1
|
Vornanen M, Badr A, Haverinen J. Cardiac arrhythmias in fish induced by natural and anthropogenic changes in environmental conditions. J Exp Biol 2024; 227:jeb247446. [PMID: 39119881 DOI: 10.1242/jeb.247446] [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] [Indexed: 08/10/2024]
Abstract
A regular heartbeat is essential for maintaining the homeostasis of the vertebrate body. However, environmental pollutants, oxygen deficiency and extreme temperatures can impair heart function in fish. In this Review, we provide an integrative view of the molecular origins of cardiac arrhythmias and their functional consequences, from the level of ion channels to cardiac electrical activity in living fish. First, we describe the current knowledge of the cardiac excitation-contraction coupling of fish, as the electrical activity of the heart and intracellular Ca2+ regulation act as a platform for cardiac arrhythmias. Then, we compile findings on cardiac arrhythmias in fish. Although fish can experience several types of cardiac arrhythmia under stressful conditions, the most typical arrhythmia in fish - both under heat stress and in the presence of toxic substances - is atrioventricular block, which is the inability of the action potential to progress from the atrium to the ventricle. Early and delayed afterdepolarizations are less common in fish hearts than in the hearts of endotherms, perhaps owing to the excitation-contraction coupling properties of the fish heart. In fish hearts, Ca2+-induced Ca2+ release from the sarcoplasmic reticulum plays a smaller role than Ca2+ influx through the sarcolemma. Environmental changes and ion channel toxins can induce arrhythmias in fish and weaken their tolerance to environmental stresses. Although different from endotherm hearts in many respects, fish hearts can serve as a translational model for studying human cardiac arrhythmias, especially for human neonates.
Collapse
Affiliation(s)
- Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, PO Box 111, 80101 Joensuu, Finland
| | - Ahmed Badr
- Department of Environmental and Biological Sciences, University of Eastern Finland, PO Box 111, 80101 Joensuu, Finland
- Department of Zoology, Faculty of Science, Sohag University, 82524 Sohag, Egypt
| | - Jaakko Haverinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, PO Box 111, 80101 Joensuu, Finland
| |
Collapse
|
2
|
Quigley KS, Gianaros PJ, Norman GJ, Jennings JR, Berntson GG, de Geus EJC. Publication guidelines for human heart rate and heart rate variability studies in psychophysiology-Part 1: Physiological underpinnings and foundations of measurement. Psychophysiology 2024; 61:e14604. [PMID: 38873876 PMCID: PMC11539922 DOI: 10.1111/psyp.14604] [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: 05/11/2022] [Revised: 12/22/2023] [Accepted: 04/04/2024] [Indexed: 06/15/2024]
Abstract
This Committee Report provides methodological, interpretive, and reporting guidance for researchers who use measures of heart rate (HR) and heart rate variability (HRV) in psychophysiological research. We provide brief summaries of best practices in measuring HR and HRV via electrocardiographic and photoplethysmographic signals in laboratory, field (ambulatory), and brain-imaging contexts to address research questions incorporating measures of HR and HRV. The Report emphasizes evidence for the strengths and weaknesses of different recording and derivation methods for measures of HR and HRV. Along with this guidance, the Report reviews what is known about the origin of the heartbeat and its neural control, including factors that produce and influence HRV metrics. The Report concludes with checklists to guide authors in study design and analysis considerations, as well as guidance on the reporting of key methodological details and characteristics of the samples under study. It is expected that rigorous and transparent recording and reporting of HR and HRV measures will strengthen inferences across the many applications of these metrics in psychophysiology. The prior Committee Reports on HR and HRV are several decades old. Since their appearance, technologies for human cardiac and vascular monitoring in laboratory and daily life (i.e., ambulatory) contexts have greatly expanded. This Committee Report was prepared for the Society for Psychophysiological Research to provide updated methodological and interpretive guidance, as well as to summarize best practices for reporting HR and HRV studies in humans.
Collapse
Affiliation(s)
- Karen S. Quigley
- Department of Psychology, Northeastern University, Boston,
Massachusetts, USA
| | - Peter J. Gianaros
- Department of Psychology, University of Pittsburgh,
Pittsburgh, Pennsylvania, USA
| | - Greg J. Norman
- Department of Psychology, The University of Chicago,
Chicago, Illinois, USA
| | - J. Richard Jennings
- Department of Psychiatry & Psychology, University of
Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gary G. Berntson
- Department of Psychology & Psychiatry, The Ohio State
University, Columbus, Ohio, USA
| | - Eco J. C. de Geus
- Department of Biological Psychology, Vrije Universiteit
Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Wulkan F, Romagnuolo R, Qiang B, Valdman Sadikov T, Kim KP, Quesnel E, Jiang W, Andharia N, Weyers JJ, Ghugre NR, Ozcan B, Alibhai FJ, Laflamme MA. Stem cell-derived cardiomyocytes expressing a dominant negative pacemaker HCN4 channel do not reduce the risk of graft-related arrhythmias. Front Cardiovasc Med 2024; 11:1374881. [PMID: 39045008 PMCID: PMC11263024 DOI: 10.3389/fcvm.2024.1374881] [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: 01/22/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
Background Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) show tremendous promise for cardiac regeneration following myocardial infarction (MI), but their transplantation gives rise to transient ventricular tachycardia (VT) in large-animal MI models, representing a major hurdle to translation. Our group previously reported that these arrhythmias arise from a focal mechanism whereby graft tissue functions as an ectopic pacemaker; therefore, we hypothesized that hPSC-CMs engineered with a dominant negative form of the pacemaker ion channel HCN4 (dnHCN4) would exhibit reduced automaticity and arrhythmogenic risk following transplantation. Methods We used CRISPR/Cas9-mediated gene-editing to create transgenic dnHCN4 hPSC-CMs, and their electrophysiological behavior was evaluated in vitro by patch-clamp recordings and optical mapping. Next, we transplanted WT and homozygous dnHCN4 hPSC-CMs in a pig MI model and compared post-transplantation outcomes including the incidence of spontaneous arrhythmias and graft structure by immunohistochemistry. Results In vitro dnHCN4 hPSC-CMs exhibited significantly reduced automaticity and pacemaker funny current (I f ) density relative to wildtype (WT) cardiomyocytes. Following transplantation with either dnHCN4 or WT hPSC-CMs, all recipient hearts showed transmural infarct scar that was partially remuscularized by scattered islands of human myocardium. However, in contrast to our hypothesis, both dnHCN4 and WT hPSC-CM recipients exhibited frequent episodes of ventricular tachycardia (VT). Conclusions While genetic silencing of the pacemaker ion channel HCN4 suppresses the automaticity of hPSC-CMs in vitro, this intervention is insufficient to reduce VT risk post-transplantation in the pig MI model, implying more complex mechanism(s) are operational in vivo.
Collapse
Affiliation(s)
- Fanny Wulkan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Beiping Qiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | | | | | - Elya Quesnel
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Wenlei Jiang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Naaz Andharia
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Jill J. Weyers
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nilesh R. Ghugre
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Schulich Heart Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Bilgehan Ozcan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Faisal J. Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Michael A. Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Ripplinger CM, Glukhov AV, Kay MW, Boukens BJ, Chiamvimonvat N, Delisle BP, Fabritz L, Hund TJ, Knollmann BC, Li N, Murray KT, Poelzing S, Quinn TA, Remme CA, Rentschler SL, Rose RA, Posnack NG. Guidelines for assessment of cardiac electrophysiology and arrhythmias in small animals. Am J Physiol Heart Circ Physiol 2022; 323:H1137-H1166. [PMID: 36269644 PMCID: PMC9678409 DOI: 10.1152/ajpheart.00439.2022] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 01/09/2023]
Abstract
Cardiac arrhythmias are a major cause of morbidity and mortality worldwide. Although recent advances in cell-based models, including human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM), are contributing to our understanding of electrophysiology and arrhythmia mechanisms, preclinical animal studies of cardiovascular disease remain a mainstay. Over the past several decades, animal models of cardiovascular disease have advanced our understanding of pathological remodeling, arrhythmia mechanisms, and drug effects and have led to major improvements in pacing and defibrillation therapies. There exist a variety of methodological approaches for the assessment of cardiac electrophysiology and a plethora of parameters may be assessed with each approach. This guidelines article will provide an overview of the strengths and limitations of several common techniques used to assess electrophysiology and arrhythmia mechanisms at the whole animal, whole heart, and tissue level with a focus on small animal models. We also define key electrophysiological parameters that should be assessed, along with their physiological underpinnings, and the best methods with which to assess these parameters.
Collapse
Affiliation(s)
- Crystal M Ripplinger
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
| | - Alexey V Glukhov
- Department of Medicine, Cardiovascular Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Bastiaan J Boukens
- Department Physiology, University Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Medical Biology, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Nipavan Chiamvimonvat
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California
- Department of Internal Medicine, University of California Davis School of Medicine, Davis, California
- Veterans Affairs Northern California Healthcare System, Mather, California
| | - Brian P Delisle
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Larissa Fabritz
- University Center of Cardiovascular Science, University Heart and Vascular Center, University Hospital Hamburg-Eppendorf with DZHK Hamburg/Kiel/Luebeck, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Thomas J Hund
- Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
- Department of Biomedical Engineering, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Na Li
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Katherine T Murray
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Steven Poelzing
- Virginia Tech Carilon School of Medicine, Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech, Roanoke, Virginia
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Stacey L Rentschler
- Cardiovascular Division, Department of Medicine, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri
| | - Robert A Rose
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nikki G Posnack
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, District of Columbia
- Department of Pediatrics, George Washington University School of Medicine, Washington, District of Columbia
| |
Collapse
|
5
|
Kolesnikova EE, Golovina IV, Soldatov AA, Gavruseva TV. Synchronized Activity of Oxidoreductases in the Brain and Heart Compartments of the Scorpionfish Scorpaena porcus under Acute Hypoxia. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022060114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
6
|
Frequency-Dependent Properties of the Hyperpolarization-Activated Cation Current, I f, in Adult Mouse Heart Primary Pacemaker Myocytes. Int J Mol Sci 2022; 23:ijms23084299. [PMID: 35457119 PMCID: PMC9024942 DOI: 10.3390/ijms23084299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
A number of distinct electrophysiological mechanisms that modulate the myogenic spontaneous pacemaker activity in the sinoatrial node (SAN) of the mammalian heart have been investigated extensively. There is agreement that several (3 or 4) different transmembrane ionic current changes (referred to as the voltage clock) are involved; and that the resulting net current interacts with direct and indirect effects of changes in intracellular Ca2+ (the calcium clock). However, significant uncertainties, and important knowledge gaps, remain concerning the functional roles in SAN spontaneous pacing of many of the individual ion channel- or exchanger-mediated transmembrane current changes. We report results from patch clamp studies and mathematical modeling of the hyperpolarization-activated current, If, in the generation/modulation of the diastolic depolarization, or pacemaker potential, produced by individual myocytes that were enzymatically isolated from the adult mouse sinoatrial node (SAN). Amphotericin-mediated patch microelectrode recordings at 35 °C were made under control conditions and in the presence of 5 or 10 nM isoproterenol (ISO). These sets of results were complemented and integrated with mathematical modeling of the current changes that take place in the range of membrane potentials (−70 to −50 mV), which corresponds to the ‘pacemaker depolarization’ in the adult mouse SAN. Our results reveal a very small, but functionally important, approximately steady-state or time-independent current generated by residual activation of If channels that are expressed in these pacemaker myocytes. Recordings of the pacemaker depolarization and action potential, combined with measurements of changes in If, and the well-known increases in the L-type Ca2+ current, ICaL, demonstrated that ICaL activation, is essential for myogenic pacing. Moreover, after being enhanced (approximately 3-fold) by 5 or 10 nM ISO, ICaL contributes significantly to the positive chronotropic effect. Our mathematical model has been developed in an attempt to better understand the underlying mechanisms for the pacemaker depolarization and action potential in adult mouse SAN myocytes. After being updated with our new experimental data describing If, our simulations reveal a novel functional component of If in adult mouse SAN. Computational work carried out with this model also confirms that in the presence of ISO the residual activation of If and opening of ICaL channels combine to generate a net current change during the slow diastolic depolarization phase that is essential for the observed accelerated pacemaking rate of these SAN myocytes.
Collapse
|
7
|
Takeuchi A, Matsuoka S. Physiological and Pathophysiological Roles of Mitochondrial Na +-Ca 2+ Exchanger, NCLX, in Hearts. Biomolecules 2021; 11:biom11121876. [PMID: 34944520 PMCID: PMC8699148 DOI: 10.3390/biom11121876] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022] Open
Abstract
It has been over 10 years since SLC24A6/SLC8B1, coding the Na+/Ca2+/Li+ exchanger (NCLX), was identified as the gene responsible for mitochondrial Na+-Ca2+ exchange, a major Ca2+ efflux system in cardiac mitochondria. This molecular identification enabled us to determine structure–function relationships, as well as physiological/pathophysiological contributions, and our understandings have dramatically increased. In this review, we provide an overview of the recent achievements in relation to NCLX, focusing especially on its heart-specific characteristics, biophysical properties, and spatial distribution in cardiomyocytes, as well as in cardiac mitochondria. In addition, we discuss the roles of NCLX in cardiac functions under physiological and pathophysiological conditions—the generation of rhythmicity, the energy metabolism, the production of reactive oxygen species, and the opening of mitochondrial permeability transition pores.
