1
|
Boutonnet M, Bünemann M, Perroy J. The voltage sensitivity of G-protein coupled receptors: Unraveling molecular mechanisms and physiological implications. Pharmacol Ther 2024:108741. [PMID: 39489434 DOI: 10.1016/j.pharmthera.2024.108741] [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: 07/16/2024] [Revised: 09/11/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
In the landscape of proteins controlled by membrane voltage (Vm), like voltage-gated ionotropic channels, the emergence of the voltage sensitivity within the vast family of G-protein coupled receptors (GPCRs) marked a significant milestone at the onset of the 21st century. Since its discovery, extensive research has been devoted to understanding the intricate relationship between Vm and GPCRs. Approximately 30 GPCRs out of a family comprising more than 800 receptors have been implicated in Vm-dependent positive and negative regulation. GPCRs stand out as the quintessential regulators of synaptic transmission in neurons, where they encounter substantial variations in Vm. However, the molecular mechanism underlying the Vm sensor of GPCRs remains enigmatic, hindered by the scarcity of mutant GPCRs insensitive to Vm yet functionally intact, impeding a comprehensive understanding of this unique property in physiology. Nevertheless, two decades of dedicated research have furnished numerous insights into the molecular aspects of GPCR Vm-sensing, accompanied by recently proposed physiological roles as well as pharmacological potential, which we encapsulate in this review. The Vm sensitivity of GPCRs emerges as a pivotal attribute, shedding light on previously unforeseen roles in synaptic transmission and extending beyond, underscoring its significance in cellular signaling and physiological processes.
Collapse
Affiliation(s)
- Marin Boutonnet
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Julie Perroy
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
2
|
Si M, Darvish A, Paulhus K, Kumar P, Hamilton KA, Glasscock E. Epilepsy-associated Kv1.1 channel subunits regulate intrinsic cardiac pacemaking in mice. J Gen Physiol 2024; 156:e202413578. [PMID: 39037413 PMCID: PMC11261506 DOI: 10.1085/jgp.202413578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/11/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
The heartbeat originates from spontaneous action potentials in specialized pacemaker cells within the sinoatrial node (SAN) of the right atrium. Voltage-gated potassium channels in SAN myocytes mediate outward K+ currents that regulate cardiac pacemaking by controlling action potential repolarization, influencing the time between heartbeats. Gene expression studies have identified transcripts for many types of voltage-gated potassium channels in the SAN, but most remain of unknown functional significance. One such gene is Kcna1, which encodes epilepsy-associated voltage-gated Kv1.1 K+ channel α-subunits that are important for regulating action potential firing in neurons and cardiomyocytes. Here, we investigated the functional contribution of Kv1.1 to cardiac pacemaking at the whole heart, SAN, and SAN myocyte levels by performing Langendorff-perfused isolated heart preparations, multielectrode array recordings, patch clamp electrophysiology, and immunocytochemistry using Kcna1 knockout (KO) and wild-type (WT) mice. Our results showed that either genetic or pharmacological ablation of Kv1.1 significantly decreased the SAN firing rate, primarily by impairing SAN myocyte action potential repolarization. Voltage-clamp electrophysiology and immunocytochemistry revealed that Kv1.1 exerts its effects despite contributing only a small outward K+ current component, which we term IKv1.1, and despite apparently being present in low abundance at the protein level in SAN myocytes. These findings establish Kv1.1 as the first identified member of the Kv1 channel family to play a role in sinoatrial function, thereby rendering it a potential candidate and therapeutic targeting of sinus node dysfunction. Furthermore, our results demonstrate that small currents generated via low-abundance channels can still have significant impacts on cardiac pacemaking.
Collapse
Affiliation(s)
- Man Si
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| | - Ahmad Darvish
- School of Biological and Physical Science, Northwestern State University, Natchitoches, LA, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, Shreveport, LA, USA
| | - Kelsey Paulhus
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| | - Praveen Kumar
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
| | - Kathryn A. Hamilton
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, Shreveport, LA, USA
| | - Edward Glasscock
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, Shreveport, LA, USA
| |
Collapse
|
3
|
Nguyen H, Glaaser IW, Slesinger PA. Direct modulation of G protein-gated inwardly rectifying potassium (GIRK) channels. Front Physiol 2024; 15:1386645. [PMID: 38903913 PMCID: PMC11187414 DOI: 10.3389/fphys.2024.1386645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/08/2024] [Indexed: 06/22/2024] Open
Abstract
Ion channels play a pivotal role in regulating cellular excitability and signal transduction processes. Among the various ion channels, G-protein-coupled inwardly rectifying potassium (GIRK) channels serve as key mediators of neurotransmission and cellular responses to extracellular signals. GIRK channels are members of the larger family of inwardly-rectifying potassium (Kir) channels. Typically, GIRK channels are activated via the direct binding of G-protein βγ subunits upon the activation of G-protein-coupled receptors (GPCRs). GIRK channel activation requires the presence of the lipid signaling molecule, phosphatidylinositol 4,5-bisphosphate (PIP2). GIRK channels are also modulated by endogenous proteins and other molecules, including RGS proteins, cholesterol, and SNX27 as well as exogenous compounds, such as alcohol. In the last decade or so, several groups have developed novel drugs and small molecules, such as ML297, GAT1508 and GiGA1, that activate GIRK channels in a G-protein independent manner. Here, we aim to provide a comprehensive overview focusing on the direct modulation of GIRK channels by G-proteins, PIP2, cholesterol, and novel modulatory compounds. These studies offer valuable insights into the underlying molecular mechanisms of channel function, and have potential implications for both basic research and therapeutic development.
Collapse
Affiliation(s)
| | | | - Paul A. Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
4
|
Hennis K, Piantoni C, Biel M, Fenske S, Wahl-Schott C. Pacemaker Channels and the Chronotropic Response in Health and Disease. Circ Res 2024; 134:1348-1378. [PMID: 38723033 PMCID: PMC11081487 DOI: 10.1161/circresaha.123.323250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Loss or dysregulation of the normally precise control of heart rate via the autonomic nervous system plays a critical role during the development and progression of cardiovascular disease-including ischemic heart disease, heart failure, and arrhythmias. While the clinical significance of regulating changes in heart rate, known as the chronotropic effect, is undeniable, the mechanisms controlling these changes remain not fully understood. Heart rate acceleration and deceleration are mediated by increasing or decreasing the spontaneous firing rate of pacemaker cells in the sinoatrial node. During the transition from rest to activity, sympathetic neurons stimulate these cells by activating β-adrenergic receptors and increasing intracellular cyclic adenosine monophosphate. The same signal transduction pathway is targeted by positive chronotropic drugs such as norepinephrine and dobutamine, which are used in the treatment of cardiogenic shock and severe heart failure. The cyclic adenosine monophosphate-sensitive hyperpolarization-activated current (If) in pacemaker cells is passed by hyperpolarization-activated cyclic nucleotide-gated cation channels and is critical for generating the autonomous heartbeat. In addition, this current has been suggested to play a central role in the chronotropic effect. Recent studies demonstrate that cyclic adenosine monophosphate-dependent regulation of HCN4 (hyperpolarization-activated cyclic nucleotide-gated cation channel isoform 4) acts to stabilize the heart rate, particularly during rapid rate transitions induced by the autonomic nervous system. The mechanism is based on creating a balance between firing and recently discovered nonfiring pacemaker cells in the sinoatrial node. In this way, hyperpolarization-activated cyclic nucleotide-gated cation channels may protect the heart from sinoatrial node dysfunction, secondary arrhythmia of the atria, and potentially fatal tachyarrhythmia of the ventricles. Here, we review the latest findings on sinoatrial node automaticity and discuss the physiological and pathophysiological role of HCN pacemaker channels in the chronotropic response and beyond.
Collapse
Affiliation(s)
- Konstantin Hennis
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| | - Chiara Piantoni
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research (M.B., S.F.), Ludwig-Maximilians-Universität München, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (M.B., S.F.)
| | - Stefanie Fenske
- Department of Pharmacy, Center for Drug Research (M.B., S.F.), Ludwig-Maximilians-Universität München, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (M.B., S.F.)
| | - Christian Wahl-Schott
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
5
|
An X, Cho H. Increased GIRK channel activity prevents arrhythmia in mice with heart failure by enhancing ventricular repolarization. Sci Rep 2023; 13:22479. [PMID: 38110503 PMCID: PMC10728207 DOI: 10.1038/s41598-023-50088-2] [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: 06/26/2023] [Accepted: 12/15/2023] [Indexed: 12/20/2023] Open
Abstract
Ventricular arrhythmia causing sudden cardiac death is the leading mode of death in patients with heart failure. Yet, the mechanisms that prevent ventricular arrhythmias in heart failure are not well characterized. Using a mouse model of heart failure created by transverse aorta constriction, we show that GIRK channel, an important regulator of cardiac action potentials, is constitutively active in failing ventricles in contrast to normal cells. Evidence is presented indicating that the tonic activation of M2 muscarinic acetylcholine receptors by endogenously released acetylcholine contributes to the constitutive GIRK activity. This constitutive GIRK activity prevents the action potential prolongation in heart failure ventricles. Consistently, GIRK channel blockade with tertiapin-Q induces QT interval prolongation and increases the incidence of arrhythmia in heart failure, but not in control mice. These results suggest that constitutive GIRK channels comprise a key mechanism to protect against arrhythmia by providing repolarizing currents in heart failure ventricles.
Collapse
Affiliation(s)
- Xue An
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea.
| |
Collapse
|
6
|
Orsi JB, Araujo LS, Scariot PPM, Polisel EEC, Cardoso LO, Gobatto CA, Manchado-Gobatto FB. Critical Velocity, Maximal Lactate Steady State, and Muscle MCT1 and MCT4 after Exhaustive Running in Mice. Int J Mol Sci 2023; 24:15753. [PMID: 37958736 PMCID: PMC10648804 DOI: 10.3390/ijms242115753] [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: 08/22/2023] [Revised: 09/30/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023] Open
Abstract
Although the critical velocity (CV) protocol has been used to determine the aerobic capacity in rodents, there is a lack of studies that compare CV with maximal lactate steady state intensity (iMLSS) in mice. As a consequence, their physiological and molecular responses after exercise until exhaustion at CV intensity remain unclear. Thus, we aimed to compare and correlate CV with iMLSS in running mice, following different mathematical models for CV estimation. We also evaluated their physiological responses and muscle MCT1 and MCT4 after running until exhaustion at CV. Thirty C57BL/6J mice were divided into two groups (exercised-E and control-C). Group E was submitted to a CV protocol (4 days), using linear (lin1 and lin2) and hyperbolic (hyp) mathematical models to determine the distance, velocity, and time to exhaustion (tlim) of each predictive CV trial, followed by an MLSS protocol. After a running effort until exhaustion at CV intensity, the mice were immediately euthanized, while group C was euthanized at rest. No differences were observed between iMLSS (21.1 ± 1.1 m.min-1) and CV estimated by lin1 (21.0 ± 0.9 m.min-1, p = 0.415), lin2 (21.3 ± 0.9 m.min-1, p = 0.209), and hyp (20.6 ± 0.9 m.min-1, p = 0.914). According to the results, CV was significantly correlated with iMLSS. After running until exhaustion at CV (tlim = 28.4 ± 8,29 min), group E showed lower concentrations of hepatic and gluteal glycogen than group C, but no difference in the content of MCT1 (p = 0.933) and MCT4 (p = 0.123) in soleus muscle. Significant correlations were not found between MCT1 and MCT4 and tlim at CV intensity. Our results reinforce that CV is a valid and non-invasive protocol to estimate the maximal aerobic capacity in mice and that the content of MCT1 and MCT4 was not decisive in determining the tlim at CV, at least when measured immediately after the running effort.
Collapse
Affiliation(s)
- Juan B Orsi
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Pedro Zaccaria Street, 1.300, Jardim Santa Luíza, Limeira 13484-350, São Paulo, Brazil
| | - Lara S Araujo
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Pedro Zaccaria Street, 1.300, Jardim Santa Luíza, Limeira 13484-350, São Paulo, Brazil
| | - Pedro P M Scariot
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Pedro Zaccaria Street, 1.300, Jardim Santa Luíza, Limeira 13484-350, São Paulo, Brazil
| | - Emanuel E C Polisel
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Pedro Zaccaria Street, 1.300, Jardim Santa Luíza, Limeira 13484-350, São Paulo, Brazil
| | - Luisa O Cardoso
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Pedro Zaccaria Street, 1.300, Jardim Santa Luíza, Limeira 13484-350, São Paulo, Brazil
| | - Claudio A Gobatto
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Pedro Zaccaria Street, 1.300, Jardim Santa Luíza, Limeira 13484-350, São Paulo, Brazil
| | - Fúlvia B Manchado-Gobatto
- Laboratory of Applied Sport Physiology, School of Applied Sciences, University of Campinas, Pedro Zaccaria Street, 1.300, Jardim Santa Luíza, Limeira 13484-350, São Paulo, Brazil
| |
Collapse
|
7
|
Delgado-Betancourt V, Chinda K, Mesirca P, Barrère C, Covinhes A, Gallot L, Vincent A, Bidaud I, Kumphune S, Nargeot J, Piot C, Wickman K, Mangoni ME, Barrère-Lemaire S. Heart rate reduction after genetic ablation of L-type Ca v1.3 channels induces cardioprotection against ischemia-reperfusion injury. Front Cardiovasc Med 2023; 10:1134503. [PMID: 37593151 PMCID: PMC10429177 DOI: 10.3389/fcvm.2023.1134503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 06/21/2023] [Indexed: 08/19/2023] Open
Abstract
Background Acute myocardial infarction (AMI) is the major cause of cardiovascular mortality worldwide. Most ischemic episodes are triggered by an increase in heart rate, which induces an imbalance between myocardial oxygen delivery and consumption. Developing drugs that selectively reduce heart rate by inhibiting ion channels involved in heart rate control could provide more clinical benefits. The Cav1.3-mediated L-type Ca2+ current (ICav1.3) play important roles in the generation of heart rate. Therefore, they can constitute relevant targets for selective control of heart rate and cardioprotection during AMI. Objective We aimed to investigate the relationship between heart rate and infarct size using mouse strains knockout for Cav1.3 (Cav1.3-/-) L-type calcium channel and of the cardiac G protein gated potassium channel (Girk4-/-) in association with the funny (f)-channel inhibitor ivabradine. Methods Wild-type (WT), Cav1.3+/-, Cav1.3-/- and Girk4-/- mice were used as models of respectively normal heart rate, moderate heart rate reduction, bradycardia, and mild tachycardia, respectively. Mice underwent a surgical protocol of myocardial IR (40 min ischemia and 60 min reperfusion). Heart rate was recorded by one-lead surface ECG recording, and infarct size measured by triphenyl tetrazolium chloride staining. In addition, Cav1.3-/- and WT hearts perfused on a Langendorff system were subjected to the same ischemia-reperfusion protocol ex vivo, without or with atrial pacing, and the coronary flow was recorded. Results Cav1.3-/- mice presented reduced infarct size (-29%), while Girk4-/- displayed increased infarct size (+30%) compared to WT mice. Consistently, heart rate reduction in Cav1.3+/- or by the f-channel blocker ivabradine was associated with significant decrease in infarct size (-27% and -32%, respectively) in comparison to WT mice. Conclusion Our results show that decreasing heart rate allows to protect the myocardium against IR injury in vivo and reveal a close relationship between basal heart rate and IR injury. In addition, this study suggests that targeting Cav1.3 channels could constitute a relevant target for reducing infarct size, since maximal heart rate dependent cardioprotective effect is already observed in Cav1.3+/- mice.
