1
|
Pfenniger A, Yoo S, Arora R. Oxidative stress and atrial fibrillation. J Mol Cell Cardiol 2024; 196:141-151. [PMID: 39307416 DOI: 10.1016/j.yjmcc.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in clinical practice. Though the pathogenesis of AF is complex and is not completely understood, many studies suggest that oxidative stress is a major mechanism in pathophysiology of AF. Through multiple mechanisms, reactive oxygen species (ROS) lead to the formation of an AF substrate that facilitates the development and maintenance of AF. In this review article, we provide an update on the different mechanisms by which oxidative stress promotes atrial remodeling. We then discuss several therapeutic strategies targeting oxidative stress for the prevention or treatment of AF. Considering the complex biology of ROS induced remodeling, and the evolution of ROS sources and compartmentalization during AF progression, there is a definite need for improvement in timing, targeting and reduction of off-target effects of therapeutic strategies targeting oxidative injury in AF.
Collapse
Affiliation(s)
- Anna Pfenniger
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Shin Yoo
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Rishi Arora
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America.
| |
Collapse
|
2
|
Fakuade FE, Hubricht D, Möller V, Sobitov I, Liutkute A, Döring Y, Seibertz F, Gerloff M, Pronto JRD, Haghighi F, Brandenburg S, Alhussini K, Ignatyeva N, Bonhoff Y, Kestel S, El-Essawi A, Jebran AF, Großmann M, Danner BC, Baraki H, Schmidt C, Sossalla S, Kutschka I, Bening C, Maack C, Linke WA, Heijman J, Lehnart SE, Kensah G, Ebert A, Mason FE, Voigt N. Impaired Intracellular Calcium Buffering Contributes to the Arrhythmogenic Substrate in Atrial Myocytes From Patients With Atrial Fibrillation. Circulation 2024; 150:544-559. [PMID: 38910563 PMCID: PMC11319087 DOI: 10.1161/circulationaha.123.066577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/31/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Alterations in the buffering of intracellular Ca2+, for which myofilament proteins play a key role, have been shown to promote cardiac arrhythmia. It is interesting that although studies report atrial myofibrillar degradation in patients with persistent atrial fibrillation (persAF), the intracellular Ca2+ buffering profile in persAF remains obscure. Therefore, we aimed to investigate the intracellular buffering of Ca2+ and its potential arrhythmogenic role in persAF. METHODS Transmembrane Ca2+ fluxes (patch-clamp) and intracellular Ca2+ signaling (fluo-3-acetoxymethyl ester) were recorded simultaneously in myocytes from right atrial biopsies of sinus rhythm (Ctrl) and patients with persAF, alongside human atrial subtype induced pluripotent stem cell-derived cardiac myocytes (iPSC-CMs). Protein levels were quantified by immunoblotting of human atrial tissue and induced pluripotent stem cell-derived cardiac myocytes. Mouse whole heart and atrial electrophysiology were measured on a Langendorff system. RESULTS Cytosolic Ca2+ buffering was decreased in atrial myocytes of patients with persAF because of a depleted amount of Ca2+ buffers. In agreement, protein levels of selected Ca2+ binding myofilament proteins, including cTnC (cardiac troponin C), a major cytosolic Ca2+ buffer, were significantly lower in patients with persAF. Small interfering RNA (siRNA)-mediated knockdown of cTnC (si-cTNC) in atrial iPSC-CM phenocopied the reduced cytosolic Ca2+ buffering observed in persAF. Si-cTnC treated atrial iPSC-CM exhibited a higher predisposition to spontaneous Ca2+ release events and developed action potential alternans at low stimulation frequencies. Last, indirect reduction of cytosolic Ca2+ buffering using blebbistatin in an ex vivo mouse whole heart model increased vulnerability to tachypacing-induced atrial arrhythmia, validating the direct mechanistic link between impaired cytosolic Ca2+ buffering and atrial arrhythmogenesis. CONCLUSIONS Our findings suggest that loss of myofilament proteins, particularly reduced cTnC protein levels, causes diminished cytosolic Ca2+ buffering in persAF, thereby potentiating the occurrence of spontaneous Ca2+ release events and atrial fibrillation susceptibility. Strategies targeting intracellular buffering may represent a promising therapeutic lead in persAF management.
Collapse
Affiliation(s)
- Funsho E. Fakuade
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Dominik Hubricht
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Vanessa Möller
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Izzatullo Sobitov
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Aiste Liutkute
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Yannic Döring
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Fitzwilliam Seibertz
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Marcus Gerloff
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Julius Ryan D. Pronto
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Fereshteh Haghighi
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Sören Brandenburg
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Khaled Alhussini
- Department of Thoracic and Cardiovascular Surgery (K.A., C.B.), University Clinic Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Nadezda Ignatyeva
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Yara Bonhoff
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Stefanie Kestel
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Aschraf El-Essawi
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
- Department of Thoracic and Cardiovascular Surgery, Klinikum Braunschweig, Germany (A.E.-E.)
| | - Ahmad Fawad Jebran
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Marius Großmann
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Bernhard C. Danner
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Hassina Baraki
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Constanze Schmidt
- Department of Cardiology, University Hospital Heidelberg, Germany (C.S.)
