1
|
Bains S, Giammarino L, Nimani S, Alerni N, Tester DJ, Kim CSJ, Christoforou N, Louradour J, Horváth A, Beslac O, Barbieri M, Matas L, Hof TS, Lopez R, Perez-Feliz S, Parodi C, Garcia Casalta LG, Jurgensen J, Barry MA, Bego M, Keyes L, Owens J, Pinkstaff J, Koren G, Zehender M, Brunner M, Casoni D, Praz F, Haeberlin A, Brooks G, Ackerman MJ, Odening KE. KCNQ1 suppression-replacement gene therapy in transgenic rabbits with type 1 long QT syndrome. Eur Heart J 2024; 45:3751-3763. [PMID: 39115049 PMCID: PMC11439107 DOI: 10.1093/eurheartj/ehae476] [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: 01/24/2024] [Revised: 05/20/2024] [Accepted: 07/12/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND AND AIMS Type 1 long QT syndrome (LQT1) is caused by pathogenic variants in the KCNQ1-encoded Kv7.1 potassium channels, which pathologically prolong ventricular action potential duration (APD). Herein, the pathologic phenotype in transgenic LQT1 rabbits is rescued using a novel KCNQ1 suppression-replacement (SupRep) gene therapy. METHODS KCNQ1-SupRep gene therapy was developed by combining into a single construct a KCNQ1 shRNA (suppression) and an shRNA-immune KCNQ1 cDNA (replacement), packaged into adeno-associated virus serotype 9, and delivered in vivo via an intra-aortic root injection (1E10 vg/kg). To ascertain the efficacy of SupRep, 12-lead electrocardiograms were assessed in adult LQT1 and wild-type (WT) rabbits and patch-clamp experiments were performed on isolated ventricular cardiomyocytes. RESULTS KCNQ1-SupRep treatment of LQT1 rabbits resulted in significant shortening of the pathologically prolonged QT index (QTi) towards WT levels. Ventricular cardiomyocytes isolated from treated LQT1 rabbits demonstrated pronounced shortening of APD compared to LQT1 controls, leading to levels similar to WT (LQT1-UT vs. LQT1-SupRep, P < .0001, LQT1-SupRep vs. WT, P = ns). Under β-adrenergic stimulation with isoproterenol, SupRep-treated rabbits demonstrated a WT-like physiological QTi and APD90 behaviour. CONCLUSIONS This study provides the first animal-model, proof-of-concept gene therapy for correction of LQT1. In LQT1 rabbits, treatment with KCNQ1-SupRep gene therapy normalized the clinical QTi and cellular APD90 to near WT levels both at baseline and after isoproterenol. If similar QT/APD correction can be achieved with intravenous administration of KCNQ1-SupRep gene therapy in LQT1 rabbits, these encouraging data should compel continued development of this gene therapy for patients with LQT1.
Collapse
Affiliation(s)
- Sahej Bains
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine, and Molecular Pharmacology & Experimental Therapeutics, Divisions of Heart Rhythm Services and Pediatric Cardiology, Windland Smith Rice Genetic Heart Rhythm Clinic and The Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Guggenheim 501, Rochester, MN 55905, USA
| | - Lucilla Giammarino
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Saranda Nimani
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Nicolo Alerni
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - David J Tester
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine, and Molecular Pharmacology & Experimental Therapeutics, Divisions of Heart Rhythm Services and Pediatric Cardiology, Windland Smith Rice Genetic Heart Rhythm Clinic and The Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Guggenheim 501, Rochester, MN 55905, USA
| | - C S John Kim
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine, and Molecular Pharmacology & Experimental Therapeutics, Divisions of Heart Rhythm Services and Pediatric Cardiology, Windland Smith Rice Genetic Heart Rhythm Clinic and The Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Guggenheim 501, Rochester, MN 55905, USA
| | | | - Julien Louradour
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - András Horváth
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Olgica Beslac
- Department of Cardiology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Miriam Barbieri
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Lluis Matas
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Thomas S Hof
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Ruben Lopez
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| | - Stefanie Perez-Feliz
- Department of Cardiology, University Heart Center, University Hospital Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Chiara Parodi
- Experimental Surgical Facility, Experimental Animal Center, University of Bern, Bern, Switzerland
| | - Luisana G Garcia Casalta
- Experimental Surgical Facility, Experimental Animal Center, University of Bern, Bern, Switzerland
| | - Jacqulyn Jurgensen
- Department of Virology & Gene Therapy, Vector and Vaccine Engineering Laboratory, Mayo Clinic, Rochester, USA
| | - Michael A Barry
- Department of Virology & Gene Therapy, Vector and Vaccine Engineering Laboratory, Mayo Clinic, Rochester, USA
| | - Mariana Bego
- Formerly from Biomedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Lisa Keyes
- Formerly from Biomedicine Design, Pfizer Inc., Cambridge, MA, USA
| | - Jane Owens
- Formerly from the Rare Disease Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - Jason Pinkstaff
- Drug Safety Research & Development, Pfizer Inc., Cambridge, MA, USA
| | - Gideon Koren
- Cardiovascular Research Center, Brown University, Providence, USA
| | - Manfred Zehender
- Department of Cardiology, University Heart Center, University Hospital Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Michael Brunner
- Department of Cardiology, University Heart Center, University Hospital Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
- Department of Cardiology and Intensive Care, St. Josefskrankenhaus Freiburg, Freiburg, Germany
| | - Daniela Casoni
- Experimental Surgical Facility, Experimental Animal Center, University of Bern, Bern, Switzerland
| | - Fabien Praz
- Department of Cardiology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Andreas Haeberlin
- Department of Cardiology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Gabriel Brooks
- Formerly from the Rare Disease Research Unit, Pfizer Inc., Cambridge, MA, USA
| | - Michael J Ackerman
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine, and Molecular Pharmacology & Experimental Therapeutics, Divisions of Heart Rhythm Services and Pediatric Cardiology, Windland Smith Rice Genetic Heart Rhythm Clinic and The Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Guggenheim 501, Rochester, MN 55905, USA
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| |
Collapse
|
2
|
Zhao Z, Zang X, Niu K, Song W, Wang X, Mügge A, Aweimer A, Hamdani N, Zhou X, Zhao Y, Akin I, El-Battrawy I. Impacts of gene variants on drug effects-the foundation of genotype-guided pharmacologic therapy for long QT syndrome and short QT syndrome. EBioMedicine 2024; 103:105108. [PMID: 38653189 PMCID: PMC11041837 DOI: 10.1016/j.ebiom.2024.105108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 03/20/2024] [Accepted: 03/24/2024] [Indexed: 04/25/2024] Open
Abstract
The clinical significance of optimal pharmacotherapy for inherited arrhythmias such as short QT syndrome (SQTS) and long QT syndrome (LQTS) has been increasingly recognised. The advancement of gene technology has opened up new possibilities for identifying genetic variations and investigating the pathophysiological roles and mechanisms of genetic arrhythmias. Numerous variants in various genes have been proven to be causative in genetic arrhythmias. Studies have demonstrated that the effectiveness of certain drugs is specific to the patient or genotype, indicating the important role of gene-variants in drug response. This review aims to summarize the reported data on the impact of different gene-variants on drug response in SQTS and LQTS, as well as discuss the potential mechanisms by which gene-variants alter drug response. These findings may provide valuable information for future studies on the influence of gene variants on drug efficacy and the development of genotype-guided or precision treatment for these diseases.