Collapse
Affiliation(s)
- Viviana Delgado-Betancourt
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Kroekkiat Chinda
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Christian Barrère
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Aurélie Covinhes
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Laura Gallot
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Sarawut Kumphune
- Biomedical Engineering Institute (BMEi), Chiang Mai University, Chiang Mai, Thailand
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Christophe Piot
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | - Matteo Elia Mangoni
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| | - Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université Montpellier, CNRS, INSERM, Montpellier, France
- LabEx Ion Channel Science & Therapeutics (ICST), Université de Nice, Valbonne, France
| |
Collapse
|
8
|
Shrestha N, Zorn-Pauly K, Mesirca P, Koyani CN, Wölkart G, Di Biase V, Torre E, Lang P, Gorischek A, Schreibmayer W, Arnold R, Maechler H, Mayer B, von Lewinski D, Torrente AG, Mangoni ME, Pelzmann B, Scheruebel S. Lipopolysaccharide-induced sepsis impairs M2R-GIRK signaling in the mouse sinoatrial node. Proc Natl Acad Sci U S A 2023; 120:e2210152120. [PMID: 37406102 PMCID: PMC10334783 DOI: 10.1073/pnas.2210152120] [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: 06/14/2022] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
Sepsis has emerged as a global health burden associated with multiple organ dysfunction and 20% mortality rate in patients. Numerous clinical studies over the past two decades have correlated the disease severity and mortality in septic patients with impaired heart rate variability (HRV), as a consequence of impaired chronotropic response of sinoatrial node (SAN) pacemaker activity to vagal/parasympathetic stimulation. However, the molecular mechanism(s) downstream to parasympathetic inputs have not been investigated yet in sepsis, particularly in the SAN. Based on electrocardiography, fluorescence Ca2+ imaging, electrophysiology, and protein assays from organ to subcellular level, we report that impaired muscarinic receptor subtype 2-G protein-activated inwardly-rectifying potassium channel (M2R-GIRK) signaling in a lipopolysaccharide-induced proxy septic mouse model plays a critical role in SAN pacemaking and HRV. The parasympathetic responses to a muscarinic agonist, namely IKACh activation in SAN cells, reduction in Ca2+ mobilization of SAN tissues, lowering of heart rate and increase in HRV, were profoundly attenuated upon lipopolysaccharide-induced sepsis. These functional alterations manifested as a direct consequence of reduced expression of key ion-channel components (GIRK1, GIRK4, and M2R) in the mouse SAN tissues and cells, which was further evident in the human right atrial appendages of septic patients and likely not mediated by the common proinflammatory cytokines elevated in sepsis.
Collapse
Affiliation(s)
- Niroj Shrestha
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics, Medical University of Graz, 8010Graz, Austria
| | - Klaus Zorn-Pauly
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics, Medical University of Graz, 8010Graz, Austria
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 34094Montpellier, France
- Laboratory of Excellence in Ion Channels Science and Therapeutics, 34094Montpellier, France
| | - Chintan N. Koyani
- Division of Cardiology, Medical University of Graz, 8036Graz, Austria
| | - Gerald Wölkart
- Department of Pharmacology and Toxicology, University of Graz, 8010Graz, Austria
| | - Valentina Di Biase
- Institute of Pharmacology, Medical University of Innsbruck, 6020Innsbruck, Austria
| | - Eleonora Torre
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 34094Montpellier, France
- Laboratory of Excellence in Ion Channels Science and Therapeutics, 34094Montpellier, France
| | - Petra Lang
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics, Medical University of Graz, 8010Graz, Austria
| | - Astrid Gorischek
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics, Medical University of Graz, 8010Graz, Austria
| | - Wolfgang Schreibmayer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics, Medical University of Graz, 8010Graz, Austria
| | - Robert Arnold
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics, Medical University of Graz, 8010Graz, Austria
| | - Heinrich Maechler
- Division of Cardiac Surgery, Medical University of Graz, 8036Graz, Austria
| | - Bernd Mayer
- Department of Pharmacology and Toxicology, University of Graz, 8010Graz, Austria
| | - Dirk von Lewinski
- Division of Cardiology, Medical University of Graz, 8036Graz, Austria
| | - Angelo G. Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 34094Montpellier, France
- Laboratory of Excellence in Ion Channels Science and Therapeutics, 34094Montpellier, France
| | - Matteo E. Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 34094Montpellier, France
- Laboratory of Excellence in Ion Channels Science and Therapeutics, 34094Montpellier, France
| | - Brigitte Pelzmann
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics, Medical University of Graz, 8010Graz, Austria
| | - Susanne Scheruebel
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical Physics and Biophysics, Medical University of Graz, 8010Graz, Austria
| |
Collapse
|
9
|
Meyer KM, Malhotra N, Kwak JS, El Refaey M. Relevance of KCNJ5 in Pathologies of Heart Disease. Int J Mol Sci 2023; 24:10849. [PMID: 37446026 PMCID: PMC10341679 DOI: 10.3390/ijms241310849] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Abnormalities in G-protein-gated inwardly rectifying potassium (GIRK) channels have been implicated in diseased states of the cardiovascular system; however, the role of GIRK4 (Kir3.4) in cardiac physiology and pathophysiology has yet to be completely understood. Within the heart, the KACh channel, consisting of two GIRK1 and two GIRK4 subunits, plays a major role in modulating the parasympathetic nervous system's influence on cardiac physiology. Being that GIRK4 is necessary for the functional KACh channel, KCNJ5, which encodes GIRK4, it presents as a therapeutic target for cardiovascular pathology. Human variants in KCNJ5 have been identified in familial hyperaldosteronism type III, long QT syndrome, atrial fibrillation, and sinus node dysfunction. Here, we explore the relevance of KCNJ5 in each of these diseases. Further, we address the limitations and complexities of discerning the role of KCNJ5 in cardiovascular pathophysiology, as identical human variants of KCNJ5 have been identified in several diseases with overlapping pathophysiology.
Collapse
Affiliation(s)
- Karisa M. Meyer
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH 43210, USA; (K.M.M.); (N.M.); (J.s.K.)
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Nipun Malhotra
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH 43210, USA; (K.M.M.); (N.M.); (J.s.K.)
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jung seo Kwak
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH 43210, USA; (K.M.M.); (N.M.); (J.s.K.)
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Mona El Refaey
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH 43210, USA; (K.M.M.); (N.M.); (J.s.K.)
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
10
|
Peukert S, Gulgeze Efthymiou HB, Mo R, Peng Y, Ma F, Barbe G, Bebernitz G, Fridrich C, Buono C, Williams ET, Daniels T, Li L, Zhang X, Adachi Y, Abe M, Taggart AKP. Discovery of a brain-sparing GIRK1/4 inhibitor for pharmacological cardioversion of atrial fibrillation. Bioorg Med Chem Lett 2023; 85:129237. [PMID: 36924945 DOI: 10.1016/j.bmcl.2023.129237] [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: 01/20/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia, and a significant risk factor for ischemic stroke and heart failure. Marketed anti-arrhythmic drugs can restore sinus rhythm, but with limited efficacy and significant toxicities, including potential to induce ventricular arrhythmia. Atrial-selective ion channel drugs are expected to restore and maintain sinus rhythm without risk of ventricular arrhythmia. One such atrial-selective channel target is GIRK1/4 (G-protein regulated inwardly rectifying potassium channel 1/4). Here we describe 14b, a potent GIRK1/4 inhibitor developed to cardiovert AF to sinus rhythm while minimizing central nervous system exposure - an issue with preceding GIRK1/4 clinical candidates.
Collapse
Affiliation(s)
- Stefan Peukert
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | | | - Ruowei Mo
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Yunshan Peng
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Fupeng Ma
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Guillaume Barbe
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | | - Cary Fridrich
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Chiara Buono
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Eric T Williams
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Thomas Daniels
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Lisha Li
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Xia Zhang
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Yuichiro Adachi
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Mie Abe
- Former Novartis Employee, USA
| | - Andrew K P Taggart
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| |
Collapse
|
11
|
Campos-Ríos A, Rueda-Ruzafa L, Lamas JA. The Relevance of GIRK Channels in Heart Function. MEMBRANES 2022; 12:1119. [PMID: 36363674 PMCID: PMC9698958 DOI: 10.3390/membranes12111119] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 06/16/2023]
Abstract
Among the large number of potassium-channel families implicated in the control of neuronal excitability, G-protein-gated inwardly rectifying potassium channels (GIRK/Kir3) have been found to be a main factor in heart control. These channels are activated following the modulation of G-protein-coupled receptors and, although they have been implicated in different neurological diseases in both human and animal studies of the central nervous system, the therapeutic potential of different subtypes of these channel families in cardiac conditions has remained untapped. As they have emerged as a promising potential tool to treat a variety of conditions that disrupt neuronal homeostasis, many studies have started to focus on these channels as mediators of cardiac dynamics, thus leading to research into their implication in cardiovascular conditions. Our aim is to review the latest advances in GIRK modulation in the heart and their role in the cardiovascular system.
Collapse
Affiliation(s)
- Ana Campos-Ríos
- CINBIO, Laboratory of Neuroscience, University of Vigo, 36310 Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), 15706 Vigo, Spain
| | - Lola Rueda-Ruzafa
- Department of Nursing Science, Physiotherapy and Medicine, Faculty of Health Sciences, University of Almeria, 04120 Almeria, Spain
| | - José Antonio Lamas
- CINBIO, Laboratory of Neuroscience, University of Vigo, 36310 Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), 15706 Vigo, Spain
| |
Collapse
|
12
|
Cui M, Xu K, Gada K, Shalomov B, Ban M, Eptaminitaki GC, Kawano T, Plant LD, Dascal N, Logothetis DE. A novel small molecule selective activator of homomeric GIRK4 channels. J Biol Chem 2022; 298:102009. [PMID: 35525275 PMCID: PMC9194863 DOI: 10.1016/j.jbc.2022.102009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 11/26/2022] Open
Abstract
G protein–sensitive inwardly rectifying potassium (GIRK) channels are important pharmaceutical targets for neuronal, cardiac, and endocrine diseases. Although a number of GIRK channel modulators have been discovered in recent years, most lack selectivity. GIRK channels function as either homomeric (i.e., GIRK2 and GIRK4) or heteromeric (e.g., GIRK1/2, GIRK1/4, and GIRK2/3) tetramers. Activators, such as ML297, ivermectin, and GAT1508, have been shown to activate heteromeric GIRK1/2 channels better than GIRK1/4 channels with varying degrees of selectivity but not homomeric GIRK2 and GIRK4 channels. In addition, VU0529331 was discovered as the first homomeric GIRK channel activator, but it shows weak selectivity for GIRK2 over GIRK4 (or G4) homomeric channels. Here, we report the first highly selective small-molecule activator targeting GIRK4 homomeric channels, 3hi2one-G4 (3-[2-(3,4-dimethoxyphenyl)-2-oxoethyl]-3-hydroxy-1-(1-naphthylmethyl)-1,3-dihydro-2H-indol-2-one). We show that 3hi2one-G4 does not activate GIRK2, GIRK1/2, or GIRK1/4 channels. Using molecular modeling, mutagenesis, and electrophysiology, we analyzed the binding site of 3hi2one-G4 formed by the transmembrane 1, transmembrane 2, and slide helix regions of the GIRK4 channel, near the phosphatidylinositol-4,5-bisphosphate binding site, and show that it causes channel activation by strengthening channel–phosphatidylinositol-4,5-bisphosphate interactions. We also identify slide helix residue L77 in GIRK4, corresponding to residue I82 in GIRK2, as a major determinant of isoform-specific selectivity. We propose that 3hi2one-G4 could serve as a useful pharmaceutical probe in studying GIRK4 channel function and may also be pursued in drug optimization studies to tackle GIRK4-related diseases such as primary aldosteronism and late-onset obesity.
Collapse
Affiliation(s)
- Meng Cui
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, 02115, USA; Center for Drug Discovery, Northeastern University, Boston, MA 02115, USA.