- German Center for Cardiovascular Research Partner Site Heidelberg/Mannheim, Heidelberg University (C.S.)
| | - Samuel Sossalla
- Department of Cardiology, University Hospital Giessen & Kerckhoff Clinic, Germany (S.S.)
- Department of Cardiology, Bad Nauheim & German Center for Cardiovascular Research Partner Site Rhine-Main, Germany (S.S.)
| | - Ingo Kutschka
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Constanze Bening
- Department of Thoracic and Cardiovascular Surgery (K.A., C.B.), University Clinic Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center Würzburg (K.A., C.B., C.M.), University Clinic Würzburg, Germany
| | - Wolfgang A. Linke
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
- Institute of Physiology II, University of Münster, Germany (W.A.L.)
| | - Jordi Heijman
- Gottfried Schatz Research Center, Division of Medical Physics and Biophysics, Medical University of Graz, Austria (J.H.)
- Department of Cardiology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands (J.H.)
| | - Stephan E. Lehnart
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - George Kensah
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Thoracic and Cardiovascular Surgery (F.H., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., G.K.), University Medical Center Göttingen, Germany
| | - Antje Ebert
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Department of Cardiology and Pneumology (S.B., N.I., W.A.L., S.E.L., A.E.), Heart Research Center Göttingen, University Medical Center Göttingen, Germany
| | - Fleur E. Mason
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| | - Niels Voigt
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (F.E.F., A.L., F.S., F.H., S.E.L., A.E., N.V.), Georg-August-University Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Germany (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., F.H., S.B., N.I., Y.B., S.K., A.E.-E., A.F.J., M. Großmann, B.C.D., H.B., I.K., W.A.L., S.E.L., G.K., A.E., F.E.M., N.V.)
- Institute of Pharmacology and Toxicology (F.E.F., D.H., V.M., I.S., A.L., Y.D., F.S., M. Gerloff, J.R.D.P., Y.B., S.K., F.E.M., N.V.), University Medical Center Göttingen, Germany
| |
Collapse
|
3
|
Dewenter M, Seitz T, Steinbrecher JH, Westenbrink BD, Ling H, Lehnart SE, Wehrens XH, Backs J, Brown JH, Maier LS, Neef S. Ca2+/calmodulin-dependent kinase IIδC-induced chronic heart failure does not depend on sarcoplasmic reticulum Ca2+ leak. ESC Heart Fail 2024; 11:2191-2199. [PMID: 38616546 PMCID: PMC11287324 DOI: 10.1002/ehf2.14772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 03/07/2024] [Indexed: 04/16/2024] Open
Abstract
AIMS Hyperactivity of Ca2+/calmodulin-dependent protein kinase II (CaMKII) has emerged as a central cause of pathologic remodelling in heart failure. It has been suggested that CaMKII-induced hyperphosphorylation of the ryanodine receptor 2 (RyR2) and consequently increased diastolic Ca2+ leak from the sarcoplasmic reticulum (SR) is a crucial mechanism by which increased CaMKII activity leads to contractile dysfunction. We aim to evaluate the relevance of CaMKII-dependent RyR2 phosphorylation for CaMKII-induced heart failure development in vivo. METHODS AND RESULTS We crossbred CaMKIIδC overexpressing [transgenic (TG)] mice with RyR2-S2814A knock-in mice that are resistant to CaMKII-dependent RyR2 phosphorylation. Ca2+-spark measurements on isolated ventricular myocytes confirmed the severe diastolic SR Ca2+ leak previously reported in CaMKIIδC TG [4.65 ± 0.73 mF/F0 vs. 1.88 ± 0.30 mF/F0 in wild type (WT)]. Crossing in the S2814A mutation completely prevented SR Ca2+-leak induction in the CaMKIIδC TG, both regarding Ca2+-spark size and frequency, demonstrating that the CaMKIIδC-induced SR Ca2+ leak entirely depends on the CaMKII-specific RyR2-S2814 phosphorylation. Yet, the RyR2-S2814A mutation did not affect the massive contractile dysfunction (ejection fraction = 12.17 ± 2.05% vs. 45.15 ± 3.46% in WT), cardiac hypertrophy (heart weight/tibia length = 24.84 ± 3.00 vs. 9.81 ± 0.50 mg/mm in WT), or severe premature mortality (median survival of 12 weeks) associated with cardiac CaMKIIδC overexpression. In the face of a prevented SR Ca2+ leak, the phosphorylation status of other critical CaMKII downstream targets that can drive heart failure, including transcriptional regulator histone deacetylase 4, as well as markers of pathological gene expression including Xirp2, Il6, and Col1a1, was equally increased in hearts from CaMKIIδC TG on a RyR WT and S2814A background. CONCLUSIONS S2814 phosphoresistance of RyR2 prevents the CaMKII-dependent SR Ca2+ leak induction but does not prevent the cardiomyopathic phenotype caused by enhanced CaMKIIδC activity. Our data indicate that additional mechanisms-independent of SR Ca2+ leak-are critical for the maladaptive effects of chronically increased CaMKIIδC activity with respect to heart failure.