Collapse
Affiliation(s)
- Zhihan Zhao
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Xiaobiao Zang
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Kerun Niu
- Department of Orthopaedic, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Weifeng Song
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Xianqing Wang
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Andreas Mügge
- Department of Cardiology and Angiology, Bergmannsheil University Hospitals, Ruhr University of Bochum, 44789, Bochum, Germany
| | - Assem Aweimer
- Institute of Physiology, Department of Cellular and Translational Physiology, Medical Faculty and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - Nazha Hamdani
- Institute of Physiology, Department of Cellular and Translational Physiology, Medical Faculty and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- HCEMM-Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, University of Budapest, Budapest, Hungary
- Department of Physiology, Cardiovascular Research Institute Maastricht University Maastricht, Maastricht, the Netherlands
| | - Xiaobo Zhou
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Medical Centre Mannheim, Heidelberg University, 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, Sichuan, China
| | - Yonghui Zhao
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Medical Centre Mannheim, Heidelberg University, Germany
| | - Ibrahim El-Battrawy
- Department of Cardiology and Angiology, Bergmannsheil University Hospitals, Ruhr University of Bochum, 44789, Bochum, Germany
- Institute of Physiology, Department of Cellular and Translational Physiology, Medical Faculty and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
3
|
Jiang Y, Senyuk V, Ma K, Chen H, Qin X, Li S, Liu Y, Gentile S, Minshall RD. Pharmacological Activation of Potassium Channel Kv11.1 with NS1643 Attenuates Triple Negative Breast Cancer Cell Migration by Promoting the Dephosphorylation of Caveolin-1. Cells 2022; 11:2461. [PMID: 35954304 PMCID: PMC9368491 DOI: 10.3390/cells11152461] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 11/17/2022] Open
Abstract
The prevention of metastasis is a central goal of cancer therapy. Caveolin-1 (Cav-1) is a structural membrane and scaffolding protein shown to be a key regulator of late-stage breast cancer metastasis. However, therapeutic strategies targeting Cav-1 are still lacking. Here, we demonstrate that the pharmacological activation of potassium channel Kv11.1, which is uniquely expressed in MDA-MB-231 triple negative breast cancer cells (TNBCs) but not in normal MCF-10A cells, induces the dephosphorylation of Cav-1 Tyr-14 by promoting the Ca2+-dependent stimulation of protein tyrosine phosphatase 1B (PTP1B). Consequently, the dephosphorylation of Cav-1 resulted in its disassociation from β-catenin, which enabled the accumulation of β-catenin at cell borders, where it facilitated the formation of cell-cell adhesion complexes via interactions with R-cadherin and desmosomal proteins. Kv11.1 activation-dependent Cav-1 dephosphorylation induced with NS1643 also reduced cell migration and invasion, consistent with its ability to regulate focal adhesion dynamics. Thus, this study sheds light on a novel pharmacological mechanism of promoting Cav-1 dephosphorylation, which may prove to be effective at reducing metastasis and promoting contact inhibition.
Collapse
Affiliation(s)
- Ying Jiang
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Vitalyi Senyuk
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ke Ma
- Research Resources Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hui Chen
- Research Resources Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Xiang Qin
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Shun Li
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yiyao Liu
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Saverio Gentile
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- UI Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, USA
- UI Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
4
|
Sanchez-Conde FG, Jimenez-Vazquez EN, Auerbach DS, Jones DK. The ERG1 K+ Channel and Its Role in Neuronal Health and Disease. Front Mol Neurosci 2022; 15:890368. [PMID: 35600076 PMCID: PMC9113952 DOI: 10.3389/fnmol.2022.890368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
The ERG1 potassium channel, encoded by KCNH2, has long been associated with cardiac electrical excitability. Yet, a growing body of work suggests that ERG1 mediates physiology throughout the human body, including the brain. ERG1 is a regulator of neuronal excitability, ERG1 variants are associated with neuronal diseases (e.g., epilepsy and schizophrenia), and ERG1 serves as a potential therapeutic target for neuronal pathophysiology. This review summarizes the current state-of-the-field regarding the ERG1 channel structure and function, ERG1’s relationship to the mammalian brain and highlights key questions that have yet to be answered.
Collapse
Affiliation(s)
| | - Eric N. Jimenez-Vazquez
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - David S. Auerbach
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, United States
- *Correspondence: David S. Auerbach,
| | - David K. Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- David K. Jones,
| |
Collapse
|
5
|
Castiglione A, Hornyik T, Wülfers EM, Giammarino L, Edler I, Jowais JJ, Rieder M, Perez-Feliz S, Koren G, Bősze Z, Varró A, Zehender M, Brunner M, Bode C, Liin SI, Larsson HP, Baczkó I, Odening KE. Docosahexaenoic acid normalizes QT interval in long QT type 2 transgenic rabbit models in a genotype-specific fashion. Europace 2022; 24:511-522. [PMID: 34601592 PMCID: PMC9125797 DOI: 10.1093/europace/euab228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022] Open
Abstract
AIM Long QT syndrome (LQTS) is a cardiac channelopathy predisposing to ventricular arrhythmias and sudden cardiac death. Since current therapies often fail to prevent arrhythmic events in certain LQTS subtypes, new therapeutic strategies are needed. Docosahexaenoic acid (DHA) is a polyunsaturated fatty acid, which enhances the repolarizing IKs current. METHODS AND RESULTS We investigated the effects of DHA in wild type (WT) and transgenic long QT Type 1 (LQT1; loss of IKs), LQT2 (loss of IKr), LQT5 (reduction of IKs), and LQT2-5 (loss of IKr and reduction of IKs) rabbits. In vivo ECGs were recorded at baseline and after 10 µM/kg DHA to assess changes in heart-rate corrected QT (QTc) and short-term variability of QT (STVQT). Ex vivo monophasic action potentials were recorded in Langendorff-perfused rabbit hearts, and action potential duration (APD75) and triangulation were assessed. Docosahexaenoic acid significantly shortened QTc in vivo only in WT and LQT2 rabbits, in which both α- and β-subunits of IKs-conducting channels are functionally intact. In LQT2, this led to a normalization of QTc and of its short-term variability. Docosahexaenoic acid had no effect on QTc in LQT1, LQT5, and LQT2-5. Similarly, ex vivo, DHA shortened APD75 in WT and normalized it in LQT2, and additionally decreased AP triangulation in LQT2. CONCLUSIONS Docosahexaenoic acid exerts a genotype-specific beneficial shortening/normalizing effect on QTc and APD75 and reduces pro-arrhythmia markers STVQT and AP triangulation through activation of IKs in LQT2 rabbits but has no effects if either α- or β-subunits to IKs are functionally impaired. Docosahexaenoic acid could represent a new genotype-specific therapy in LQT2.