| | - Keman Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, 02115, USA
| | - Kirin Gada
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, 02115, USA
| | - Boris Shalomov
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Michelle Ban
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, 02115, USA
| | - Giasemi C Eptaminitaki
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, 02115, USA
| | - Takeharu Kawano
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, 02115, USA
| | - Leigh D Plant
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, 02115, USA; Center for Drug Discovery, Northeastern University, Boston, MA 02115, USA
| | - Nathan Dascal
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Diomedes E Logothetis
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, 02115, USA; Chemistry and Chemical Biology, College of Science, Northeastern University, Boston, MA 02115, USA; Center for Drug Discovery, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Shalomov B, Handklo-Jamal R, Reddy HP, Theodor N, Bera AK, Dascal N. A revised mechanism of action of hyperaldosteronism-linked mutations in cytosolic domains of GIRK4 (KCNJ5). J Physiol 2021; 600:1419-1437. [PMID: 34957562 DOI: 10.1113/jp282690] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 12/21/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Mutations in GIRK4 (KCNJ5) G-protein gated channels cause primary aldosteronism, a major cause of secondary hypertension. The primary mechanism is believed to be loss of K+ selectivity. R52H and E246K, aldosteronism-causing mutations in cytosolic N- and C- termini of GIRK4, were reported to cause loss of K+ selectivity. We show that R52H, E246K and G247R mutations render homotetrameric GIRK channels non-functional. In heterotetrameric context with GIRK1, these mutations impair membrane expression, interaction with Gβγ and open probability, but do not alter K+ selectivity or inward rectification. In human aldosterone-secreting cell line, a GIRK4 opener and overexpression of heterotetrameric GIRK1/4WT , but not over-expression of GIRK1/4 mutants, reduced aldosterone secretion. Aldosteronism-causing mutations in cytosolic domain of GIRK4 are loss-of-function mutations rather than gain-of-function, selectivity-loss mutations. Deciphering of exact biophysical mechanism that impairs the channel is crucial for setting the course of treatment. ABSTRACT G-protein gated, inwardly rectifying potassium channels (GIRK) mediate inhibitory transmission in brain and heart, and are present in adrenal cortex. GIRK4 (KCNJ5) subunits are abundant in the heart and adrenal cortex. Multiple mutations of KCNJ5 cause primary aldosteronism (PA). Mutations in the pore region of GIRK4 cause loss of K+ selectivity, Na+ influx, and depolarization of zona glomerulosa cells followed by hypersecretion of aldosterone. The concept of selectivity loss has been extended to mutations in cytosolic domains of GIRK4 channels, remote from the pore. We expressed aldosteronism-linked GIRK4R52H , GIRK4E246K , and GIRK4G247R mutants in Xenopus oocytes. Whole-cell currents of heterotetrameric GIRK1/4R52H and GIRK1/4E246K channels were greatly reduced compared to GIRK1/4WT . Nevertheless, all heterotetrameric mutants retained full K+ selectivity and inward rectification. When expressed as homotetramers, only GIRK4WT , but none of the mutants, produced whole-cell currents. Confocal imaging, single channel and Förster Resonance Energy Transfer (FRET) analyses showed: 1) reduction of membrane abundance of all mutated channels, especially as homotetramers, 2) impaired interaction with Gβγ subunits, and 3) reduced open probability of GIRK1/4R52H . VU0529331, a GIRK4 opener, activated homotetrameric GIRK4G247R channels, but not GIRK4R52H and GIRK4E246K . In human adrenocortical carcinoma cell line (HAC15), VU0529331 and over-expression of heterotetrameric GIRK1/4WT , but not over-expression of GIRK1/4 mutants, reduced aldosterone secretion. Our results suggest that, contrary to pore mutants of GIRK4, non-pore mutants R52H and E246K mutants are loss-of-function rather than gain-of-function/selectivity-loss mutants. Hence, GIRK4 openers may be a potential course of treatment for patients with cytosolic N- and C-terminal mutations. Abstract Figure: There are two mutations types in KCNJ5 (GIRK4) that can cause excessive secretion of aldosterone, leading to primary aldosteronism. Mutations of the first type render the channel non-selective to monovalent cations and often constitutively active, thus depolarizing the zona granulosa cells. This previously described mechanism underlies the disease-causing effects of mutations of amino acid residues located in the pore region (red color). Blockers of the channel may be useful as potential treatment to reduce aldosterone secretion. Here we show that mutations of the second type, located in the cytosolic domain remote from the pore, act by a different mechanism. They do not alter channel's ion selectivity or rectification but cause poor expression or poor activation by Gβγ, resulting in a reduction in cell's K+ conductance and depolarization. In this case, GIRK4 openers can potentially be useful to prevent the excessive aldosterone secretion. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Boris Shalomov
- Department of Physiology and Pharmacology, School of Medicine, and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Reem Handklo-Jamal
- Department of Physiology and Pharmacology, School of Medicine, and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Haritha P Reddy
- Department of Physiology and Pharmacology, School of Medicine, and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.,Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Neta Theodor
- Department of Physiology and Pharmacology, School of Medicine, and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Amal K Bera
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Nathan Dascal
- Department of Physiology and Pharmacology, School of Medicine, and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| |
Collapse
|
14
|
Takla M, Edling CE, Zhang K, Saadeh K, Tse G, Salvage SC, Huang CL, Jeevaratnam K. Transcriptional profiles of genes related to electrophysiological function in Scn5a +/- murine hearts. Physiol Rep 2021; 9:e15043. [PMID: 34617689 PMCID: PMC8495800 DOI: 10.14814/phy2.15043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 11/24/2022] Open
Abstract
The Scn5a gene encodes the major pore-forming Nav 1.5 (α) subunit, of the voltage-gated Na+ channel in cardiomyocytes. The key role of Nav 1.5 in action potential initiation and propagation in both atria and ventricles predisposes organisms lacking Scn5a or carrying Scn5a mutations to cardiac arrhythmogenesis. Loss-of-function Nav 1.5 genetic abnormalities account for many cases of the human arrhythmic disorder Brugada syndrome (BrS) and related conduction disorders. A murine model with a heterozygous Scn5a deletion recapitulates many electrophysiological phenotypes of BrS. This study examines the relationships between its Scn5a+/- genotype, resulting transcriptional changes, and the consequent phenotypic presentations of BrS. Of 62 selected protein-coding genes related to cardiomyocyte electrophysiological or homeostatic function, concentrations of mRNA transcribed from 15 differed significantly from wild type (WT). Despite halving apparent ventricular Scn5a transcription heterozygous deletion did not significantly downregulate its atrial expression, raising possibilities of atria-specific feedback mechanisms. Most of the remaining 14 genes whose expression differed significantly between WT and Scn5a+/- animals involved Ca2+ homeostasis specifically in atrial tissue, with no overlap with any ventricular changes. All statistically significant changes in expression were upregulations in the atria and downregulations in the ventricles. This investigation demonstrates the value of future experiments exploring for and clarifying links between transcriptional control of Scn5a and of genes whose protein products coordinate Ca2+ regulation and examining their possible roles in BrS.
Collapse
Affiliation(s)
- Michael Takla
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- Christ’s CollegeUniversity of CambridgeCambridgeUK
| | | | - Kevin Zhang
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- School of MedicineImperial College LondonLondonUK
| | - Khalil Saadeh
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- Clinical SchoolUniversity of CambridgeCambridgeUK
| | - Gary Tse
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- Second Hospital of Tianjin Medical UniversityTianjinChina
| | | | - Christopher L.‐H. Huang
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
15
|
Anderson A, Vo BN, de Velasco EMF, Hopkins CR, Weaver CD, Wickman K. Characterization of VU0468554, a New Selective Inhibitor of Cardiac G Protein-Gated Inwardly Rectifying K + Channels. Mol Pharmacol 2021; 100:540-547. [PMID: 34503975 DOI: 10.1124/molpharm.121.000311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022] Open
Abstract
G protein-gated inwardly rectifying K+ (GIRK) channels are critical mediators of excitability in the heart and brain. Enhanced GIRK-channel activity has been implicated in the pathogenesis of supraventricular arrhythmias, including atrial fibrillation. The lack of selective pharmacological tools has impeded efforts to investigate the therapeutic potential of cardiac GIRK-channel interventions in arrhythmias. Here, we characterize a recently identified GIRK-channel inhibitor, VU0468554. Using whole-cell electrophysiological approaches and primary cultures of sinoatrial nodal cells and hippocampal neurons, we show that VU0468554 more effectively inhibits the cardiac GIRK channel than the neuronal GIRK channel. Concentration-response experiments suggest that VU0468554 inhibits Gβγ-activated GIRK channels in noncompetitive and potentially uncompetitive fashion. In contrast, VU0468554 competitively inhibits GIRK-channel activation by ML297, a GIRK-channel activator containing the same chemical scaffold as VU0468554. In the isolated heart model, VU0468554 partially reversed carbachol-induced bradycardia in hearts from wild-type mice but not Girk4-/- mice. Collectively, these data suggest that VU0468554 represents a promising new pharmacological tool for targeting cardiac GIRK channels with therapeutic implications for relevant cardiac arrhythmias. SIGNIFICANCE STATEMENT: Although cardiac GIRK-channel inhibition shows promise for the treatment of supraventricular arrhythmias, the absence of subtype-selective channel inhibitors has hindered exploration into this therapeutic strategy. This study utilizes whole-cell patch-clamp electrophysiology to characterize the new GIRK-channel inhibitor VU0468554 in human embryonic kidney 293T cells and primary cultures. We report that VU0468554 exhibits a favorable pharmacodynamic profile for cardiac over neuronal GIRK channels and partially reverses GIRK-mediated bradycardia in the isolated mouse heart model.
Collapse
Affiliation(s)
- Allison Anderson
- Graduate Program in Pharmacology (A.A., B.N.V.) and Department of Pharmacology (E.M.F.d.V., K.W.), University of Minnesota, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.); and Departments of Pharmacology and Chemistry and Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (C.D.W.)
| | - Baovi N Vo
- Graduate Program in Pharmacology (A.A., B.N.V.) and Department of Pharmacology (E.M.F.d.V., K.W.), University of Minnesota, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.); and Departments of Pharmacology and Chemistry and Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (C.D.W.)
| | - Ezequiel Marron Fernandez de Velasco
- Graduate Program in Pharmacology (A.A., B.N.V.) and Department of Pharmacology (E.M.F.d.V., K.W.), University of Minnesota, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.); and Departments of Pharmacology and Chemistry and Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (C.D.W.)
| | - Corey R Hopkins
- Graduate Program in Pharmacology (A.A., B.N.V.) and Department of Pharmacology (E.M.F.d.V., K.W.), University of Minnesota, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.); and Departments of Pharmacology and Chemistry and Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (C.D.W.)
| | - C David Weaver
- Graduate Program in Pharmacology (A.A., B.N.V.) and Department of Pharmacology (E.M.F.d.V., K.W.), University of Minnesota, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.); and Departments of Pharmacology and Chemistry and Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (C.D.W.)
| | - Kevin Wickman
- Graduate Program in Pharmacology (A.A., B.N.V.) and Department of Pharmacology (E.M.F.d.V., K.W.), University of Minnesota, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska (C.R.H.); and Departments of Pharmacology and Chemistry and Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee (C.D.W.)
| |
Collapse
|
16
|
Matos-Nieves A, Manivannan S, Majumdar U, McBride KL, White P, Garg V. A Multi-Omics Approach Using a Mouse Model of Cardiac Malformations for Prioritization of Human Congenital Heart Disease Contributing Genes. Front Cardiovasc Med 2021; 8:683074. [PMID: 34504875 PMCID: PMC8421733 DOI: 10.3389/fcvm.2021.683074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/22/2021] [Indexed: 01/22/2023] Open
Abstract
Congenital heart disease (CHD) is the most common type of birth defect, affecting ~1% of all live births. Malformations of the cardiac outflow tract (OFT) account for ~30% of all CHD and include a range of CHDs from bicuspid aortic valve (BAV) to tetralogy of Fallot (TOF). We hypothesized that transcriptomic profiling of a mouse model of CHD would highlight disease-contributing genes implicated in congenital cardiac malformations in humans. To test this hypothesis, we utilized global transcriptional profiling differences from a mouse model of OFT malformations to prioritize damaging, de novo variants identified from exome sequencing datasets from published cohorts of CHD patients. Notch1 +/- ; Nos3 -/- mice display a spectrum of cardiac OFT malformations ranging from BAV, semilunar valve (SLV) stenosis to TOF. Global transcriptional profiling of the E13.5 Notch1 +/- ; Nos3 -/- mutant mouse OFTs and wildtype controls was performed by RNA sequencing (RNA-Seq). Analysis of the RNA-Seq dataset demonstrated genes belonging to the Hif1α, Tgf-β, Hippo, and Wnt signaling pathways were differentially expressed in the mutant OFT. Mouse to human comparative analysis was then performed to determine if patients with TOF and SLV stenosis display an increased burden of damaging, genetic variants in gene homologs that were dysregulated in Notch1 +/- ; Nos3 -/- OFT. We found an enrichment of de novo variants in the TOF population among the 1,352 significantly differentially expressed genes in Notch1 +/- ; Nos3 -/- mouse OFT but not the SLV population. This association was not significant when comparing only highly expressed genes in the murine OFT to de novo variants in the TOF population. These results suggest that transcriptomic datasets generated from the appropriate temporal, anatomic and cellular tissues from murine models of CHD may provide a novel approach for the prioritization of disease-contributing genes in patients with CHD.
Collapse
Affiliation(s)
- Adrianna Matos-Nieves
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Sathiyanarayanan Manivannan
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Uddalak Majumdar
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
| | - Kim L. McBride
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
| | - Peter White
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Vidu Garg
- Center for Cardiovascular Research and Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, Ohio State University, Columbus, OH, United States
| |
Collapse
|
17
|
Kir Channel Molecular Physiology, Pharmacology, and Therapeutic Implications. Handb Exp Pharmacol 2021; 267:277-356. [PMID: 34345939 DOI: 10.1007/164_2021_501] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
For the past two decades several scholarly reviews have appeared on the inwardly rectifying potassium (Kir) channels. We would like to highlight two efforts in particular, which have provided comprehensive reviews of the literature up to 2010 (Hibino et al., Physiol Rev 90(1):291-366, 2010; Stanfield et al., Rev Physiol Biochem Pharmacol 145:47-179, 2002). In the past decade, great insights into the 3-D atomic resolution structures of Kir channels have begun to provide the molecular basis for their functional properties. More recently, computational studies are beginning to close the time domain gap between in silico dynamic and patch-clamp functional studies. The pharmacology of these channels has also been expanding and the dynamic structural studies provide hope that we are heading toward successful structure-based drug design for this family of K+ channels. In the present review we focus on placing the physiology and pharmacology of this K+ channel family in the context of atomic resolution structures and in providing a glimpse of the promising future of therapeutic opportunities.
Collapse
|
18
|
Choi S, Baudot M, Vivas O, Moreno CM. Slowing down as we age: aging of the cardiac pacemaker's neural control. GeroScience 2021; 44:1-17. [PMID: 34292477 PMCID: PMC8811107 DOI: 10.1007/s11357-021-00420-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022] Open
Abstract
The cardiac pacemaker ignites and coordinates the contraction of the whole heart, uninterruptedly, throughout our entire life. Pacemaker rate is constantly tuned by the autonomous nervous system to maintain body homeostasis. Sympathetic and parasympathetic terminals act over the pacemaker cells as the accelerator and the brake pedals, increasing or reducing the firing rate of pacemaker cells to match physiological demands. Despite the remarkable reliability of this tissue, the pacemaker is not exempt from the detrimental effects of aging. Mammals experience a natural and continuous decrease in the pacemaker rate throughout the entire lifespan. Why the pacemaker rhythm slows with age is poorly understood. Neural control of the pacemaker is remodeled from birth to adulthood, with strong evidence of age-related dysfunction that leads to a downshift of the pacemaker. Such evidence includes remodeling of pacemaker tissue architecture, alterations in the innervation, changes in the sympathetic acceleration and the parasympathetic deceleration, and alterations in the responsiveness of pacemaker cells to adrenergic and cholinergic modulation. In this review, we revisit the main evidence on the neural control of the pacemaker at the tissue and cellular level and the effects of aging on shaping this neural control.
Collapse
Affiliation(s)
- Sabrina Choi
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Matthias Baudot
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Oscar Vivas
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Claudia M Moreno
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
19
|
Ahern BM, Sebastian A, Levitan BM, Goh J, Andres DA, Satin J. L-type channel inactivation balances the increased peak calcium current due to absence of Rad in cardiomyocytes. J Gen Physiol 2021; 153:212476. [PMID: 34269819 PMCID: PMC8289690 DOI: 10.1085/jgp.202012854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
The L-type Ca2+ channel (LTCC) provides trigger calcium to initiate cardiac contraction in a graded fashion that is regulated by L-type calcium current (ICa,L) amplitude and kinetics. Inactivation of LTCC is controlled to fine-tune calcium flux and is governed by voltage-dependent inactivation (VDI) and calcium-dependent inactivation (CDI). Rad is a monomeric G protein that regulates ICa,L and has recently been shown to be critical to β-adrenergic receptor (β-AR) modulation of ICa,L. Our previous work showed that cardiomyocyte-specific Rad knockout (cRadKO) resulted in elevated systolic function, underpinned by an increase in peak ICa,L, but without pathological remodeling. Here, we sought to test whether Rad-depleted LTCC contributes to the fight-or-flight response independently of β-AR function, resulting in ICa,L kinetic modifications to homeostatically balance cardiomyocyte function. We recorded whole-cell ICa,L from ventricular cardiomyocytes from inducible cRadKO and control (CTRL) mice. The kinetics of ICa,L stimulated with isoproterenol in CTRL cardiomyocytes were indistinguishable from those of unstimulated cRadKO cardiomyocytes. CDI and VDI are both enhanced in cRadKO cardiomyocytes without differences in action potential duration or QT interval. To confirm that Rad loss modulates LTCC independently of β-AR stimulation, we crossed a β1,β2-AR double-knockout mouse with cRadKO, resulting in a Rad-inducible triple-knockout mouse. Deletion of Rad in cardiomyocytes that do not express β1,β2-AR still yielded modulated ICa,L and elevated basal heart function. Thus, in the absence of Rad, increased Ca2+ influx is homeostatically balanced by accelerated CDI and VDI. Our results indicate that the absence of Rad can modulate the LTCC without contribution of β1,β2-AR signaling and that Rad deletion supersedes β-AR signaling to the LTCC to enhance in vivo heart function.
