1
|
Hohendanner F, Prabhu A, Wilck N, Stangl V, Pieske B, Stangl K, Althoff TF. G q-Mediated Arrhythmogenic Signaling Promotes Atrial Fibrillation. Biomedicines 2023; 11:biomedicines11020526. [PMID: 36831062 PMCID: PMC9953645 DOI: 10.3390/biomedicines11020526] [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: 01/27/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is promoted by various stimuli like angiotensin II, endothelin-1, epinephrine/norepinephrine, vagal activation, or mechanical stress, all of which activate receptors coupled to G-proteins of the Gαq/Gα11-family (Gq). Besides pro-fibrotic and pro-inflammatory effects, Gq-mediated signaling induces inositol trisphosphate receptor (IP3R)-mediated intracellular Ca2+ mobilization related to delayed after-depolarisations and AF. However, direct evidence of arrhythmogenic Gq-mediated signaling is absent. METHODS AND RESULTS To define the role of Gq in AF, transgenic mice with tamoxifen-inducible, cardiomyocyte-specific Gαq/Gα11-deficiency (Gq-KO) were created and exposed to intracardiac electrophysiological studies. Baseline electrophysiological properties, including heart rate, sinus node recovery time, and atrial as well as AV nodal effective refractory periods, were comparable in Gq-KO and control mice. However, inducibility and mean duration of AF episodes were significantly reduced in Gq-KO mice-both before and after vagal stimulation. To explore underlying mechanisms, left atrial cardiomyocytes were isolated from Gq-KO and control mice and electrically stimulated to study Ca2+-mobilization during excitation-contraction coupling using confocal microscopy. Spontaneous arrhythmogenic Ca2+ waves and sarcoplasmic reticulum content-corrected Ca2+ sparks were less frequent in Gq-KO mice. Interestingly, nuclear but not cytosolic Ca2+ transient amplitudes were significantly decreased in Gq-KO mice. CONCLUSION Gq-signaling promotes arrhythmogenic atrial Ca2+-release and AF in mice. Targeting this pathway, ideally using Gq-selective, biased receptor ligands, may be a promising approach for the treatment and prevention of AF. Importantly, the atrial-specific expression of the Gq-effector IP3R confers atrial selectivity mitigating the risk of life-threatening ventricular pro-arrhythmic effects.
Collapse
Affiliation(s)
- Felix Hohendanner
- Department of Cardiology and German Heart Center, Campus Virchow-Klinikum, Charité–University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13316 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Ashok Prabhu
- Department of Cardiology and German Heart Center, Campus Virchow-Klinikum, Charité–University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nicola Wilck
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13316 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a Cooperation of Charité–Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine (MDC), 13125 Berlin, Germany
- Department of Nephrology and Medical Intensive Care Medicine, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Verena Stangl
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13316 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Cardiology and Angiology, Charité Campus Mitte, Charité–University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Burkert Pieske
- Department of Cardiology and German Heart Center, Campus Virchow-Klinikum, Charité–University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13316 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Karl Stangl
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13316 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Cardiology and Angiology, Charité Campus Mitte, Charité–University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Till F. Althoff
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13316 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Cardiology and Angiology, Charité Campus Mitte, Charité–University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Arrhythmia Section, Cardiovascular Institute (ICCV), Hospital Clínic, Universitat de Barcelona, C/Villarroel N° 170, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-2275551; Fax: +34-93-4513045
| |
Collapse
|
2
|
Bose SJ, Read MJ, Akerman E, Capel RA, Ayagama T, Russell A, Terrar DA, Zaccolo M, Burton RAB. Inhibition of adenylyl cyclase 1 by ST034307 inhibits IP 3-evoked changes in sino-atrial node beat rate. Front Pharmacol 2022; 13:951897. [PMID: 36105228 PMCID: PMC9465815 DOI: 10.3389/fphar.2022.951897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Atrial arrhythmias, such as atrial fibrillation (AF), are a major mortality risk and a leading cause of stroke. The IP3 signalling pathway has been proposed as an atrial-specific target for AF therapy, and atrial IP3 signalling has been linked to the activation of calcium sensitive adenylyl cyclases AC1 and AC8. We investigated the involvement of AC1 in the response of intact mouse atrial tissue and isolated guinea pig atrial and sino-atrial node (SAN) cells to the α-adrenoceptor agonist phenylephrine (PE) using the selective AC1 inhibitor ST034307. The maximum rate change of spontaneously beating mouse right atrial tissue exposed to PE was reduced from 14.5% to 8.2% (p = 0.005) in the presence of 1 μM ST034307, whereas the increase in tension generated in paced left atrial tissue in the presence of PE was not inhibited by ST034307 (Control = 14.2%, ST034307 = 16.3%; p > 0.05). Experiments were performed using isolated guinea pig atrial and SAN cells loaded with Fluo-5F-AM to record changes in calcium transients (CaT) generated by 10 μM PE in the presence and absence of 1 μM ST034307. ST034307 significantly reduced the beating rate of SAN cells (0.34-fold decrease; p = 0.003) but did not inhibit changes in CaT amplitude in response to PE in atrial cells. The results presented here demonstrate pharmacologically the involvement of AC1 in the downstream response of atrial pacemaker activity to α-adrenoreceptor stimulation and IP3R calcium release.
