent, Belgium
| | - László Környei
- Gottsegen György National Institute of Cardiology, Haller u. 9, 1096 Budapest, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, PO Box 427, Szeged 6720, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 12, 6720 Szeged, Hungary
- MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Dóm tér 12, 6720 Szeged, Hungary
| | - Róbert Sepp
- 2nd Department of Internal Medicine and Cardiology Centre, University of Szeged, Semmelweis u. 8, 6725 Szeged, Hungary
| | - Balázs Ördög
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, PO Box 427, Szeged 6720, Hungary
| |
Collapse
|
21
|
Clerx M, Mirams GR, Rogers AJ, Narayan SM, Giles WR. Immediate and Delayed Response of Simulated Human Atrial Myocytes to Clinically-Relevant Hypokalemia. Front Physiol 2021; 12:651162. [PMID: 34122128 PMCID: PMC8188899 DOI: 10.3389/fphys.2021.651162] [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: 01/08/2021] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Although plasma electrolyte levels are quickly and precisely regulated in the mammalian cardiovascular system, even small transient changes in K+, Na+, Ca2+, and/or Mg2+ can significantly alter physiological responses in the heart, blood vessels, and intrinsic (intracardiac) autonomic nervous system. We have used mathematical models of the human atrial action potential (AP) to explore the electrophysiological mechanisms that underlie changes in resting potential (Vr) and the AP following decreases in plasma K+, [K+]o, that were selected to mimic clinical hypokalemia. Such changes may be associated with arrhythmias and are commonly encountered in patients (i) in therapy for hypertension and heart failure; (ii) undergoing renal dialysis; (iii) with any disease with acid-base imbalance; or (iv) post-operatively. Our study emphasizes clinically-relevant hypokalemic conditions, corresponding to [K+]o reductions of approximately 1.5 mM from the normal value of 4 to 4.5 mM. We show how the resulting electrophysiological responses in human atrial myocytes progress within two distinct time frames: (i) Immediately after [K+]o is reduced, the K+-sensing mechanism of the background inward rectifier current (IK1) responds. Specifically, its highly non-linear current-voltage relationship changes significantly as judged by the voltage dependence of its region of outward current. This rapidly alters, and sometimes even depolarizes, Vr and can also markedly prolong the final repolarization phase of the AP, thus modulating excitability and refractoriness. (ii) A second much slower electrophysiological response (developing 5-10 minutes after [K+]o is reduced) results from alterations in the intracellular electrolyte balance. A progressive shift in intracellular [Na+]i causes a change in the outward electrogenic current generated by the Na+/K+ pump, thereby modifying Vr and AP repolarization and changing the human atrial electrophysiological substrate. In this study, these two effects were investigated quantitatively, using seven published models of the human atrial AP. This highlighted the important role of IK1 rectification when analyzing both the mechanisms by which [K+]o regulates Vr and how the AP waveform may contribute to "trigger" mechanisms within the proarrhythmic substrate. Our simulations complement and extend previous studies aimed at understanding key factors by which decreases in [K+]o can produce effects that are known to promote atrial arrhythmias in human hearts.
Collapse
Affiliation(s)
- Michael Clerx
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Gary R Mirams
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Albert J Rogers
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Sanjiv M Narayan
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Wayne R Giles
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
22
|
Huang M, Liao Z, Li X, Yang Z, Fan X, Li Y, Zhao Z, Lang S, Cyganek L, Zhou X, Akin I, Borggrefe M, El-Battrawy I. Effects of Antiarrhythmic Drugs on hERG Gating in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes From a Patient With Short QT Syndrome Type 1. Front Pharmacol 2021; 12:675003. [PMID: 34025432 PMCID: PMC8138577 DOI: 10.3389/fphar.2021.675003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Aims: The short QT syndrome type 1 (SQT1) is linked to hERG channel mutations (e.g., N588K). Drug effects on hERG channel gating kinetics in SQT1-cells have not been investigated. Methods: This study used hiPSC-CMs of a healthy donor and a SQT1-patient carrying the N588K mutation and patch clamp to examine the drug effects on hERG channel gating kinetics. Results: Ajmaline, amiodarone, ivabradine, flecainide, quinidine, mexiletine and ranolazine inhibited the hERG channel current (IKr) less strongly in hiPSC-CMs from the SQTS1-patient (SQT1-hiPSC-CMs) comparing with cells from the healthy donor (donor-hiPSC-CMs). Quinidine and mexiletine reduced, but ajmaline, amiodarone, ivabradine and ranolazine increased the time to peak of IKr similarly in SQT1-hiPSC-CMs and donor-hiPSC-CMs. Although regarding the shift of activation and inactivation curves, tested drugs showed differential effects in donor- and SQT1-hiPSC-CMs, quinidine, ajmaline, ivabradine and mexiletine but not amiodarone, flecainide and ranolazine reduced the window current in SQT1-hiPSC-CMs. Quinidine, ajmaline, ivabradine and mexiletine differentially changed the time constant of recovery from inactivation, but all of them increased the time constant of deactivation in SQT1-hiPSC-CMs. Conclusion: The window current-reducing and deactivation-slowing effects may be important for the antiarrhythmic effect of ajmaline, ivabradine, quinidine and mexiletine in SQT1-cells. This information may be helpful for selecting drugs for treating SQT1-patients with hERG channel mutation.