Collapse
Affiliation(s)
- Brooke M Ahern
- Department of Physiology, University of Kentucky, Lexington, KY
| | | | - Bryana M Levitan
- Department of Physiology, University of Kentucky, Lexington, KY.,Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY
| | - Jensen Goh
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
20
|
Hupfeld J, Ernst M, Knyrim M, Binas S, Kloeckner U, Rabe S, Quarch K, Misiak D, Fuszard M, Grossmann C, Gekle M, Schreier B. miR-208b Reduces the Expression of Kcnj5 in a Cardiomyocyte Cell Line. Biomedicines 2021; 9:719. [PMID: 34201741 PMCID: PMC8301481 DOI: 10.3390/biomedicines9070719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRs) contribute to different aspects of cardiovascular pathology, among them cardiac hypertrophy and atrial fibrillation. Cardiac miR expression was analyzed in a mouse model with structural and electrical remodeling. Next-generation sequencing revealed that miR-208b-3p was ~25-fold upregulated. Therefore, the aim of our study was to evaluate the impact of miR-208b on cardiac protein expression. First, an undirected approach comparing whole RNA sequencing data to miR-walk 2.0 miR-208b 3'-UTR targets revealed 58 potential targets of miR-208b being regulated. We were able to show that miR-208b mimics bind to the 3' untranslated region (UTR) of voltage-gated calcium channel subunit alpha1 C and Kcnj5, two predicted targets of miR-208b. Additionally, we demonstrated that miR-208b mimics reduce GIRK1/4 channel-dependent thallium ion flux in HL-1 cells. In a second undirected approach we performed mass spectrometry to identify the potential targets of miR-208b. We identified 40 potential targets by comparison to miR-walk 2.0 3'-UTR, 5'-UTR and CDS targets. Among those targets, Rock2 and Ran were upregulated in Western blots of HL-1 cells by miR-208b mimics. In summary, miR-208b targets the mRNAs of proteins involved in the generation of cardiac excitation and propagation, as well as of proteins involved in RNA translocation (Ran) and cardiac hypertrophic response (Rock2).
Collapse
Affiliation(s)
- Julia Hupfeld
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Maximilian Ernst
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Maria Knyrim
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Stephanie Binas
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Udo Kloeckner
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Sindy Rabe
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Katja Quarch
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Danny Misiak
- Institute of Molecular Medicine, Medical Faculty of the Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, 06120 Halle (Saale), Germany;
| | - Matthew Fuszard
- Zentrum für Medizinische Grundlagenforschung, Core Facility—Proteomic Mass Spectrometry, Proteinzentrum Charles Tanford, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| | - Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Medical Faculty of the Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (J.H.); (M.E.); (M.K.); (S.B.); (U.K.); (S.R.); (K.Q.); (C.G.); (M.G.)
| |
Collapse
|
21
|
DiFrancesco ML, Mesirca P, Bidaud I, Isbrandt D, Mangoni ME. The funny current in genetically modified mice. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:39-50. [PMID: 34129872 DOI: 10.1016/j.pbiomolbio.2021.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 12/27/2022]
Abstract
Since its first description in 1979, the hyperpolarization-activated funny current (If) has been the object of intensive research aimed at understanding its role in cardiac pacemaker activity and its modulation by the sympathetic and parasympathetic branches of the autonomic nervous system. If was described in isolated tissue strips of the rabbit sinoatrial node using the double-electrode voltage-clamp technique. Since then, the rabbit has been the principal animal model for studying pacemaker activity and If for more than 20 years. In 2001, the first study describing the electrophysiological properties of mouse sinoatrial pacemaker myocytes and those of If was published. It was soon followed by the description of murine myocytes of the atrioventricular node and the Purkinje fibres. The sinoatrial node of genetically modified mice has become a very popular model for studying the mechanisms of cardiac pacemaker activity. This field of research benefits from the impressive advancement of in-vivo exploration techniques of physiological parameters, imaging, genetics, and large-scale genomic approaches. The present review discusses the influence of mouse genetic on the most recent knowledge of the funny current's role in the physiology and pathophysiology of cardiac pacemaker activity. Genetically modified mice have provided important insights into the role of If in determining intrinsic automaticity in vivo and in myocytes of the conduction system. In addition, gene targeting of f-(HCN) channel isoforms have contributed to elucidating the current's role in the regulation of heart rate by the parasympathetic nervous system. This review is dedicated to Dario DiFrancesco on his retirement.
Collapse
Affiliation(s)
- Mattia L DiFrancesco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France; LabEx Ion Channels Science and Therapeutics (ICST), France.
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France; LabEx Ion Channels Science and Therapeutics (ICST), France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France; LabEx Ion Channels Science and Therapeutics (ICST), France
| | - Dirk Isbrandt
- Deutsches Zentrum für Neurodegenerative Erktankungen (DZNE), Bonn, Germany; University of Cologne, Institute for Molecular and Behavioral Neuroscience, Cologne, Germany
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France; LabEx Ion Channels Science and Therapeutics (ICST), France.
| |
Collapse
|
22
|
Impaired regulation of heart rate and sinoatrial node function by the parasympathetic nervous system in type 2 diabetic mice. Sci Rep 2021; 11:12465. [PMID: 34127743 PMCID: PMC8203800 DOI: 10.1038/s41598-021-91937-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/31/2021] [Indexed: 01/01/2023] Open
Abstract
Heart rate (HR) and sinoatrial node (SAN) function are modulated by the autonomic nervous system. HR regulation by the parasympathetic nervous system (PNS) is impaired in diabetes mellitus (DM), which is denoted cardiovascular autonomic neuropathy. Whether blunted PNS effects on HR in type 2 DM are related to impaired responsiveness of the SAN to PNS agonists is unknown. This was investigated in type 2 diabetic db/db mice in vivo and in isolated SAN myocytes. The PNS agonist carbachol (CCh) had a smaller inhibitory effect on HR, while HR recovery time after CCh removal was accelerated in db/db mice. In isolated SAN myocytes CCh reduced spontaneous action potential firing frequency but this effect was reduced in db/db mice due to blunted effects on diastolic depolarization slope and maximum diastolic potential. Impaired effects of CCh occurred due to enhanced desensitization of the acetylcholine-activated K+ current (IKACh) and faster IKACh deactivation. IKACh alterations were reversed by inhibition of regulator of G-protein signaling 4 (RGS4) and by the phospholipid PIP3. SAN expression of RGS4 was increased in db/db mice. Impaired PNS regulation of HR in db/db mice occurs due to reduced responsiveness of SAN myocytes to PNS agonists in association with enhanced RGS4 activity.
Collapse
|
23
|
Tatsi C, Maria AG, Malloy C, Lin L, London E, Settas N, Flippo C, Keil M, Hannah-Shmouni F, Hoffman DA, Stratakis CA. Cushing Syndrome in a Pediatric Patient With a KCNJ5 Variant and Successful Treatment With Low-dose Ketoconazole. J Clin Endocrinol Metab 2021; 106:1606-1616. [PMID: 33630995 PMCID: PMC8118581 DOI: 10.1210/clinem/dgab118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT Pathogenic variants in KCNJ5, encoding the GIRK4 (Kir3.4) potassium channel, have been implicated in the pathogenesis of familial hyperaldosteronism type-III (FH-III) and sporadic primary aldosteronism (PA). In addition to aldosterone, glucocorticoids are often found elevated in PA in association with KCNJ5 pathogenic variants, albeit at subclinical levels. However, to date no GIRK4 defects have been linked to Cushing syndrome (CS). PATIENT We present the case of a 10-year-old child who presented with CS at an early age due to bilateral adrenocortical hyperplasia (BAH). The patient was placed on low-dose ketoconazole (KZL), which controlled hypercortisolemia and CS-related signs. Discontinuation of KZL for even 6 weeks led to recurrent CS. RESULTS Screening for known genes causing cortisol-producing BAHs (PRKAR1A, PRKACA, PRKACB, PDE11A, PDE8B, ARMC5) failed to identify any gene defects. Whole-exome sequencing showed a novel KCNJ5 pathogenic variant (c.506T>C, p.L169S) inherited from her father. In vitro studies showed that the p.L169S variant affects conductance of the Kir3.4 channel without affecting its expression or membrane localization. Although there were no effects on steroidogenesis in vitro, there were modest changes in protein kinase A activity. In silico analysis of the mutant channel proposed mechanisms for the altered conductance. CONCLUSION We present a pediatric patient with CS due to BAH and a germline defect in KCNJ5. Molecular investigations of this KCNJ5 variant failed to show a definite cause of her CS. However, this KCNJ5 variant differed in its function from KCNJ5 defects leading to PA. We speculate that GIRK4 (Kir3.4) may play a role in early human adrenocortical development and zonation and participate in the pathogenesis of pediatric BAH.
Collapse
Affiliation(s)
- Christina Tatsi
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
- Internal medicine and Pediatric Endocrinology Inter-institute Training Programs, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
- Correspondence: Christina Tatsi MD, MHSc, PhD, 10 Center Drive, Building 10, NIH-Clinical Research Center, Room 1-3330, MSC1103, Bethesda, MD 20892,USA.
| | - Andrea G Maria
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Cole Malloy
- Section on Molecular Neurophysiology and Biophysics, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Lin Lin
- Section on Molecular Neurophysiology and Biophysics, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Edra London
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Nick Settas
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Chelsi Flippo
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
- Internal medicine and Pediatric Endocrinology Inter-institute Training Programs, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Meg Keil
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
- Internal medicine and Pediatric Endocrinology Inter-institute Training Programs, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Fady Hannah-Shmouni
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
- Internal medicine and Pediatric Endocrinology Inter-institute Training Programs, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Dax A Hoffman
- Section on Molecular Neurophysiology and Biophysics, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
- Internal medicine and Pediatric Endocrinology Inter-institute Training Programs, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892,USA
| |
Collapse
|
24
|
Wallace MJ, El Refaey M, Mesirca P, Hund TJ, Mangoni ME, Mohler PJ. Genetic Complexity of Sinoatrial Node Dysfunction. Front Genet 2021; 12:654925. [PMID: 33868385 PMCID: PMC8047474 DOI: 10.3389/fgene.2021.654925] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
The pacemaker cells of the cardiac sinoatrial node (SAN) are essential for normal cardiac automaticity. Dysfunction in cardiac pacemaking results in human sinoatrial node dysfunction (SND). SND more generally occurs in the elderly population and is associated with impaired pacemaker function causing abnormal heart rhythm. Individuals with SND have a variety of symptoms including sinus bradycardia, sinus arrest, SAN block, bradycardia/tachycardia syndrome, and syncope. Importantly, individuals with SND report chronotropic incompetence in response to stress and/or exercise. SND may be genetic or secondary to systemic or cardiovascular conditions. Current management of patients with SND is limited to the relief of arrhythmia symptoms and pacemaker implantation if indicated. Lack of effective therapeutic measures that target the underlying causes of SND renders management of these patients challenging due to its progressive nature and has highlighted a critical need to improve our understanding of its underlying mechanistic basis of SND. This review focuses on current information on the genetics underlying SND, followed by future implications of this knowledge in the management of individuals with SND.
Collapse
Affiliation(s)
- Michael J. Wallace
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mona El Refaey
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Pietro Mesirca
- CNRS, INSERM, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France
- Laboratory of Excellence ICST, Montpellier, France
| | - Thomas J. Hund
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Matteo E. Mangoni
- CNRS, INSERM, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France
- Laboratory of Excellence ICST, Montpellier, France
| | - Peter J. Mohler
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
25
|
Sebastian S, Nobles M, Tsisanova E, Ludwig A, Munroe PB, Tinker A. The role of resistance to inhibitors of cholinesterase 8b in the control of heart rate. Physiol Genomics 2021; 53:150-159. [PMID: 33719582 DOI: 10.1152/physiolgenomics.00157.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have assessed the role of ric-b8 in the control of heart rate after the gene was implicated in a recent genome-wide association study of resting heart rate. We developed a novel murine model in which it was possible to conditionally delete ric-8b in the sinoatrial (SA) node after the addition of tamoxifen. Despite this, we were unable to obtain homozygotes and thus studied heterozygotes. Haploinsufficiency of ric-8b in the sinoatrial node induced by the addition of tamoxifen in adult animals leads to mice with a reduced heart rate. However, other electrocardiographic intervals (e.g., PR and QRS) were normal, and there was no apparent arrhythmia such as heart block. The positive chronotropic response to isoprenaline was abrogated, whereas the response to carbachol was unchanged. The pacemaker current If (funny current) has an important role in regulating heart rate, and its function is modulated by both isoprenaline and carbachol. Using a heterologous system expressing HCN4, we show that ric-8b can modulate the HCN4 current. Overexpression of ric-8b led to larger HCN4 currents, whereas silencing ric-8b led to smaller currents. Ric-8b modulates heart rate responses in vivo likely via its actions on the stimulatory G-protein.
Collapse
Affiliation(s)
- Sonia Sebastian
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Muriel Nobles
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Elena Tsisanova
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Patricia B Munroe
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Andrew Tinker
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
26
|
Bidaud I, D'Souza A, Forte G, Torre E, Greuet D, Thirard S, Anderson C, Chung You Chong A, Torrente AG, Roussel J, Wickman K, Boyett MR, Mangoni ME, Mesirca P. Genetic Ablation of G Protein-Gated Inwardly Rectifying K + Channels Prevents Training-Induced Sinus Bradycardia. Front Physiol 2021; 11:519382. [PMID: 33551824 PMCID: PMC7857143 DOI: 10.3389/fphys.2020.519382] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 12/17/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Endurance athletes are prone to bradyarrhythmias, which in the long-term may underscore the increased incidence of pacemaker implantation reported in this population. Our previous work in rodent models has shown training-induced sinus bradycardia to be due to microRNA (miR)-mediated transcriptional remodeling of the HCN4 channel, leading to a reduction of the "funny" (I f) current in the sinoatrial node (SAN). Objective: To test if genetic ablation of G-protein-gated inwardly rectifying potassium channel, also known as I KACh channels prevents sinus bradycardia induced by intensive exercise training in mice. Methods: Control wild-type (WT) and mice lacking GIRK4 (Girk4 -/-), an integral subunit of I KACh were assigned to trained or sedentary groups. Mice in the trained group underwent 1-h exercise swimming twice a day for 28 days, 7 days per week. We performed electrocardiogram recordings and echocardiography in both groups at baseline, during and after the training period. At training cessation, mice were euthanized and SAN tissues were isolated for patch clamp recordings in isolated SAN cells and molecular profiling by quantitative PCR (qPCR) and western blotting. Results: At swimming cessation trained WT mice presented with a significantly lower resting HR that was reversible by acute I KACh block whereas Girk4 -/- mice failed to develop a training-induced sinus bradycardia. In line with HR reduction, action potential rate, density of I f, as well as of T- and L-type Ca2+ currents (I CaT and I CaL ) were significantly reduced only in SAN cells obtained from WT-trained mice. I f reduction in WT mice was concomitant with downregulation of HCN4 transcript and protein, attributable to increased expression of corresponding repressor microRNAs (miRs) whereas reduced I CaL in WT mice was associated with reduced Cav1.3 protein levels. Strikingly, I KACh ablation suppressed all training-induced molecular remodeling observed in WT mice. Conclusion: Genetic ablation of cardiac I KACh in mice prevents exercise-induced sinus bradycardia by suppressing training induced remodeling of inward currents I f, I CaT and I CaL due in part to the prevention of miR-mediated transcriptional remodeling of HCN4 and likely post transcriptional remodeling of Cav1.3. Strategies targeting cardiac I KACh may therefore represent an alternative to pacemaker implantation for bradyarrhythmias seen in some veteran athletes.