Collapse
Affiliation(s)
- Ayako Takeuchi
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
- Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan
- Correspondence: ; Tel.: +81-776-61-8311
| | - Satoshi Matsuoka
- Department of Integrative and Systems Physiology, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
- Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan
| |
Collapse
|
8
|
Petkova MA, Dobrzynski H. Do human sinoatrial node cells have t-tubules? TRANSLATIONAL RESEARCH IN ANATOMY 2021. [DOI: 10.1016/j.tria.2021.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
9
|
Monfredi O, Keim-Malpass J, Moorman JR. Continuous cardiorespiratory monitoring is a dominant source of predictive signal in machine learning for risk stratification and clinical decision support . Physiol Meas 2021; 42. [PMID: 34580243 DOI: 10.1088/1361-6579/ac2130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022]
Abstract
Beaulieu-Jones and coworkers propose a litmus test for the field of predictive analytics-performance improvements must be demonstrated to be the result of non-clinician-initiated data, otherwise, there should be caution in assuming that predictive models could improve clinical decision-making (Beaulieu-Joneset al2021). They demonstrate substantial prognostic information in unsorted physician orders made before the first midnight of hospital admission, and we are persuaded that it is fair to ask-if the physician thought of it first, what exactly is machine learning for in-patient risk stratification learning about? While we want predictive analytics to represent the leading indicators of a patient's illness, does it instead merely reflect the lagging indicators of clinicians' actions? We propose that continuous cardiorespiratory monitoring-'routine telemetry data,' in Beaulieu-Jones' terms-represents the most valuable non-clinician-initiated predictive signal present in patient data, and the value added to patient care justifies the efforts and expense required. Here, we present a clinical and a physiological point of view to support our contention.
Collapse
Affiliation(s)
- Oliver Monfredi
- Center for Advanced Medical Analytics, University of Virginia, United States of America.,Cardiovascular Division, Department of Internal Medicine, School of Medicine, University of Virginia, United States of America
| | - Jessica Keim-Malpass
- Center for Advanced Medical Analytics, University of Virginia, United States of America.,School of Nursing, University of Virginia, United States of America
| | - J Randall Moorman
- Center for Advanced Medical Analytics, University of Virginia, United States of America.,Cardiovascular Division, Department of Internal Medicine, School of Medicine, University of Virginia, United States of America
| |
Collapse
|
10
|
Qi Z, Wang T, Chen X, Wong CK, Ding Q, Sauer H, Chen ZF, Long C, Yao X, Cai Z, Tsang SY. Extracellular and Intracellular Angiotensin II Regulate the Automaticity of Developing Cardiomyocytes via Different Signaling Pathways. Front Mol Biosci 2021; 8:699827. [PMID: 34513920 PMCID: PMC8425478 DOI: 10.3389/fmolb.2021.699827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/27/2021] [Indexed: 12/02/2022] Open
Abstract
Angiotensin II (Ang II) plays an important role in regulating various physiological processes. However, little is known about the existence of intracellular Ang II (iAng II), whether iAng II would regulate the automaticity of early differentiating cardiomyocytes, and the underlying mechanism involved. Here, iAng II was detected by immunocytochemistry and ultra-high performance liquid chromatography combined with electrospray ionization triple quadrupole tandem mass spectrometry in mouse embryonic stem cell–derived cardiomyocytes (mESC-CMs) and neonatal rat ventricular myocytes. Expression of AT1R-YFP in mESC-CMs revealed that Ang II type 1 receptors were located on the surface membrane, while immunostaining of Ang II type 2 receptors (AT2R) revealed that AT2R were predominately located on the nucleus and the sarcoplasmic reticulum. While extracellular Ang II increased spontaneous action potentials (APs), dual patch clamping revealed that intracellular delivery of Ang II or AT2R activator C21 decreased spontaneous APs. Interestingly, iAng II was found to decrease the caffeine-induced increase in spontaneous APs and caffeine-induced calcium release, suggesting that iAng II decreased spontaneous APs via the AT2R- and ryanodine receptor–mediated pathways. This is the first study that provides evidence of the presence and function of iAng II in regulating the automaticity behavior of ESC-CMs and may therefore shed light on the role of iAng II in fate determination.
Collapse
Affiliation(s)
- Zenghua Qi
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,Institute of Environmental Health and Pollution Control, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Tao Wang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, SAR China
| | - Xiangmao Chen
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Chun Kit Wong
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Qianqian Ding
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Zhi-Feng Chen
- Institute of Environmental Health and Pollution Control, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, SAR China
| | - Suk Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| |
Collapse
|
11
|
Buchhorn R, Baumann C, Willaschek C. Pathophysiological mechanisms of bradycardia in patients with anorexia nervosa. Health Sci Rep 2021; 4:e331. [PMID: 34322602 PMCID: PMC8299991 DOI: 10.1002/hsr2.331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 04/29/2021] [Accepted: 07/04/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The purpose of this investigation was to examine heart rate variability (HRV), interbeat interval (IBI), and their interrelationship in healthy controls, bradycardic hyperpolarization-activated cyclic nucleotide-gated channel 4 (HCN4) mutation carriers, and patients with anorexia nervosa (AN). We tested the hypothesis that neural mechanisms cause bradycardia in patients with AN. Therefore, we assumed that saturation of the HRV/IBI relationship as a consequence of sustained parasympathetic control of the sinus node is exclusively detectable in patients with AN. METHODS Patients with AN between the ages of 12 and 16 years admitted to our hospital due to malnutrition were grouped and included in the present investigation (N = 20). A matched-pair group with healthy children and adolescents was created. Groups were matched for age and sex. A 24-hour Holter electrocardiography (ECG) was performed in controls and patients. More specifically, all patients underwent two 24-hour Holter ECG examinations (admission; refeeding treatment). Additionally, the IBI was recorded during the night in HCN4 mutation carriers (N = 4). HRV parameters were analyzed in 5-minute sequences during the night and plotted against mean corresponding IBI length. HRV, IBI, and their interrelationship were examined using Spearman's rank correlation analyses, Mann-Whitney U tests, and Wilcoxon signed-rank tests. RESULTS The relationship between IBI and HRV showed signs of saturation in patients with AN. Furthermore, signs of HRV saturation were present in two HCN4 mutation carriers. In contrast, signs of HRV saturation were not present in controls. CONCLUSIONS The existence of HRV saturation does not support the existence of parasympathetically mediated bradycardia. Nonneural mechanisms, such as HCN4 downregulation, may be responsible for bradycardia and HRV saturation in patients with AN.
Collapse
Affiliation(s)
- Reiner Buchhorn
- Department of PediatricsCaritas‐Krankenhaus Bad MergentheimBad MergentheimGermany
| | | | - Christian Willaschek
- Department of PediatricsCaritas‐Krankenhaus Bad MergentheimBad MergentheimGermany
| |
Collapse
|
12
|
Vinogradova TM, Lakatta EG. Dual Activation of Phosphodiesterase 3 and 4 Regulates Basal Cardiac Pacemaker Function and Beyond. Int J Mol Sci 2021. [PMID: 34445119 DOI: 10.3390/ijms22168414.pmid:34445119;pmcid:pmc8395138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
The sinoatrial (SA) node is the physiological pacemaker of the heart, and resting heart rate in humans is a well-known risk factor for cardiovascular disease and mortality. Consequently, the mechanisms of initiating and regulating the normal spontaneous SA node beating rate are of vital importance. Spontaneous firing of the SA node is generated within sinoatrial nodal cells (SANC), which is regulated by the coupled-clock pacemaker system. Normal spontaneous beating of SANC is driven by a high level of cAMP-mediated PKA-dependent protein phosphorylation, which rely on the balance between high basal cAMP production by adenylyl cyclases and high basal cAMP degradation by cyclic nucleotide phosphodiesterases (PDEs). This diverse class of enzymes includes 11 families and PDE3 and PDE4 families dominate in both the SA node and cardiac myocardium, degrading cAMP and, consequently, regulating basal cardiac pacemaker function and excitation-contraction coupling. In this review, we will demonstrate similarities between expression, distribution, and colocalization of various PDE subtypes in SANC and cardiac myocytes of different species, including humans, focusing on PDE3 and PDE4. Here, we will describe specific targets of the coupled-clock pacemaker system modulated by dual PDE3 + PDE4 activation and provide evidence that concurrent activation of PDE3 + PDE4, operating in a synergistic manner, regulates the basal cardiac pacemaker function and provides control over normal spontaneous beating of SANCs through (PDE3 + PDE4)-dependent modulation of local subsarcolemmal Ca2+ releases (LCRs).
Collapse
Affiliation(s)
- Tatiana M Vinogradova
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institute of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institute of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| |
Collapse
|
13
|
Dual Activation of Phosphodiesterase 3 and 4 Regulates Basal Cardiac Pacemaker Function and Beyond. Int J Mol Sci 2021; 22:ijms22168414. [PMID: 34445119 PMCID: PMC8395138 DOI: 10.3390/ijms22168414] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 11/17/2022] Open
Abstract
The sinoatrial (SA) node is the physiological pacemaker of the heart, and resting heart rate in humans is a well-known risk factor for cardiovascular disease and mortality. Consequently, the mechanisms of initiating and regulating the normal spontaneous SA node beating rate are of vital importance. Spontaneous firing of the SA node is generated within sinoatrial nodal cells (SANC), which is regulated by the coupled-clock pacemaker system. Normal spontaneous beating of SANC is driven by a high level of cAMP-mediated PKA-dependent protein phosphorylation, which rely on the balance between high basal cAMP production by adenylyl cyclases and high basal cAMP degradation by cyclic nucleotide phosphodiesterases (PDEs). This diverse class of enzymes includes 11 families and PDE3 and PDE4 families dominate in both the SA node and cardiac myocardium, degrading cAMP and, consequently, regulating basal cardiac pacemaker function and excitation-contraction coupling. In this review, we will demonstrate similarities between expression, distribution, and colocalization of various PDE subtypes in SANC and cardiac myocytes of different species, including humans, focusing on PDE3 and PDE4. Here, we will describe specific targets of the coupled-clock pacemaker system modulated by dual PDE3 + PDE4 activation and provide evidence that concurrent activation of PDE3 + PDE4, operating in a synergistic manner, regulates the basal cardiac pacemaker function and provides control over normal spontaneous beating of SANCs through (PDE3 + PDE4)-dependent modulation of local subsarcolemmal Ca2+ releases (LCRs).
Collapse
|
14
|
Regulation of sinus node pacemaking and atrioventricular node conduction by HCN channels in health and disease. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:61-85. [PMID: 34197836 DOI: 10.1016/j.pbiomolbio.2021.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/02/2021] [Accepted: 06/14/2021] [Indexed: 12/19/2022]
Abstract
The funny current, If, was first recorded in the heart 40 or more years ago by Dario DiFrancesco and others. Since then, we have learnt that If plays an important role in pacemaking in the sinus node, the innate pacemaker of the heart, and more recently evidence has accumulated to show that If may play an important role in action potential conduction through the atrioventricular (AV) node. Evidence has also accumulated to show that regulation of the transcription and translation of the underlying Hcn genes plays an important role in the regulation of sinus node pacemaking and AV node conduction under normal physiological conditions - in athletes, during the circadian rhythm, in pregnancy, and during postnatal development - as well as pathological states - ageing, heart failure, pulmonary hypertension, diabetes and atrial fibrillation. There may be yet more pathological conditions involving changes in the expression of the Hcn genes. Here, we review the role of If and the underlying HCN channels in physiological and pathological changes of the sinus and AV nodes and we begin to explore the signalling pathways (microRNAs, transcription factors, GIRK4, the autonomic nervous system and inflammation) involved in this regulation. This review is dedicated to Dario DiFrancesco on his retirement.
Collapse
|
15
|
Hu W, Clark RB, Giles WR, Shibata E, Zhang H. Physiological Roles of the Rapidly Activated Delayed Rectifier K + Current in Adult Mouse Heart Primary Pacemaker Activity. Int J Mol Sci 2021; 22:4761. [PMID: 33946248 PMCID: PMC8124469 DOI: 10.3390/ijms22094761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/01/2023] Open
Abstract
Robust, spontaneous pacemaker activity originating in the sinoatrial node (SAN) of the heart is essential for cardiovascular function. Anatomical, electrophysiological, and molecular methods as well as mathematical modeling approaches have quite thoroughly characterized the transmembrane fluxes of Na+, K+ and Ca2+ that produce SAN action potentials (AP) and 'pacemaker depolarizations' in a number of different in vitro adult mammalian heart preparations. Possible ionic mechanisms that are responsible for SAN primary pacemaker activity are described in terms of: (i) a Ca2+-regulated mechanism based on a requirement for phasic release of Ca2+ from intracellular stores and activation of an inward current-mediated by Na+/Ca2+ exchange; (ii) time- and voltage-dependent activation of Na+ or Ca2+ currents, as well as a cyclic nucleotide-activated current, If; and/or (iii) a combination of (i) and (ii). Electrophysiological studies of single spontaneously active SAN myocytes in both adult mouse and rabbit hearts consistently reveal significant expression of a rapidly activating time- and voltage-dependent K+ current, often denoted IKr, that is selectively expressed in the leading or primary pacemaker region of the adult mouse SAN. The main goal of the present study was to examine by combined experimental and simulation approaches the functional or physiological roles of this K+ current in the pacemaker activity. Our patch clamp data of mouse SAN myocytes on the effects of a pharmacological blocker, E4031, revealed that a rapidly activating K+ current is essential for action potential (AP) repolarization, and its deactivation during the pacemaker potential contributes a small but significant component to the pacemaker depolarization. Mathematical simulations using a murine SAN AP model confirm that well known biophysical properties of a delayed rectifier K+ current can contribute to its role in generating spontaneous myogenic activity.