Collapse
Affiliation(s)
- Mengying Huang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhenxing Liao
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,North Sichuan Medical College, Nanchong, China
| | - Xin Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Yang
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,North Sichuan Medical College, Nanchong, China
| | - Xuehui Fan
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,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
| | - Yingrui Li
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Zhihan Zhao
- 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 Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany.,Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaobo Zhou
- First Department of Medicine, Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany.,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.,DZHK (German Center for Cardiovascular Research), Partner Sites, Heidelberg-Mannheim and Göttingen, 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 Sites, Heidelberg-Mannheim and Göttingen, Mannheim, 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 Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| | - 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 Sites, Heidelberg-Mannheim and Göttingen, Mannheim, Germany
| |
Collapse
|
23
|
Weaver CD, Denton JS. Next-generation inward rectifier potassium channel modulators: discovery and molecular pharmacology. Am J Physiol Cell Physiol 2021; 320:C1125-C1140. [PMID: 33826405 DOI: 10.1152/ajpcell.00548.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inward rectifying potassium (Kir) channels play important roles in both excitable and nonexcitable cells of various organ systems and could represent valuable new drug targets for cardiovascular, metabolic, immune, and neurological diseases. In nonexcitable epithelial cells of the kidney tubule, for example, Kir1.1 (KCNJ1) and Kir4.1 (KCNJ10) are linked to sodium reabsorption in the thick ascending limb of Henle's loop and distal convoluted tubule, respectively, and have been explored as novel-mechanism diuretic targets for managing hypertension and edema. G protein-coupled Kir channels (Kir3) channels expressed in the central nervous system are critical effectors of numerous signal transduction pathways underlying analgesia, addiction, and respiratory-depressive effects of opioids. The historical dearth of pharmacological tool compounds for exploring the therapeutic potential of Kir channels has led to a molecular target-based approach using high-throughput screen (HTS) of small-molecule libraries and medicinal chemistry to develop "next-generation" Kir channel modulators that are both potent and specific for their targets. In this article, we review recent efforts focused specifically on discovery and improvement of target-selective molecular probes. The reader is introduced to fluorescence-based thallium flux assays that have enabled much of this work and then provided with an overview of progress made toward developing modulators of Kir1.1 (VU590, VU591), Kir2.x (ML133), Kir3.X (ML297, GAT1508, GiGA1, VU059331), Kir4.1 (VU0134992), and Kir7.1 (ML418). We discuss what is known about the small molecules' molecular mechanisms of action, in vitro and in vivo pharmacology, and then close with our view of what critical work remains to be done.
Collapse
Affiliation(s)
- C David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Department of Chemistry, Vanderbilt University, Nashville, Tennessee.,Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee
| | - Jerod S Denton
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee.,Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee.,Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
24
|
Cano J, Zorio E, Mazzanti A, Arnau MÁ, Trenor B, Priori SG, Saiz J, Romero L. Ranolazine as an Alternative Therapy to Flecainide for SCN5A V411M Long QT Syndrome Type 3 Patients. Front Pharmacol 2020; 11:580481. [PMID: 33519442 PMCID: PMC7845660 DOI: 10.3389/fphar.2020.580481] [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: 07/06/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
The prolongation of the QT interval represents the main feature of the long QT syndrome (LQTS), a life-threatening genetic disease. The heterozygous SCN5A V411M mutation of the human sodium channel leads to a LQTS type 3 with severe proarrhythmic effects due to an increase in the late component of the sodium current (INaL). The two sodium blockers flecainide and ranolazine are equally recommended by the current 2015 ESC guidelines to treat patients with LQTS type 3 and persistently prolonged QT intervals. However, awareness of pro-arrhythmic effects of flecainide in LQTS type 3 patients arose upon the study of the SCN5A E1784K mutation. Regarding SCN5A V411M individuals, flecainide showed good results albeit in a reduced number of patients and no evidence supporting the use of ranolazine has ever been released. Therefore, we ought to compare the effect of ranolazine and flecainide in a SCN5A V411M model using an in-silico modeling and simulation approach. We collected clinical data of four patients. Then, we fitted four Markovian models of the human sodium current (INa) to experimental and clinical data. Two of them correspond to the wild type and the heterozygous SCN5A V411M scenarios, and the other two mimic the effects of flecainide and ranolazine on INa. Next, we inserted them into three isolated cell action potential (AP) models for endocardial, midmyocardial and epicardial cells and in a one-dimensional tissue model. The SCN5A V411M mutation produced a 15.9% APD90 prolongation in the isolated endocardial cell model, which corresponded to a 14.3% of the QT interval prolongation in a one-dimensional strand model, in keeping with clinical observations. Although with different underlying mechanisms, flecainide and ranolazine partially countered this prolongation at the isolated endocardial model by reducing the APD90 by 8.7 and 4.3%, and the QT interval by 7.2 and 3.2%, respectively. While flecainide specifically targeted the mutation-induced increase in peak INaL, ranolazine reduced it during the entire AP. Our simulations also suggest that ranolazine could prevent early afterdepolarizations triggered by the SCN5A V411M mutation during bradycardia, as flecainide. We conclude that ranolazine could be used to treat SCN5A V411M patients, specifically when flecainide is contraindicated.
Collapse
Affiliation(s)
- Jordi Cano
- Centro de Investigación e Innovación en Bioingeniería (CI2B), Universitat Politècnica de València, Valencia, España
| | - Esther Zorio
- Unidad de Cardiopatías Familiares y Muerte Súbita, Servicio de Cardiología, Hospital Universitario y Politécnico La Fe, Valencia, España.,Center for Biomedical Network Research on Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Andrea Mazzanti
- Molecular Cardiology, IRCCS, Istituti Clinici Scientifici Maugeri, Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Miguel Ángel Arnau
- Unidad de Cardiopatías Familiares y Muerte Súbita, Servicio de Cardiología, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - Beatriz Trenor
- Centro de Investigación e Innovación en Bioingeniería (CI2B), Universitat Politècnica de València, Valencia, España
| | - Silvia G Priori
- Molecular Cardiology, IRCCS, Istituti Clinici Scientifici Maugeri, Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Javier Saiz
- Centro de Investigación e Innovación en Bioingeniería (CI2B), Universitat Politècnica de València, Valencia, España
| | - Lucia Romero
- Centro de Investigación e Innovación en Bioingeniería (CI2B), Universitat Politècnica de València, Valencia, España
| |
Collapse
|
25
|
Zahedi K, Brooks M, Barone S, Rahmati N, Murray Stewart T, Dunworth M, Destefano-Shields C, Dasgupta N, Davidson S, Lindquist DM, Fuller CE, Smith RD, Cleveland JL, Casero RA, Soleimani M. Ablation of polyamine catabolic enzymes provokes Purkinje cell damage, neuroinflammation, and severe ataxia. J Neuroinflammation 2020; 17:301. [PMID: 33054763 PMCID: PMC7559641 DOI: 10.1186/s12974-020-01955-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Polyamine catabolism plays a key role in maintaining intracellular polyamine pools, yet its physiological significance is largely unexplored. Here, we report that the disruption of polyamine catabolism leads to severe cerebellar damage and ataxia, demonstrating the fundamental role of polyamine catabolism in the maintenance of cerebellar function and integrity. METHODS Mice with simultaneous deletion of the two principal polyamine catabolic enzymes, spermine oxidase and spermidine/spermine N1-acetyltransferase (Smox/Sat1-dKO), were generated by the crossbreeding of Smox-KO (Smox-/-) and Sat1-KO (Sat1-/-) animals. Development and progression of tissue injury was monitored using imaging, behavioral, and molecular analyses. RESULTS Smox/Sat1-dKO mice are normal at birth, but develop progressive cerebellar damage and ataxia. The cerebellar injury in Smox/Sat1-dKO mice is associated with Purkinje cell loss and gliosis, leading to neuroinflammation and white matter demyelination during the latter stages of the injury. The onset of tissue damage in Smox/Sat1-dKO mice is not solely dependent on changes in polyamine levels as cerebellar injury was highly selective. RNA-seq analysis and confirmatory studies revealed clear decreases in the expression of Purkinje cell-associated proteins and significant increases in the expression of transglutaminases and markers of neurodegenerative microgliosis and astrocytosis. Further, the α-Synuclein expression, aggregation, and polyamination levels were significantly increased in the cerebellum of Smox/Sat1-dKO mice. Finally, there were clear roles of transglutaminase-2 (TGM2) in the cerebellar pathologies manifest in Smox/Sat1-dKO mice, as pharmacological inhibition of transglutaminases reduced the severity of ataxia and cerebellar injury in Smox/Sat1-dKO mice. CONCLUSIONS These results indicate that the disruption of polyamine catabolism, via coordinated alterations in tissue polyamine levels, elevated transglutaminase activity and increased expression, polyamination, and aggregation of α-Synuclein, leads to severe cerebellar damage and ataxia. These studies indicate that polyamine catabolism is necessary to Purkinje cell survival, and for sustaining the functional integrity of the cerebellum.