Collapse
Affiliation(s)
- Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx Ion Channels Science and Therapeutics, Montpellier, France
| | - Alicia D'Souza
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Gabriella Forte
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Eleonora Torre
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx Ion Channels Science and Therapeutics, Montpellier, France
| | - Denis Greuet
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Steeve Thirard
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Cali Anderson
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Antony Chung You Chong
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx Ion Channels Science and Therapeutics, Montpellier, France
| | - Angelo G Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx Ion Channels Science and Therapeutics, Montpellier, France
| | - Julien Roussel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | - Mark R Boyett
- Division of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx Ion Channels Science and Therapeutics, Montpellier, France
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.,LabEx Ion Channels Science and Therapeutics, Montpellier, France
| |
Collapse
|
27
|
Zhao Y, Gameiro-Ros I, Glaaser IW, Slesinger PA. Advances in Targeting GIRK Channels in Disease. Trends Pharmacol Sci 2021; 42:203-215. [PMID: 33468322 DOI: 10.1016/j.tips.2020.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/30/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022]
Abstract
G protein-gated inwardly rectifying potassium (GIRK) channels are essential regulators of cell excitability in the brain. While they are implicated in a variety of neurological diseases in both human and animal model studies, their therapeutic potential has been largely untapped. Here, we review recent advances in the development of small molecule compounds that specifically modulate GIRK channels and compare them with first-generation compounds that exhibit off-target activity. We describe the method of discovery of these small molecule modulators, their chemical features, and their effects in vivo. These studies provide a promising outlook on the future development of subunit-specific GIRK modulators to regulate neuronal excitability in a brain region-specific manner.
Collapse
Affiliation(s)
- Yulin Zhao
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Isabel Gameiro-Ros
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ian W Glaaser
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul A Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
28
|
Liang D, Xue J, Geng L, Zhou L, Lv B, Zeng Q, Xiong K, Zhou H, Xie D, Zhang F, Liu J, Liu Y, Li L, Yang J, Xue Z, Chen YH. Cellular and molecular landscape of mammalian sinoatrial node revealed by single-cell RNA sequencing. Nat Commun 2021; 12:287. [PMID: 33436583 PMCID: PMC7804277 DOI: 10.1038/s41467-020-20448-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Bioelectrical impulses intrinsically generated within the sinoatrial node (SAN) trigger the contraction of the heart in mammals. Though discovered over a century ago, the molecular and cellular features of the SAN that underpin its critical function in the heart are uncharted territory. Here, we identify four distinct transcriptional clusters by single-cell RNA sequencing in the mouse SAN. Functional analysis of differentially expressed genes identifies a core cell cluster enriched in the electrogenic genes. The similar cellular features are also observed in the SAN from both rabbit and cynomolgus monkey. Notably, Vsnl1, a core cell cluster marker in mouse, is abundantly expressed in SAN, but is barely detectable in atrium or ventricle, suggesting that Vsnl1 is a potential SAN marker. Importantly, deficiency of Vsnl1 not only reduces the beating rate of human induced pluripotent stem cell - derived cardiomyocytes (hiPSC-CMs) but also the heart rate of mice. Furthermore, weighted gene co-expression network analysis (WGCNA) unveiled the core gene regulation network governing the function of the SAN in mice. Overall, these findings reveal the whole transcriptome profiling of the SAN at single-cell resolution, representing an advance toward understanding of both the biology and the pathology of SAN. The spontaneous bioelectrical activity of pacemaker cells in sinoatrial node (SAN) triggers the heartbeats. Here, the authors perform single-cell RNA sequencing in the mouse SAN and identify molecular and cellular features of the SAN conserved in rabbit and cynomolgus monkey, identifying a new potential SAN marker.
Collapse
Affiliation(s)
- Dandan Liang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Jinfeng Xue
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200092, China
| | - Li Geng
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Liping Zhou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Bo Lv
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qiao Zeng
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ke Xiong
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Huixing Zhou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Duanyang Xie
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Fulei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Jie Liu
- Translational Center of Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yi Liu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China
| | - Li Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jian Yang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China.,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Zhigang Xue
- Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai, 200092, China. .,Reproductive Medicine Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Yi-Han Chen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China. .,Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China. .,Institute of Medical Genetics, Tongji University, Shanghai, 200092, China. .,Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
29
|
Mesirca P, Fedorov VV, Hund TJ, Torrente AG, Bidaud I, Mohler PJ, Mangoni ME. Pharmacologic Approach to Sinoatrial Node Dysfunction. Annu Rev Pharmacol Toxicol 2021; 61:757-778. [PMID: 33017571 PMCID: PMC7790915 DOI: 10.1146/annurev-pharmtox-031120-115815] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The spontaneous activity of the sinoatrial node initiates the heartbeat. Sino-atrial node dysfunction (SND) and sick sinoatrial (sick sinus) syndrome are caused by the heart's inability to generate a normal sinoatrial node action potential. In clinical practice, SND is generally considered an age-related pathology, secondary to degenerative fibrosis of the heart pacemaker tissue. However, other forms of SND exist, including idiopathic primary SND, which is genetic, and forms that are secondary to cardiovascular or systemic disease. The incidence of SND in the general population is expected to increase over the next half century, boosting the need to implant electronic pacemakers. During the last two decades, our knowledge of sino-atrial node physiology and of the pathophysiological mechanisms underlying SND has advanced considerably. This review summarizes the current knowledge about SND mechanisms and discusses the possibility of introducing new pharmacologic therapies for treating SND.
Collapse
Affiliation(s)
- Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Vadim V Fedorov
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio 43210, USA
| | - Thomas J Hund
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Angelo G Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Peter J Mohler
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio 43210, USA
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| |
Collapse
|
30
|
Torrente AG, Mesirca P, Bidaud I, Mangoni ME. Channelopathies of voltage-gated L-type Cav1.3/α 1D and T-type Cav3.1/α 1G Ca 2+ channels in dysfunction of heart automaticity. Pflugers Arch 2020; 472:817-830. [PMID: 32601767 DOI: 10.1007/s00424-020-02421-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/12/2020] [Accepted: 06/19/2020] [Indexed: 10/24/2022]
Abstract
The heart automaticity is a fundamental physiological function in vertebrates. The cardiac impulse is generated in the sinus node by a specialized population of spontaneously active myocytes known as "pacemaker cells." Failure in generating or conducting spontaneous activity induces dysfunction in cardiac automaticity. Several families of ion channels are involved in the generation and regulation of the heart automaticity. Among those, voltage-gated L-type Cav1.3 (α1D) and T-type Cav3.1 (α1G) Ca2+ channels play important roles in the spontaneous activity of pacemaker cells. Ca2+ channel channelopathies specifically affecting cardiac automaticity are considered rare. Recent research on familial disease has identified mutations in the Cav1.3-encoding CACNA1D gene that underlie congenital sinus node dysfunction and deafness (OMIM # 614896). In addition, both Cav1.3 and Cav3.1 channels have been identified as pathophysiological targets of sinus node dysfunction and heart block, caused by congenital autoimmune disease of the cardiac conduction system. The discovery of channelopathies linked to Cav1.3 and Cav3.1 channels underscores the importance of Ca2+ channels in the generation and regulation of heart's automaticity.
Collapse
Affiliation(s)
- Angelo G Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx Ion Channels Science and Therapeutics (ICST), Montpellier, France
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx Ion Channels Science and Therapeutics (ICST), Montpellier, France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx Ion Channels Science and Therapeutics (ICST), Montpellier, France
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141, rue de la cardonille, 34094, Montpellier, France. .,LabEx Ion Channels Science and Therapeutics (ICST), Montpellier, France.
| |
Collapse
|
31
|
Anderson A, Masuho I, Marron Fernandez de Velasco E, Nakano A, Birnbaumer L, Martemyanov KA, Wickman K. GPCR-dependent biasing of GIRK channel signaling dynamics by RGS6 in mouse sinoatrial nodal cells. Proc Natl Acad Sci U S A 2020; 117:14522-14531. [PMID: 32513692 PMCID: PMC7322085 DOI: 10.1073/pnas.2001270117] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
How G protein-coupled receptors (GPCRs) evoke specific biological outcomes while utilizing a limited array of G proteins and effectors is poorly understood, particularly in native cell systems. Here, we examined signaling evoked by muscarinic (M2R) and adenosine (A1R) receptor activation in the mouse sinoatrial node (SAN), the cardiac pacemaker. M2R and A1R activate a shared pool of cardiac G protein-gated inwardly rectifying K+ (GIRK) channels in SAN cells from adult mice, but A1R-GIRK responses are smaller and slower than M2R-GIRK responses. Recordings from mice lacking Regulator of G protein Signaling 6 (RGS6) revealed that RGS6 exerts a GPCR-dependent influence on GIRK-dependent signaling in SAN cells, suppressing M2R-GIRK coupling efficiency and kinetics and A1R-GIRK signaling amplitude. Fast kinetic bioluminescence resonance energy transfer assays in transfected HEK cells showed that RGS6 prefers Gαo over Gαi as a substrate for its catalytic activity and that M2R signals preferentially via Gαo, while A1R does not discriminate between inhibitory G protein isoforms. The impact of atrial/SAN-selective ablation of Gαo or Gαi2 was consistent with these findings. Gαi2 ablation had minimal impact on M2R-GIRK and A1R-GIRK signaling in SAN cells. In contrast, Gαo ablation decreased the amplitude and slowed the kinetics of M2R-GIRK responses, while enhancing the sensitivity and prolonging the deactivation rate of A1R-GIRK signaling. Collectively, our data show that differences in GPCR-G protein coupling preferences, and the Gαo substrate preference of RGS6, shape A1R- and M2R-GIRK signaling dynamics in mouse SAN cells.
Collapse
Affiliation(s)
- Allison Anderson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| | | | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA 90095
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
- Biomedical Research Institute, Catholic University of Argentina, C1107AAZ Buenos Aires, Argentina
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455;
| |
Collapse
|
32
|
Inhibition of G protein-gated K + channels by tertiapin-Q rescues sinus node dysfunction and atrioventricular conduction in mouse models of primary bradycardia. Sci Rep 2020; 10:9835. [PMID: 32555258 PMCID: PMC7300035 DOI: 10.1038/s41598-020-66673-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Sinus node (SAN) dysfunction (SND) manifests as low heart rate (HR) and is often accompanied by atrial tachycardia or atrioventricular (AV) block. The only currently available therapy for chronic SND is the implantation of an electronic pacemaker. Because of the growing burden of SND in the population, new pharmacological therapies of chronic SND and heart block are desirable. We developed a collection of genetically modified mouse strains recapitulating human primary SND associated with different degrees of AV block. These mice were generated with genetic ablation of L-type Cav1.3 (Cav1.3-/-), T-type Cav3.1 (Cav3.1-/-), or both (Cav1.3-/-/Cav3.1-/-). We also studied mice haplo-insufficient for the Na+ channel Nav1.5 (Nav1.5+/) and mice in which the cAMP-dependent regulation of hyperpolarization-activated f-(HCN4) channels has been abolished (HCN4-CNBD). We analysed, by telemetric ECG recording, whether pharmacological inhibition of the G-protein-activated K+ current (IKACh) by the peptide tertiapin-Q could improve HR and AV conduction in these mouse strains. Tertiapin-Q significantly improved the HR of Cav1.3-/- (19%), Cav1.3-/-/Cav3.1-/- (23%) and HCN4-CNBD (14%) mice. Tertiapin-Q also improved cardiac conduction of Nav1.5+/- mice by 24%. Our data suggest that the development of pharmacological IKACh inhibitors for the management of SND and conduction disease is a viable approach.
Collapse
|
33
|
Kuß J, Stallmeyer B, Goldstein M, Rinné S, Pees C, Zumhagen S, Seebohm G, Decher N, Pott L, Kienitz MC, Schulze-Bahr E. Familial Sinus Node Disease Caused by a Gain of GIRK (G-Protein Activated Inwardly Rectifying K + Channel) Channel Function. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 12:e002238. [PMID: 30645171 DOI: 10.1161/circgen.118.002238] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Inherited forms of sinus node dysfunction (SND) clinically include bradycardia, sinus arrest, and chronotropic incompetence and may serve as disease models to understand sinus node physiology and impulse generation. Recently, a gain-of-function mutation in the G-protein gene GNB2 led to enhanced activation of the GIRK (G-protein activated inwardly rectifying K+ channel). Thus, human cardiac GIRK channels are important for heart rate regulation and subsequently, genes encoding their subunits Kir3.1 and Kir3.4 ( KCNJ3 and KCNJ5) are potential candidates for inherited SND in human. METHODS We performed a combined approach of targeted sequencing of KCNJ3 and KCNJ5 in 52 patients with idiopathic SND and subsequent whole exome sequencing of additional family members in a genetically affected patient. A putative novel disease-associated gene variant was functionally analyzed by voltage-clamp experiments using various heterologous cell expression systems (Xenopus oocytes, CHO cells, and rat atrial cardiomyocytes). RESULTS In a 3-generation family with SND we identified a novel variant in KCNJ5 which leads to an amino acid substitution (p.Trp101Cys) in the first transmembrane domain of the Kir3.4 subunit of the cardiac GIRK channel. The identified variant cosegregated with the disease in the family and was absent in the Exome Variant Server and Exome Aggregation Consortium databases. Expression of mutant Kir3.4 (±native Kir3.1) in different heterologous cell expression systems resulted in increased GIRK currents ( IK,ACh) and a reduced inward rectification which was not compensated by intracellular spermidine. Moreover, in silico modeling of heterotetrameric mutant GIRK channels indicates a structurally altered binding site for spermine. CONCLUSIONS For the first time, an inherited gain-of-function mutation in the human GIRK3.4 causes familial human SND. The increased activity of GIRK channels is likely to lead to a sustained hyperpolarization of pacemaker cells and thereby reduces heart rate. Modulation of human GIRK channels may pave a way for further treatment of cardiac pacemaking.
Collapse
Affiliation(s)
- Johanna Kuß
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Germany (J.K., B.S., S.Z., G.S., E.S.-B.)
| | - Birgit Stallmeyer
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Germany (J.K., B.S., S.Z., G.S., E.S.-B.)
| | - Matthias Goldstein
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Germany (M.G., S.R., N.D.)
| | - Susanne Rinné
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Germany (M.G., S.R., N.D.)
| | - Christiane Pees
- Department of Pediatric Cardiology, University Children's Hospital Vienna, Austria (C.P.)
| | - Sven Zumhagen
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Germany (J.K., B.S., S.Z., G.S., E.S.-B.)
| | - Guiscard Seebohm
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Germany (J.K., B.S., S.Z., G.S., E.S.-B.)