Collapse
Affiliation(s)
- Wei Hu
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
| | - Robert B. Clark
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.)
| | - Wayne R. Giles
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (R.B.C.); (W.R.G.)
| | - Erwin Shibata
- Department of Physiology, Carver School of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Henggui Zhang
- Biological Physics Group, Department of Physics and Astronomy, The University of Manchester, Manchester M13 9PL, UK;
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
16
|
Pineda S, Nikolova-Krstevski V, Leimena C, Atkinson AJ, Altekoester AK, Cox CD, Jacoby A, Huttner IG, Ju YK, Soka M, Ohanian M, Trivedi G, Kalvakuri S, Birker K, Johnson R, Molenaar P, Kuchar D, Allen DG, van Helden DF, Harvey RP, Hill AP, Bodmer R, Vogler G, Dobrzynski H, Ocorr K, Fatkin D. Conserved Role of the Large Conductance Calcium-Activated Potassium Channel, K Ca1.1, in Sinus Node Function and Arrhythmia Risk. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003144. [PMID: 33629867 DOI: 10.1161/circgen.120.003144] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND KCNMA1 encodes the α-subunit of the large-conductance Ca2+-activated K+ channel, KCa1.1, and lies within a linkage interval for atrial fibrillation (AF). Insights into the cardiac functions of KCa1.1 are limited, and KCNMA1 has not been investigated as an AF candidate gene. METHODS The KCNMA1 gene was sequenced in 118 patients with familial AF. The role of KCa1.1 in normal cardiac structure and function was evaluated in humans, mice, zebrafish, and fly. A novel KCNMA1 variant was functionally characterized. RESULTS A complex KCNMA1 variant was identified in 1 kindred with AF. To evaluate potential disease mechanisms, we first evaluated the distribution of KCa1.1 in normal hearts using immunostaining and immunogold electron microscopy. KCa1.1 was seen throughout the atria and ventricles in humans and mice, with strong expression in the sinus node. In an ex vivo murine sinoatrial node preparation, addition of the KCa1.1 antagonist, paxilline, blunted the increase in beating rate induced by adrenergic receptor stimulation. Knockdown of the KCa1.1 ortholog, kcnma1b, in zebrafish embryos resulted in sinus bradycardia with dilatation and reduced contraction of the atrium and ventricle. Genetic inactivation of the Drosophila KCa1.1 ortholog, slo, systemically or in adult stages, also slowed the heartbeat and produced fibrillatory cardiac contractions. Electrophysiological characterization of slo-deficient flies revealed bursts of action potentials, reflecting increased events of fibrillatory arrhythmias. Flies with cardiac-specific overexpression of the human KCNMA1 mutant also showed increased heart period and bursts of action potentials, similar to the KCa1.1 loss-of-function models. CONCLUSIONS Our data point to a highly conserved role of KCa1.1 in sinus node function in humans, mice, zebrafish, and fly and suggest that KCa1.1 loss of function may predispose to AF.
Collapse
Affiliation(s)
- Santiago Pineda
- Development, Aging & Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA (S.P., S.K., K.B., R.B., G.V., K.O.)
| | - Vesna Nikolova-Krstevski
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.).,Faculty of Medicine, UNSW Sydney, Kensington (V.N.-K., I.G.H., R.J., R.P.H., A.P.H., D.F.)
| | - Christiana Leimena
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.)
| | - Andrew J Atkinson
- Institute of Cardiovascular Sciences, University of Manchester, United Kingdom (A.J.A., H.D.)
| | - Ann-Kristin Altekoester
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.)
| | - Charles D Cox
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.)
| | - Arie Jacoby
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.)
| | - Inken G Huttner
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.).,Faculty of Medicine, UNSW Sydney, Kensington (V.N.-K., I.G.H., R.J., R.P.H., A.P.H., D.F.)
| | - Yue-Kun Ju
- Bosch Institute, University of Sydney, Camperdown (Y.-K.J., D.G.A.)
| | - Magdalena Soka
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.)
| | - Monique Ohanian
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.)
| | - Gunjan Trivedi
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.)
| | - Sreehari Kalvakuri
- Development, Aging & Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA (S.P., S.K., K.B., R.B., G.V., K.O.)
| | - Katja Birker
- Development, Aging & Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA (S.P., S.K., K.B., R.B., G.V., K.O.)
| | - Renee Johnson
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.).,Faculty of Medicine, UNSW Sydney, Kensington (V.N.-K., I.G.H., R.J., R.P.H., A.P.H., D.F.)
| | - Peter Molenaar
- Faculty of Health, Queensland University of Technology (P.M.).,School of Medicine, University of Queensland, Prince Charles Hospital, Brisbane, Queensland, Australia (P.M.)
| | - Dennis Kuchar
- Cardiology Department, St Vincent's Hospital, Darlinghurst (D.K., D.F.)
| | - David G Allen
- Bosch Institute, University of Sydney, Camperdown (Y.-K.J., D.G.A.)
| | - Dirk F van Helden
- University of Newcastle and Hunter Medical Research Institute, NSW, Australia (D.F.v.H.)
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.).,Faculty of Medicine, UNSW Sydney, Kensington (V.N.-K., I.G.H., R.J., R.P.H., A.P.H., D.F.)
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.).,Faculty of Medicine, UNSW Sydney, Kensington (V.N.-K., I.G.H., R.J., R.P.H., A.P.H., D.F.)
| | - Rolf Bodmer
- Development, Aging & Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA (S.P., S.K., K.B., R.B., G.V., K.O.)
| | - Georg Vogler
- Development, Aging & Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA (S.P., S.K., K.B., R.B., G.V., K.O.)
| | - Halina Dobrzynski
- Institute of Cardiovascular Sciences, University of Manchester, United Kingdom (A.J.A., H.D.).,Jagiellonian University Medical College, Cracow, Poland (H.D.)
| | - Karen Ocorr
- Development, Aging & Regeneration Program, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA (S.P., S.K., K.B., R.B., G.V., K.O.)
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Darlinghurst (V.N.-K., C.L., A.-K.A., C.D.C., A.J., I.G.H., M.S., M.O., G.T., R.J., R.P.H., A.P.H., D.F.).,Faculty of Medicine, UNSW Sydney, Kensington (V.N.-K., I.G.H., R.J., R.P.H., A.P.H., D.F.).,Cardiology Department, St Vincent's Hospital, Darlinghurst (D.K., D.F.)
| |
Collapse
|
17
|
Konstantinidis K, Bezzerides VJ, Lai L, Isbell HM, Wei AC, Wu Y, Viswanathan MC, Blum ID, Granger JM, Heims-Waldron D, Zhang D, Luczak ED, Murphy KR, Lu F, Gratz DH, Manta B, Wang Q, Wang Q, Kolodkin AL, Gladyshev VN, Hund TJ, Pu WT, Wu MN, Cammarato A, Bianchet MA, Shea MA, Levine RL, Anderson ME. MICAL1 constrains cardiac stress responses and protects against disease by oxidizing CaMKII. J Clin Invest 2021; 130:4663-4678. [PMID: 32749237 PMCID: PMC7456244 DOI: 10.1172/jci133181] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 05/29/2020] [Indexed: 01/22/2023] Open
Abstract
Oxidant stress can contribute to health and disease. Here we show that invertebrates and vertebrates share a common stereospecific redox pathway that protects against pathological responses to stress, at the cost of reduced physiological performance, by constraining Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity. MICAL1, a methionine monooxygenase thought to exclusively target actin, and MSRB, a methionine reductase, control the stereospecific redox status of M308, a highly conserved residue in the calmodulin-binding (CaM-binding) domain of CaMKII. Oxidized or mutant M308 (M308V) decreased CaM binding and CaMKII activity, while absence of MICAL1 in mice caused cardiac arrhythmias and premature death due to CaMKII hyperactivation. Mimicking the effects of M308 oxidation decreased fight-or-flight responses in mice, strikingly impaired heart function in Drosophila melanogaster, and caused disease protection in human induced pluripotent stem cell-derived cardiomyocytes with catecholaminergic polymorphic ventricular tachycardia, a CaMKII-sensitive genetic arrhythmia syndrome. Our studies identify a stereospecific redox pathway that regulates cardiac physiological and pathological responses to stress across species.
Collapse
Affiliation(s)
- Klitos Konstantinidis
- Division of Cardiology.,Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Lo Lai
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Holly M Isbell
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - An-Chi Wei
- Department of Electrical Engineering, Graduate Institute of Biomedical and Bioinformatics, National Taiwan University, Taipei City, Taiwan
| | - Yuejin Wu
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Meera C Viswanathan
- Division of Cardiology.,Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ian D Blum
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan M Granger
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Donghui Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA.,State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Elizabeth D Luczak
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kevin R Murphy
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fujian Lu
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Daniel H Gratz
- Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Bruno Manta
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qiang Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qinchuan Wang
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alex L Kolodkin
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Maryland, USA
| | - Vadim N Gladyshev
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas J Hund
- Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Mark N Wu
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Maryland, USA.,Department of Genetic Medicine
| | - Anthony Cammarato
- Division of Cardiology.,Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Physiology, and
| | - Mario A Bianchet
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, Maryland, USA
| | - Madeline A Shea
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Rodney L Levine
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Mark E Anderson
- Division of Cardiology.,Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Physiology, and
| |
Collapse
|
18
|
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
|
19
|
Clancy CE, Santana LF. Evolving Discovery of the Origin of the Heartbeat: A New Perspective on Sinus Rhythm. JACC Clin Electrophysiol 2020; 6:932-934. [PMID: 32819527 DOI: 10.1016/j.jacep.2020.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/01/2022]
Affiliation(s)
- Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California-Davis, Davis, California, USA.
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California-Davis, Davis, California, USA.
| |
Collapse
|
20
|
Macartney MJ, McLennan PL, Peoples GE. Heart rate variability during cardiovascular reflex testing: the importance of underlying heart rate. J Basic Clin Physiol Pharmacol 2020; 32:145-153. [PMID: 33141106 DOI: 10.1515/jbcpp-2020-0245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/04/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES Heart rate variability (HRV) is often measured during clinical and experimental cardiovascular reflex tests (CRT), as a reflection of cardiac autonomic modulation, despite limited characterization of the rapid responses that occur. Therefore, we evaluated the responsiveness of HRV indices in 20 healthy young adults (age, 27 ± 6 y; mass, 76.9 ± 16.8 kg; height, 1.79 ± 0.12 m) during four separate established CRT. METHODS These included the [I] orthostatic challenge, [II] isometric handgrip, [III] cold pressor and [IV] cold diving reflex tests. Electrocardiogram was recorded throughout, with HRV derived from RR intervals at rest and from each CRT. On a separate day, a subgroup of participants (n=9) completed the same protocol for a second time. RESULTS The maximal slope of heart rate change (dTdt) was significantly different between all CRT, with the orthostatic challenge producing the fastest increase (2.56 ± 0.48) and the cold pressor the fastest reduction (-1.93 ± 0.68) in heart rate. Overall HRV, reflected by Poincaré plot ratio (SD1:SD2), was significantly reduced during all CRT ([I], -0.41 ± 0.12; [II], -0.19 ± 0.05; [III], -0.36 ± 0.12; [IV], -0.44 ± 0.11; p<0.05) relative to baseline and this was reproducible in time-series. However, when HRV indices were correlated to mean-RR an exponential growth-like relationship was evident (R2 ranging from: 0.52-0.62). CONCLUSIONS These unique outcomes demonstrate that short-term alterations in HRV are evident during CRT, while indicating the importance of adjusting for, or at least reporting, underlying heart rate when interpreting such measures.