Collapse
Affiliation(s)
- Kamyar Zahedi
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA.
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA.
- Research Services, Veterans Affairs Medical Center, Albuquerque, NM, 87108, USA.
- Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, 915 Camino de Salud, Bldg. 289, IDTC 3315, Albuquerque, NM, 87113, USA.
- Present Address: Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, Albuquerque, NM, 87131, USA.
| | - Marybeth Brooks
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
- Present Address: Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, Albuquerque, NM, 87131, USA
| | - Sharon Barone
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
- Research Services, Veterans Affairs Medical Center, Albuquerque, NM, 87108, USA
- Present Address: Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, Albuquerque, NM, 87131, USA
| | - Negah Rahmati
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Tracy Murray Stewart
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Matthew Dunworth
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Christina Destefano-Shields
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Nupur Dasgupta
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Steve Davidson
- Department of Anesthesiology and Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Diana M Lindquist
- Department of Radiology, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Christine E Fuller
- Upstate Medical University Department of Pathology, Syracuse, NY, 13219, USA
| | - Roger D Smith
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL, USA
| | - Robert A Casero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Manoocher Soleimani
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, 45220, USA.
- Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA.
- Research Services, Veterans Affairs Medical Center, Albuquerque, NM, 87108, USA.
- Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, 915 Camino de Salud, Bldg. 289, IDTC 3315, Albuquerque, NM, 87113, USA.
- Present Address: Department of Internal Medicine, Division of Nephrology, University of New Mexico College of Medicine, Albuquerque, NM, 87131, USA.
| |
Collapse
|
26
|
Sachdeva S, Gupta SK, Naik N. Every face tells a story-unravelling a case of bidirectional ventricular tachycardia. Indian Pacing Electrophysiol J 2020; 20:199-202. [PMID: 32615315 PMCID: PMC7517587 DOI: 10.1016/j.ipej.2020.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/02/2020] [Accepted: 06/05/2020] [Indexed: 11/29/2022] Open
Abstract
Bidirectional ventricular tachycardia is a rare form of tachycardia. We hereby report a case of bidirectional ventricular tachycardia in an 8-year-old boy wherein careful clinical exami-nation led to the diagnosis of Andersen Tawil syndrome. The case also demonstrates the efficacy of flecainide in managing bidirectional ventricular tachycardia in the setting of Andersen Tawil syndrome.
Collapse
Affiliation(s)
- Sakshi Sachdeva
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India.
| | - Saurabh Kumar Gupta
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India.
| | - Nitish Naik
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
27
|
Clinical Importance of the Human Umbilical Artery Potassium Channels. Cells 2020; 9:cells9091956. [PMID: 32854241 PMCID: PMC7565333 DOI: 10.3390/cells9091956] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Potassium (K+) channels are usually predominant in the membranes of vascular smooth muscle cells (SMCs). These channels play an important role in regulating the membrane potential and vessel contractility-a role that depends on the vascular bed. Thus, the activity of K+ channels represents one of the main mechanisms regulating the vascular tone in physiological and pathophysiological conditions. Briefly, the activation of K+ channels in SMC leads to hyperpolarization and vasorelaxation, while its inhibition induces depolarization and consequent vascular contraction. Currently, there are four different types of K+ channels described in SMCs: voltage-dependent K+ (KV) channels, calcium-activated K+ (KCa) channels, inward rectifier K+ (Kir) channels, and 2-pore domain K+ (K2P) channels. Due to the fundamental role of K+ channels in excitable cells, these channels are promising therapeutic targets in clinical practice. Therefore, this review discusses the basic properties of the various types of K+ channels, including structure, cellular mechanisms that regulate their activity, and new advances in the development of activators and blockers of these channels. The vascular functions of these channels will be discussed with a focus on vascular SMCs of the human umbilical artery. Then, the clinical importance of K+ channels in the treatment and prevention of cardiovascular diseases during pregnancy, such as gestational hypertension and preeclampsia, will be explored.
Collapse
|
28
|
Towards the Development of AgoKirs: New Pharmacological Activators to Study K ir2.x Channel and Target Cardiac Disease. Int J Mol Sci 2020; 21:ijms21165746. [PMID: 32796537 PMCID: PMC7461056 DOI: 10.3390/ijms21165746] [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: 07/10/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022] Open
Abstract
Inward rectifier potassium ion channels (IK1-channels) of the Kir2.x family are responsible for maintaining a stable negative resting membrane potential in excitable cells, but also play a role in processes of non-excitable tissues, such as bone development. IK1-channel loss-of-function, either congenital or acquired, has been associated with cardiac disease. Currently, basic research and specific treatment are hindered by the absence of specific and efficient Kir2.x channel activators. However, twelve different compounds, including approved drugs, show off-target IK1 activation. Therefore, these compounds contain valuable information towards the development of agonists of Kir channels, AgoKirs. We reviewed the mechanism of IK1 channel activation of these compounds, which can be classified as direct or indirect activators. Subsequently, we examined the most viable starting points for rationalized drug development and possible safety concerns with emphasis on cardiac and skeletal muscle adverse effects of AgoKirs. Finally, the potential value of AgoKirs is discussed in view of the current clinical applications of potentiators and activators in cystic fibrosis therapy.