Collapse
Affiliation(s)
- Alessandro Castiglione
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Faculty, University of Freiburg, Freiburg, Germany
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Buehlplatz 5, CH-3012 Bern, Switzerland
- Department of Translational Cardiology/Electrophysiology, Institute of Physiology, University of Bern, Bern, Switzerland
| | - Tibor Hornyik
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Faculty, University of Freiburg, Freiburg, Germany
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Buehlplatz 5, CH-3012 Bern, Switzerland
- Department of Translational Cardiology/Electrophysiology, Institute of Physiology, University of Bern, Bern, Switzerland
- Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg—Bad Krozingen, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Eike M Wülfers
- Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg—Bad Krozingen, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Lucilla Giammarino
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Buehlplatz 5, CH-3012 Bern, Switzerland
- Department of Translational Cardiology/Electrophysiology, Institute of Physiology, University of Bern, Bern, Switzerland
| | - Iask Edler
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jessica J Jowais
- Department of Physiology and Biophysics, University of Miami, Miami, FL, USA
| | - Marina Rieder
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Faculty, University of Freiburg, Freiburg, Germany
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Buehlplatz 5, CH-3012 Bern, Switzerland
- Department of Translational Cardiology/Electrophysiology, Institute of Physiology, University of Bern, Bern, Switzerland
| | - Stefanie Perez-Feliz
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Faculty, University of Freiburg, Freiburg, Germany
- Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg—Bad Krozingen, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Gideon Koren
- Division of Cardiology, Cardiovascular Research Center, Brown University, Providence, RI, USA
| | - Zsuzsanna Bősze
- Animal Biotechnology Department, NARIC Agricultural Biotechnology Institute, Gödöllő, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Manfred Zehender
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Michael Brunner
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Cardiology and Medical Intensive Care, St. Josefskrankenhaus, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Sara I Liin
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hans Peter Larsson
- Department of Physiology and Biophysics, University of Miami, Miami, FL, USA
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Katja E Odening
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Faculty, University of Freiburg, Freiburg, Germany
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Buehlplatz 5, CH-3012 Bern, Switzerland
- Department of Translational Cardiology/Electrophysiology, Institute of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Crotti L, Odening KE, Sanguinetti MC. Heritable arrhythmias associated with abnormal function of cardiac potassium channels. Cardiovasc Res 2021; 116:1542-1556. [PMID: 32227190 DOI: 10.1093/cvr/cvaa068] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Cardiomyocytes express a surprisingly large number of potassium channel types. The primary physiological functions of the currents conducted by these channels are to maintain the resting membrane potential and mediate action potential repolarization under basal conditions and in response to changes in the concentrations of intracellular sodium, calcium, and ATP/ADP. Here, we review the diversity and functional roles of cardiac potassium channels under normal conditions and how heritable mutations in the genes encoding these channels can lead to distinct arrhythmias. We briefly review atrial fibrillation and J-wave syndromes. For long and short QT syndromes, we describe their genetic basis, clinical manifestation, risk stratification, traditional and novel therapeutic approaches, as well as insights into disease mechanisms provided by animal and cellular models.
Collapse
Affiliation(s)
- Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano, IRCCS, Milan, Italy.,Department of Cardiovascular, Neural and Metabolic Sciences, Istituto Auxologico Italiano, IRCCS, San Luca Hospital, Milan, Italy.,Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Katja E Odening
- Department of Cardiology and Angiology I, Heart Center University of Freiburg, Medical Faculty, Freiburg, Germany.,Institute of Experimental Cardiovascular Medicine, Heart Center University of Freiburg, Medical Faculty, Freiburg, Germany.,Department of Cardiology, Translational Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland
| | - Michael C Sanguinetti
- Department of Internal Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
7
|
Potassium and Chloride Ion Channels in Cancer: A Novel Paradigm for Cancer Therapeutics. Rev Physiol Biochem Pharmacol 2021; 183:135-155. [PMID: 34291318 DOI: 10.1007/112_2021_62] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cancer is a collection of diseases caused by specific changes at the genomic level that support cell proliferation indefinitely. Traditionally, ion channels are known to control a variety of cellular processes including electrical signal generation and transmission, secretion, and contraction by controlling ionic gradients. However, recent studies had brought to light important facts on ion channels in cancer biology.In this review we discuss the mechanism linking potassium or chloride ion channel activity to biochemical pathways controlling proliferation in cancer cells and the potential advantages of targeting ion channels as an anticancer therapeutic option.
Collapse
|
8
|
Targeting of Potassium Channels in Cardiac Arrhythmias. Trends Pharmacol Sci 2021; 42:491-506. [PMID: 33858691 DOI: 10.1016/j.tips.2021.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Cardiomyocytes are endowed with a complex repertoire of ion channels, responsible for the generation of action potentials (APs), travelling waves of electrical excitation, propagating throughout the heart and leading to cardiac contractions. Cardiac AP waveforms are shaped by a striking diversity of K+ channels. The pivotal role of K+ channels in cardiac health and disease is underscored by the dramatic impact that K+ channel dysfunction has on cardiac arrhythmias. The development of drugs targeted to specific K+ channels is expected to provide an optimized approach to antiarrhythmic therapy. Here, we review the functional roles of cardiac potassium channels under normal and diseased states. We survey current antiarrhythmic drugs (AADs) targeted to voltage-gated and Ca2+-activated K+ channels and highlight future research opportunities.
Collapse
|
9
|
Su S, Sun J, Wang Y, Xu Y. Cardiac hERG K + Channel as Safety and Pharmacological Target. Handb Exp Pharmacol 2021; 267:139-166. [PMID: 33829343 DOI: 10.1007/164_2021_455] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The human ether-á-go-go related gene (hERG, KCNH2) encodes the pore-forming subunit of the potassium channel responsible for a fast component of the cardiac delayed rectifier potassium current (IKr). Outward IKr is an important determinant of cardiac action potential (AP) repolarization and effectively controls the duration of the QT interval in humans. Dysfunction of hERG channel can cause severe ventricular arrhythmias and thus modulators of the channel, including hERG inhibitors and activators, continue to attract intense pharmacological interest. Certain inhibitors of hERG channel prolong the action potential duration (APD) and effective refractory period (ERP) to suppress premature ventricular contraction and are used as class III antiarrhythmic agents. However, a reduction of the hERG/IKr current has been recognized as a predominant mechanism responsible for the drug-induced delayed repolarization known as acquired long QT syndromes (LQTS), which is linked to an increased risk for "torsades de pointes" (TdP) ventricular arrhythmias and sudden cardiac death. Many drugs of different classes and structures have been identified to carry TdP risk. Hence, assessing hERG/IKr blockade of new drug candidates is mandatory in the drug development process according to the regulatory agencies. In contrast, several hERG channel activators have been shown to enhance IKr and shorten the APD and thus might have potential antiarrhythmic effects against pathological LQTS. However, these activators may also be proarrhythmic due to excessive shortening of APD and the ERP.
Collapse
Affiliation(s)
- Shi Su
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Jinglei Sun
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Yi Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei, China.
| |
Collapse
|
10
|
Hornyik T, Rieder M, Castiglione A, Major P, Baczko I, Brunner M, Koren G, Odening KE. Transgenic rabbit models for cardiac disease research. Br J Pharmacol 2021; 179:938-957. [PMID: 33822374 DOI: 10.1111/bph.15484] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022] Open
Abstract
To study the pathophysiology of human cardiac diseases and to develop novel treatment strategies, complex interactions of cardiac cells on cellular, tissue and on level of the whole heart need to be considered. As in vitro cell-based models do not depict the complexity of the human heart, animal models are used to obtain insights that can be translated to human diseases. Mice are the most commonly used animals in cardiac research. However, differences in electrophysiological and mechanical cardiac function and a different composition of electrical and contractile proteins limit the transferability of the knowledge gained. Moreover, the small heart size and fast heart rate are major disadvantages. In contrast to rodents, electrophysiological, mechanical and structural cardiac characteristics of rabbits resemble the human heart more closely, making them particularly suitable as an animal model for cardiac disease research. In this review, various methodological approaches for the generation of transgenic rabbits for cardiac disease research, such as pronuclear microinjection, the sleeping beauty transposon system and novel genome-editing methods (ZFN and CRISPR/Cas9)will be discussed. In the second section, we will introduce the different currently available transgenic rabbit models for monogenic cardiac diseases (such as long QT syndrome, short-QT syndrome and hypertrophic cardiomyopathy) in detail, especially in regard to their utility to increase the understanding of pathophysiological disease mechanisms and novel treatment options.