Collapse
Affiliation(s)
- Michael J Macartney
- Graduate Medicine, School of Medicine, University of Wollongong, Wollongong, Australia
- Centre for Medical and Exercise Physiology, Faculty of Science Medicine and Health, University of Wollongong, Wollongong, Australia
| | - Peter L McLennan
- Graduate Medicine, School of Medicine, University of Wollongong, Wollongong, Australia
- Centre for Medical and Exercise Physiology, Faculty of Science Medicine and Health, University of Wollongong, Wollongong, Australia
| | - Gregory E Peoples
- Graduate Medicine, School of Medicine, University of Wollongong, Wollongong, Australia
- Centre for Medical and Exercise Physiology, Faculty of Science Medicine and Health, University of Wollongong, Wollongong, Australia
| |
Collapse
|
21
|
Baudot M, Torre E, Bidaud I, Louradour J, Torrente AG, Fossier L, Talssi L, Nargeot J, Barrère-Lemaire S, Mesirca P, Mangoni ME. Concomitant genetic ablation of L-type Ca v1.3 (α 1D) and T-type Ca v3.1 (α 1G) Ca 2+ channels disrupts heart automaticity. Sci Rep 2020; 10:18906. [PMID: 33144668 PMCID: PMC7642305 DOI: 10.1038/s41598-020-76049-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/15/2020] [Indexed: 12/02/2022] Open
Abstract
Cardiac automaticity is set by pacemaker activity of the sinus node (SAN). In addition to the ubiquitously expressed cardiac voltage-gated L-type Cav1.2 Ca2+ channel isoform, pacemaker cells within the SAN and the atrioventricular node co-express voltage-gated L-type Cav1.3 and T-type Cav3.1 Ca2+ channels (SAN-VGCCs). The role of SAN-VGCCs in automaticity is incompletely understood. We used knockout mice carrying individual genetic ablation of Cav1.3 (Cav1.3−/−) or Cav3.1 (Cav3.1−/−) channels and double mutant Cav1.3−/−/Cav3.1−/− mice expressing only Cav1.2 channels. We show that concomitant loss of SAN-VGCCs prevents physiological SAN automaticity, blocks impulse conduction and compromises ventricular rhythmicity. Coexpression of SAN-VGCCs is necessary for impulse formation in the central SAN. In mice lacking SAN-VGCCs, residual pacemaker activity is predominantly generated in peripheral nodal and extranodal sites by f-channels and TTX-sensitive Na+ channels. In beating SAN cells, ablation of SAN-VGCCs disrupted late diastolic local intracellular Ca2+ release, which demonstrates an important role for these channels in supporting the sarcoplasmic reticulum based “Ca2+clock” mechanism during normal pacemaking. These data implicate an underappreciated role for co-expression of SAN-VGCCs in heart automaticity and define an integral role for these channels in mechanisms that control the heartbeat.
Collapse
Affiliation(s)
- Matthias Baudot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France
| | - Eleonora Torre
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France.,Department of Biotechnology and Biosciences, Università Degli Studi di Milano-Bicocca, Milan, Italy
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France
| | - Julien Louradour
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France
| | - Angelo G Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France
| | - Lucile Fossier
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France
| | - Leïla Talssi
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France
| | - Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx ICST, Montpellier, France
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France. .,LabEx ICST, Montpellier, France.
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, Inserm, 141, rue de la cardonille, 34094, Montpellier, France. .,LabEx ICST, Montpellier, France.
| |
Collapse
|
22
|
Vornanen M. Effects of acute warming on cardiac and myotomal sarco(endo)plasmic reticulum ATPase (SERCA) of thermally acclimated brown trout (Salmo trutta). J Comp Physiol B 2020; 191:43-53. [PMID: 32980918 PMCID: PMC7819936 DOI: 10.1007/s00360-020-01313-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/21/2020] [Accepted: 09/09/2020] [Indexed: 11/24/2022]
Abstract
At high temperatures, ventricular beating rate collapses and depresses cardiac output in fish. The role of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) in thermal tolerance of ventricular function was examined in brown trout (Salmo trutta) by measuring heart SERCA and comparing it to that of the dorsolateral myotomal muscle. Activity of SERCA was measured from crude homogenates of cold-acclimated (+ 3 °C, c.a.) and warm-acclimated (+ 13 °C, w.a.) brown trout as cyclopiazonic acid (20 µM) sensitive Ca2+-ATPase between + 3 and + 33 °C. Activity of the heart SERCA was significantly higher in c.a. than w.a. trout and increased strongly between + 3 and + 23 °C with linear Arrhenius plots but started to plateau between + 23 and + 33 °C in both acclimation groups. The rate of thermal inactivation of the heart SERCA at + 35 °C was similar in c.a. and w.a. fish. Activity of the muscle SERCA was less temperature dependent and more heat resistant than that of the heart SERCA and showed linear Arrhenius plots between + 3 and + 33 °C in both c.a. and w.a. fish. SERCA activity of the c.a. muscle was slightly higher than that of w.a. muscle. The rate of thermal inactivation at + 40 °C was similar for both c.a. and w.a. muscle SERCA at + 40 °C. Although the heart SERCA is more sensitive to high temperatures than the muscle SERCA, it is unlikely to be a limiting factor for heart rate, because its heat tolerance, unlike that of the ventricular beating rate, was not changed by temperature acclimation.
Collapse
Affiliation(s)
- Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, P.O. Box 111, 80101, Joensuu, Finland.
| |
Collapse
|
23
|
Lensen IS, Monfredi OJ, Andris RT, Lake DE, Moorman JR. Heart rate fragmentation gives novel insights into non-autonomic mechanisms governing beat-to-beat control of the heart's rhythm. JRSM Cardiovasc Dis 2020; 9:2048004020948732. [PMID: 32922768 PMCID: PMC7457638 DOI: 10.1177/2048004020948732] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022] Open
Abstract
To demonstrate how heart rate fragmentation gives novel insights into
non-autonomic mechanisms of beat-to-beat variability in cycle length, and
predicts survival of cardiology clinic patients, over and above traditional
clinical risk factors and measures of heart rate variability. Approach: We studied 2893 patients seen by cardiologists with
clinical data including 24-hour Holter monitoring. Novel measures of heart
rate fragmentation alongside canonical time and frequency domain measures of
heart rate variability, as well as an existing local dynamics score were
calculated. A proportional hazards model was utilized to relate the results
to survival. Main results: The novel heart rate fragmentation measures were
validated and characterized with respect to the effects of age, ectopy and
atrial fibrillation. Correlations between parameters were determined.
Critically, heart rate fragmentation results could not be accounted for by
undersampling respiratory sinus arrhythmia. Increased heart rate
fragmentation was associated with poorer survival (p ≪ 0.01 in the
univariate model). In multivariable analyses, increased heart rate
fragmentation and more abnormal local dynamics (p 0.045), along with
increased clinical risk factors (age (p ≪ 0.01), tobacco use (p ≪ 0.01) and
history of heart failure (p 0.019)) and lower low- to high-frequency ratio
(p 0.022) were all independent predictors of 2-year mortality. Significance: Analysis of continuous ECG data with heart rate
fragmentation indices yields information regarding non-autonomic control of
beat-to-beat variability in cycle length that is independent of and additive
to established parameters for investigating heart rate variability, and
predicts mortality in concert with measures of local dynamics, frequency
content of heart rate, and clinical risk factors.
Collapse
Affiliation(s)
- Irene S Lensen
- University of Technology Eindhoven, Noord-Brabant, Netherlands
| | | | | | | | | |
Collapse
|
24
|
Yano YF, Tada H, Arakawa E, Voegeli W, Ina T, Uruga T, Matsushita T. Periodic Elastic Motion in a Self-Assembled Monolayer under Spontaneous Oscillations of Surface Tension: Molecules in a Scrum Push Back a Marangoni Flow. J Phys Chem Lett 2020; 11:6330-6336. [PMID: 32663403 DOI: 10.1021/acs.jpclett.0c01205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Regularly recurring phenomena are a common and important part of life. Such rhythmic behaviors are often seen in nonliving systems under far-from-equilibrium conditions. The study of simple nonliving systems provides clues for improving our understanding of the origin of biological rhythms. Here, we focus on the spontaneous oscillation of surface tension associated with an intermittent Marangoni convective flow generated by two types of surfactants, those that are partially soluble (long chain alcohols) and insoluble (lipids) in water. In this system, we find that the collective motions of two surfactants interact with each other in a systematic manner to control a stable periodic motion: the alcohol molecules (donor) produce a Marangoni flow, and the lipid molecules (acceptor) in a monolayer push the flow back. The shape of the surface tension oscillation can be explained by the viscoelastic properties of the acceptor surfactant, whereas the period of the surface tension oscillation has been explained by the physical properties of the donor surfactant. A recently developed time-resolved X-ray surface scattering technique enables the dynamic structure of the water surface under flow to be determined. We have repeatedly observed that lipid molecules at the air-water interface become regularly oriented normal to the surface at every onset of the Marangoni convective flow.
Collapse
Affiliation(s)
- Yohko F Yano
- Department of Physics, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka 577-8502, Japan
| | - Hiroki Tada
- Department of Physics, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka 577-8502, Japan
| | - Etsuo Arakawa
- Department of Physics, Tokyo Gakugei University, 4-1-1 Nukuikita-machi, Koganei, Tokyo 184-8501, Japan
| | - Wolfgang Voegeli
- Department of Physics, Tokyo Gakugei University, 4-1-1 Nukuikita-machi, Koganei, Tokyo 184-8501, Japan
| | - Toshiaki Ina
- Japan Synchrotron Radiation Research Institute, 1-1-1 Kouto, Sayo-cyo, Sayo-gun, Hyogo 679-5198, Japan
| | - Tomoya Uruga
- Japan Synchrotron Radiation Research Institute, 1-1-1 Kouto, Sayo-cyo, Sayo-gun, Hyogo 679-5198, Japan
| | - Tadashi Matsushita
- Photon Factory, Institute of Materials Structure Science, KEK, 1-1 Oho, Tsukuba, Ibaraki 305-0801, Japan
| |
Collapse
|
25
|
Monfredi O, Lakatta EG. Complexities in cardiovascular rhythmicity: perspectives on circadian normality, ageing and disease. Cardiovasc Res 2020; 115:1576-1595. [PMID: 31150049 DOI: 10.1093/cvr/cvz112] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/06/2019] [Accepted: 05/25/2019] [Indexed: 12/13/2022] Open
Abstract
Biological rhythms exist in organisms at all levels of complexity, in most organs and at myriad time scales. Our own biological rhythms are driven by energy emitted by the sun, interacting via our retinas with brain stem centres, which then send out complex messages designed to synchronize the behaviour of peripheral non-light sensing organs, to ensure optimal physiological responsiveness and performance of the organism based on the time of day. Peripheral organs themselves have autonomous rhythmic behaviours that can act independently from central nervous system control but is entrainable. Dysregulation of biological rhythms either through environment or disease has far-reaching consequences on health that we are only now beginning to appreciate. In this review, we focus on cardiovascular rhythms in health, with ageing and under disease conditions.
Collapse
Affiliation(s)
- Oliver Monfredi
- Division of Medicine, Department of Cardiology, The Johns Hopkins Hospital, 1800 Orleans Street, Baltimore, MD, USA.,Laboratory of Cardiovascular Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD, USA
| |
Collapse
|
26
|
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
|
27
|
Marchant JL, Farrell AP. Membrane and calcium clock mechanisms contribute variably as a function of temperature to setting cardiac pacemaker rate in zebrafish Danio rerio. JOURNAL OF FISH BIOLOGY 2019; 95:1265-1274. [PMID: 31429079 DOI: 10.1111/jfb.14126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/19/2019] [Indexed: 06/10/2023]
Abstract
Here, we show that heart rate in zebrafish Danio rerio is dependent upon two pacemaking mechanisms and it possesses a limited ability to reset the cardiac pacemaker with temperature acclimation. Electrocardiogram recordings, taken from individual, anaesthetised zebrafish that had been acclimated to 18, 23 or 28°C were used to follow the response of maximum heart rate (fHmax ) to acute warming from 18°C until signs of cardiac failure appeared (up to c. 40°C). Because fHmax was similar across the acclimation groups at almost all equivalent test temperatures, warm acclimation was limited to one significant effect, the 23°C acclimated zebrafish had a significantly higher (21%) peak fHmax and reached a higher (3°C) test temperature than the 18°C acclimated zebrafish. Using zatebradine to block the membrane hyperpolarisation-activated cyclic nucleotide-gated channels (HCN) and examine the contribution of the membrane clock mechanisms to cardiac pacemaking, f Hmax was significantly reduced (by at least 40%) at all acute test temperatures and significantly more so at most test temperatures for zebrafish acclimated to 28°C vs. 23°C. Thus, HCN channels and the membrane clock were not only important, but could be modified by thermal acclimation. Using a combination of ryanodine (to block sarcoplasmic calcium release) and thapsigargin (to block sarcoplasmic calcium reuptake) to examine the contribution of sarcoplasmic reticular handling of calcium and the calcium clock, f Hmax was again consistently reduced independent of the test temperature and acclimation temperature, but to a significantly lesser degree than zatebradine for zebrafish acclimated to both 28 and 18°C. Thus, the calcium clock mechanism plays an additional role in setting pacemaker activity that was independent of temperature. In conclusion, the zebrafish cardiac pacemaker has a limited temperature acclimation ability compared with known effects for other fishes and involves two pacemaking mechanisms, one of which was independent of temperature.