Collapse
Affiliation(s)
- Samuel J. Bose
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Matthew J. Read
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Emily Akerman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Rebecca A. Capel
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Thamali Ayagama
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Angela Russell
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, United Kingdom
| | - Derek A. Terrar
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
3
|
Electroacupuncture Ameliorates Acute Myocardial Ischemic Injury and Long QT Interval in Mice through the α1A-Adrenergic Receptor: Electrophysiological, Morphological, and Molecular Evidence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1984706. [PMID: 35814274 PMCID: PMC9262560 DOI: 10.1155/2022/1984706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/10/2022] [Indexed: 11/18/2022]
Abstract
Acute myocardial ischemia (AMI) is a condition caused by a decrease in blood flow to the heart that can sometimes predispose to acquired long QT syndrome (LQTS), thereby resulting in sudden cardiac death. Recent evidence indicates that electroacupuncture (EA) can alleviate MI injury, but its specific mechanism remains unclear. This study was aimed at investigating the efficacy of EA, which utilizes α1A-adrenergic receptors (α1A-AR) in alleviating MI injury as well as the resulting LQTS. The AMI model was established by ligating the left anterior descending arteries (LAD) of both the wild-type and α1A gene-knockout mice and treating them with EA for three consecutive days. A PowerLab 16 physiological recorder was used to collect the electrocardiogram (ECG) while the serum creatine kinase isoenzymes (CK-MB), lactate dehydrogenase (LDH), and norepinephrine (NE) levels in myocardial tissue were determined by using the enzyme-linked immunosorbent assay (ELISA) kit. Moreover, TTC staining was used to observe the myocardial ischemic area, while H&E and TUNEL staining determined the pathological morphology of the myocardium. Quantitative real-time PCR (qRT-PCR) was used to detect the α1A mRNA, and Western blot was used to detect the specific proteins, such as α1A, cleaved caspase-3, Gq, PLC, p-PKCα, and p-hERG. Our results showed that EA could effectively reduce elevated ST-segment, shorten the extended QT interval, and reduce the serum myocardial enzyme content and the degree of pathological injury in wild mice with MI. EA can also decrease the expression of α1A-AR, PLC, p-PKCα, and NE content in myocardial tissues of wild mice, while those of p-hERG increased in ischemic myocardial tissue. These findings suggested that α1A-AR is involved in the development of MI as well as LQTS. Additionally, EA treatment improves the cardiac function and ischemic long QT interval and plays an important role in reducing the hERG inhibition through the α1A-AR-mediated Gq/PLC/PKCα pathway and myocardial apoptosis. Hence, it is suggested that α1A-AR might become a potential target for EA in treating AMI treatment of myocardial ischemia injury and acquired long QT intervals caused by MI.