Collapse
Affiliation(s)
- Tibor Hornyik
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland.,Department of Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marina Rieder
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland
| | - Alessandro Castiglione
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland
| | - Peter Major
- Institute for Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, Hungary
| | - Istvan Baczko
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Michael Brunner
- Department of Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Cardiology and Medical Intensive Care, St. Josefskrankenhaus, Freiburg, Germany
| | - Gideon Koren
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland.,Department of Cardiology and Angiology I, University Heart Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Odening KE. Mechanism-based QT/APD-shortening therapy for potassium-channel-based long-QT syndromes? Acta Physiol (Oxf) 2020; 229:e13481. [PMID: 32330372 DOI: 10.1111/apha.13481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Katja E Odening
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, and Institute of Physiology, University of Bern, Bern, Switzerland.,Department of Cardiology and Angiology I, Medical Faculty, Heart Center University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Breuer EK, Fukushiro-Lopes D, Dalheim A, Burnette M, Zartman J, Kaja S, Wells C, Campo L, Curtis KJ, Romero-Moreno R, Littlepage LE, Niebur GL, Hoskins K, Nishimura MI, Gentile S. Potassium channel activity controls breast cancer metastasis by affecting β-catenin signaling. Cell Death Dis 2019; 10:180. [PMID: 30792401 PMCID: PMC6385342 DOI: 10.1038/s41419-019-1429-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
Potassium ion channels are critical in the regulation of cell motility. The acquisition of cell motility is an essential parameter of cancer metastasis. However, the role of K+ channels in cancer metastasis has been poorly studied. High expression of the hG1 gene, which encodes for Kv11.1 channel associates with good prognosis in estrogen receptor-negative breast cancer (BC). We evaluated the efficacy of the Kv11.1 activator NS1643 in arresting metastasis in a triple negative breast cancer (TNBC) mouse model. NS1643 significantly reduces the metastatic spread of breast tumors in vivo by inhibiting cell motility, reprogramming epithelial–mesenchymal transition via attenuation of Wnt/β-catenin signaling and suppressing cancer cell stemness. Our findings provide important information regarding the clinical relevance of potassium ion channel expression in breast tumors and the mechanisms by which potassium channel activity can modulate tumor biology. Findings suggest that Kv11.1 activators may represent a novel therapeutic approach for the treatment of metastatic estrogen receptor-negative BC. Ion channels are critical factor for cell motility but little is known about their role in metastasis. Stimulation of the Kv11.1 channel suppress the metastatic phenotype in TNBC. This work could represent a paradigm-shifting approach to reducing mortality by targeting a pathway that is central to the development of metastases.
Collapse
Affiliation(s)
- Eun-Kyoung Breuer
- Department of Radiation Oncology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Daniela Fukushiro-Lopes
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Annika Dalheim
- Department of Surgery, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Miranda Burnette
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Jeremiah Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Simon Kaja
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA.,Department of Ophthalmology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.,Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA
| | - Claire Wells
- Division of Cancer Studies, King's College London, Rm. 2.34 A New Hunts House, Guy's Campus, London, SE1 1 UL, UK
| | - Loredana Campo
- Department of Radiation Oncology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Kimberly J Curtis
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Ricardo Romero-Moreno
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Laurie E Littlepage
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Glen L Niebur
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA.,Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Kent Hoskins
- Division of Hematology Oncology, Department of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Michael I Nishimura
- Department of Surgery, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - Saverio Gentile
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, 60153, USA. .,Division of Hematology Oncology, Department of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
13
|
Perez-Neut M, Haar L, Rao V, Santha S, Lansu K, Rana B, Jones WK, Gentile S. Activation of hERG3 channel stimulates autophagy and promotes cellular senescence in melanoma. Oncotarget 2017; 7:21991-2004. [PMID: 26942884 PMCID: PMC5008339 DOI: 10.18632/oncotarget.7831] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/15/2016] [Indexed: 01/07/2023] Open
Abstract
Ion channels play a major factor in maintaining cellular homeostasis but very little is known about the role of these proteins in cancer biology. In this work we have discovered that, the Kv11.3 (hERG3) a plasma-membrane potassium channel plays a critical role in the regulation of autophagy in a cancer cell model. We have found that pharmacologic stimulation of the Kv11.3 channel with a small molecule activator, NS1643 induced autophagy via activation of an AMPK-dependent signaling pathway in melanoma cell line. In addition, we have found that NS1643 produced a strong inhibition of cell proliferation by activating a cellular senescence program. Furthermore, inhibition of autophagy via siRNA targeting AMPK or treatment with hydroxychloroquine an autophagy inhibitor activates apoptosis in NS1643-treated cells. Thus, we propose that, Kv11.3 is a novel mediator of autophagy, autophagy can be a survival mechanism contributing to cellular senescence, and that use of a combinatorial pharmacologic approach of Kv11.3 activator with inhibitors of autophagy represents a novel therapeutic approach against melanoma.
Collapse
Affiliation(s)
- Mathew Perez-Neut
- Department of Molecular Pharmacology & Therapeutics, Loyola University, Chicago, IL-60153, USA
| | - Lauren Haar
- Department of Molecular Pharmacology & Therapeutics, Loyola University, Chicago, IL-60153, USA
| | - Vidhya Rao
- Department of Molecular Pharmacology & Therapeutics, Loyola University, Chicago, IL-60153, USA
| | - Sreevidya Santha
- Department of Molecular Pharmacology & Therapeutics, Loyola University, Chicago, IL-60153, USA
| | - Katherine Lansu
- Department of Molecular Pharmacology & Therapeutics, Loyola University, Chicago, IL-60153, USA
| | - Basabi Rana
- Department of Molecular Pharmacology & Therapeutics, Loyola University, Chicago, IL-60153, USA
| | - Walter K Jones
- Department of Molecular Pharmacology & Therapeutics, Loyola University, Chicago, IL-60153, USA
| | - Saverio Gentile
- Department of Molecular Pharmacology & Therapeutics, Loyola University, Chicago, IL-60153, USA
| |
Collapse
|
14
|
Fukushiro-Lopes DF, Hegel AD, Rao V, Wyatt D, Baker A, Breuer EK, Osipo C, Zartman JJ, Burnette M, Kaja S, Kouzoukas D, Burris S, Jones WK, Gentile S. Preclinical study of a Kv11.1 potassium channel activator as antineoplastic approach for breast cancer. Oncotarget 2017; 9:3321-3337. [PMID: 29423049 PMCID: PMC5790466 DOI: 10.18632/oncotarget.22925] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/09/2017] [Indexed: 12/04/2022] Open
Abstract
Potassium ion (K+) channels have been recently found to play a critical role in cancer biology. Despite that pharmacologic manipulation of ion channels is recognized as an important therapeutic approach, very little is known about the effects of targeting of K+ channels in cancer. In this study, we demonstrate that use of the Kv11.1 K+ channel activator NS1643 inhibits tumor growth in an in vivo model of breast cancer. Tumors exposed to NS1643 had reduced levels of proliferation markers, high expression levels of senescence markers, increased production of ROS and DNA damage compared to tumors of untreated mice. Importantly, mice treated with NS1643 did not exhibit significant cardiac dysfunction. In conclusion, pharmacological stimulation of Kv11.1 activity produced arrested TNBC-derived tumor growth by generating DNA damage and senescence without significant side effects. We propose that use of Kv11.1 channels activators could be considered as a possible pharmacological strategy against breast tumors.