Collapse
|
29
|
Walsh KB. Screening Technologies for Inward Rectifier Potassium Channels: Discovery of New Blockers and Activators. SLAS DISCOVERY 2020; 25:420-433. [PMID: 32292089 DOI: 10.1177/2472555220905558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
K+ channels play a critical role in maintaining the normal electrical activity of excitable cells by setting the cell resting membrane potential and by determining the shape and duration of the action potential. In nonexcitable cells, K+ channels establish electrochemical gradients necessary for maintaining salt and volume homeostasis of body fluids. Inward rectifier K+ (Kir) channels typically conduct larger inward currents than outward currents, resulting in an inwardly rectifying current versus voltage relationship. This property of inward rectification results from the voltage-dependent block of the channels by intracellular polyvalent cations and makes these channels uniquely designed for maintaining the resting potential near the K+ equilibrium potential (EK). The Kir family of channels consist of seven subfamilies of channels (Kir1.x through Kir7.x) that include the classic inward rectifier (Kir2.x) channel, the G-protein-gated inward rectifier K+ (GIRK) (Kir3.x), and the adenosine triphosphate (ATP)-sensitive (KATP) (Kir 6.x) channels as well as the renal Kir1.1 (ROMK), Kir4.1, and Kir7.1 channels. These channels not only function to regulate electrical/electrolyte transport activity, but also serve as effector molecules for G-protein-coupled receptors (GPCRs) and as molecular sensors for cell metabolism. Of significance, Kir channels represent promising pharmacological targets for treating a number of clinical conditions, including cardiac arrhythmias, anxiety, chronic pain, and hypertension. This review provides a brief background on the structure, function, and pharmacology of Kir channels and then focuses on describing and evaluating current high-throughput screening (HTS) technologies, such as membrane potential-sensitive fluorescent dye assays, ion flux measurements, and automated patch clamp systems used for Kir channel drug discovery.
Collapse
Affiliation(s)
- Kenneth B Walsh
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
30
|
Mazzanti A, Guz D, Trancuccio A, Pagan E, Kukavica D, Chargeishvili T, Olivetti N, Biernacka EK, Sacilotto L, Sarquella-Brugada G, Campuzano O, Nof E, Anastasakis A, Sansone VA, Jimenez-Jaimez J, Cruz F, Sánchez-Quiñones J, Hernandez-Afonso J, Fuentes ME, Średniawa B, Garoufi A, Andršová I, Izquierdo M, Marinov R, Danon A, Expósito-García V, Garcia-Fernandez A, Muñoz-Esparza C, Ortíz M, Zienciuk-Krajka A, Tavazzani E, Monteforte N, Bloise R, Marino M, Memmi M, Napolitano C, Zorio E, Monserrat L, Bagnardi V, Priori SG. Natural History and Risk Stratification in Andersen-Tawil Syndrome Type 1. J Am Coll Cardiol 2020; 75:1772-1784. [PMID: 32299589 DOI: 10.1016/j.jacc.2020.02.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/25/2020] [Accepted: 02/11/2020] [Indexed: 11/25/2022]
|
31
|
The intriguing effect of ethanol and nicotine on acetylcholine-sensitive potassium current IKAch: Insight from a quantitative model. PLoS One 2019; 14:e0223448. [PMID: 31600261 PMCID: PMC6786802 DOI: 10.1371/journal.pone.0223448] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/20/2019] [Indexed: 02/01/2023] Open
Abstract
Recent experimental work has revealed unusual features of the effect of certain drugs on cardiac inwardly rectifying potassium currents, including the constitutively active and acetylcholine-induced components of acetylcholine-sensitive current (IKAch). These unusual features have included alternating susceptibility of the current components to activation and inhibition induced by ethanol or nicotine applied at various concentrations, and significant correlation between the drug effect and the current magnitude measured under drug-free conditions. To explain these complex drug effects, we have developed a new type of quantitative model to offer a possible interpretation of the effect of ethanol and nicotine on the IKAch channels. The model is based on a description of IKAch as a sum of particular currents related to the populations of channels formed by identical assemblies of different α-subunits. Assuming two different channel populations in agreement with the two reported functional IKAch-channels (GIRK1/4 and GIRK4), the model was able to simulate all the above-mentioned characteristic features of drug-channel interactions and also the dispersion of the current measured in different cells. The formulation of our model equations allows the model to be incorporated easily into the existing integrative models of electrical activity of cardiac cells involving quantitative description of IKAch. We suppose that the model could also help make sense of certain observations related to the channels that do not show inward rectification. This new ionic channel model, based on a concept we call population type, may allow for the interpretation of complex interactions of drugs with ionic channels of various types, which cannot be done using the ionic channel models available so far.
Collapse
|
32
|
Sato A, Takano T, Chinushi M, Minamino T. Usefulness of the intravenous flecainide challenge test before oral flecainide treatment in a patient with Andersen-Tawil syndrome. BMJ Case Rep 2019; 12:12/7/e229628. [PMID: 31311787 DOI: 10.1136/bcr-2019-229628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Andersen-Tawil syndrome (ATS) is an inherited disorder characterised by the triad of ventricular arrhythmias (VAs), periodic paralysis and dysmorphic features. A 31-year-old woman diagnosed with ATS caused by a KCNJ2 mutation (p.R228ins) was urgently admitted to our hospital following an episode of syncope during exercise. Electrocardiography revealed frequent premature ventricular complexes and non-sustained ventricular tachycardias (VTs) with pleomorphic QRS patterns. During the intravenous flecainide test (30 mg), the frequent VAs were inhibited completely. After oral flecainide (100 mg) was started, VAs, except for a brief bigeminy, were suppressed during the exercise test. On 24-hour Holter recordings, the VAs decreased from 50 133 to 13 363 beats/day (-73%). Sustained VT and syncope were not observed during a 3-year follow-up period. Intravenous flecainide challenge test may be useful in predicting the efficacy of oral flecainide treatment for patients with ATS.