Collapse
|
4
|
Pfenniger A, Yoo S, Arora R. Nucleoplasmic Ca 2+: The 'Mastermind' Behind Pathological Atrial Remodeling? Circ Res 2021; 128:636-638. [PMID: 34314193 PMCID: PMC9278521 DOI: 10.1161/circresaha.121.318800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anna Pfenniger
- Department of Medicine, Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, IL
| | - Shin Yoo
- Department of Medicine, Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, IL
| | - Rishi Arora
- Department of Medicine, Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
5
|
Luo Y, Liu X, Ma R, Wang Y, Zimering M, Pan Z. Circulating IgGs in Type 2 Diabetes with Atrial Fibrillation Induce IP 3-Mediated Calcium Elevation in Cardiomyocytes. iScience 2020; 23:101036. [PMID: 32315831 PMCID: PMC7170991 DOI: 10.1016/j.isci.2020.101036] [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: 08/30/2019] [Revised: 01/16/2020] [Accepted: 04/01/2020] [Indexed: 11/06/2022] Open
Abstract
Higher risk of cardiac arrhythmias including atrial fibrillation (AF) associates with type 2 diabetes mellitus (T2DM) with the underlying mechanism largely unknown. The present study reported a subset of circulating immunoglobulin G autoantibodies (IgGs) from patients with T2DM with AF (T2DM/AF)-induced intracellular calcium elevation in both human induced pluripotent stem cell (iPSC)-derived and mouse atrial cardiomyocytes, whereas (identical concentrations of) IgGs from patients with T2DM without AF could not. The IgG-evoked intracellular calcium elevation was insensitive to verapamil, mibefradil, or BTP-2, indicating calcium source from neither voltage-gated calcium channels nor store-operated calcium entry. On the other hand, pharmacological antagonism or genetic knockdown of inositol triphosphate (IP3) receptor significantly decreased T2DM/AF IgG-induced intracellular calcium elevation. Furthermore, pharmacological blockage of G protein-coupled receptor (GPCR), heterotrimeric G protein or phospholipase C dampened IgG-induced intracellular calcium elevation. Taken together, circulating IgGs from patients with T2DM/AF stimulated arrhythmogenic intracellular calcium elevation through IP3 pathway in atrial cardiomyocytes. Identification of cardiomyocyte-targeting IgGs in T2DM atrial fibrillation patients Induction of arrhythmogenic Ca2+ signaling by these IgGs Independent of voltage-gated or store-operated Ca2+ channels Involvement of GPCR-IP3-IP3R axis in IgG-evoked intracellular Ca2+ elevation
Collapse
Affiliation(s)
- Yanhong Luo
- Davis Heart and Lung Research Institute, Ohio State University-Wexner Medical Center, Columbus, OH 43210, USA; Department of Endocrinology, The Children's Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Xian Liu
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX 76010, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76010, USA
| | - Ruilian Ma
- Division of Regenerative Medicine Research, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Yigang Wang
- Division of Regenerative Medicine Research, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Mark Zimering
- Endocrinology, Veterans Affairs New Jersey Healthcare System, East Orange, NJ 07018, USA; Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| | - Zui Pan
- Davis Heart and Lung Research Institute, Ohio State University-Wexner Medical Center, Columbus, OH 43210, USA; Department of Kinesiology, University of Texas at Arlington, Arlington, TX 76010, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76010, USA.
| |
Collapse
|
6
|
Denham NC, Pearman CM, Caldwell JL, Madders GWP, Eisner DA, Trafford AW, Dibb KM. Calcium in the Pathophysiology of Atrial Fibrillation and Heart Failure. Front Physiol 2018; 9:1380. [PMID: 30337881 PMCID: PMC6180171 DOI: 10.3389/fphys.2018.01380] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022] Open
Abstract
Atrial fibrillation (AF) is commonly associated with heart failure. A bidirectional relationship exists between the two-AF exacerbates heart failure causing a significant increase in heart failure symptoms, admissions to hospital and cardiovascular death, while pathological remodeling of the atria as a result of heart failure increases the risk of AF. A comprehensive understanding of the pathophysiology of AF is essential if we are to break this vicious circle. In this review, the latest evidence will be presented showing a fundamental role for calcium in both the induction and maintenance of AF. After outlining atrial electrophysiology and calcium handling, the role of calcium-dependent afterdepolarizations and atrial repolarization alternans in triggering AF will be considered. The atrial response to rapid stimulation will be discussed, including the short-term protection from calcium overload in the form of calcium signaling silencing and the eventual progression to diastolic calcium leak causing afterdepolarizations and the development of an electrical substrate that perpetuates AF. The role of calcium in the bidirectional relationship between heart failure and AF will then be covered. The effects of heart failure on atrial calcium handling that promote AF will be reviewed, including effects on both atrial myocytes and the pulmonary veins, before the aspects of AF which exacerbate heart failure are discussed. Finally, the limitations of human and animal studies will be explored allowing contextualization of what are sometimes discordant results.