Collapse
Affiliation(s)
- Daniela F Fukushiro-Lopes
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Alexandra D Hegel
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Vidhya Rao
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.,Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA
| | - Debra Wyatt
- Department of Pathology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Andrew Baker
- Department of Pathology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Eun-Kyoung Breuer
- Department of Pathology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Clodia Osipo
- Department of Pathology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Jeremiah J Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Miranda Burnette
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Simon Kaja
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.,Department of Ophthalmology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.,Research Service, Edward Hines Jr. VA Hospital, Hines, IL, USA
| | - Dimitrios Kouzoukas
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Sarah Burris
- Cardiovascular Research Institute, Loyola University Chicago, Maywood, IL, USA
| | - W Keith Jones
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Saverio Gentile
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
15
|
Sala L, Bellin M, Mummery CL. Integrating cardiomyocytes from human pluripotent stem cells in safety pharmacology: has the time come? Br J Pharmacol 2017; 174:3749-3765. [PMID: 27641943 PMCID: PMC5647193 DOI: 10.1111/bph.13577] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 07/27/2016] [Accepted: 08/11/2016] [Indexed: 12/20/2022] Open
Abstract
Cardiotoxicity is a severe side effect of drugs that induce structural or electrophysiological changes in heart muscle cells. As a result, the heart undergoes failure and potentially lethal arrhythmias. It is still a major reason for drug failure in preclinical and clinical phases of drug discovery. Current methods for predicting cardiotoxicity are based on guidelines that combine electrophysiological analysis of cell lines expressing ion channels ectopically in vitro with animal models and clinical trials. Although no new cases of drugs linked to lethal arrhythmias have been reported since the introduction of these guidelines in 2005, their limited predictive power likely means that potentially valuable drugs may not reach clinical practice. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are now emerging as potentially more predictive alternatives, particularly for the early phases of preclinical research. However, these cells are phenotypically immature and culture and assay methods not standardized, which could be a hurdle to the development of predictive computational models and their implementation into the drug discovery pipeline, in contrast to the ambitions of the comprehensive pro-arrhythmia in vitro assay (CiPA) initiative. Here, we review present and future preclinical cardiotoxicity screening and suggest possible hPSC-CM-based strategies that may help to move the field forward. Coordinated efforts by basic scientists, companies and hPSC banks to standardize experimental conditions for generating reliable and reproducible safety indices will be helpful not only for cardiotoxicity prediction but also for precision medicine. LINKED ARTICLES This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.
Collapse
Affiliation(s)
- Luca Sala
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenZAThe Netherlands
| | - Milena Bellin
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenZAThe Netherlands
| | - Christine L Mummery
- Department of Anatomy and EmbryologyLeiden University Medical CenterLeidenZAThe Netherlands
- Department of Applied Stem Cell TechnologiesUniversity of TwenteEnschedeThe Netherlands
| |
Collapse
|
16
|
Huo J, Guo X, Lu Q, Qiang H, Liu P, Bai L, Huang CLH, Zhang Y, Ma A. NS1643 enhances ionic currents in a G604S-WT hERG co-expression system associated with long QT syndrome 2. Clin Exp Pharmacol Physiol 2017; 44:1125-1133. [PMID: 28741726 DOI: 10.1111/1440-1681.12820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/27/2017] [Accepted: 07/17/2017] [Indexed: 12/19/2022]
Affiliation(s)
- JianHua Huo
- Department of Cardiovascular Medicine; First Affiliated Hospital of Xi'an Jiaotong University; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University); Ministry of Education; Xi'an Shaanxi China
| | - Xueyan Guo
- Shaanxi Provincial People's Hospital; Xi'an Shaanxi China
| | - Qun Lu
- Department of Cardiovascular Medicine; First Affiliated Hospital of Xi'an Jiaotong University; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University); Ministry of Education; Xi'an Shaanxi China
| | - Hua Qiang
- Department of Cardiovascular Medicine; First Affiliated Hospital of Xi'an Jiaotong University; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University); Ministry of Education; Xi'an Shaanxi China
| | - Ping Liu
- Department of Cardiovascular Medicine; First Affiliated Hospital of Xi'an Jiaotong University; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University); Ministry of Education; Xi'an Shaanxi China
| | - Ling Bai
- Department of Cardiovascular Medicine; First Affiliated Hospital of Xi'an Jiaotong University; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University); Ministry of Education; Xi'an Shaanxi China
| | | | - Yanmin Zhang
- Department of Pediatric Cardiology; Childrens Research Institute; affiliate children's hospital of Xi'an Jiaotong University; Xi'an Shaanxi China
| | - Aiqun Ma
- Department of Cardiovascular Medicine; First Affiliated Hospital of Xi'an Jiaotong University; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University); Ministry of Education; Xi'an Shaanxi China
| |
Collapse
|
17
|
Gentile S. hERG1 potassium channel in cancer cells: a tool to reprogram immortality. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:649-655. [PMID: 27649700 DOI: 10.1007/s00249-016-1169-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 07/21/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022]
Abstract
It has been well established that changes in ion fluxes across cellular membranes as a function of time is fundamental in maintaining cellular homeostasis of every living cell. Consequently, dysregulation of ion channels activity is a critical event in pathological conditions of several tissues, including cancer. Nevertheless, the role of ion channels in cancer biology is still not well understood and very little is known about the possible therapeutic opportunities offered by the use of the vast collection of drugs that target ion channels. In this review, we focus on the recent advances in understanding the role of the voltage-gated hERG1 potassium channel in cancer and on the effects of pharmacologic manipulation of the hERG1 in cancer cells aiming to provide insights into the biochemical signaling and cellular processes that are altered by using these drugs.
Collapse
|
18
|
Sala L, Yu Z, Ward-van Oostwaard D, van Veldhoven JP, Moretti A, Laugwitz KL, Mummery CL, IJzerman AP, Bellin M. A new hERG allosteric modulator rescues genetic and drug-induced long-QT syndrome phenotypes in cardiomyocytes from isogenic pairs of patient induced pluripotent stem cells. EMBO Mol Med 2016; 8:1065-81. [PMID: 27470144 PMCID: PMC5009811 DOI: 10.15252/emmm.201606260] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Long-QT syndrome (LQTS) is an arrhythmogenic disorder characterised by prolongation of the QT interval in the electrocardiogram, which can lead to sudden cardiac death. Pharmacological treatments are far from optimal for congenital forms of LQTS, while the acquired form, often triggered by drugs that (sometimes inadvertently) target the cardiac hERG channel, is still a challenge in drug development because of cardiotoxicity. Current experimental models in vitro fall short in predicting proarrhythmic properties of new drugs in humans. Here, we leveraged a series of isogenically matched, diseased and genetically engineered, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from patients to test a novel hERG allosteric modulator for treating congenital LQTS, drug-induced LQTS or a combination of the two. By slowing IK r deactivation and positively shifting IK r inactivation, the small molecule LUF7346 effectively rescued all of these conditions, demonstrating in a human system that allosteric modulation of hERG may be useful as an approach to treat inherited and drug-induced LQTS Furthermore, our study provides experimental support of the value of isogenic pairs of patient hiPSC-CMs as platforms for testing drug sensitivities and performing safety pharmacology.