Collapse
Affiliation(s)
- James L Marchant
- Zoology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anthony P Farrell
- Zoology Department, University of British Columbia, Vancouver, British Columbia, Canada
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
28
|
Huang JH, Chen YC, Lu YY, Lin YK, Chen SA, Chen YJ. Arginine vasopressin modulates electrical activity and calcium homeostasis in pulmonary vein cardiomyocytes. J Biomed Sci 2019; 26:71. [PMID: 31530276 PMCID: PMC6747756 DOI: 10.1186/s12929-019-0564-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
Background Atrial fibrillation (AF) frequently coexists with congestive heart failure (HF) and arginine vasopressin (AVP) V1 receptor antagonists are used to treat hyponatremia in HF. However, the role of AVP in HF-induced AF still remains unclear. Pulmonary veins (PVs) are central in the genesis of AF. The purpose of this study was to determine if AVP is directly involved in the regulation of PV electrophysiological properties and calcium (Ca2+) homeostasis as well as the identification of the underlying mechanisms. Methods Patch clamp, confocal microscopy with Fluo-3 fluorescence, and Western blot analyses were used to evaluate the electrophysiological characteristics, Ca2+ homeostasis, and Ca2+ regulatory proteins in isolated rabbit single PV cardiomyocytes incubated with and without AVP (1 μM), OPC 21268 (0.1 μM, AVP V1 antagonist), or OPC 41061 (10 nM, AVP V2 antagonist) for 4–6 h. Results AVP (0.1 and 1 μM)-treated PV cardiomyocytes had a faster beating rate (108 to 152%) than the control cells. AVP (1 μM) treated PV cardiomyocytes had higher late sodium (Na+) and Na+/Ca2+ exchanger (NCX) currents than control PV cardiomyocytes. AVP (1 μM) treated PV cardiomyocytes had smaller Ca2+i transients, and sarcoplasmic reticulum (SR) Ca2+ content as well as higher Ca2+ leak. However, combined AVP (1 μM) and OPC 21268 (0.1 μM) treated PV cardiomyocytes had a slower PV beating rate, larger Ca2+i transients and SR Ca2+ content, smaller late Na+ and NCX currents than AVP (1 μM)-treated PV cardiomyocytes. Western blot experiments showed that AVP (1 μM) treated PV cardiomyocytes had higher expression of NCX and p-CaMKII, and a higher ratio of p-CaMKII/CaMKII. Conclusions AVP increases PV arrhythmogenesis with dysregulated Ca2+ homeostasis through vasopressin V1 signaling.
Collapse
Affiliation(s)
- Jen-Hung Huang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, 111 Hsin-Lung Road, Sec. 3, Taipei, 116, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, and Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan.,School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, 111 Hsin-Lung Road, Sec. 3, Taipei, 116, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ann Chen
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, 111 Hsin-Lung Road, Sec. 3, Taipei, 116, Taiwan. .,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
29
|
Kawada T, Yamamoto H, Uemura K, Hayama Y, Nishikawa T, Sugimachi M. Intravenous ivabradine augments the dynamic heart rate response to moderate vagal nerve stimulation in anesthetized rats. Am J Physiol Heart Circ Physiol 2019; 317:H597-H606. [DOI: 10.1152/ajpheart.00288.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Ivabradine is a selective bradycardic agent that reduces the heart rate (HR) by inhibiting the hyperpolarization-activated cyclic nucleotide-gated channels. Although its cardiovascular effect is thought to be minimal except for inducing bradycardia, ivabradine could interact with cardiovascular regulation by the autonomic nervous system. We tested whether ivabradine modifies dynamic characteristics of peripheral vagal HR control. In anesthetized Wistar-Kyoto rats ( n = 7), the right vagal nerve was sectioned and stimulated for 10 min according to a binary white noise sequence with a switching interval of 500 ms. The efferent vagal nerve stimulation (VNS) trials were performed using three different rates (10, 20, and 40 Hz), and were designated as V0–10, V0–20, and V0–40, respectively. The transfer function from the VNS to the HR was estimated before and after the intravenous administration of ivabradine (2 mg/kg). Ivabradine increased the asymptotic dynamic gain in V0–20 [from 3.88 (1.78–5.79) to 6.62 (3.12–8.31) beats·min−1·Hz−1, P < 0.01, median (range)] but not in V0–10 or V0–40. Ivabradine increased the corner frequency in V0–10 [from 0.032 (0.026–0.041) to 0.064 (0.029–0.090) Hz, P < 0.01] and V0–20 [from 0.040 (0.037–0.056) to 0.068 (0.051–0.100) Hz, P < 0.01] but not in V0–40. In conclusion, ivabradine augmented the dynamic HR response to moderate VNS. At high VNS, however, ivabradine did not significantly augment the dynamic HR response, possibly because ivabradine reduced the baseline HR and limited the range for the bradycardic response to high VNS. NEW & NOTEWORTHY Ivabradine is considered to be a pure bradycardic agent that has little effect on cardiovascular function except inducing bradycardia. The present study demonstrated that ivabradine interacts with the dynamic vagal heart rate control in a manner that augments the heart rate response to moderate-intensity efferent vagal nerve stimulation.
Collapse
Affiliation(s)
- Toru Kawada
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Hiromi Yamamoto
- Division of Cardiology, Department of Medicine, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Kazunori Uemura
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yohsuke Hayama
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Takuya Nishikawa
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Masaru Sugimachi
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center, Osaka, Japan
| |
Collapse
|
30
|
Zhang Y, Wang Y, Yanni J, Qureshi MA, Logantha SJRJ, Kassab S, Boyett MR, Gardiner NJ, Sun H, Howarth FC, Dobrzynski H. Electrical Conduction System Remodeling in Streptozotocin-Induced Diabetes Mellitus Rat Heart. Front Physiol 2019; 10:826. [PMID: 31338036 PMCID: PMC6628866 DOI: 10.3389/fphys.2019.00826] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 06/13/2019] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular complications are common in type 1 diabetes mellitus (TIDM) and there is an increased risk of arrhythmias as a result of dysfunction of the cardiac conduction system (CCS). We have previously shown that, in vivo, there is a decrease in the heart rate and prolongation of the QRS complex in streptozotocin-induced type 1 diabetic rats indicating dysfunction of the CCS. The aim of this study was to investigate the function of the ex vivo CCS and key proteins that are involved in pacemaker mechanisms in TIDM. RR interval, PR interval and QRS complex duration were significantly increased in diabetic rats. The beating rate of the isolated sinoatrial node (SAN) preparation was significantly decreased in diabetic rats. The funny current density and cell capacitance were significantly decreased in diabetic nodal cells. Western blot showed that proteins involved in the function of the CCS were significantly decreased in diabetic rats, namely: HCN4, Cav1.3, Cav3.1, Cx45, and NCX1 in the SAN; RyR2 and NCX1 in the atrioventricular junction and Cx40, Cx43, Cx45, and RyR2 in the Purkinje network. We conclude that there are complex functional and cellular changes in the CCS in TIDM. The changes in the proteins involved in the function of this electrical system are expected to adversely affect action potential generation and propagation, and these changes are likely to be arrhythmogenic.
Collapse
Affiliation(s)
- Yu Zhang
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom.,Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Yanwen Wang
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Joseph Yanni
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Mohammed Anwar Qureshi
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sunil Jit R J Logantha
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Sarah Kassab
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Mark R Boyett
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Natalie J Gardiner
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Frank Christopher Howarth
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
31
|
Swan AH, Gruscheski L, Boland LA, Brand T. The Popeye domain containing gene family encoding a family of cAMP-effector proteins with important functions in striated muscle and beyond. J Muscle Res Cell Motil 2019; 40:169-183. [PMID: 31197601 PMCID: PMC6726836 DOI: 10.1007/s10974-019-09523-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022]
Abstract
The Popeye domain containing (POPDC) gene family encodes a novel class of membrane-bound cyclic AMP effector proteins. POPDC proteins are abundantly expressed in cardiac and skeletal muscle. Consistent with its predominant expression in striated muscle, Popdc1 and Popdc2 null mutants in mouse and zebrafish develop cardiac arrhythmia and muscular dystrophy. Likewise, mutations in POPDC genes in patients have been associated with cardiac arrhythmia and muscular dystrophy phenotypes. A membrane trafficking function has been identified in this context. POPDC proteins have also been linked to tumour formation. Here, POPDC1 plays a role as a tumour suppressor by limiting c-Myc and WNT signalling. Currently, a common functional link between POPDC's role in striated muscle and as a tumour suppressor is lacking. We also discuss several alternative working models to better understand POPDC protein function.
Collapse
Affiliation(s)
- Alexander H Swan
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
- Institute of Chemical Biology, Imperial College London, London, UK
| | - Lena Gruscheski
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
| | - Lauren A Boland
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK
| | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, 4th Floor ICTEM Building, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
32
|
Zhang W, Zhu B, Ren J, Lu F, Qi Y, Weng W, Gao R. Two methods for modeling of sick sinus syndrome in rats: Ischemia reperfusion and sodium hydroxide induced injury. Biomed Pharmacother 2019; 111:778-784. [PMID: 30612002 DOI: 10.1016/j.biopha.2018.11.091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/13/2018] [Accepted: 11/25/2018] [Indexed: 01/23/2023] Open
Abstract
The Sick Sinus Syndrome (SSS) is a serious life-threatening heart disease. It is important to establish a credible and stable sinus node damage model. In this study, we use two methods to construct an SSS damage model in rats. One is to inject sodium hydroxide to the SSS area through internal jugular vein. Another is to cause ischemia-reperfusion injury on the SSS area. 43 healthy SD rats were randomly divided into 4 groups, namely ischemia-reperfusion injury group (IRIG), inject sodium hydroxide group (ISHG), and propranolol group (PG) and the control group (CG). The achievement ratio of modeling was 67% in the IRIG and 83% in the ISHG. The HR significantly decreased after operation in the IRIG and ISHG compared with pre-operation (P<0.01). The HR was reduced by above 30% in these 2 groups after modeling, while the reduction was better maintained in IRIG. Additionally, the sinoatrial node recovery time (SNRT) and sinoatrial conduction time (SACT) were significantly prolonged compared with pre-modeling in 2 groups (P < 0.01). Morphology results showed blurry in structure and boundaries with pale cytoplasm. It is speculated that IRIG and ISHG modeling might influence the calcium concentration and damage the sinus node function by decrease the expression of HCN4 and SCN5A, which impaired the driving ability of sinus node and leading to apoptosis. Ischemia reperfusion injury and sodium hydroxide injury could construct stable SSS models which could represent clinic pathological damage. Thus, both methods could be used for further studies of the SSS mechanisms and drugs.
Collapse
Affiliation(s)
- Wantong Zhang
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Baochen Zhu
- Beijing University of Chinese Medicine, 100029, China
| | - Jianxun Ren
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Fang Lu
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Yi Qi
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Weiliang Weng
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China
| | - Rui Gao
- China Academy of Chinese Medicine Sciences, Xiyuan hospital, 100091, China.
| |
Collapse
|
33
|
Canine and human sinoatrial node: differences and similarities in the structure, function, molecular profiles, and arrhythmia. J Vet Cardiol 2018; 22:2-19. [PMID: 30559056 DOI: 10.1016/j.jvc.2018.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/02/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
The sinoatrial node (SAN) is the primary pacemaker in canine and human hearts. The SAN in both species has a unique three-dimensional heterogeneous structure characterized by small pacemaker myocytes enmeshed within fibrotic strands, which partially insulate the cells from aberrant atrial activation. The SAN pacemaker tissue expresses a unique signature of proteins and receptors that mediate SAN automaticity, ion channel currents, and cell-to-cell communication, which are predominantly similar in both species. Recent intramural optical mapping, integrated with structural and molecular studies, has revealed the existence of up to five specialized SAN conduction pathways that preferentially conduct electrical activation to atrial tissues. The intrinsic heart rate, intranodal leading pacemaker shifts, and changes in conduction in response to physiological and pathophysiological stimuli are similar. Structural and/or functional impairments due to cardiac diseases including heart failure cause SAN dysfunctions (SNDs) in both species. These dysfunctions are usually manifested as severe bradycardia, tachy-brady arrhythmias, and conduction abnormalities including exit block and SAN reentry, which could lead to atrial tachycardia and fibrillation, cardiac arrest, and heart failure. Pharmaceutical drugs and implantable pacemakers are only partially successful in managing SNDs, emphasizing a critical need to develop targeted mechanism-based therapies to treat SNDs. Because several structural and functional characteristics are similar between the canine and human SAN, research in these species may be mutually beneficial for developing novel treatment approaches. This review describes structural, functional, and molecular similarities and differences between the canine and human SAN, with special emphasis on arrhythmias and unique causal mechanisms of SND in diseased hearts.