Collapse
Affiliation(s)
- Akinori Sato
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Toshiki Takano
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masaomi Chinushi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Graduate School of Health Science, Niigata University School of Medicine, Niigata, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
33
|
Jackson WF. Boosting the signal: Endothelial inward rectifier K + channels. Microcirculation 2018; 24. [PMID: 27652592 DOI: 10.1111/micc.12319] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/12/2016] [Indexed: 12/19/2022]
Abstract
Endothelial cells express a diverse array of ion channels including members of the strong inward rectifier family composed of KIR 2 subunits. These two-membrane spanning domain channels are modulated by their lipid environment, and exist in macromolecular signaling complexes with receptors, protein kinases and other ion channels. Inward rectifier K+ channel (KIR ) currents display a region of negative slope conductance at membrane potentials positive to the K+ equilibrium potential that allows outward current through the channels to be activated by membrane hyperpolarization, permitting KIR to amplify hyperpolarization induced by other K+ channels and ion transporters. Increases in extracellular K+ concentration activate KIR allowing them to sense extracellular K+ concentration and transduce this change into membrane hyperpolarization. These properties position KIR to participate in the mechanism of action of hyperpolarizing vasodilators and contribute to cell-cell conduction of hyperpolarization along the wall of microvessels. The expression of KIR in capillaries in electrically active tissues may allow KIR to sense extracellular K+ , contributing to functional hyperemia. Understanding the regulation of expression and function of microvascular endothelial KIR will improve our understanding of the control of blood flow in the microcirculation in health and disease and may provide new targets for the development of therapeutics in the future.
Collapse
Affiliation(s)
- William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
34
|
Different responses to exercise between Andersen–Tawil syndrome and catecholaminergic polymorphic ventricular tachycardia. Europace 2017; 20:1675-1682. [DOI: 10.1093/europace/eux351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/31/2017] [Indexed: 11/15/2022] Open
|
35
|
Utrilla RG, Nieto-Marín P, Alfayate S, Tinaquero D, Matamoros M, Pérez-Hernández M, Sacristán S, Ondo L, de Andrés R, Díez-Guerra FJ, Tamargo J, Delpón E, Caballero R. Kir2.1-Nav1.5 Channel Complexes Are Differently Regulated than Kir2.1 and Nav1.5 Channels Alone. Front Physiol 2017; 8:903. [PMID: 29184507 PMCID: PMC5694551 DOI: 10.3389/fphys.2017.00903] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/25/2017] [Indexed: 12/27/2022] Open
Abstract
Cardiac Kir2.1 and Nav1.5 channels generate the inward rectifier K+ (IK1) and the Na+ (INa) currents, respectively. There is a mutual interplay between the ventricular INa and IK1 densities, because Nav1.5 and Kir2.1 channels exhibit positive reciprocal modulation. Here we compared some of the biological properties of Nav1.5 and Kir2.1 channels when they are expressed together or separately to get further insights regarding their putative interaction. First we demonstrated by proximity ligation assays (PLAs) that in the membrane of ventricular myocytes Nav1.5 and Kir2.1 proteins are in close proximity to each other (<40 nm apart). Furthermore, intracellular dialysis with anti-Nav1.5 and anti-Kir2.1 antibodies suggested that these channels form complexes. Patch-clamp experiments in heterologous transfection systems demonstrated that the inhibition of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) decreased the INa and the IK1 generated by Nav1.5 and Kir2.1 channels when they were coexpressed, but not the IK1 generated by Kir2.1 channels alone, suggesting that complexes, but not Kir2.1 channels, are a substrate of CaMKII. Furthermore, inhibition of CaMKII precluded the interaction between Nav1.5 and Kir2.1 channels. Inhibition of 14-3-3 proteins did not modify the INa and IK1 densities generated by each channel separately, whereas it decreased the INa and IK1 generated when they were coexpressed. However, inhibition of 14-3-3 proteins did not abolish the Nav1.5-Kir2.1 interaction. Inhibition of dynamin-dependent endocytosis reduced the internalization of Kir2.1 but not of Nav1.5 or Kir2.1-Nav1.5 complexes. Inhibition of cytoskeleton-dependent vesicular trafficking via the dynein/dynactin motor increased the IK1, but reduced the INa, thus suggesting that the dynein/dynactin motor is preferentially involved in the backward and forward traffic of Kir2.1 and Nav1.5, respectively. Conversely, the dynein/dynactin motor participated in the forward movement of Kir2.1-Nav1.5 complexes. Ubiquitination by Nedd4-2 ubiquitin-protein ligase promoted the Nav1.5 degradation by the proteasome, but not that of Kir2.1 channels. Importantly, the Kir2.1-Nav1.5 complexes were degraded following this route as demonstrated by the overexpression of Nedd4-2 and the inhibition of the proteasome with MG132. These results suggested that Kir2.1 and Nav1.5 channels closely interact with each other leading to the formation of a pool of complexed channels whose biology is similar to that of the Nav1.5 channels.
Collapse
Affiliation(s)
- Raquel G Utrilla
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Paloma Nieto-Marín
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Silvia Alfayate
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - David Tinaquero
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Marcos Matamoros
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Marta Pérez-Hernández
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | | | - Lorena Ondo
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Raquel de Andrés
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - F Javier Díez-Guerra
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Tamargo
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain.,CIBERCV, Madrid, Spain
| |
Collapse
|
36
|
Šimurda J, Šimurdová M, Bébarová M. Inward rectifying potassium currents resolved into components: modeling of complex drug actions. Pflugers Arch 2017; 470:315-325. [DOI: 10.1007/s00424-017-2071-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/12/2017] [Accepted: 09/18/2017] [Indexed: 10/18/2022]
|
37
|
El-Sherif N, Turitto G, Boutjdir M. Congenital Long QT syndrome and torsade de pointes. Ann Noninvasive Electrocardiol 2017; 22. [PMID: 28670758 DOI: 10.1111/anec.12481] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022] Open
Abstract
Since its initial description by Jervell and Lange-Nielsen in 1957, the congenital long QT syndrome (LQTS) has been the most investigated cardiac ion channelopathy. A prolonged QT interval in the surface electrocardiogram is the sine qua non of the LQTS and is a surrogate measure of the ventricular action potential duration (APD). Congenital as well as acquired alterations in certain cardiac ion channels can affect their currents in such a way as to increase the APD and hence the QT interval. The inhomogeneous lengthening of the APD across the ventricular wall results in dispersion of APD. This together with the tendency of prolonged APD to be associated with oscillations at the plateau level, termed early afterdepolarizations (EADs), provides the substrate of ventricular tachyarrhythmia associated with LQTS, usually referred to as torsade de pointes (TdP) VT. This review will discuss the genetic, molecular, and phenotype characteristics of congenital LQTS as well as current management strategies and future directions in the field.