Collapse
Affiliation(s)
- Nathan C. Denham
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | - Katharine M. Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
7
|
Calvo D, Filgueiras-Rama D, Jalife J. Mechanisms and Drug Development in Atrial Fibrillation. Pharmacol Rev 2018; 70:505-525. [PMID: 29921647 PMCID: PMC6010660 DOI: 10.1124/pr.117.014183] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation is a highly prevalent cardiac arrhythmia and the most important cause of embolic stroke. Although genetic studies have identified an increasing assembly of AF-related genes, the impact of these genetic discoveries is yet to be realized. In addition, despite more than a century of research and speculation, the molecular and cellular mechanisms underlying AF have not been established, and therapy for AF, particularly persistent AF, remains suboptimal. Current antiarrhythmic drugs are associated with a significant rate of adverse events, particularly proarrhythmia, which may explain why many highly symptomatic AF patients are not receiving any rhythm control therapy. This review focuses on recent advances in AF research, including its epidemiology, genetics, and pathophysiological mechanisms. We then discuss the status of antiarrhythmic drug therapy for AF today, reviewing molecular mechanisms, and the possible clinical use of some of the new atrial-selective antifibrillatory agents, as well as drugs that target atrial remodeling, inflammation and fibrosis, which are being tested as upstream therapies to prevent AF perpetuation. Altogether, the objective is to highlight the magnitude and endemic dimension of AF, which requires a significant effort to develop new and effective antiarrhythmic drugs, but also improve AF prevention and treatment of risk factors that are associated with AF complications.
Collapse
Affiliation(s)
- David Calvo
- Department of Cardiology, Arrhythmia Unit, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain (D.C.); Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.F.-R., J.J.); Department of Cardiology, Arrhythmia Unit, Hospital Clínico Universitario San Carlos, Madrid, Spain (D.F.-R.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (D.F.-R., J.J.); and Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan (J.J.)
| | - David Filgueiras-Rama
- Department of Cardiology, Arrhythmia Unit, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain (D.C.); Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.F.-R., J.J.); Department of Cardiology, Arrhythmia Unit, Hospital Clínico Universitario San Carlos, Madrid, Spain (D.F.-R.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (D.F.-R., J.J.); and Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan (J.J.)
| | - José Jalife
- Department of Cardiology, Arrhythmia Unit, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain (D.C.); Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.F.-R., J.J.); Department of Cardiology, Arrhythmia Unit, Hospital Clínico Universitario San Carlos, Madrid, Spain (D.F.-R.); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (D.F.-R., J.J.); and Center for Arrhythmia Research, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan (J.J.)
| |
Collapse
|
8
|
Finlay M, Harmer SC, Tinker A. The control of cardiac ventricular excitability by autonomic pathways. Pharmacol Ther 2017; 174:97-111. [PMID: 28223225 DOI: 10.1016/j.pharmthera.2017.02.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Central to the genesis of ventricular cardiac arrhythmia are variations in determinants of excitability. These involve individual ionic channels and transporters in cardiac myocytes but also tissue factors such as variable conduction of the excitation wave, fibrosis and source-sink mismatch. It is also known that in certain diseases and particularly the channelopathies critical events occur with specific stressors. For example, in hereditary long QT syndrome due to mutations in KCNQ1 arrhythmic episodes are provoked by exercise and in particular swimming. Thus not only is the static substrate important but also how this is modified by dynamic signalling events associated with common physiological responses. In this review, we examine the regulation of ventricular excitability by signalling pathways from a cellular and tissue perspective in an effort to identify key processes, effectors and potential therapeutic approaches. We specifically focus on the autonomic nervous system and related signalling pathways.
Collapse
Affiliation(s)
- Malcolm Finlay
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK
| | - Stephen C Harmer
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK
| | - Andrew Tinker
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK.
| |
Collapse
|
9
|
Berridge MJ. The Inositol Trisphosphate/Calcium Signaling Pathway in Health and Disease. Physiol Rev 2016; 96:1261-96. [DOI: 10.1152/physrev.00006.2016] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Many cellular functions are regulated by calcium (Ca2+) signals that are generated by different signaling pathways. One of these is the inositol 1,4,5-trisphosphate/calcium (InsP3/Ca2+) signaling pathway that operates through either primary or modulatory mechanisms. In its primary role, it generates the Ca2+ that acts directly to control processes such as metabolism, secretion, fertilization, proliferation, and smooth muscle contraction. Its modulatory role occurs in excitable cells where it modulates the primary Ca2+ signal generated by the entry of Ca2+ through voltage-operated channels that releases Ca2+ from ryanodine receptors (RYRs) on the internal stores. In carrying out this modulatory role, the InsP3/Ca2+ signaling pathway induces subtle changes in the generation and function of the voltage-dependent primary Ca2+ signal. Changes in the nature of both the primary and modulatory roles of InsP3/Ca2+ signaling are a contributory factor responsible for the onset of a large number human diseases.
Collapse
Affiliation(s)
- Michael J. Berridge
- Laboratory of Molecular Signalling, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| |
Collapse
|