Collapse
|
34
|
Balcazar D, Regge V, Santalla M, Meyer H, Paululat A, Mattiazzi A, Ferrero P. SERCA is critical to control the Bowditch effect in the heart. Sci Rep 2018; 8:12447. [PMID: 30127403 PMCID: PMC6102201 DOI: 10.1038/s41598-018-30638-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/16/2018] [Indexed: 11/08/2022] Open
Abstract
The Bowditch effect or staircase phenomenon is the increment or reduction of contractile force when heart rate increases, defined as either a positive or negative staircase. The healthy and failing human heart both show positive or negative staircase, respectively, but the causes of these distinct cardiac responses are unclear. Different experimental approaches indicate that while the level of Ca2+ in the sarcoplasmic reticulum is critical, the molecular mechanisms are unclear. Here, we demonstrate that Drosophila melanogaster shows a negative staircase which is associated to a slight but significant frequency-dependent acceleration of relaxation (FDAR) at the highest stimulation frequencies tested. We further showed that the type of staircase is oppositely modified by two distinct SERCA mutations. The dominant conditional mutation SERCAA617T induced positive staircase and arrhythmia, while SERCAE442K accentuated the negative staircase of wild type. At the stimulation frequencies tested, no significant FDAR could be appreciated in mutant flies. The present results provide evidence that two individual mutations directly modify the type of staircase occurring within the heart and suggest an important role of SERCA in regulating the Bowditch effect.
Collapse
Affiliation(s)
- Darío Balcazar
- Centro de Investigaciones Cardiovasculares - CONICET/Universidad Nacional de la Plata, La Plata, Argentina
| | - Victoria Regge
- Centro de Investigaciones Cardiovasculares - CONICET/Universidad Nacional de la Plata and Departamento de Ciencias Básicas y Experimentales -UNNOBA, La Plata, Argentina
| | - Manuela Santalla
- Centro de Investigaciones Cardiovasculares - CONICET/Universidad Nacional de la Plata and Departamento de Ciencias Básicas y Experimentales -UNNOBA, La Plata, Argentina
| | - Heiko Meyer
- University of Osnabrück, Biology, Department of Zoology and Developmental Biology, Barbarastraße 11, 49076, Osnabrück, Germany
| | - Achim Paululat
- University of Osnabrück, Biology, Department of Zoology and Developmental Biology, Barbarastraße 11, 49076, Osnabrück, Germany
| | - Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares - CONICET/Universidad Nacional de la Plata, La Plata, Argentina
| | - Paola Ferrero
- Centro de Investigaciones Cardiovasculares - CONICET/Universidad Nacional de la Plata and Departamento de Ciencias Básicas y Experimentales -UNNOBA, La Plata, Argentina.
| |
Collapse
|
35
|
Wilders R, Verkerk AO. Long QT Syndrome and Sinus Bradycardia-A Mini Review. Front Cardiovasc Med 2018; 5:106. [PMID: 30123799 PMCID: PMC6085426 DOI: 10.3389/fcvm.2018.00106] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022] Open
Abstract
Congenital long-QT syndrome (LQTS) is an inherited cardiac disorder characterized by the prolongation of ventricular repolarization, susceptibility to Torsades de Pointes (TdP), and a risk for sudden death. Various types of congenital LQTS exist, all due to specific defects in ion channel-related genes. Interestingly, almost all of the ion channels affected by the various types of LQTS gene mutations are also expressed in the human sinoatrial node (SAN). It is therefore not surprising that LQTS is frequently associated with a change in basal heart rate (HR). However, current data on how the LQTS-associated ion channel defects result in impaired human SAN pacemaker activity are limited. In this mini-review, we provide an overview of known LQTS mutations with effects on HR and the underlying changes in expression and kinetics of ion channels. Sinus bradycardia has been reported in relation to a large number of LQTS mutations. However, the occurrence of both QT prolongation and sinus bradycardia on a family basis is almost completely limited to LQTS types 3 and 4 (LQT3 and Ankyrin-B syndrome, respectively). Furthermore, a clear causative role of this sinus bradycardia in cardiac events seems reserved to mutations underlying LQT3.
Collapse
Affiliation(s)
- Ronald Wilders
- Department of Medical Biology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam University Medical Centers, Amsterdam, Netherlands.,Department of Experimental Cardiology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
36
|
Tsutsui K, Monfredi OJ, Sirenko-Tagirova SG, Maltseva LA, Bychkov R, Kim MS, Ziman BD, Tarasov KV, Tarasova YS, Zhang J, Wang M, Maltsev AV, Brennan JA, Efimov IR, Stern MD, Maltsev VA, Lakatta EG. A coupled-clock system drives the automaticity of human sinoatrial nodal pacemaker cells. Sci Signal 2018; 11:eaap7608. [PMID: 29895616 PMCID: PMC6138244 DOI: 10.1126/scisignal.aap7608] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The spontaneous rhythmic action potentials generated by the sinoatrial node (SAN), the primary pacemaker in the heart, dictate the regular and optimal cardiac contractions that pump blood around the body. Although the heart rate of humans is substantially slower than that of smaller experimental animals, current perspectives on the biophysical mechanisms underlying the automaticity of sinoatrial nodal pacemaker cells (SANCs) have been gleaned largely from studies of animal hearts. Using human SANCs, we demonstrated that spontaneous rhythmic local Ca2+ releases generated by a Ca2+ clock were coupled to electrogenic surface membrane molecules (the M clock) to trigger rhythmic action potentials, and that Ca2+-cAMP-protein kinase A (PKA) signaling regulated clock coupling. When these clocks became uncoupled, SANCs failed to generate spontaneous action potentials, showing a depolarized membrane potential and disorganized local Ca2+ releases that failed to activate the M clock. β-Adrenergic receptor (β-AR) stimulation, which increases cAMP concentrations and clock coupling in other species, restored spontaneous, rhythmic action potentials in some nonbeating "arrested" human SANCs by increasing intracellular Ca2+ concentrations and synchronizing diastolic local Ca2+ releases. When β-AR stimulation was withdrawn, the clocks again became uncoupled, and SANCs reverted to a nonbeating arrested state. Thus, automaticity of human pacemaker cells is driven by a coupled-clock system driven by Ca2+-cAMP-PKA signaling. Extreme clock uncoupling led to failure of spontaneous action potential generation, which was restored by recoupling of the clocks. Clock coupling and action potential firing in some of these arrested cells can be restored by β-AR stimulation-induced augmentation of Ca2+-cAMP-PKA signaling.
Collapse
Affiliation(s)
- Kenta Tsutsui
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Oliver J Monfredi
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
- Department of Cardiovascular Electrophysiology, Johns Hopkins Hospital, 1800 Orleans Street, Baltimore, MD 21287, USA
| | | | - Larissa A Maltseva
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Rostislav Bychkov
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Mary S Kim
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Bruce D Ziman
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Kirill V Tarasov
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Yelena S Tarasova
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Jing Zhang
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Alexander V Maltsev
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Jaclyn A Brennan
- Department of Biomedical Engineering, George Washington University, Washington, DC 20052, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, George Washington University, Washington, DC 20052, USA
| | - Michael D Stern
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Victor A Maltsev
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD 21224, USA.
| |
Collapse
|
37
|
Duverger JE, Jacquemet V, Vinet A, Comtois P. In silico study of multicellular automaticity of heterogeneous cardiac cell monolayers: Effects of automaticity strength and structural linear anisotropy. PLoS Comput Biol 2018. [PMID: 29529023 PMCID: PMC5877903 DOI: 10.1371/journal.pcbi.1005978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The biological pacemaker approach is an alternative to cardiac electronic pacemakers. Its main objective is to create pacemaking activity from added or modified distribution of spontaneous cells in the myocardium. This paper aims to assess how automaticity strength of pacemaker cells (i.e. their ability to maintain robust spontaneous activity with fast rate and to drive neighboring quiescent cells) and structural linear anisotropy, combined with density and spatial distribution of pacemaker cells, may affect the macroscopic behavior of the biological pacemaker. A stochastic algorithm was used to randomly distribute pacemaker cells, with various densities and spatial distributions, in a semi-continuous mathematical model. Simulations of the model showed that stronger automaticity allows onset of spontaneous activity for lower densities and more homogeneous spatial distributions, displayed more central foci, less variability in cycle lengths and synchronization of electrical activation for similar spatial patterns, but more variability in those same variables for dissimilar spatial patterns. Compared to their isotropic counterparts, in silico anisotropic monolayers had less central foci and displayed more variability in cycle lengths and synchronization of electrical activation for both similar and dissimilar spatial patterns. The present study established a link between microscopic structure and macroscopic behavior of the biological pacemaker, and may provide crucial information for optimized biological pacemaker therapies. Implantation of electronic pacemakers is a standard treatment to pathologically slow heart rhythm. Despite improving quality of life, those devices display many shortcomings. Bioengineered tissue pacemakers may be a therapeutic alternative, but associated design methods usually lack control of the way cells with spontaneous activity are scattered throughout the tissue. Our study is the first to use a mathematical model to rigorously define and thoroughly characterize how pacemaker cells scattering at the microscopic level may affect macroscopic behaviors of the bioengineered tissue pacemaker. Automaticity strength (ability of pacemaker cell to drive its non-pacemaker neighbors) and anisotropy (preferential orientation of cell shape) are also implemented and give unparalleled insights on how effects of uncontrollable scattered pacemaker cells may be modulated by available experimental techniques. Our model is a powerful tool to aid in optimized bioengineered pacemaker therapies.
Collapse
Affiliation(s)
- James Elber Duverger
- Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology / Institute of Biomedical Engineering, Université de Montréal, Montreal, Quebec, Canada
| | - Vincent Jacquemet
- Department of Pharmacology and Physiology / Institute of Biomedical Engineering, Université de Montréal, Montreal, Quebec, Canada
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Alain Vinet
- Department of Pharmacology and Physiology / Institute of Biomedical Engineering, Université de Montréal, Montreal, Quebec, Canada
- Research Centre, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | - Philippe Comtois
- Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Pharmacology and Physiology / Institute of Biomedical Engineering, Université de Montréal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
38
|
Monfredi O, Tsutsui K, Ziman B, Stern MD, Lakatta EG, Maltsev VA. Electrophysiological heterogeneity of pacemaker cells in the rabbit intercaval region, including the SA node: insights from recording multiple ion currents in each cell. Am J Physiol Heart Circ Physiol 2018; 314:H403-H414. [PMID: 28916636 PMCID: PMC5899256 DOI: 10.1152/ajpheart.00253.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 09/13/2017] [Accepted: 09/13/2017] [Indexed: 11/22/2022]
Abstract
Cardiac pacemaker cells, including cells of the sinoatrial node, are heterogeneous in size, morphology, and electrophysiological characteristics. The exact extent to which these cells differ electrophysiologically is unclear yet is critical to understanding their functioning. We examined major ionic currents in individual intercaval pacemaker cells (IPCs) sampled from the paracristal, intercaval region (including the sinoatrial node) that were spontaneously beating after enzymatic isolation from rabbit hearts. The beating rate was measured at baseline and after inhibition of the Ca2+ pump with cyclopiazonic acid. Thereafter, in each cell, we consecutively measured the density of funny current ( If), delayed rectifier K+ current ( IK) (a surrogate of repolarization capacity), and L-type Ca2+ current ( ICa,L) using whole cell patch clamp. The ionic current densities varied to a greater extent than previously appreciated, with some IPCs demonstrating very small or zero If . The density of none of the currents was correlated with cell size, while ICa,L and If densities were related to baseline beating rates. If density was correlated with IK density but not with that of ICa,L. Inhibition of Ca2+ cycling had a greater beating rate slowing effect in IPCs with lower If densities. Our numerical model simulation indicated that 1) IPCs with small (or zero) If or small ICa,L can operate via a major contribution of Ca2+ clock, 2) If-Ca2+-clock interplay could be important for robust pacemaking function, and 3) coupled If- IK function could regulate maximum diastolic potential. Thus, we have demonstrated marked electrophysiological heterogeneity of IPCs. This heterogeneity is manifested in basal beating rate and response to interference of Ca2+ cycling, which is linked to If. NEW & NOTEWORTHY In the present study, a hitherto unrecognized range of heterogeneity of ion currents in pacemaker cells from the intercaval region is demonstrated. Relationships between basal beating rate and L-type Ca2+ current and funny current ( If) density are uncovered, along with a positive relationship between If and delayed rectifier K+ current. Links are shown between the response to Ca2+ cycling blockade and If density.
Collapse
Affiliation(s)
- Oliver Monfredi
- Laboratory of Cardiovascular Science, Biomedical Research Center, National Institute on Aging, National Institutes of Health , Baltimore, Maryland
- Department of Cardiovascular Electrophysiology, The Johns Hopkins Hospital , Baltimore, Maryland
- Division of Cardiovascular Sciences, University of Manchester , Manchester , United Kingdom
| | - Kenta Tsutsui
- Laboratory of Cardiovascular Science, Biomedical Research Center, National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| | - Bruce Ziman
- Laboratory of Cardiovascular Science, Biomedical Research Center, National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| | - Michael D Stern
- Laboratory of Cardiovascular Science, Biomedical Research Center, National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Biomedical Research Center, National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| | - Victor A Maltsev
- Laboratory of Cardiovascular Science, Biomedical Research Center, National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| |
Collapse
|
39
|
Abstract
Electrogenesis in the heart begins in the sinoatrial node and proceeds down the conduction system to originate the heartbeat. Conduction system disorders lead to slow heart rates that are insufficient to support the circulation, necessitating implantation of electronic pacemakers. The typical electronic pacemaker consists of a subcutaneous generator and battery module attached to one or more endocardial leads. New leadless pacemakers can be implanted directly into the right ventricular apex, providing single-chamber pacing without a subcutaneous generator. Modern pacemakers are generally reliable, and their programmability provides options for different pacing modes tailored to specific clinical needs. Advances in device technology will probably include alternative energy sources and dual-chamber leadless pacing in the not-too-distant future. Although effective, current electronic devices have limitations related to lead or generator malfunction, lack of autonomic responsiveness, undesirable interactions with strong magnetic fields, and device-related infections. Biological pacemakers, generated by somatic gene transfer, cell fusion, or cell transplantation, provide an alternative to electronic devices. Somatic reprogramming strategies, which involve transfer of genes encoding transcription factors to transform working myocardium into a surrogate sinoatrial node, are furthest along in the translational pipeline. Even as electronic pacemakers become smaller and less invasive, biological pacemakers might expand the therapeutic armamentarium for conduction system disorders.