Collapse
Affiliation(s)
- Nabil El-Sherif
- Downstate Medical Center, State University of New York, Brooklyn, NY, USA.,VA NY Harbor Healthcare System, Brooklyn, NY, USA
| | - Gioia Turitto
- NewYork-Presbyterian Brooklyn Methodist Hospital, New York, NY, USA
| | - Mohamed Boutjdir
- Downstate Medical Center, State University of New York, Brooklyn, NY, USA.,VA NY Harbor Healthcare System, Brooklyn, NY, USA.,NYU School of Medicine, New York, NY, USA
| |
Collapse
|
38
|
Nicotine at clinically relevant concentrations affects atrial inward rectifier potassium current sensitive to acetylcholine. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:471-481. [DOI: 10.1007/s00210-017-1341-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/10/2017] [Indexed: 10/20/2022]
|
39
|
Abstract
Long QT syndrome (LQTS) exhibits great phenotype variability among family members carrying the same mutation, which can be partially attributed to genetic factors. We functionally analyzed the KCNH2 (encoding for Kv11.1 or hERG channels) and TBX20 (encoding for the transcription factor Tbx20) variants found by next-generation sequencing in two siblings with LQTS in a Spanish family of African ancestry. Affected relatives harbor a heterozygous mutation in KCNH2 that encodes for p.T152HfsX180 Kv11.1 (hERG). This peptide, by itself, failed to generate any current when transfected into Chinese hamster ovary (CHO) cells but, surprisingly, exerted "chaperone-like" effects over native hERG channels in both CHO cells and mouse atrial-derived HL-1 cells. Therefore, heterozygous transfection of native (WT) and p.T152HfsX180 hERG channels generated a current that was indistinguishable from that generated by WT channels alone. Some affected relatives also harbor the p.R311C mutation in Tbx20. In human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), Tbx20 enhanced human KCNH2 gene expression and hERG currents (IhERG) and shortened action-potential duration (APD). However, Tbx20 did not modify the expression or activity of any other channel involved in ventricular repolarization. Conversely, p.R311C Tbx20 did not increase KCNH2 expression in hiPSC-CMs, which led to decreased IhERG and increased APD. Our results suggest that Tbx20 controls the expression of hERG channels responsible for the rapid component of the delayed rectifier current. On the contrary, p.R311C Tbx20 specifically disables the Tbx20 protranscriptional activity over KCNH2 Therefore, TBX20 can be considered a KCNH2-modifying gene.
Collapse
|
40
|
Van Ert HA, McCune EC, Orland KM, Maginot KR, Von Bergen NH, January CT, Eckhardt LL. Flecainide treats a novel KCNJ2 mutation associated with Andersen-Tawil syndrome. HeartRhythm Case Rep 2016; 3:151-154. [PMID: 28491792 PMCID: PMC5420046 DOI: 10.1016/j.hrcr.2016.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Hanora A Van Ert
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Elise C McCune
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kate M Orland
- University of Wisconsin-Madison Inherited Arrhythmia Clinic, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kathleen R Maginot
- University of Wisconsin-Madison Inherited Arrhythmia Clinic, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Nicholas H Von Bergen
- University of Wisconsin-Madison Inherited Arrhythmia Clinic, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Craig T January
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison Inherited Arrhythmia Clinic, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lee L Eckhardt
- Cellular and Molecular Arrhythmia Research Program, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin-Madison Inherited Arrhythmia Clinic, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
41
|
Filgueiras-Rama D, Jalife J. STRUCTURAL AND FUNCTIONAL BASES OF CARDIAC FIBRILLATION. DIFFERENCES AND SIMILARITIES BETWEEN ATRIA AND VENTRICLES. JACC Clin Electrophysiol 2016; 2:1-3. [PMID: 27042693 DOI: 10.1016/j.jacep.2015.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Evidence accumulated over the last 25 years suggests that, whether in the atria or ventricles, fibrillation may be explained by the self-organization of the cardiac electrical activity into rapidly spinning rotors giving way to spiral waves that break intermittently and result in fibrillatory conduction. The dynamics and frequency of such rotors depend on the ion channel composition, excitability and refractory properties of the tissues involved, as well as on the thickness and respective three-dimensional fiber structure of the atrial and ventricular chambers. Therefore, improving the understanding of fibrillation has required the use of multidisciplinary research approaches, including optical mapping, patch clamping and molecular biology, and the application of concepts derived from the theory of wave propagation in excitable media. Moreover, translation of such concepts to the clinic has recently opened new opportunities to apply novel mechanistic approaches to therapy, particularly during atrial fibrillation ablation. Here we review the current understanding of the manner in which the underlying myocardial structure and function influence rotor initiation and maintenance during cardiac fibrillation. We also examine relevant underlying differences and similarities between atrial fibrillation and ventricular fibrillation and evaluate the latest clinical mapping technologies used to identify rotors in either arrhythmia. Altogether, the data being discussed have significantly improved our understanding of the cellular and structural bases of cardiac fibrillation and pointed toward potentially exciting new avenues for more efficient and effective identification and therapy of the most complex cardiac arrhythmias.
Collapse
Affiliation(s)
- David Filgueiras-Rama
- Fundación Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC)., Myocardial Pathophysiology Area, Madrid, Spain; Hospital Clínico San Carlos, Cardiology department, Madrid, Spain
| | - José Jalife
- Fundación Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC)., Myocardial Pathophysiology Area, Madrid, Spain; Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, EEUU
| |
Collapse
|
42
|
Matamoros M, Pérez-Hernández M, Guerrero-Serna G, Amorós I, Barana A, Núñez M, Ponce-Balbuena D, Sacristán S, Gómez R, Tamargo J, Caballero R, Jalife J, Delpón E. Nav1.5 N-terminal domain binding to α1-syntrophin increases membrane density of human Kir2.1, Kir2.2 and Nav1.5 channels. Cardiovasc Res 2016; 110:279-90. [PMID: 26786162 DOI: 10.1093/cvr/cvw009] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/13/2016] [Indexed: 01/11/2023] Open
Abstract
AIMS Cardiac excitability and refractoriness are largely determined by the function and number of inward rectifier K⁺ channels (Kir2.1-2.3), which are differentially expressed in the atria and ventricles, and Nav1.5 channels. We have focused on how Nav1.5 and Kir2.x function within a macromolecular complex by elucidating the molecular determinants that govern Nav1.5/Kir2.x reciprocal modulation. METHODS AND RESULTS The results demonstrate that there is an unexpected 'internal' PDZ-like binding domain located at the N-terminus of the Nav1.5 channel that mediates its binding to α1-syntrophin. Nav1.5 N-terminal domain, by itself (the 132 aa peptide) (Nter), exerts a 'chaperone-like' effect that increases sodium (I(Na)) and inward rectifier potassium (I(K1)) currents by enhancing the expression of Nav1.5, Kir2.1, and Kir2.2 channels as demonstrated in Chinese hamster ovary (CHO) cells and in rat cardiomyocytes. Site-directed mutagenesis analysis demonstrates that the Nter chaperone-like effect is determined by Serine 20. Nav1.5-Kir2.x reciprocal positive interactions depend on a specific C-terminal PDZ-binding domain sequence (SEI), which is present in Kir2.1 and Kir2.2 channels but not in Kir2.3. Therefore, in human atrial myocytes, the presence of Kir2.3 isoforms precludes reciprocal I(K1)-INa density modulation. Moreover, results in rat and human atrial myocytes demonstrate that binding to α1-syntrophin is necessary for the Nav1.5-Kir2.x-positive reciprocal modulation. CONCLUSIONS The results demonstrate the critical role of the N-terminal domain of Nav1.5 channels in Nav1.5-Kir2.x-reciprocal interactions and suggest that the molecular mechanisms controlling atrial and ventricular cellular excitability may be different.