Collapse
Affiliation(s)
- Luca Sala
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Zhiyi Yu
- Gorlaeus Laboratories, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Jacobus Pd van Veldhoven
- Gorlaeus Laboratories, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Alessandra Moretti
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Karl-Ludwig Laugwitz
- I. Department of Medicine (Cardiology), Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Gorlaeus Laboratories, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
19
|
Lang CN, Koren G, Odening KE. Transgenic rabbit models to investigate the cardiac ion channel disease long QT syndrome. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 121:142-56. [PMID: 27210307 DOI: 10.1016/j.pbiomolbio.2016.05.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/01/2016] [Indexed: 12/13/2022]
Abstract
Long QT syndrome (LQTS) is a rare inherited channelopathy caused mainly by different mutations in genes encoding for cardiac K(+) or Na(+) channels, but can also be caused by commonly used ion-channel-blocking and QT-prolonging drugs, thus affecting a much larger population. To develop novel diagnostic and therapeutic strategies to improve the clinical management of these patients, a thorough understanding of the pathophysiological mechanisms of arrhythmogenesis and potential pharmacological targets is needed. Drug-induced and genetic animal models of various species have been generated and have been instrumental for identifying pro-arrhythmic triggers and important characteristics of the arrhythmogenic substrate in LQTS. However, due to species differences in features of cardiac electrical function, these different models do not entirely recapitulate all aspects of the human disease. In this review, we summarize advantages and shortcomings of different drug-induced and genetically mediated LQTS animal models - focusing on mouse and rabbit models since these represent the most commonly used small animal models for LQTS that can be subjected to genetic manipulation. In particular, we highlight the different aspects of arrhythmogenic mechanisms, pro-arrhythmic triggering factors, anti-arrhythmic agents, and electro-mechanical dysfunction investigated in transgenic LQTS rabbit models and their translational application for the clinical management of LQTS patients in detail. Transgenic LQTS rabbits have been instrumental to increase our understanding of the role of spatial and temporal dispersion of repolarization to provide an arrhythmogenic substrate, genotype-differences in the mechanisms for early afterdepolarization formation and arrhythmia maintenance, mechanisms of hormonal modification of arrhythmogenesis and regional heterogeneities in electro-mechanical dysfunction in LQTS.
Collapse
Affiliation(s)
- C N Lang
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany
| | - G Koren
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - K E Odening
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Medical Center - University of Freiburg, Germany; Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
20
|
Electro-mechanical dysfunction in long QT syndrome: Role for arrhythmogenic risk prediction and modulation by sex and sex hormones. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 120:255-69. [PMID: 26718598 DOI: 10.1016/j.pbiomolbio.2015.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/26/2015] [Accepted: 12/15/2015] [Indexed: 12/24/2022]
Abstract
Long QT syndrome (LQTS) is a congenital arrhythmogenic channelopathy characterized by impaired cardiac repolarization. Increasing evidence supports the notion that LQTS is not purely an "electrical" disease but rather an "electro-mechanical" disease with regionally heterogeneously impaired electrical and mechanical cardiac function. In the first part, this article reviews current knowledge on electro-mechanical (dys)function in LQTS, clinical consequences of the observed electro-mechanical dysfunction, and potential underlying mechanisms. Since several novel imaging techniques - Strain Echocardiography (SE) and Magnetic Resonance Tissue Phase Mapping (TPM) - are applied in clinical and experimental settings to assess the (regional) mechanical function, advantages of these non-invasive techniques and their feasibility in the clinical routine are particularly highlighted. The second part provides novel insights into sex differences and sex hormone effects on electro-mechanical cardiac function in a transgenic LQT2 rabbit model. Here we demonstrate that female LQT2 rabbits exhibit a prolonged time to diastolic peak - as marker for contraction duration and early relaxation - compared to males. Chronic estradiol-treatment enhances these differences in time to diastolic peak even more and additionally increases the risk for ventricular arrhythmia. Importantly, time to diastolic peak is particularly prolonged in rabbits exhibiting ventricular arrhythmia - regardless of hormone treatment - contrasting with a lack of differences in QT duration between symptomatic and asymptomatic LQT2 rabbits. This indicates the potential added value of the assessment of mechanical dysfunction in future risk stratification of LQTS patients.
Collapse
|
21
|
Osadchii OE. Flecainide attenuates rate adaptation of ventricular repolarization in guinea-pig heart. SCAND CARDIOVASC J 2015; 50:28-35. [DOI: 10.3109/14017431.2015.1099721] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Oleg E. Osadchii
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Department of Health Science and Technology, University of Aalborg, Aalborg, Denmark
| |
Collapse
|
22
|
Humphries ESA, Dart C. Neuronal and Cardiovascular Potassium Channels as Therapeutic Drug Targets: Promise and Pitfalls. JOURNAL OF BIOMOLECULAR SCREENING 2015; 20:1055-73. [PMID: 26303307 PMCID: PMC4576507 DOI: 10.1177/1087057115601677] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 07/26/2015] [Accepted: 07/28/2015] [Indexed: 12/21/2022]
Abstract
Potassium (K(+)) channels, with their diversity, often tissue-defined distribution, and critical role in controlling cellular excitability, have long held promise of being important drug targets for the treatment of dysrhythmias in the heart and abnormal neuronal activity within the brain. With the exception of drugs that target one particular class, ATP-sensitive K(+) (KATP) channels, very few selective K(+) channel activators or inhibitors are currently licensed for clinical use in cardiovascular and neurological disease. Here we review what a range of human genetic disorders have told us about the role of specific K(+) channel subunits, explore the potential of activators and inhibitors of specific channel populations as a therapeutic strategy, and discuss possible reasons for the difficulty in designing clinically relevant K(+) channel modulators.
Collapse
Affiliation(s)
| | - Caroline Dart
- Institute of Integrative Biology, University of Liverpool, UK
| |
Collapse
|
23
|
Ziupa D, Beck J, Franke G, Perez Feliz S, Hartmann M, Koren G, Zehender M, Bode C, Brunner M, Odening KE. Pronounced effects of HERG-blockers E-4031 and erythromycin on APD, spatial APD dispersion and triangulation in transgenic long-QT type 1 rabbits. PLoS One 2014; 9:e107210. [PMID: 25244401 PMCID: PMC4170956 DOI: 10.1371/journal.pone.0107210] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/04/2014] [Indexed: 12/13/2022] Open
Abstract
Background Prolongation of action potential duration (APD), increased spatial APD dispersion, and triangulation are major factors promoting drug-induced ventricular arrhythmia. Preclinical identification of HERG/IKr-blocking drugs and their pro-arrhythmic potential, however, remains a challenge. We hypothesize that transgenic long-QT type 1 (LQT1) rabbits lacking repolarizing IKs current may help to sensitively detect HERG/IKr-blocking properties of drugs. Methods Hearts of adult female transgenic LQT1 and wild type littermate control (LMC) rabbits were Langendorff-perfused with increasing concentrations of HERG/IKr-blockers E-4031 (0.001–0.1 µM, n = 9/7) or erythromycin (1–300 µM, n = 9/7) and APD, APD dispersion, and triangulation were analyzed. Results At baseline, APD was longer in LQT1 than in LMC rabbits in LV apex and RV mid. Erythromycin and E-4031 prolonged APD in LQT1 and LMC rabbits in all positions. However, erythromycin-induced percentaged APD prolongation related to baseline (%APD) was more pronounced in LQT1 at LV base-lateral and RV mid positions (100 µM, LQT1, +40.6±9.7% vs. LMC, +24.1±10.0%, p<0.05) and E-4031-induced %APD prolongation was more pronounced in LQT1 at LV base-lateral (0.01 µM, LQT1, +29.6±10.6% vs. LMC, +19.1±3.8%, p<0.05) and LV base-septal positions. Moreover, erythromycin significantly increased spatial APD dispersion only in LQT1 and increased triangulation only in LQT1 in LV base-septal and RV mid positions. Similarly, E-4031 increased triangulation only in LQT1 in LV apex and base-septal positions. Conclusions E-4031 and erythromycin prolonged APD and increased triangulation more pronouncedly in LQT1 than in LMC rabbits. Moreover, erythromycin increased APD dispersion only in LQT1, indicating that transgenic LQT1 rabbits could serve as sensitive model to detect HERG/IKr-blocking properties of drugs.