Collapse
Affiliation(s)
- Eugenio Cingolani
- Cedars-Sinai Heart Institute, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| | - Joshua I Goldhaber
- Cedars-Sinai Heart Institute, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| | - Eduardo Marbán
- Cedars-Sinai Heart Institute, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| |
Collapse
|
40
|
Howarth FC, Qureshi MA, Jayaprakash P, Parekh K, Oz M, Dobrzynski H, Adrian TE. The Pattern of mRNA Expression Is Changed in Sinoatrial Node from Goto-Kakizaki Type 2 Diabetic Rat Heart. J Diabetes Res 2018; 2018:8454078. [PMID: 30246030 PMCID: PMC6139199 DOI: 10.1155/2018/8454078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/16/2018] [Accepted: 08/12/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND In vivo experiments in Goto-Kakizaki (GK) type 2 diabetic rats have demonstrated reductions in heart rate from a young age. The expression of genes encoding more than 70 proteins that are associated with the generation and conduction of electrical activity in the GK sinoatrial node (SAN) have been evaluated to further clarify the molecular basis of the low heart rate. MATERIALS AND METHODS Heart rate and expression of genes were evaluated with an extracellular electrode and real-time RT-PCR, respectively. Rats aged 12-13 months were employed in these experiments. RESULTS Isolated spontaneous heart rate was reduced in GK heart (161 ± 12 bpm) compared to controls (229 ± 11 bpm). There were many differences in expression of mRNA, and some of these differences were of particular interest. Compared to control SAN, expression of some genes were downregulated in GK-SAN: gap junction, Gja1 (Cx43), Gja5 (Cx40), Gjc1 (Cx45), and Gjd3 (Cx31.9); cell membrane transport, Trpc1 (TRPC1) and Trpc6 (TRPC6); hyperpolarization-activated cyclic nucleotide-gated channels, Hcn1 (HCN1) and Hcn4 (HCN4); calcium channels, Cacna1d (Cav1.3), Cacna1g (Cav3.1), Cacna1h (Cav3.2), Cacna2d1 (Cavα2δ1), Cacna2d3 (Cavα2δ3), and Cacng4 (Cav γ 4); and potassium channels, Kcna2 (Kv1.2), Kcna4 (Kv1.4), Kcna5 (Kv1.5), Kcnb1 (Kv2.1), Kcnd3 (Kv4.3), Kcnj2 (Kir2.1), Kcnk1 (TWIK1), Kcnk5 (K2P5.1), Kcnk6 (TWIK2), and Kcnn2 (SK2) whilst others were upregulated in GK-SAN: Ryr2 (RYR2) and Nppb (BNP). CONCLUSIONS This study provides new insight into the changing expression of genes in the sinoatrial node of diabetic heart.
Collapse
MESH Headings
- Action Potentials
- Animals
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/physiopathology
- Disease Models, Animal
- Gene Expression Regulation
- Heart Rate/genetics
- Isolated Heart Preparation
- Male
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Wistar
- Sinoatrial Node/metabolism
- Sinoatrial Node/physiopathology
Collapse
Affiliation(s)
- F. C. Howarth
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - M. A. Qureshi
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - P. Jayaprakash
- Department of Pharmacology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - K. Parekh
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - M. Oz
- Department of Pharmacology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - H. Dobrzynski
- Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - T. E. Adrian
- Department of Basic Medical Sciences, Mohammed Bin Rashid University of Medicine & Health Sciences, Dubai, UAE
| |
Collapse
|
41
|
Cook RF, Bussey CT, Mellor KM, Cragg PA, Lamberts RR. β1-Adrenoceptor, but not β2-adrenoceptor, subtype regulates heart rate in type 2 diabetic ratsin vivo. Exp Physiol 2017; 102:911-923. [DOI: 10.1113/ep086293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/16/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Rosalind F. Cook
- Department of Physiology, Otago School of Medical Sciences, HeartOtago; University of Otago; Dunedin New Zealand
| | - Carol T. Bussey
- Department of Physiology, Otago School of Medical Sciences, HeartOtago; University of Otago; Dunedin New Zealand
| | - Kimberley M. Mellor
- Department of Physiology, Faculty of Medical and Health Sciences; University of Auckland; Auckland New Zealand
| | - Patricia A. Cragg
- Department of Physiology, Otago School of Medical Sciences, HeartOtago; University of Otago; Dunedin New Zealand
| | - Regis R. Lamberts
- Department of Physiology, Otago School of Medical Sciences, HeartOtago; University of Otago; Dunedin New Zealand
| |
Collapse
|
42
|
Fabbri A, Fantini M, Wilders R, Severi S. Computational analysis of the human sinus node action potential: model development and effects of mutations. J Physiol 2017; 595:2365-2396. [PMID: 28185290 PMCID: PMC5374121 DOI: 10.1113/jp273259] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 02/02/2017] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS We constructed a comprehensive mathematical model of the spontaneous electrical activity of a human sinoatrial node (SAN) pacemaker cell, starting from the recent Severi-DiFrancesco model of rabbit SAN cells. Our model is based on electrophysiological data from isolated human SAN pacemaker cells and closely matches the action potentials and calcium transient that were recorded experimentally. Simulated ion channelopathies explain the clinically observed changes in heart rate in corresponding mutation carriers, providing an independent qualitative validation of the model. The model shows that the modulatory role of the 'funny current' (If ) in the pacing rate of human SAN pacemaker cells is highly similar to that of rabbit SAN cells, despite its considerably lower amplitude. The model may prove useful in the design of experiments and the development of heart-rate modulating drugs. ABSTRACT The sinoatrial node (SAN) is the normal pacemaker of the mammalian heart. Over several decades, a large amount of data on the ionic mechanisms underlying the spontaneous electrical activity of SAN pacemaker cells has been obtained, mostly in experiments on single cells isolated from rabbit SAN. This wealth of data has allowed the development of mathematical models of the electrical activity of rabbit SAN pacemaker cells. The present study aimed to construct a comprehensive model of the electrical activity of a human SAN pacemaker cell using recently obtained electrophysiological data from human SAN pacemaker cells. We based our model on the recent Severi-DiFrancesco model of a rabbit SAN pacemaker cell. The action potential and calcium transient of the resulting model are close to the experimentally recorded values. The model has a much smaller 'funny current' (If ) than do rabbit cells, although its modulatory role is highly similar. Changes in pacing rate upon the implementation of mutations associated with sinus node dysfunction agree with the clinical observations. This agreement holds for both loss-of-function and gain-of-function mutations in the HCN4, SCN5A and KCNQ1 genes, underlying ion channelopathies in If , fast sodium current and slow delayed rectifier potassium current, respectively. We conclude that our human SAN cell model can be a useful tool in the design of experiments and the development of drugs that aim to modulate heart rate.
Collapse
Affiliation(s)
- Alan Fabbri
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”University of BolognaCesenaItaly
| | - Matteo Fantini
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”University of BolognaCesenaItaly
| | - Ronald Wilders
- Department of Anatomy, Embryology and Physiology, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Stefano Severi
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”University of BolognaCesenaItaly
| |
Collapse
|
43
|
Klassen MP, Peters CJ, Zhou S, Williams HH, Jan LY, Jan YN. Age-dependent diastolic heart failure in an in vivo Drosophila model. eLife 2017; 6. [PMID: 28328397 PMCID: PMC5362267 DOI: 10.7554/elife.20851] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 02/11/2017] [Indexed: 12/13/2022] Open
Abstract
While the signals and complexes that coordinate the heartbeat are well established, how the heart maintains its electromechanical rhythm over a lifetime remains an open question with significant implications to human health. Reasoning that this homeostatic challenge confronts all pulsatile organs, we developed a high resolution imaging and analysis toolset for measuring cardiac function in intact, unanesthetized Drosophila melanogaster. We demonstrate that, as in humans, normal aging primarily manifests as defects in relaxation (diastole) while preserving contractile performance. Using this approach, we discovered that a pair of two-pore potassium channel (K2P) subunits, largely dispensable early in life, are necessary for terminating contraction (systole) in aged animals, where their loss culminates in fibrillatory cardiac arrest. As the pumping function of its heart is acutely dispensable for survival, Drosophila represents a uniquely accessible model for understanding the signaling networks maintaining cardiac performance during normal aging.
Collapse
Affiliation(s)
- Matthew P Klassen
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Christian J Peters
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Shiwei Zhou
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Hannah H Williams
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Lily Yeh Jan
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Yuh Nung Jan
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
44
|
Rezazadeh S, Duff HJ. Genetic Determinants of Hereditary Bradyarrhythmias: A Contemporary Review of a Diverse Group of Disorders. Can J Cardiol 2017; 33:758-767. [PMID: 28545623 DOI: 10.1016/j.cjca.2017.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/03/2017] [Accepted: 03/10/2017] [Indexed: 11/25/2022] Open
Abstract
Bradyarrhythmia is a common clinical presentation. Although the majority of cases are acquired, genetic screening of families with bradyarrhythmia has led to the discovery of a growing number of causative hereditary mutations. These mutations can interfere with any of the steps required for the occurrence of each cardiac cycle, including generation of an action potential in the sinoatrial node, successful exit of the action potential from the node, propagation of the action potential throughout the atria until the depolarization waves reach the atrioventricular node, and finally transmission of the action potential to the ventricles through the His-Purkinje system. As expected, channelopathies are the predominant culprit for hereditary bradyarrhythmias, because they play a crucial role in action potential generation and propagation. Interestingly, there are an increasing number of genes that encode for various regulatory or structural cellular components that have been linked to hereditary bradyarrhythmias. Furthermore, population-based genetic screening has revealed that age-related conduction defects may in fact be caused by genetic predispositions rather than the simple process of aging. With recent advances in genetic testing and the creation of animal models, not only have we discovered new culprit genes but it has also has become evident that there are still significant gaps in our knowledge of cardiac pathophysiology. In this review, we discuss the clinical presentations of known hereditary bradyarrhythmias and their associated conditions in addition to detailing our current molecular understanding of the mechanisms by which they are manifested.
Collapse
Affiliation(s)
- Saman Rezazadeh
- Department of Cardiac Sciences and Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Henry J Duff
- Department of Cardiac Sciences and Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
45
|
Weirich J. [Remodeling of the aging heart : Sinus node dysfunction and atrial fibrillation]. Herzschrittmacherther Elektrophysiol 2017; 28:29-38. [PMID: 28204916 DOI: 10.1007/s00399-017-0485-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022]
Abstract
The incidence of both sinus node dysfunction (SND) and atrial fibrillation (AF) increases with age. SND and AF frequently coexist. Likewise, they are often associated with cardiovascular diseases. Both arrhythmias share similar pathomechanisms such as structural and functional remodeling, i. e., degenerative fibrosis and altered Ca2+ handling, respectively. A growing body of evidence suggests an important role for the CamKII (Ca2+/calmodulin-dependent protein kinase II) in structural as well as in functional remodeling. In the sinus node, remodeling leads to degenerative fibrosis and as a consequence to sinus node arrest or to reentry (brady/tachycardia). In the atrium, remodeling sets the conditions for reentry and its triggering mechanisms, especially the conditions for triggered activity on the basis of delayed afterdepolarizations (DAD). Thus, SND and AF seem to be different phenotypes of related pathophysiological mechanisms. On the other hand, it remains controversial as to whether SND causes AF or vice versa.