Collapse
Affiliation(s)
- Marcos Matamoros
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Marta Pérez-Hernández
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Guadalupe Guerrero-Serna
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA Department of Molecular and Integrative Physiology, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA
| | - Irene Amorós
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Adriana Barana
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Mercedes Núñez
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Daniela Ponce-Balbuena
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA Department of Molecular and Integrative Physiology, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sandra Sacristán
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Ricardo Gómez
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - José Jalife
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA Department of Molecular and Integrative Physiology, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| |
Collapse
|
43
|
Abstract
In the last decade, there have been considerable advances in the understanding of the pathophysiology of malignant ventricular tachyarrhythmias (VT) and sudden cardiac death (SCD). Over 80% of SCD occurs in patients with organic heart disease. However, approximately 10%-15% of SCD occurs in the presence of structurally normal heart, and the majority of these patients are young. In this group of patients, changes in genes encoding cardiac ion channels produce modifications of the function of the channel resulting in an electrophysiological substrate of VT and SCD. Collectively, these disorders are referred to as cardiac ion channelopathies. The four major syndromes in this group are: the long QT syndrome (LQTS), the Brugada syndrome (BrS), the short QT syndrome (SQTS), and the catecholaminergic polymorphic ventricular tachycardia (CPVT). Each of these syndromes includes multiple subtypes with different and sometimes complex cardiac ion channel genetic abnormalities. Many are associated with other somatic and neurological abnormalities besides the risk of VT and SCD. The current management of cardiac ion channelopathies can be summarized as follows: (1) in symptomatic patients, the implantable cardioverter defibrillator (ICD) is the only viable option; (2) in asymptomatic patients, risk stratification is necessary, followed by either the ICD, pharmacotherapy, or a combination of both. A genotype-specific approach to pharmacotherapy requires a thorough understanding of the molecular-cellular basis of arrhythmogenesis in cardiac ion channelopathies as well as the specific drug profile.
Collapse
|
44
|
Tadros R, Cadrin-Tourigny J, Abadir S, Rivard L, Nattel S, Talajic M, Khairy P. Pharmacotherapy for inherited arrhythmia syndromes: mechanistic basis, clinical trial evidence and practical application. Expert Rev Cardiovasc Ther 2015; 13:769-82. [DOI: 10.1586/14779072.2015.1049156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Miyamoto K, Aiba T, Kimura H, Hayashi H, Ohno S, Yasuoka C, Tanioka Y, Tsuchiya T, Yoshida Y, Hayashi H, Tsuboi I, Nakajima I, Ishibashi K, Okamura H, Noda T, Ishihara M, Anzai T, Yasuda S, Miyamoto Y, Kamakura S, Kusano K, Ogawa H, Horie M, Shimizu W. Efficacy and safety of flecainide for ventricular arrhythmias in patients with Andersen-Tawil syndrome with KCNJ2 mutations. Heart Rhythm 2015; 12:596-603. [DOI: 10.1016/j.hrthm.2014.12.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Indexed: 11/24/2022]
|
46
|
Daimi H, Lozano-Velasco E, Haj Khelil A, Chibani JBE, Barana A, Amorós I, González de la Fuente M, Caballero R, Aranega A, Franco D. Regulation of SCN5A by microRNAs: miR-219 modulates SCN5A transcript expression and the effects of flecainide intoxication in mice. Heart Rhythm 2015; 12:1333-42. [PMID: 25701775 DOI: 10.1016/j.hrthm.2015.02.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND The human cardiac action potential in atrial and ventricular cells is initiated by a fast-activating, fast-inactivating sodium current generated by the SCN5A/Nav1.5 channel in association with its β1/SCN1B subunit. The role of Nav1.5 in the etiology of many cardiac diseases strongly suggests that proper regulation of cell biology and function of the channel is critical for normal cardiac function. Hence, numerous recent studies have focused on the regulatory mechanisms of Nav1.5 biosynthetic and degradation processes as well as its subcellular localization. OBJECTIVE The purpose of this study was to investigate the role of microRNAs in the Scn5a/Nav1.5 posttranscriptional regulation. METHODS Quantitative polymerase chain reaction, immunohistochemical and electrophysiological measurements of distinct microRNA gain-of-function experiments in cardiomyocytes for the assessment of Scn5a expression. RESULTS Functional studies of HL-1 cardiomyocytes and luciferase assays in fibroblasts demonstrate that Scn5a is directly (miR-98, miR-106, miR-200, and miR-219) and indirectly (miR-125 and miR-153) regulated by multiple microRNAs displaying distinct time-dependent profiles. Cotransfection experiments demonstrated that miR-219 and miR-200 have independent opposite effects on Scn5a expression modulation. Of all the microRNAs studied, only miR-219 increases Scn5a expression levels, leading to altered contraction rhythm of HL-1 cardiomyocytes. Electrophysiological analyses in HL-1 cells revealed that miR-219 increases the sodium current. In vivo administration of miR-219 does not alter normal cardiac rhythm, but abolishes some of the effects of flecainide intoxication in mice, particularly QRS prolongation. CONCLUSION This study demonstrates the involvement of multiple microRNAs in the regulation of Scn5a. Particularly, miR-219 increases Scn5a/Nav1.5 transcript and protein expression. Our data suggest that microRNAs, such as miR-219, constitute a promising therapeutical tool to treat sodium cardiac arrhythmias.