Collapse
Affiliation(s)
- David Ziupa
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Julia Beck
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Gerlind Franke
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Stefanie Perez Feliz
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Maximilian Hartmann
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Gideon Koren
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Manfred Zehender
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Christoph Bode
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Michael Brunner
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
| | - Katja E. Odening
- Heart Center University of Freiburg, Department of Cardiology and Angiology I, Freiburg, Germany
- * E-mail:
| |
Collapse
|
24
|
Mitcheson J, Arcangeli A. The Therapeutic Potential of hERG1 K+ Channels for Treating Cancer and Cardiac Arrhythmias. ION CHANNEL DRUG DISCOVERY 2014. [DOI: 10.1039/9781849735087-00258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
hERG potassium channels present pharmacologists and medicinal chemists with a dilemma. On the one hand hERG is a major reason for drugs being withdrawn from the market because of drug induced long QT syndrome and the associated risk of inducing sudden cardiac death, and yet hERG blockers are still widely used in the clinic to treat cardiac arrhythmias. Moreover, in the last decade overwhelming evidence has been provided that hERG channels are aberrantly expressed in cancer cells and that they contribute to tumour cell proliferation, resistance to apoptosis, and neoangiogenesis. Here we provide an overview of the properties of hERG channels and their role in excitable cells of the heart and nervous system as well as in cancer. We consider the therapeutic potential of hERG, not only with regard to the negative impact due to drug induced long QT syndrome, but also its future potential as a treatment in the fight against cancer.
Collapse
Affiliation(s)
- John Mitcheson
- University of Leicester, Department of Cell Physiology and Pharmacology, Medical Sciences Building University Road Leicester LE1 9HN UK
| | - Annarosa Arcangeli
- Department of Experimental Pathology and Oncology, University of Florence Viale GB Morgagni, 50 50134 Firenze Italy
| |
Collapse
|
25
|
Guo J, Durdagi S, Changalov M, Perissinotti LL, Hargreaves JM, Back TG, Noskov SY, Duff HJ. Structure driven design of novel human ether-a-go-go-related-gene channel (hERG1) activators. PLoS One 2014; 9:e105553. [PMID: 25191697 PMCID: PMC4156305 DOI: 10.1371/journal.pone.0105553] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/22/2014] [Indexed: 01/01/2023] Open
Abstract
One of the main culprits in modern drug discovery is apparent cardiotoxicity of many lead-candidates via inadvertent pharmacologic blockade of K+, Ca2+ and Na+ currents. Many drugs inadvertently block hERG1 leading to an acquired form of the Long QT syndrome and potentially lethal polymorphic ventricular tachycardia. An emerging strategy is to rely on interventions with a drug that may proactively activate hERG1 channels reducing cardiovascular risks. Small molecules-activators have a great potential for co-therapies where the risk of hERG-related QT prolongation is significant and rehabilitation of the drug is impractical. Although a number of hERG1 activators have been identified in the last decade, their binding sites, functional moieties responsible for channel activation and thus mechanism of action, have yet to be established. Here, we present a proof-of-principle study that combines de-novo drug design, molecular modeling, chemical synthesis with whole cell electrophysiology and Action Potential (AP) recordings in fetal mouse ventricular myocytes to establish basic chemical principles required for efficient activator of hERG1 channel. In order to minimize the likelihood that these molecules would also block the hERG1 channel they were computationally engineered to minimize interactions with known intra-cavitary drug binding sites. The combination of experimental and theoretical studies led to identification of functional elements (functional groups, flexibility) underlying efficiency of hERG1 activators targeting binding pocket located in the S4–S5 linker, as well as identified potential side-effects in this promising line of drugs, which was associated with multi-channel targeting of the developed drugs.
Collapse
Affiliation(s)
- Jiqing Guo
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - Serdar Durdagi
- Centre for Molecular Simulation, Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Mohamed Changalov
- Department of Chemistry, University of Calgary, Calgary, Alberta, Canada
| | - Laura L. Perissinotti
- Centre for Molecular Simulation, Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | | | - Thomas G. Back
- Department of Chemistry, University of Calgary, Calgary, Alberta, Canada
- * E-mail: (TGB); (SYN); (HJD)
| | - Sergei Y. Noskov
- Centre for Molecular Simulation, Biochemistry Research Cluster, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- * E-mail: (TGB); (SYN); (HJD)
| | - Henry J. Duff
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
- * E-mail: (TGB); (SYN); (HJD)
| |
Collapse
|
26
|
Sanguinetti MC. HERG1 channel agonists and cardiac arrhythmia. Curr Opin Pharmacol 2013; 15:22-7. [PMID: 24721650 DOI: 10.1016/j.coph.2013.11.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/06/2013] [Accepted: 11/07/2013] [Indexed: 01/25/2023]
Abstract
Type 1 human ether-a-go-go-related gene (hERG1) potassium channels are a key determinant of normal repolarization of cardiac action potentials. Loss of function mutations in hERG1 channels cause inherited long QT syndrome and increased risk of cardiac arrhythmia and sudden death. Many common medications that block hERG1 channels as an unintended side effect also increase arrhythmic risk. Routine preclinical screening for hERG1 block led to the discovery of agonists that shorten action potential duration and QT interval. Agonists have the potential to be used as pharmacotherapy for long QT syndrome, but can also be proarrhythmic. Recent studies have elucidated multiple mechanisms of action for these compounds and the structural basis for their binding to the pore domain of the hERG1 channel.
Collapse
Affiliation(s)
- Michael C Sanguinetti
- Department of Internal Medicine, Nora Eccles Harrison Cardiovascular Research & Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT 84112, USA.
| |
Collapse
|
27
|
Meng J, Shi C, Li L, Du Y, Xu Y. Compound ICA-105574 prevents arrhythmias induced by cardiac delayed repolarization. Eur J Pharmacol 2013; 718:87-97. [PMID: 24041920 DOI: 10.1016/j.ejphar.2013.09.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 08/27/2013] [Accepted: 09/04/2013] [Indexed: 01/14/2023]
Abstract
Impaired ventricular repolarization can lead to long QT syndrome (LQT), a proarrhythmic disease with high risk of developing lethal ventricular tachyarrhythmias. The compound ICA-105574 is a recently developed hERG activator and it enhances IKr current with very high potency by removing the channel inactivation. The present study was designed to investigate antiarrhythmic properties of ICA-105574. For comparison, the effects of another compound NS1643 was in-parallel assessed, which also acts primarily to attenuate channel inactivation with moderate potency. We found that both ICA-105574 and NS1643 concentration-dependently shortened action potential duration (APD) in ventricular myocytes, and QT/QTc intervals in isolated guinea-pig hearts. ICA-105574, but not NS1643, completely prevented ventricular arrhythmias in intact guinea-pig hearts caused by IKr and IKs inhibitors, although both ICA-105574 and NS1643 could reverse the drug-induced prolongation of APD in ventricular myocytes. Reversing prolongation of QT/QTc intervals and antagonizing the increases in transmural dispersion of repolarization and instability of the QT interval induced by IKr and IKs inhibitors contributed to antiarrhythmic effect of ICA-105574. Meanwhile, ICA-105574 at higher concentrations showed a potential proarrhythmic risk in normal hearts. Our results suggest that ICA-105574 has more efficient antiarrhythmic activity than NS1643. However, its potential proarrhythmic risk implies that benefits and risks should be seriously taken into consideration for further developing this type of hERG activators.