Collapse
Affiliation(s)
- Jörg Weirich
- Institut für Physiologie, Abteilung II, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Hermann-Herder-Str. 7, 79104, Freiburg, Deutschland.
| |
Collapse
|
46
|
Regan CP, Morissette P, Regan HK, Travis JJ, Gerenser P, Wen J, Fitzgerald K, Gruver S, DeGeorge JJ, Sannajust FJ. Assessment of the clinical cardiac drug-drug interaction associated with the combination of hepatitis C virus nucleotide inhibitors and amiodarone in guinea pigs and rhesus monkeys. Hepatology 2016; 64:1430-1441. [PMID: 27474787 DOI: 10.1002/hep.28752] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/05/2016] [Accepted: 07/24/2016] [Indexed: 02/04/2023]
Abstract
UNLABELLED In 2015, European and U.S. health agencies issued warning letters in response to 9 reported clinical cases of severe bradycardia/bradyarrhythmia in hepatitis C virus (HCV)-infected patients treated with sofosbuvir (SOF) in combination with other direct acting antivirals (DAAs) and the antiarrhythmic drug, amiodarone (AMIO). We utilized preclinical in vivo models to better understand this cardiac effect, the potential pharmacological mechanism(s), and to identify a clinically translatable model to assess the drug-drug interaction (DDI) cardiac risk of current and future HCV inhibitors. An anesthetized guinea pig model was used to elicit a SOF+AMIO-dependent bradycardia. Detailed cardiac electrophysiological studies in this species revealed SOF+AMIO-dependent selective nodal dysfunction, with initial, larger effects on the sinoatrial node. Further studies in conscious, rhesus monkeys revealed an emergent bradycardia and bradyarrhythmia in 3 of 4 monkeys administered SOF+AMIO, effects not observed with either agent alone. Morever, bradycardia and bradyarrhythmia were not observed in rhesus monkeys when intravenous infusion of MK-3682 was completed after AMIO pretreatment. CONCLUSIONS These are the first preclinical in vivo experiments reported to replicate the severe clinical SOF+AMIO cardiac DDI and provide potential in vivo mechanism of action. As such, these data provide a preclinical risk assessment paradigm, including a clinically relevant nonhuman primate model, with which to better understand cardiovascular DDI risk for this therapeutic class. Furthermore, these studies suggest that not all HCV DAAs and, in particular, not all HCV nonstructural protein 5B inhibitors may exhibit this cardiac DDI with amiodarone. Given the selective in vivo cardiac electrophysiological effect, these data enable targeted cellular/molecular mechanistic studies to more precisely identify cell types, receptors, and/or ion channels responsible for the clinical DDI. (Hepatology 2016;64:1430-1441).
Collapse
Affiliation(s)
- Christopher P Regan
- Department of Safety & Exploratory Pharmacology, Merck Research Laboratories, West Point, PA.
| | - Pierre Morissette
- Department of Safety & Exploratory Pharmacology, Merck Research Laboratories, West Point, PA
| | - Hillary K Regan
- Department of Safety & Exploratory Pharmacology, Merck Research Laboratories, West Point, PA
| | - Jeffery J Travis
- Department of Safety & Exploratory Pharmacology, Merck Research Laboratories, West Point, PA
| | - Pamela Gerenser
- Department of Safety & Exploratory Pharmacology, Merck Research Laboratories, West Point, PA
| | - Jianzhong Wen
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck Research Laboratories, West Point, PA
| | - Kevin Fitzgerald
- Department of Safety & Exploratory Pharmacology, Merck Research Laboratories, West Point, PA
| | - Shaun Gruver
- Department of Safety & Exploratory Pharmacology, Merck Research Laboratories, West Point, PA
| | - Joseph J DeGeorge
- Safety Assessment and Laboratory Animal Resources, Merck Research Laboratories, West Point, PA
| | - Frederick J Sannajust
- Department of Safety & Exploratory Pharmacology, Merck Research Laboratories, West Point, PA
| |
Collapse
|
47
|
Simões MR, Azevedo BF, Fiorim J, Jr Freire DD, Covre EP, Vassallo DV, dos Santos L. Chronic mercury exposure impairs the sympathovagal control of the rat heart. Clin Exp Pharmacol Physiol 2016; 43:1038-1045. [DOI: 10.1111/1440-1681.12624] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 07/07/2016] [Accepted: 07/14/2016] [Indexed: 02/04/2023]
Affiliation(s)
- MR Simões
- Department of Physiological Sciences; Federal University of Espírito Santo; Vitória ES Brazil
| | - BF Azevedo
- Department of Physiological Sciences; Federal University of Espírito Santo; Vitória ES Brazil
| | - J Fiorim
- Department of Physiological Sciences; Federal University of Espírito Santo; Vitória ES Brazil
| | - DD Jr Freire
- Department of Physiological Sciences; Federal University of Espírito Santo; Vitória ES Brazil
| | - EP Covre
- Department of Physiological Sciences; Federal University of Espírito Santo; Vitória ES Brazil
| | - DV Vassallo
- Department of Physiological Sciences; Federal University of Espírito Santo; Vitória ES Brazil
- Health Science Centre of Vitória; EMESCAM; Vitória ES Brazil
| | - L dos Santos
- Department of Physiological Sciences; Federal University of Espírito Santo; Vitória ES Brazil
| |
Collapse
|
48
|
Bround MJ, Wambolt R, Cen H, Asghari P, Albu RF, Han J, McAfee D, Pourrier M, Scott NE, Bohunek L, Kulpa JE, Chen SRW, Fedida D, Brownsey RW, Borchers CH, Foster LJ, Mayor T, Moore EDW, Allard MF, Johnson JD. Cardiac Ryanodine Receptor (Ryr2)-mediated Calcium Signals Specifically Promote Glucose Oxidation via Pyruvate Dehydrogenase. J Biol Chem 2016; 291:23490-23505. [PMID: 27621312 DOI: 10.1074/jbc.m116.756973] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Indexed: 11/06/2022] Open
Abstract
Cardiac ryanodine receptor (Ryr2) Ca2+ release channels and cellular metabolism are both disrupted in heart disease. Recently, we demonstrated that total loss of Ryr2 leads to cardiomyocyte contractile dysfunction, arrhythmia, and reduced heart rate. Acute total Ryr2 ablation also impaired metabolism, but it was not clear whether this was a cause or consequence of heart failure. Previous in vitro studies revealed that Ca2+ flux into the mitochondria helps pace oxidative metabolism, but there is limited in vivo evidence supporting this concept. Here, we studied heart-specific, inducible Ryr2 haploinsufficient (cRyr2Δ50) mice with a stable 50% reduction in Ryr2 protein. This manipulation decreased the amplitude and frequency of cytosolic and mitochondrial Ca2+ signals in isolated cardiomyocytes, without changes in cardiomyocyte contraction. Remarkably, in the context of well preserved contractile function in perfused hearts, we observed decreased glucose oxidation, but not fat oxidation, with increased glycolysis. cRyr2Δ50 hearts exhibited hyperphosphorylation and inhibition of pyruvate dehydrogenase, the key Ca2+-sensitive gatekeeper to glucose oxidation. Metabolomic, proteomic, and transcriptomic analyses revealed additional functional networks associated with altered metabolism in this model. These results demonstrate that Ryr2 controls mitochondrial Ca2+ dynamics and plays a specific, critical role in promoting glucose oxidation in cardiomyocytes. Our findings indicate that partial RYR2 loss is sufficient to cause metabolic abnormalities seen in heart disease.
Collapse
Affiliation(s)
- Michael J Bround
- From the Cardiovascular Research Group, Life Sciences Institute and.,Departments of Cellular and Physiological Sciences
| | - Rich Wambolt
- From the Cardiovascular Research Group, Life Sciences Institute and.,the Department of Pathology and Laboratory Medicine, University of British Columbia and the Centre for Heart and Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia V6Z 1Y6
| | - Haoning Cen
- From the Cardiovascular Research Group, Life Sciences Institute and.,Departments of Cellular and Physiological Sciences
| | - Parisa Asghari
- From the Cardiovascular Research Group, Life Sciences Institute and.,Departments of Cellular and Physiological Sciences
| | - Razvan F Albu
- Biochemistry and Molecular Biology, and.,the Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4
| | - Jun Han
- the University of Victoria-Genome British Columbia Proteomics Centre, Victoria, British Columbia V8Z 7X8, and
| | - Donald McAfee
- From the Cardiovascular Research Group, Life Sciences Institute and.,Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3
| | - Marc Pourrier
- From the Cardiovascular Research Group, Life Sciences Institute and.,Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3
| | - Nichollas E Scott
- Biochemistry and Molecular Biology, and.,the Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4
| | - Lubos Bohunek
- the Department of Pathology and Laboratory Medicine, University of British Columbia and the Centre for Heart and Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia V6Z 1Y6
| | | | - S R Wayne Chen
- the Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 2T9, Canada
| | - David Fedida
- From the Cardiovascular Research Group, Life Sciences Institute and.,Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia V6T 1Z3
| | | | - Christoph H Borchers
- the University of Victoria-Genome British Columbia Proteomics Centre, Victoria, British Columbia V8Z 7X8, and
| | - Leonard J Foster
- Biochemistry and Molecular Biology, and.,the Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4
| | - Thibault Mayor
- Biochemistry and Molecular Biology, and.,the Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4
| | - Edwin D W Moore
- From the Cardiovascular Research Group, Life Sciences Institute and.,Departments of Cellular and Physiological Sciences
| | - Michael F Allard
- From the Cardiovascular Research Group, Life Sciences Institute and.,the Department of Pathology and Laboratory Medicine, University of British Columbia and the Centre for Heart and Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia V6Z 1Y6
| | - James D Johnson
- From the Cardiovascular Research Group, Life Sciences Institute and .,Departments of Cellular and Physiological Sciences
| |
Collapse
|
49
|
Wilson CM, Roa JN, Cox GK, Tresguerres M, Farrell AP. Introducing a novel mechanism to control heart rate in the ancestral Pacific hagfish. ACTA ACUST UNITED AC 2016; 219:3227-3236. [PMID: 27510962 DOI: 10.1242/jeb.138198] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 08/02/2016] [Indexed: 12/15/2022]
Abstract
Although neural modulation of heart rate is well established among chordate animals, the Pacific hagfish (Eptatretus stoutii) lacks any cardiac innervation, yet it can increase its heart rate from the steady, depressed heart rate seen in prolonged anoxia to almost double its normal normoxic heart rate, an almost fourfold overall change during the 1-h recovery from anoxia. The present study sought mechanistic explanations for these regulatory changes in heart rate. We provide evidence for a bicarbonate-activated, soluble adenylyl cyclase (sAC)-dependent mechanism to control heart rate, a mechanism never previously implicated in chordate cardiac control.
Collapse
Affiliation(s)
- Christopher M Wilson
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada V6T 1Z4
| | - Jinae N Roa
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Georgina K Cox
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada V6T 1Z4
| | - Martin Tresguerres
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Anthony P Farrell
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada V6T 1Z4.,Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, British Columbia, Canada V6T 1Z4
| |
Collapse
|
50
|
Mesirca P, Bidaud I, Mangoni ME. Rescuing cardiac automaticity in L-type Cav1.3 channelopathies and beyond. J Physiol 2016; 594:5869-5879. [PMID: 27374078 DOI: 10.1113/jp270678] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 06/24/2016] [Indexed: 11/08/2022] Open
Abstract
Pacemaker activity of the sino-atrial node generates the heart rate. Disease of the sinus node and impairment of atrioventricular conduction induce an excessively low ventricular rate (bradycardia), which cannot meet the needs of the organism. Bradycardia accounts for about half of the total workload of clinical cardiologists. The 'sick sinus' syndrome (SSS) is characterized by sinus bradycardia and periods of intermittent atrial fibrillation. Several genetic or acquired risk factors or pathologies can lead to SSS. Implantation of an electronic pacemaker constitutes the only available therapy for SSS. The incidence of SSS is forecast to double over the next 50 years, with ageing of the general population thus urging the development of complementary or alternative therapeutic strategies. In recent years an increasing number of mutations affecting ion channels involved in sino-atrial automaticity have been reported to underlie inheritable SSS. L-type Cav 1.3 channels play a major role in the generation and regulation of sino-atrial pacemaker activity and atrioventricular conduction. Mutation in the CACNA1D gene encoding Cav 1.3 channels induces loss-of-function in channel activity and underlies the sino-atrial node dysfunction and deafness syndrome (SANDD). Mice lacking Cav 1.3 channels (Cav 1.3-/- ) fairly recapitulate SSS and constitute a precious model to test new therapeutic approaches to handle this disease. Work in our laboratory shows that targeting G protein-gated K+ (IKACh ) channels effectively rescues SSS of Cav 1.3-/- mice. This new concept of 'compensatory' ion channel targeting shines new light on the principles underlying the pacemaker mechanism and may open the way to new therapies for SSS.
Collapse
Affiliation(s)
- Pietro Mesirca
- Département de Physiologie, Institut de Genomique Fonctionnelle, LabEx ICST, UMR-5203, Centre national de la recherche scientifique, F-34094, Montpellier, France. .,INSERM U1191, F-34094, Montpellier, France. .,Université de Montpellier, F-34094, Montpellier, France.
| | - Isabelle Bidaud
- Département de Physiologie, Institut de Genomique Fonctionnelle, LabEx ICST, UMR-5203, Centre national de la recherche scientifique, F-34094, Montpellier, France.,INSERM U1191, F-34094, Montpellier, France.,Université de Montpellier, F-34094, Montpellier, France
| | - Matteo E Mangoni
- Département de Physiologie, Institut de Genomique Fonctionnelle, LabEx ICST, UMR-5203, Centre national de la recherche scientifique, F-34094, Montpellier, France. .,INSERM U1191, F-34094, Montpellier, France. .,Université de Montpellier, F-34094, Montpellier, France.
| |
Collapse
|