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, Germany (M.G., S.R., N.D.)
| | - Lutz Pott
- Department of Cardiovascular Medicine, Institute of Physiology, Ruhr-University Bochum, Germany (L.P., M.-C.K.)
| | - Marie-Cécile Kienitz
- Department of Cardiovascular Medicine, Institute of Physiology, Ruhr-University Bochum, Germany (L.P., M.-C.K.)
| | - Eric Schulze-Bahr
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Germany (J.K., B.S., S.Z., G.S., E.S.-B.)
| |
Collapse
|
34
|
MacDonald EA, Rose RA, Quinn TA. Neurohumoral Control of Sinoatrial Node Activity and Heart Rate: Insight From Experimental Models and Findings From Humans. Front Physiol 2020; 11:170. [PMID: 32194439 PMCID: PMC7063087 DOI: 10.3389/fphys.2020.00170] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
The sinoatrial node is perhaps one of the most important tissues in the entire body: it is the natural pacemaker of the heart, making it responsible for initiating each-and-every normal heartbeat. As such, its activity is heavily controlled, allowing heart rate to rapidly adapt to changes in physiological demand. Control of sinoatrial node activity, however, is complex, occurring through the autonomic nervous system and various circulating and locally released factors. In this review we discuss the coupled-clock pacemaker system and how its manipulation by neurohumoral signaling alters heart rate, considering the multitude of canonical and non-canonical agents that are known to modulate sinoatrial node activity. For each, we discuss the principal receptors involved and known intracellular signaling and protein targets, highlighting gaps in our knowledge and understanding from experimental models and human studies that represent areas for future research.
Collapse
Affiliation(s)
- Eilidh A. MacDonald
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Robert A. Rose
- Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - T. Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
35
|
Binas S, Knyrim M, Hupfeld J, Kloeckner U, Rabe S, Mildenberger S, Quarch K, Strätz N, Misiak D, Gekle M, Grossmann C, Schreier B. miR-221 and -222 target CACNA1C and KCNJ5 leading to altered cardiac ion channel expression and current density. Cell Mol Life Sci 2020; 77:903-918. [PMID: 31312877 PMCID: PMC7058603 DOI: 10.1007/s00018-019-03217-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/14/2019] [Accepted: 07/02/2019] [Indexed: 12/26/2022]
Abstract
MicroRNAs (miRs) contribute to different aspects of cardiovascular pathology, among others cardiac hypertrophy and atrial fibrillation. The aim of our study was to evaluate the impact of miR-221/222 on cardiac electrical remodeling. Cardiac miR expression was analyzed in a mouse model with altered electrocardiography parameters and severe heart hypertrophy. Next generation sequencing revealed 14 differentially expressed miRs in hypertrophic hearts, with miR-221 and -222 being the strongest regulated miR-cluster. This increase was restricted to cardiomyocytes and not observed in cardiac fibroblasts. Additionally, we evaluated the change of miR-221/222 in vivo in two models of pharmacologically induced heart hypertrophy (angiotensin II, isoprenaline), thereby demonstrating a stimulus-induced increase in miR-221/222 in vivo by angiotensin II but not by isoprenaline. Whole transcriptome analysis by RNA-seq and qRT-PCR validation revealed an enriched number of downregulated mRNAs coding for proteins located in the T-tubule, which are also predicted targets for miR-221/222. Among those, mRNAs were the L-type Ca2+ channel subunits as well as potassium channel subunits. We confirmed that both miRs target the 3'-untranslated regions of Cacna1c and Kcnj5. Furthermore, enhanced expression of these miRs reduced L-type Ca2+ channel and Kcnj5 channel abundance and function, which was analyzed by whole-cell patch clamp recordings or Western blot and flux measurements, respectively. miR-221 and -222 contribute to the regulation of L-type Ca2+ channels as well as Kcnj5 channels and, therefore, potentially contribute to disturbed cardiac excitation generation and propagation. Future studies will have to evaluate the pathophysiological and clinical relevance of aberrant miR-221/222 expression for electrical remodeling.
Collapse
Affiliation(s)
- Stephanie Binas
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Maria Knyrim
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Julia Hupfeld
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Udo Kloeckner
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Sindy Rabe
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Sigrid Mildenberger
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Katja Quarch
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Nicole Strätz
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Danny Misiak
- Institute of Molecular Medicine, Martin-Luther-University Halle-Wittenberg, Heinrich-Damerow-Str. 1, 06120, Halle/Saale, Germany
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany.
| |
Collapse
|
36
|
Yamada N, Asano Y, Fujita M, Yamazaki S, Inanobe A, Matsuura N, Kobayashi H, Ohno S, Ebana Y, Tsukamoto O, Ishino S, Takuwa A, Kioka H, Yamashita T, Hashimoto N, Zankov DP, Shimizu A, Asakura M, Asanuma H, Kato H, Nishida Y, Miyashita Y, Shinomiya H, Naiki N, Hayashi K, Makiyama T, Ogita H, Miura K, Ueshima H, Komuro I, Yamagishi M, Horie M, Kawakami K, Furukawa T, Koizumi A, Kurachi Y, Sakata Y, Minamino T, Kitakaze M, Takashima S. Mutant KCNJ3 and KCNJ5 Potassium Channels as Novel Molecular Targets in Bradyarrhythmias and Atrial Fibrillation. Circulation 2020; 139:2157-2169. [PMID: 30764634 DOI: 10.1161/circulationaha.118.036761] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Bradyarrhythmia is a common clinical manifestation. Although the majority of cases are acquired, genetic analysis of families with bradyarrhythmia has identified a growing number of causative gene mutations. Because the only ultimate treatment for symptomatic bradyarrhythmia has been invasive surgical implantation of a pacemaker, the discovery of novel therapeutic molecular targets is necessary to improve prognosis and quality of life. METHODS We investigated a family containing 7 individuals with autosomal dominant bradyarrhythmias of sinus node dysfunction, atrial fibrillation with slow ventricular response, and atrioventricular block. To identify the causative mutation, we conducted the family-based whole exome sequencing and genome-wide linkage analysis. We characterized the mutation-related mechanisms based on the pathophysiology in vitro. After generating a transgenic animal model to confirm the human phenotypes of bradyarrhythmia, we also evaluated the efficacy of a newly identified molecular-targeted compound to upregulate heart rate in bradyarrhythmias by using the animal model. RESULTS We identified one heterozygous mutation, KCNJ3 c.247A>C, p.N83H, as a novel cause of hereditary bradyarrhythmias in this family. KCNJ3 encodes the inwardly rectifying potassium channel Kir3.1, which combines with Kir3.4 (encoded by KCNJ5) to form the acetylcholine-activated potassium channel ( IKACh channel) with specific expression in the atrium. An additional study using a genome cohort of 2185 patients with sporadic atrial fibrillation revealed another 5 rare mutations in KCNJ3 and KCNJ5, suggesting the relevance of both genes to these arrhythmias. Cellular electrophysiological studies revealed that the KCNJ3 p.N83H mutation caused a gain of IKACh channel function by increasing the basal current, even in the absence of m2 muscarinic receptor stimulation. We generated transgenic zebrafish expressing mutant human KCNJ3 in the atrium specifically. It is interesting to note that the selective IKACh channel blocker NIP-151 repressed the increased current and improved bradyarrhythmia phenotypes in the mutant zebrafish. CONCLUSIONS The IKACh channel is associated with the pathophysiology of bradyarrhythmia and atrial fibrillation, and the mutant IKACh channel ( KCNJ3 p.N83H) can be effectively inhibited by NIP-151, a selective IKACh channel blocker. Thus, the IKACh channel might be considered to be a suitable pharmacological target for patients who have bradyarrhythmia with a gain-of-function mutation in the IKACh channel.
Collapse
Affiliation(s)
- Noriaki Yamada
- Departments of Cardiovascular Medicine (N.Y., Y.A., A.T., H. Kioka, Y.M., H.S., Y.S.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshihiro Asano
- Departments of Cardiovascular Medicine (N.Y., Y.A., A.T., H. Kioka, Y.M., H.S., Y.S.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Masashi Fujita
- Department of Onco-cardiology, Osaka International Cancer Institute, Japan (M.F.)
| | - Satoru Yamazaki
- Departments of Cell Biology (S.Y.), National Cerebral and Cardiovascular Center, Suita, Japan
| | - Atsushi Inanobe
- Pharmacology (A.I., Y.K.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Norio Matsuura
- Departments of Health and Environmental Sciences (N.M.), Kyoto University Graduate School of Medicine, Japan
| | - Hatasu Kobayashi
- Department of Biomedical Sciences, College of Life and Health Sciences Chubu University, Kasugai, Japan (H. Kobayashi)
| | - Seiko Ohno
- Bioscience and Genetics (S.O.), National Cerebral and Cardiovascular Center, Suita, Japan.,Center for Epidemiologic Research in Asia (S.O., K.M., H.U., M.H.), Shiga University of Medical Science, Otsu, Japan
| | - Yusuke Ebana
- Life Science and Bioethics Research Center (Y.E.), Tokyo Medical and Dental University, Japan
| | - Osamu Tsukamoto
- Medical Biochemistry (O.T., H. Kato, Y.N., S.T.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Saki Ishino
- Center of Medical Innovation and Translational Research (S.I.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Ayako Takuwa
- Departments of Cardiovascular Medicine (N.Y., Y.A., A.T., H. Kioka, Y.M., H.S., Y.S.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Hidetaka Kioka
- Departments of Cardiovascular Medicine (N.Y., Y.A., A.T., H. Kioka, Y.M., H.S., Y.S.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Toru Yamashita
- Pharmaceuticals Division, Nissan Chemical Corporation, Tokyo, Japan (T.Y., N.H.)
| | - Norio Hashimoto
- Pharmaceuticals Division, Nissan Chemical Corporation, Tokyo, Japan (T.Y., N.H.)
| | - Dimitar P Zankov
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology (D.P.Z., A.S., H.O.), Shiga University of Medical Science, Otsu, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology (D.P.Z., A.S., H.O.), Shiga University of Medical Science, Otsu, Japan
| | - Masanori Asakura
- Cardiovascular Division, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan (M.A.)
| | - Hiroshi Asanuma
- Department of Internal Medicine, Meiji University of Integrative Medicine, Nantan, Japan (H.A.)
| | - Hisakazu Kato
- Medical Biochemistry (O.T., H. Kato, Y.N., S.T.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuya Nishida
- Medical Biochemistry (O.T., H. Kato, Y.N., S.T.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Yohei Miyashita
- Departments of Cardiovascular Medicine (N.Y., Y.A., A.T., H. Kioka, Y.M., H.S., Y.S.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Haruki Shinomiya
- Departments of Cardiovascular Medicine (N.Y., Y.A., A.T., H. Kioka, Y.M., H.S., Y.S.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Nobu Naiki
- Departments of Cardiovascular Medicine (N.N., M.H.), Shiga University of Medical Science, Otsu, Japan
| | - Kenshi Hayashi
- Department of Cardiovascular and Internal Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (K.H., M.Y.)
| | - Takeru Makiyama
- Cardiovascular Medicine (T. Makiyama), Kyoto University Graduate School of Medicine, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology (D.P.Z., A.S., H.O.), Shiga University of Medical Science, Otsu, Japan
| | - Katsuyuki Miura
- Center for Epidemiologic Research in Asia (S.O., K.M., H.U., M.H.), Shiga University of Medical Science, Otsu, Japan.,Public Health (K.M., H.U.), Shiga University of Medical Science, Otsu, Japan
| | - Hirotsugu Ueshima
- Center for Epidemiologic Research in Asia (S.O., K.M., H.U., M.H.), Shiga University of Medical Science, Otsu, Japan.,Public Health (K.M., H.U.), Shiga University of Medical Science, Otsu, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Japan (I.K.)
| | - Masakazu Yamagishi
- Department of Cardiovascular and Internal Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan (K.H., M.Y.).,Department of Human Sciences, Osaka University of Human Sciences, Settsu, Japan (M.Y.)
| | - Minoru Horie
- Center for Epidemiologic Research in Asia (S.O., K.M., H.U., M.H.), Shiga University of Medical Science, Otsu, Japan.,Departments of Cardiovascular Medicine (N.N., M.H.), Shiga University of Medical Science, Otsu, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan (K.K.).,Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Japan (K.K.)
| | - Tetsushi Furukawa
- Department of Bioinformational Pharmacology (T.F.), Tokyo Medical and Dental University, Japan
| | - Akio Koizumi
- Public Interest Foundation Kyoto Hokenkai, Japan (A.K.)
| | - Yoshihisa Kurachi
- Pharmacology (A.I., Y.K.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasushi Sakata
- Departments of Cardiovascular Medicine (N.Y., Y.A., A.T., H. Kioka, Y.M., H.S., Y.S.), Osaka University Graduate School of Medicine, Suita, Japan
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Japan (T. Minamino)
| | - Masafumi Kitakaze
- Clinical Medicine and Development (M.K.), National Cerebral and Cardiovascular Center, Suita, Japan
| | - Seiji Takashima
- Medical Biochemistry (O.T., H. Kato, Y.N., S.T.), Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
37
|
Abstract
A progressive decline in maximum heart rate (mHR) is a fundamental aspect of aging in humans and other mammals. This decrease in mHR is independent of gender, fitness, and lifestyle, affecting in equal measure women and men, athletes and couch potatoes, spinach eaters and fast food enthusiasts. Importantly, the decline in mHR is the major determinant of the age-dependent decline in aerobic capacity that ultimately limits functional independence for many older individuals. The gradual reduction in mHR with age reflects a slowing of the intrinsic pacemaker activity of the sinoatrial node of the heart, which results from electrical remodeling of individual pacemaker cells along with structural remodeling and a blunted β-adrenergic response. In this review, we summarize current evidence about the tissue, cellular, and molecular mechanisms that underlie the reduction in pacemaker activity with age and highlight key areas for future work.
Collapse
Affiliation(s)
- Colin H Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA; , ,
| | - Emily J Sharpe
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA; , ,
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA; , ,
- Department of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| |
Collapse
|
38
|
Wei AD, Ramirez JM. Presynaptic Mechanisms and KCNQ Potassium Channels Modulate Opioid Depression of Respiratory Drive. Front Physiol 2019; 10:1407. [PMID: 31824331 PMCID: PMC6882777 DOI: 10.3389/fphys.2019.01407] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/31/2019] [Indexed: 01/02/2023] Open
Abstract
Opioid-induced respiratory depression (OIRD) is the major cause of death associated with opioid analgesics and drugs of abuse, but the underlying cellular and molecular mechanisms remain poorly understood. We investigated opioid action in vivo in unanesthetized mice and in in vitro medullary slices containing the preBötzinger Complex (preBötC), a locus critical for breathing and inspiratory rhythm generation. Although hypothesized as a primary mechanism, we found that mu-opioid receptor (MOR1)-mediated GIRK activation contributed only modestly to OIRD. Instead, mEPSC recordings from genetically identified Dbx1-derived interneurons, essential for rhythmogenesis, revealed a prevalent presynaptic mode of action for OIRD. Consistent with MOR1-mediated suppression of presynaptic release as a major component of OIRD, Cacna1a KO slices lacking P/Q-type Ca2+ channels enhanced OIRD. Furthermore, OIRD was mimicked and reversed by KCNQ potassium channel activators and blockers, respectively. In vivo whole-body plethysmography combined with systemic delivery of GIRK- and KCNQ-specific potassium channel drugs largely recapitulated these in vitro results, and revealed state-dependent modulation of OIRD. We propose that respiratory failure from OIRD results from a general reduction of synaptic efficacy, leading to a state-dependent collapse of rhythmic network activity.