Collapse
Affiliation(s)
- Houria Daimi
- Department of Experimental Biology, University of Jaén, Jáen, Spain; Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | | | - Amel Haj Khelil
- Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Jemni B E Chibani
- Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Adriana Barana
- Department of Pharmacology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Irene Amorós
- Department of Pharmacology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | - Ricardo Caballero
- Department of Pharmacology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Amelia Aranega
- Department of Experimental Biology, University of Jaén, Jáen, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaén, Jáen, Spain.
| |
Collapse
|
47
|
Insights in KIR2.1 channel structure and function by an evolutionary approach; cloning and functional characterization of the first reptilian inward rectifier channel KIR2.1, derived from the California kingsnake (Lampropeltis getula californiae). Biochem Biophys Res Commun 2014; 452:992-7. [DOI: 10.1016/j.bbrc.2014.09.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/08/2014] [Indexed: 11/21/2022]
|
48
|
Gómez R, Caballero R, Barana A, Amorós I, De Palm SH, Matamoros M, Núñez M, Pérez-Hernández M, Iriepa I, Tamargo J, Delpón E. Structural basis of drugs that increase cardiac inward rectifier Kir2.1 currents. Cardiovasc Res 2014; 104:337-46. [PMID: 25205296 DOI: 10.1093/cvr/cvu203] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS We hypothesize that some drugs, besides flecainide, increase the inward rectifier current (IK1) generated by Kir2.1 homotetramers (IKir2.1) and thus, exhibit pro- and/or antiarrhythmic effects particularly at the ventricular level. To test this hypothesis, we analysed the effects of propafenone, atenolol, dronedarone, and timolol on Kir2.x channels. METHODS AND RESULTS Currents were recorded with the patch-clamp technique using whole-cell, inside-out, and cell-attached configurations. Propafenone (0.1 nM-1 µM) did not modify either IK1 recorded in human right atrial myocytes or the current generated by homo- or heterotetramers of Kir2.2 and 2.3 channels recorded in transiently transfected Chinese hamster ovary cells. On the other hand, propafenone increased IKir2.1 (EC50 = 12.0 ± 3.0 nM) as a consequence of its interaction with Cys311, an effect which decreased inward rectification of the current. Propafenone significantly increased mean open time and opening frequency at all the voltages tested, resulting in a significant increase of the mean open probability of the channel. Timolol, which interacted with Cys311, was also able to increase IKir2.1. On the contrary, neither atenolol nor dronedarone modified IKir2.1. Molecular modelling of the Kir2.1-drugs interaction allowed identification of the pharmacophore of drugs that increase IKir2.1. CONCLUSIONS Kir2.1 channels exhibit a binding site determined by Cys311 that is responsible for drug-induced IKir2.1 increase. Drug binding decreases channel affinity for polyamines and current rectification, and can be a mechanism of drug-induced pro- and antiarrhythmic effects not considered until now.
Collapse
Affiliation(s)
- Ricardo Gómez
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Adriana Barana
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Irene Amorós
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Sue-Haida De Palm
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Marcos Matamoros
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Mercedes Núñez
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| | - Marta Pérez-Hernández
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Isabel Iriepa
- Department of Organic Chemistry, School of Pharmacy, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid 28040, Spain Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid 28040, Spain
| |
Collapse
|
49
|
KCNJ2 mutation causes an adrenergic-dependent rectification abnormality with calcium sensitivity and ventricular arrhythmia. Heart Rhythm 2014; 11:885-94. [PMID: 24561538 DOI: 10.1016/j.hrthm.2014.02.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Indexed: 11/23/2022]
Abstract
BACKGROUND KCNJ2 mutations are associated with a variety of inherited arrhythmia syndromes including catecholaminergic polymorphic ventricular tachycardia 3. OBJECTIVE To characterize the detailed cellular mechanisms of the clinically recognized KCNJ2 mutation R67Q. METHODS Kir2.1 current density was measured from COS-1 cells transiently transfected with wild-type human Kir-2.1 (WT-Kir2.1) and/or a heterozygous missense mutation in KCNJ2 (R67Q-Kir2.1) by using the whole-cell voltage clamp technique. Catecholamine activity was simulated with protein kinase A-stimulating cocktail exposure. Phosphorylation-deficient mutants, S425N-Kir2.1 and S425N-Kir2.1/R67Q-S425N-Kir2.1, were used in a separate set of experiments. HA- or Myc-Tag-WT-Kir2.1 and HA-Tag-R67Q-Kir2.1 were used for confocal imaging. RESULTS A 33-year-old woman presented with a catecholaminergic polymorphic ventricular tachycardia-like clinical phenotype and was found to have KCNJ2 missense mutation R67Q. Treatment with nadolol and flecainide resulted in the complete suppression of arrhythmias and symptom resolution. Under baseline conditions, R67Q-Kir2.1 expressed alone did not produce inward rectifier current while cells coexpressing WT-Kir2.1 and R67Q-Kir2.1 demonstrated the rectification index (RI) similar to that of WT-Kir2.1. After PKA stimulation, R67Q-Kir2.1/WT-Kir2.1 failed to increase peak outward current density; WT-Kir2.1 increased by 46% (n = 5), while R67Q-Kir2.1/WT-Kir2.1 decreased by 6% (n = 6) (P = .002). Rectification properties in R67Q-Kir2.1/WT-Kir2.1 demonstrated sensitivity to calcium with a decreased RI in the high-calcium pipette solution (RI 20.3% ± 4.1%) than in the low-calcium pipette solution (RI 36.5% ± 5.7%) (P < .05). Immunostaining of WT-Kir2.1 and R67Q-Kir2.1 individually and together showed a normal membrane expression pattern and colocalization by using the Pearson correlation coefficient. CONCLUSIONS R67Q-Kir2.1 is associated with an adrenergic-dependent clinical and cellular phenotype with rectification abnormality enhanced by increased calcium. These findings are a significant advancement of our knowledge and understanding of the phenotype-genotype relationship of arrhythmia syndromes related to KCNJ2 mutations.
Collapse
|
50
|
Druggability of the inward rectifier family: a hope for rare channelopathies? Future Med Chem 2014; 6:971-3. [PMID: 24547691 DOI: 10.4155/fmc.14.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|