Collapse
Affiliation(s)
- Jing Meng
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Hebei Medical University, Hebei Province, Shijiazhuang 050017, China; Department of Pharmaceutical Chemistry, Hebei Medical University, Shijiazhuang, China
| | | | | | | | | |
Collapse
|
28
|
Odening KE, Jung BA, Lang CN, Cabrera Lozoya R, Ziupa D, Menza M, Relan J, Franke G, Perez Feliz S, Koren G, Zehender M, Bode C, Brunner M, Sermesant M, Föll D. Spatial correlation of action potential duration and diastolic dysfunction in transgenic and drug-induced LQT2 rabbits. Heart Rhythm 2013; 10:1533-41. [PMID: 23892340 DOI: 10.1016/j.hrthm.2013.07.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Indexed: 11/24/2022]
Abstract
BACKGROUND Enhanced dispersion of action potential duration (APD) is a major contributor to long QT syndrome (LQTS)-related arrhythmias. OBJECTIVE To investigate spatial correlations of regional heterogeneities in cardiac repolarization and mechanical function in LQTS. METHODS Female transgenic LQTS type 2 (LQT2; n = 11) and wild-type littermate control (LMC) rabbits (n = 9 without E4031 and n = 10 with E4031) were subjected to phase contrast magnetic resonance imaging to assess regional myocardial velocities. In the same rabbits' hearts, monophasic APDs were assessed in corresponding segments. RESULTS In LQT2 and E4031-treated rabbits, APD was longer in all left ventricular segments (P < .01) and APD dispersion was greater than that in LMC rabbits (P < .01). In diastole, peak radial velocities (Vr) were reduced in LQT2 and E4031-treated compared to LMC rabbits in LV base and mid (LQT2: -3.36 ± 0.4 cm/s, P < .01; E4031-treated: -3.24 ± 0.6 cm/s, P < .0001; LMC: -4.42 ± 0.5 cm/s), indicating an impaired diastolic function. Regionally heterogeneous diastolic Vr correlated with APD (LQT2: correlation coefficient [CC] 0.38, P = .01; E4031-treated: CC 0.42, P < .05). Time-to-diastolic peak Vr were prolonged in LQT2 rabbits (LQT2: 196.8 ± 2.9 ms, P < .001; E4031-treated: 199.5 ± 2.2 ms, P < .0001, LMC 183.1 ± 1.5), indicating a prolonged contraction duration. Moreover, in transgenic LQT2 rabbits, diastolic time-to-diastolic peak Vr correlated with APD (CC 0.47, P = .001). In systole, peak Vr were reduced in LQT2 and E4031-treated rabbits (P < .01) but longitudinal velocities or ejection fraction did not differ. Finally, random forest machine learning algorithms enabled a differentiation between LQT2, E4031-treated, and LMC rabbits solely based on "mechanical" magnetic resonance imaging data. CONCLUSIONS The prolongation of APD led to impaired diastolic and systolic function in transgenic and drug-induced LQT2 rabbits. APD correlated with regional diastolic dysfunction, indicating that LQTS is not purely an electrical but an electromechanical disorder.
Collapse
Affiliation(s)
- Katja E Odening
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Modulation of hERG potassium channel gating normalizes action potential duration prolonged by dysfunctional KCNQ1 potassium channel. Proc Natl Acad Sci U S A 2012; 109:11866-71. [PMID: 22745159 DOI: 10.1073/pnas.1205266109] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Long QT syndrome (LQTS) is a genetic disease characterized by a prolonged QT interval in an electrocardiogram (ECG), leading to higher risk of sudden cardiac death. Among the 12 identified genes causal to heritable LQTS, ∼90% of affected individuals harbor mutations in either KCNQ1 or human ether-a-go-go related genes (hERG), which encode two repolarizing potassium currents known as I(Ks) and I(Kr). The ability to quantitatively assess contributions of different current components is therefore important for investigating disease phenotypes and testing effectiveness of pharmacological modulation. Here we report a quantitative analysis by simulating cardiac action potentials of cultured human cardiomyocytes to match the experimental waveforms of both healthy control and LQT syndrome type 1 (LQT1) action potentials. The quantitative evaluation suggests that elevation of I(Kr) by reducing voltage sensitivity of inactivation, not via slowing of deactivation, could more effectively restore normal QT duration if I(Ks) is reduced. Using a unique specific chemical activator for I(Kr) that has a primary effect of causing a right shift of V(1/2) for inactivation, we then examined the duration changes of autonomous action potentials from differentiated human cardiomyocytes. Indeed, this activator causes dose-dependent shortening of the action potential durations and is able to normalize action potentials of cells of patients with LQT1. In contrast, an I(Kr) chemical activator of primary effects in slowing channel deactivation was not effective in modulating action potential durations. Our studies provide both the theoretical basis and experimental support for compensatory normalization of action potential duration by a pharmacological agent.
Collapse
|
30
|
Duranthon V, Beaujean N, Brunner M, Odening KE, Santos AN, Kacskovics I, Hiripi L, Weinstein EJ, Bosze Z. On the emerging role of rabbit as human disease model and the instrumental role of novel transgenic tools. Transgenic Res 2012; 21:699-713. [PMID: 22382461 DOI: 10.1007/s11248-012-9599-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 02/04/2012] [Indexed: 12/19/2022]
Abstract
The laboratory rabbit (Oryctolagus cuniculus) is widely used as a model for human diseases, because of its size, which permits non-lethal monitoring of physiological changes and similar disease characteristics. Novel transgenic tools such as, the zinc finger nuclease method and the sleeping beauty transposon mediated or BAC transgenesis were recently adapted to the laboratory rabbit and opened new opportunities in precise tissue and developmental stage specific gene expression/silencing, coupled with increased transgenic efficiencies. Many facets of human development and diseases cannot be investigated in rodents. This is especially true for early prenatal development, its long-lasting effects on health and complex disorders, and some economically important diseases such as atherosclerosis or cardiovascular diseases. The first transgenic rabbits models of arrhythmogenesis mimic human cardiac diseases much better than transgenic mice and hereby underline the importance of non-mouse models. Another emerging field is epigenetic reprogramming and pathogenic mechanisms in diabetic pregnancy, where rabbit models are indispensable. Beyond that rabbit is used for decades as major source of polyclonal antibodies and recently in monoclonal antibody production. Alteration of its genome to increase the efficiency and value of the antibodies by humanization of the immunoglobulin genes, or by increasing the expression of a special receptor (Fc receptor) that augments humoral immune response is a current demand.
Collapse
Affiliation(s)
- V Duranthon
- INRA, UMR 1198 Biologie du Développement et Reproduction, 78350 Jouy en Josas, France
| | | | | | | | | | | | | | | | | |
Collapse
|