Collapse
Affiliation(s)
- Aguan D. Wei
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, United States
| | - Jan-Marino Ramirez
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
39
|
Garg P, Garg V, Shrestha R, Sanguinetti MC, Kamp TJ, Wu JC. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes as Models for Cardiac Channelopathies: A Primer for Non-Electrophysiologists. Circ Res 2019; 123:224-243. [PMID: 29976690 DOI: 10.1161/circresaha.118.311209] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Life threatening ventricular arrhythmias leading to sudden cardiac death are a major cause of morbidity and mortality. In the absence of structural heart disease, these arrhythmias, especially in the younger population, are often an outcome of genetic defects in specialized membrane proteins called ion channels. In the heart, exceptionally well-orchestrated activity of a diversity of ion channels mediates the cardiac action potential. Alterations in either the function or expression of these channels can disrupt the configuration of the action potential, leading to abnormal electrical activity of the heart that can sometimes initiate an arrhythmia. Understanding the pathophysiology of inherited arrhythmias can be challenging because of the complexity of the disorder and lack of appropriate cellular and in vivo models. Recent advances in human induced pluripotent stem cell technology have provided remarkable progress in comprehending the underlying mechanisms of ion channel disorders or channelopathies by modeling these complex arrhythmia syndromes in vitro in a dish. To fully realize the potential of induced pluripotent stem cells in elucidating the mechanistic basis and complex pathophysiology of channelopathies, it is crucial to have a basic knowledge of cardiac myocyte electrophysiology. In this review, we will discuss the role of the various ion channels in cardiac electrophysiology and the molecular and cellular mechanisms of arrhythmias, highlighting the promise of human induced pluripotent stem cell-cardiomyocytes as a model for investigating inherited arrhythmia syndromes and testing antiarrhythmic strategies. Overall, this review aims to provide a basic understanding of the electrical activity of the heart and related channelopathies, especially to clinicians or research scientists in the cardiovascular field with limited electrophysiology background.
Collapse
Affiliation(s)
- Priyanka Garg
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.).,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| | - Vivek Garg
- Stanford University School of Medicine, CA; Department of Physiology, University of California San Francisco (V.G.)
| | - Rajani Shrestha
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.).,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| | | | - Timothy J Kamp
- Department of Medicine, University of Wisconsin-Madison (T.J.K.)
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute (P.G., R.S., J.C.W.) .,Department of Medicine, Division of Cardiology (P.G., R.S., J.C.W.).,Institute for Stem Cell Biology and Regenerative Medicine (P.G., R.S., J.C.W.)
| |
Collapse
|
40
|
Veerman CC, Mengarelli I, Koopman CD, Wilders R, van Amersfoorth SC, Bakker D, Wolswinkel R, Hababa M, de Boer TP, Guan K, Milnes J, Lodder EM, Bakkers J, Verkerk AO, Bezzina CR. Genetic variation in GNB5 causes bradycardia by augmenting the cholinergic response via increased acetylcholine-activated potassium current ( I K,ACh). Dis Model Mech 2019; 12:dmm.037994. [PMID: 31208990 PMCID: PMC6679373 DOI: 10.1242/dmm.037994] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 06/06/2019] [Indexed: 12/19/2022] Open
Abstract
Mutations in GNB5, encoding the G-protein β5 subunit (Gβ5), have recently been linked to a multisystem disorder that includes severe bradycardia. Here, we investigated the mechanism underlying bradycardia caused by the recessive p.S81L Gβ5 variant. Using CRISPR/Cas9-based targeting, we generated an isogenic series of human induced pluripotent stem cell (hiPSC) lines that were either wild type, heterozygous or homozygous for the GNB5 p.S81L variant. These were differentiated into cardiomyocytes (hiPSC-CMs) that robustly expressed the acetylcholine-activated potassium channel [I(KACh); also known as IK,ACh]. Baseline electrophysiological properties of the lines did not differ. Upon application of carbachol (CCh), homozygous p.S81L hiPSC-CMs displayed an increased acetylcholine-activated potassium current (I K,ACh) density and a more pronounced decrease of spontaneous activity as compared to wild-type and heterozygous p.S81L hiPSC-CMs, explaining the bradycardia in homozygous carriers. Application of the specific I(KACh) blocker XEN-R0703 resulted in near-complete reversal of the phenotype. Our results provide mechanistic insights and proof of principle for potential therapy in patients carrying GNB5 mutations.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Christiaan C Veerman
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Isabella Mengarelli
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Charlotte D Koopman
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Ronald Wilders
- Amsterdam UMC, University of Amsterdam, Department of Medical Biology, Heart Failure Research Center, 1105 AZ Amsterdam, The Netherlands
| | - Shirley C van Amersfoorth
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Diane Bakker
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Rianne Wolswinkel
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Mariam Hababa
- Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Kaomei Guan
- Department of Pharmacology and Toxicology, Technische Universität Dresden, 01062 Dresden, Germany
| | | | - Elisabeth M Lodder
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| | - Jeroen Bakkers
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Arie O Verkerk
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands.,Amsterdam UMC, University of Amsterdam, Department of Medical Biology, Heart Failure Research Center, 1105 AZ Amsterdam, The Netherlands
| | - Connie R Bezzina
- Amsterdam UMC, University of Amsterdam, Department of Experimental Cardiology, Heart Center, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
41
|
Vo BN, Abney KK, Anderson A, Marron Fernandez de Velasco E, Benneyworth MA, Daniels JS, Morrison RD, Hopkins CR, Weaver CD, Wickman K. VU0810464, a non-urea G protein-gated inwardly rectifying K + (K ir 3/GIRK) channel activator, exhibits enhanced selectivity for neuronal K ir 3 channels and reduces stress-induced hyperthermia in mice. Br J Pharmacol 2019; 176:2238-2249. [PMID: 30924523 DOI: 10.1111/bph.14671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/16/2019] [Accepted: 02/20/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE G protein-gated inwardly rectifying K+ (Kir 3) channels moderate the activity of excitable cells and have been implicated in neurological disorders and cardiac arrhythmias. Most neuronal Kir 3 channels consist of Kir 3.1 and Kir 3.2 subtypes, while cardiac Kir 3 channels consist of Kir 3.1 and Kir 3.4 subtypes. Previously, we identified a family of urea-containing Kir 3 channel activators, but these molecules exhibit suboptimal pharmacokinetic properties and modest selectivity for Kir 3.1/3.2 relative to Kir 3.1/3.4 channels. Here, we characterize a non-urea activator, VU0810464, which displays nanomolar potency as a Kir 3.1/3.2 activator, improved selectivity for neuronal Kir 3 channels, and improved brain penetration. EXPERIMENTAL APPROACH We used whole-cell electrophysiology to measure the efficacy and potency of VU0810464 in neurons and the selectivity of VU0810464 for neuronal and cardiac Kir 3 channel subtypes. We tested VU0810464 in vivo in stress-induced hyperthermia and elevated plus maze paradigms. Parallel studies with ML297, the prototypical activator of Kir 3.1-containing Kir 3 channels, were performed to permit direct comparisons. KEY RESULTS VU0810464 and ML297 exhibited comparable efficacy and potency as neuronal Kir 3 channel activators, but VU0810464 was more selective for neuronal Kir 3 channels. VU0810464, like ML297, reduced stress-induced hyperthermia in a Kir 3-dependent manner in mice. ML297, but not VU0810464, decreased anxiety-related behaviour as assessed with the elevated plus maze test. CONCLUSION AND IMPLICATIONS VU0810464 represents a new class of Kir 3 channel activator with enhanced selectivity for Kir 3.1/3.2 channels. VU0810464 may be useful for examining Kir 3.1/3.2 channel contributions to complex behaviours and for probing the potential of Kir 3 channel-dependent manipulations to treat neurological disorders.
Collapse
Affiliation(s)
- Baovi N Vo
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Kristopher K Abney
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN
| | - Allison Anderson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | | | | | | | - Ryan D Morrison
- Research and Development, Precera Bioscience, Inc., Franklin, TN
| | - Corey R Hopkins
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
42
|
Kozek KA, Du Y, Sharma S, Prael FJ, Spitznagel BD, Kharade SV, Denton JS, Hopkins CR, Weaver CD. Discovery and Characterization of VU0529331, a Synthetic Small-Molecule Activator of Homomeric G Protein-Gated, Inwardly Rectifying, Potassium (GIRK) Channels. ACS Chem Neurosci 2019; 10:358-370. [PMID: 30136838 PMCID: PMC6528656 DOI: 10.1021/acschemneuro.8b00287] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
G protein-gated, inwardly rectifying, potassium (GIRK) channels are important regulators of cellular excitability throughout the body. GIRK channels are heterotetrameric and homotetrameric combinations of the Kir3.1-4 (GIRK1-4) subunits. Different subunit combinations are expressed throughout the central nervous system (CNS) and the periphery, and most of these combinations contain a GIRK1 subunit. For example, the predominance of GIRK channels in the CNS are composed of GIRK1 and GIRK2 subunits, while the GIRK channels in cardiac atrial myocytes are made up mostly of GIRK1 and GIRK4 subunits. Although the vast majority of GIRK channels contain a GIRK1 subunit, discrete populations of cells that express non-GIRK1-containing GIRK (non-GIRK1/X) channels do exist. For instance, dopaminergic neurons in the ventral tegmental area of the brain, associated with addiction and reward, do not express the GIRK1 subunit. Targeting these non-GIRK1/X channels with subunit-selective pharmacological probes could lead to important insights into how GIRK channels are involved in reward and addiction. Such insights may, in turn, reveal therapeutic opportunities for the treatment or prevention of addiction. Previously, our laboratory discovered small molecules that can specifically modulate the activity of GIRK1-containing GIRK channels. However, efforts to generate compounds active on non-GIRK1/X channels from these scaffolds have been unsuccessful. Recently, ivermectin was shown to modulate non-GIRK1/X channels, and historically, ivermectin is known to modulate a wide variety of neuronal channels and receptors. Further, ivermectin is a complex natural product, which makes it a challenging starting point for development of more selective, effective, and potent compounds. Thus, while ivermectin provides proof-of-concept as a non-GIRK1/X channel activator, it is of limited utility. Therefore, we sought to discover a synthetic small molecule that would serve as a starting point for the development of non-GIRK1/X channel modulators. To accomplish this, we used a high-throughput thallium flux assay to screen a 100 000-compound library in search of activators of homomeric GIRK2 channels. Using this approach, we discovered VU0529331, the first synthetic small molecule reported to activate non-GIRK1/X channels, to our knowledge. This discovery represents the first step toward developing potent and selective non-GIRK1/X channel probes. Such molecules will help elucidate the role of GIRK channels in addiction, potentially establishing a foundation for future development of therapies utilizing targeted GIRK channel modulation.
Collapse
Affiliation(s)
- Krystian A. Kozek
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Medical Scientist Training Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Yu Du
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Swagat Sharma
- Department of Pharmaceutical Sciences, Center for Drug Discovery, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Francis J. Prael
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Brittany D. Spitznagel
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Sujay V. Kharade
- Department of Anesthesiology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jerod S. Denton
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Anesthesiology, Vanderbilt University, Nashville, Tennessee, USA
| | - Corey R. Hopkins
- Department of Pharmaceutical Sciences, Center for Drug Discovery, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - C. David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
- vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
43
|
Moss R, Sachse FB, Moreno-Galindo EG, Navarro-Polanco RA, Tristani-Firouzi M, Seemann G. Modeling effects of voltage dependent properties of the cardiac muscarinic receptor on human sinus node function. PLoS Comput Biol 2018; 14:e1006438. [PMID: 30303952 PMCID: PMC6197694 DOI: 10.1371/journal.pcbi.1006438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 10/22/2018] [Accepted: 08/15/2018] [Indexed: 11/25/2022] Open
Abstract
The cardiac muscarinic receptor (M2R) regulates heart rate, in part, by modulating the acetylcholine (ACh) activated K+ current IK,ACh through dissociation of G-proteins, that in turn activate KACh channels. Recently, M2Rs were noted to exhibit intrinsic voltage sensitivity, i.e. their affinity for ligands varies in a voltage dependent manner. The voltage sensitivity of M2R implies that the affinity for ACh (and thus the ACh effect) varies throughout the time course of a cardiac electrical cycle. The aim of this study was to investigate the contribution of M2R voltage sensitivity to the rate and shape of the human sinus node action potentials in physiological and pathophysiological conditions. We developed a Markovian model of the IK,ACh modulation by voltage and integrated it into a computational model of human sinus node. We performed simulations with the integrated model varying ACh concentration and voltage sensitivity. Low ACh exerted a larger effect on IK,ACh at hyperpolarized versus depolarized membrane voltages. This led to a slowing of the pacemaker rate due to an attenuated slope of phase 4 depolarization with only marginal effect on action potential duration and amplitude. We also simulated the theoretical effects of genetic variants that alter the voltage sensitivity of M2R. Modest negative shifts in voltage sensitivity, predicted to increase the affinity of the receptor for ACh, slowed the rate of phase 4 depolarization and slowed heart rate, while modest positive shifts increased heart rate. These simulations support our hypothesis that altered M2R voltage sensitivity contributes to disease and provide a novel mechanistic foundation to study clinical disorders such as atrial fibrillation and inappropriate sinus tachycardia. Heart rate regulation is dependent upon a delicate interplay between parasympathetic and sympathetic nerve activity at the level of the sinus node. Acetylcholine slows the heart rate by activating the M2 muscarinic receptor (M2R) that, in turn, opens the acetylcholine-activated potassium channel (IK,ACh) to slow the firing of the sinus node. Surprisingly, the M2R is sensitive to membrane potential and undergoes conformational changes throughout the cardiac action potential that alter the affinity for acetylcholine, with secondary consequences for IK,ACh activity. Here, we investigated the contribution of M2R voltage sensitivity to the rate and shape of the human sinus node action potential in physiological and pathophysiological conditions, using a Markovian model of the IK,ACh channel integrated into a computational model of human sinus node. The computational model allowed us to assess the effects of potential genetic variants that alter specific properties of voltage sensitivity. Our results indicate that alterations in the M2R voltage sensitivity play a significant role in the physiology and pathophysiology of the human sinus node and atria. Our computational model is relevant for future studies aimed at the design and development of anti-arrhythmic agents that specifically target the unique voltage-sensitive properties of M2R.
Collapse
Affiliation(s)
- Robin Moss
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Freiburg, Germany
- Faculty of Medicine, Albert-Ludwigs University of Freiburg, Freiburg, Germany
- Institute of Biomedical Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Eloy G Moreno-Galindo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, Mexico
| | | | - Martin Tristani-Firouzi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Gunnar Seemann
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Freiburg, Germany
- Faculty of Medicine, Albert-Ludwigs University of Freiburg, Freiburg, Germany
- Institute of Biomedical Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
44
|
Salazar-Fajardo PD, Aréchiga-Figueroa IA, López-Serrano AL, Rodriguez-Elias JC, Alamilla J, Sánchez-Chapula JA, Tristani-Firouzi M, Navarro-Polanco RA, Moreno-Galindo EG. The voltage-sensitive cardiac M 2 muscarinic receptor modulates the inward rectification of the G protein-coupled, ACh-gated K + current. Pflugers Arch 2018; 470:1765-1776. [PMID: 30155776 DOI: 10.1007/s00424-018-2196-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/28/2018] [Accepted: 08/13/2018] [Indexed: 11/25/2022]
Abstract
The acetylcholine (ACh)-gated inwardly rectifying K+ current (IKACh) plays a vital role in cardiac excitability by regulating heart rate variability and vulnerability to atrial arrhythmias. These crucial physiological contributions are determined principally by the inwardly rectifying nature of IKACh. Here, we investigated the relative contribution of two distinct mechanisms of IKACh inward rectification measured in atrial myocytes: a rapid component due to KACh channel block by intracellular Mg2+ and polyamines; and a time- and concentration-dependent mechanism. The time- and ACh concentration-dependent inward rectification component was eliminated when IKACh was activated by GTPγS, a compound that bypasses the muscarinic-2 receptor (M2R) and directly stimulates trimeric G proteins to open KACh channels. Moreover, the time-dependent component of IKACh inward rectification was also eliminated at ACh concentrations that saturate the receptor. These observations indicate that the time- and concentration-dependent rectification mechanism is an intrinsic property of the receptor, M2R; consistent with our previous work demonstrating that voltage-dependent conformational changes in the M2R alter the receptor affinity for ACh. Our analysis of the initial and time-dependent components of IKACh indicate that rapid Mg2+-polyamine block accounts for 60-70% of inward rectification, with M2R voltage sensitivity contributing 30-40% at sub-saturating ACh concentrations. Thus, while both inward rectification mechanisms are extrinsic to the KACh channel, to our knowledge, this is the first description of extrinsic inward rectification of ionic current attributable to an intrinsic voltage-sensitive property of a G protein-coupled receptor.
Collapse
Affiliation(s)
- Pedro D Salazar-Fajardo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965, Colonia Villa San Sebastián, C.P, 28045, Colima, COL, Mexico
| | - Iván A Aréchiga-Figueroa
- CONACyT, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, Mexico
| | - Ana Laura López-Serrano
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965, Colonia Villa San Sebastián, C.P, 28045, Colima, COL, Mexico
| | - Julio C Rodriguez-Elias
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965, Colonia Villa San Sebastián, C.P, 28045, Colima, COL, Mexico
| | - Javier Alamilla
- CONACyT, Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima, COL, Mexico
| | - José A Sánchez-Chapula
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965, Colonia Villa San Sebastián, C.P, 28045, Colima, COL, Mexico
| | - Martin Tristani-Firouzi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Ricardo A Navarro-Polanco
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965, Colonia Villa San Sebastián, C.P, 28045, Colima, COL, Mexico.
| | - Eloy G Moreno-Galindo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965, Colonia Villa San Sebastián, C.P, 28045, Colima, COL, Mexico.
| |
Collapse
|
45
|
Lee SW, Anderson A, Guzman PA, Nakano A, Tolkacheva EG, Wickman K. Atrial GIRK Channels Mediate the Effects of Vagus Nerve Stimulation on Heart Rate Dynamics and Arrhythmogenesis. Front Physiol 2018; 9:943. [PMID: 30072916 PMCID: PMC6060443 DOI: 10.3389/fphys.2018.00943] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/27/2018] [Indexed: 01/09/2023] Open
Abstract
Diminished parasympathetic influence is central to the pathogenesis of cardiovascular diseases, including heart failure and hypertension. Stimulation of the vagus nerve has shown promise in treating cardiovascular disease, prompting renewed interest in understanding the signaling pathway(s) that mediate the vagal influence on cardiac physiology. Here, we evaluated the contribution of G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels to the effect of vagus nerve stimulation (VNS) on heart rate (HR), HR variability (HRV), and arrhythmogenesis in anesthetized mice. As parasympathetic fibers innervate both atria and ventricle, and GIRK channels contribute to the cholinergic impact on atrial and ventricular myocytes, we collected in vivo electrocardiogram recordings from mice lacking either atrial or ventricular GIRK channels, during VNS. VNS decreased HR and increased HRV in control mice, in a muscarinic receptor-dependent manner. This effect was preserved in mice lacking ventricular GIRK channels, but was nearly completely absent in mice lacking GIRK channels in the atria. In addition, atrial-specific ablation of GIRK channels conferred resistance to arrhythmic episodes induced by VNS. These data indicate that atrial GIRK channels are the primary mediators of the impact of VNS on HR, HRV, and arrhythmogenesis in the anesthetized mouse.
Collapse
Affiliation(s)
- Steven W. Lee
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Allison Anderson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | - Pilar A. Guzman
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
46
|
Kulkarni K, Xie X, Fernandez de Velasco EM, Anderson A, Martemyanov KA, Wickman K, Tolkacheva EG. The influences of the M2R-GIRK4-RGS6 dependent parasympathetic pathway on electrophysiological properties of the mouse heart. PLoS One 2018; 13:e0193798. [PMID: 29668674 PMCID: PMC5905881 DOI: 10.1371/journal.pone.0193798] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/20/2018] [Indexed: 02/07/2023] Open
Abstract
A large body of work has established the prominent roles of the atrial M2R-IKACh signaling pathway, and the negative regulatory protein RGS6, in modulating critical aspects of parasympathetic influence on cardiac function, including pace-making, heart rate (HR) variability (HRV), and atrial arrhythmogenesis. Despite increasing evidence of its innervation of the ventricles, and the expression of M2R, IKACh channel subunits, and RGS6 in ventricle, the effects of parasympathetic modulation on ventricular electrophysiology are less clear. The main objective of our study was to investigate the contribution of M2R-IKACh signaling pathway elements in murine ventricular electrophysiology, using in-vivo ECG measurements, isolated whole-heart optical mapping and constitutive knockout mice lacking IKACh (Girk4–/–) or RGS6 (Rgs6-/-). Consistent with previous findings, mice lacking GIRK4 exhibited diminished HR and HRV responses to the cholinergic agonist carbachol (CCh), and resistance to CCh-induced arrhythmic episodes. In line with its role as a negative regulator of atrial M2R-IKACh signaling, loss of RGS6 correlated with a mild resting bradycardia, enhanced HR and HRV responses to CCh, and increased propensity for arrhythmic episodes. Interestingly, ventricles from mice lacking GIRK4 or RGS6 both exhibited increased action potential duration (APD) at baseline, and APD was prolonged by CCh across all genotypes. Similarly, CCh significantly increased the slope of APD restitution in all genotypes. There was no impact of genotype or CCh on either conduction velocity or heterogeneity. Our data suggests that altered parasympathetic signaling through the M2R-IKACh pathway can affect ventricular electrophysiological properties distinct from its influence on atrial physiology.
Collapse
Affiliation(s)
- Kanchan Kulkarni
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Xueyi Xie
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | | | - Allison Anderson
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
47
|
Giudicessi JR, Wilde AAM, Ackerman MJ. The genetic architecture of long QT syndrome: A critical reappraisal. Trends Cardiovasc Med 2018; 28:453-464. [PMID: 29661707 DOI: 10.1016/j.tcm.2018.03.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 12/19/2022]
Abstract
Collectively, the completion of the Human Genome Project and subsequent development of high-throughput next-generation sequencing methodologies have revolutionized genomic research. However, the rapid sequencing and analysis of thousands upon thousands of human exomes and genomes has taught us that most genes, including those known to cause heritable cardiovascular disorders such as long QT syndrome, harbor an unexpected background rate of rare, and presumably innocuous, non-synonymous genetic variation. In this Review, we aim to reappraise the genetic architecture underlying both the acquired and congenital forms of long QT syndrome by examining how the clinical phenotype associated with and background genetic variation in long QT syndrome-susceptibility genes impacts the clinical validity of existing gene-disease associations and the variant classification and reporting strategies that serve as the foundation for diagnostic long QT syndrome genetic testing.
Collapse
Affiliation(s)
- John R Giudicessi
- Department of Cardiovascular Medicine (Cardiovascular Diseases Fellowship and Clinician-Investigator Training Programs), Mayo Clinic, Rochester, MN, United States
| | - Arthur A M Wilde
- Department of Medicine (Division of Cardiology), Columbia University Irving Medical Center, New York, NY, United States; Department of Clinical & Experimental Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael J Ackerman
- Departments of Cardiovascular Medicine (Division of Heart Rhythm Services), Pediatrics (Division of Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
48
|
Kozasa Y, Nakashima N, Ito M, Ishikawa T, Kimoto H, Ushijima K, Makita N, Takano M. HCN4 pacemaker channels attenuate the parasympathetic response and stabilize the spontaneous firing of the sinoatrial node. J Physiol 2018; 596:809-825. [PMID: 29315578 PMCID: PMC5830425 DOI: 10.1113/jp275303] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/02/2018] [Indexed: 01/01/2023] Open
Abstract
Key points The contribution of HCN4 pacemaker channels in the autonomic regulation of the sino‐atrial node (SAN) has been a matter of debate. The transgenic overexpression of HCN4 did not induce tachycardia, but reduced heart rate variability, while the conditional knockdown of HCN4 gave rise to sinus arrhythmia. The response of the SAN to β‐adrenergic stimulation was not affected by overexpression or knockdown of HCN4 channels. When HCN4 channels were knocked down, the parasympathetic response examined by cervical vagus nerve stimulation (CVNS) was enhanced; the CVNS induced complete sinus pause. The overexpression of HCN4 attenuated bradycardia induced by CVNS only during β‐adrenergic stimulation. We concluded that HCN4 pacemaker channels stabilize the spontaneous firing by attenuating the parasympathetic response of the SAN.
Abstract The heart rate is dynamically controlled by the sympathetic and parasympathetic nervous systems that regulate the sinoatrial node (SAN). HCN4 pacemaker channels are the well‐known causative molecule of congenital sick sinus syndrome. Although HCN4 channels are activated by cAMP, the sympathetic response of the SAN was preserved in patients carrying loss‐of‐function mutations of the HCN4 gene. In order to clarify the contribution of HCN4 channels in the autonomic regulation of the SAN, we developed novel gain‐of‐function mutant mice in which the expression level of HCN4 channels could be reversibly changed from zero to ∼3 times that in wild‐type mice, using tetracycline transactivator and the tetracycline responsive element. We recorded telemetric ECGs in freely moving conscious mice and analysed the heart rate variability. We also evaluated the response of the SAN to cervical vagus nerve stimulation (CVNS). The conditional overexpression of HCN4 did not induce tachycardia, but reduced heart rate variability. The HCN4 overexpression also attenuated bradycardia induced by the CVNS only during the β‐adrenergic stimulation. In contrast, the knockdown of HCN4 gave rise to sinus arrhythmia, and enhanced the parasympathetic response; complete sinus pause was induced by the CVNS. In vitro, we compared the effects of acetylcholine on the spontaneous action potentials of single pacemaker cells, and found that similar phenotypic changes were induced by genetic manipulation of HCN4 expression both in the presence and absence of β‐adrenergic stimulation. Our study suggests that HCN4 channels attenuate the vagal response of the SAN, and thereby stabilize the spontaneous firing of the SAN. The contribution of HCN4 pacemaker channels in the autonomic regulation of the sino‐atrial node (SAN) has been a matter of debate. The transgenic overexpression of HCN4 did not induce tachycardia, but reduced heart rate variability, while the conditional knockdown of HCN4 gave rise to sinus arrhythmia. The response of the SAN to β‐adrenergic stimulation was not affected by overexpression or knockdown of HCN4 channels. When HCN4 channels were knocked down, the parasympathetic response examined by cervical vagus nerve stimulation (CVNS) was enhanced; the CVNS induced complete sinus pause. The overexpression of HCN4 attenuated bradycardia induced by CVNS only during β‐adrenergic stimulation. We concluded that HCN4 pacemaker channels stabilize the spontaneous firing by attenuating the parasympathetic response of the SAN.
Collapse
Affiliation(s)
- Yuko Kozasa
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.,Department of Anesthesiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Noriyuki Nakashima
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Masayuki Ito
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, 187-8502, Japan
| | - Taisuke Ishikawa
- Department of Molecular Physiology, Nagasaki University, Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Hiroki Kimoto
- Department of Molecular Physiology, Nagasaki University, Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Kazuo Ushijima
- Department of Anesthesiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Naomasa Makita
- Department of Molecular Physiology, Nagasaki University, Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Makoto Takano
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| |
Collapse
|
49
|
Expression and relevance of the G protein-gated K + channel in the mouse ventricle. Sci Rep 2018; 8:1192. [PMID: 29352184 PMCID: PMC5775354 DOI: 10.1038/s41598-018-19719-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/08/2018] [Indexed: 12/31/2022] Open
Abstract
The atrial G protein-gated inwardly rectifying K+ (GIRK) channel is a critical mediator of parasympathetic influence on cardiac physiology. Here, we probed the details and relevance of the GIRK channel in mouse ventricle. mRNAs for the atrial GIRK channel subunits (GIRK1, GIRK4), M2 muscarinic receptor (M2R), and RGS6, a negative regulator of atrial GIRK-dependent signaling, were detected in mouse ventricle at relatively low levels. The cholinergic agonist carbachol (CCh) activated small GIRK currents in adult wild-type ventricular myocytes that exhibited relatively slow kinetics and low CCh sensitivity; these currents were absent in ventricular myocytes from Girk1-/- or Girk4-/- mice. While loss of GIRK channels attenuated the CCh-induced shortening of action potential duration and suppression of ventricular myocyte excitability, selective ablation of GIRK channels in ventricle had no effect on heart rate, heart rate variability, or electrocardiogram parameters at baseline or after CCh injection. Additionally, loss of ventricular GIRK channels did not impact susceptibility to ventricular arrhythmias. These data suggest that the mouse ventricular GIRK channel is a GIRK1/GIRK4 heteromer, and show that while it contributes to the cholinergic suppression of ventricular myocyte excitability, this influence does not substantially impact cardiac physiology or ventricular arrhythmogenesis in the mouse.
Collapse
|
50
|
Nobles M, Montaigne D, Sebastian S, Birnbaumer L, Tinker A. Differential effects of inhibitory G protein isoforms on G protein-gated inwardly rectifying K + currents in adult murine atria. Am J Physiol Cell Physiol 2018; 314:C616-C626. [PMID: 29342363 DOI: 10.1152/ajpcell.00271.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
G protein-gated inwardly rectifying K+ (GIRK) channels are the major inwardly rectifying K+ currents in cardiac atrial myocytes and an important determinant of atrial electrophysiology. Inhibitory G protein α-subunits can both mediate activation via acetylcholine but can also suppress basal currents in the absence of agonist. We studied this phenomenon using whole cell patch clamping in murine atria from mice with global genetic deletion of Gαi2, combined deletion of Gαi1/Gαi3, and littermate controls. We found that mice with deletion of Gαi2 had increased basal and agonist-activated currents, particularly in the right atria while in contrast those with Gαi1/Gαi3 deletion had reduced currents. Mice with global genetic deletion of Gαi2 had decreased action potential duration. Tissue preparations of the left atria studied with a multielectrode array from Gαi2 knockout mice showed a shorter effective refractory period, with no change in conduction velocity, than littermate controls. Transcriptional studies revealed increased expression of GIRK channel subunit genes in Gαi2 knockout mice. Thus different G protein isoforms have differential effects on GIRK channel behavior and paradoxically Gαi2 act to increase basal and agonist-activated GIRK currents. Deletion of Gαi2 is potentially proarrhythmic in the atria.
Collapse
Affiliation(s)
- Muriel Nobles
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry , London , United Kingdom
| | - David Montaigne
- Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Université Lille 2 , Lille , France.,Institut National de la Santé et de la Recherche Médicale, U1011, Lille , France.,European Genomic Institute for Diabetes , Lille , France.,Institut Pasteur de Lille , Lille , France
| | - Sonia Sebastian
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry , London , United Kingdom
| | - Lutz Birnbaumer
- Division of Intramural Research, National Institute of Environmental Health Sciences , Research Triangle Park, North Carolina.,Institute of Biomedical Research, Catholic University of Argentina , Buenos Aires , Argentina
| | - Andrew Tinker
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry , London , United Kingdom
| |
Collapse
|