Collapse
Affiliation(s)
- Matthias Dewenter
- Medical Faculty Heidelberg, Institute of Experimental CardiologyHeidelberg UniversityHeidelbergGermany
- Department of Internal Medicine 8Heidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Sites Heidelberg/Mannheim and GöttingenHeidelberg/Mannheim and GöttingenGermany
| | - Tilmann Seitz
- DZHK (German Centre for Cardiovascular Research), Partner Sites Heidelberg/Mannheim and GöttingenHeidelberg/Mannheim and GöttingenGermany
- Department of Cardiology and Pneumology, University Medical Center Göttingen (UMG)Georg August University of GöttingenGöttingenGermany
| | - Julia H. Steinbrecher
- DZHK (German Centre for Cardiovascular Research), Partner Sites Heidelberg/Mannheim and GöttingenHeidelberg/Mannheim and GöttingenGermany
- Department of Cardiology and Pneumology, University Medical Center Göttingen (UMG)Georg August University of GöttingenGöttingenGermany
| | - B. Daan Westenbrink
- Department of PharmacologyUniversity of California San DiegoSan DiegoCAUSA
- Department of Cardiology, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Haiyun Ling
- Department of PharmacologyUniversity of California San DiegoSan DiegoCAUSA
- Genomics Institute of the Novartis Research FoundationSan DiegoCAUSA
| | - Stephan E. Lehnart
- DZHK (German Centre for Cardiovascular Research), Partner Sites Heidelberg/Mannheim and GöttingenHeidelberg/Mannheim and GöttingenGermany
- Department of Cardiology and Pneumology, University Medical Center Göttingen (UMG)Georg August University of GöttingenGöttingenGermany
| | - Xander H.T. Wehrens
- Cardiovascular Research Institute and Department of Molecular Physiology and BiophysicsBaylor College of MedicineHoustonTXUSA
| | - Johannes Backs
- Medical Faculty Heidelberg, Institute of Experimental CardiologyHeidelberg UniversityHeidelbergGermany
- Department of Internal Medicine 8Heidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner Sites Heidelberg/Mannheim and GöttingenHeidelberg/Mannheim and GöttingenGermany
- Molecular Medicine Partnership UnitHeidelberg University & EMBLHeidelbergGermany
- Helmholtz Institute for Translational AngioCardioScience (HI‐TAC)—a branch of the MDC at Heidelberg UniversityHeidelbergGermany
| | - Joan Heller Brown
- Department of PharmacologyUniversity of California San DiegoSan DiegoCAUSA
| | - Lars S. Maier
- Department of Internal Medicine IIUniversity Hospital RegensburgFranz‐Josef‐Strauss‐Allee 11RegensburgGermany
| | - Stefan Neef
- Department of Internal Medicine IIUniversity Hospital RegensburgFranz‐Josef‐Strauss‐Allee 11RegensburgGermany
| |
Collapse
|
4
|
Chen J, Qin H, Hao J, Wang Q, Chen S, Yang G, Li M, Zhu X, Wang D, Chen H, Cui C, Chen M. Cardiac-specific overexpression of CREM-IbΔC-X via CRISPR/Cas9 in mice presents a new model of atrial cardiomyopathy with spontaneous atrial fibrillation. Transl Res 2024; 267:54-66. [PMID: 38199433 DOI: 10.1016/j.trsl.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/13/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Atrial cardiomyopathy (ACM) forms the substrate for atrial fibrillation (AF) and underlies the potential for atrial thrombus formation and subsequent stroke. However, generating stable animal models that accurately replicate the entire progression of atrial lesions, particularly the onset of AF, presents significant challenges. In the present study, we found that the isoform of CRE-binding protein modulator (CREM-IbΔC-X), which is involved in the regulation of cardiac development and atrial rhythm, was highly expressed in atrial biopsies from patients with AF. Building upon this finding, we employed CRISPR/Cas9 technology to create a mouse model with cardiac-specific overexpression of CREM-IbΔC-X (referred to as CS-CREM mice). This animal model effectively illustrated the development of ACM through electrophysiological and structural remodelings over time. Proteomics and Chip-qPCR analysis of atrial samples revealed significant upregulation of cell-matrix adhesion and extracellular matrix structural components, alongside significant downregulation of genes related to atrial functions in the CS-CREM mice. Furthermore, the corresponding responses to anti-arrhythmia drugs, i.e., amiodarone and propafenone, suggested that CS-CREM mice could serve as an ideal in vivo model for drug testing. Our study introduced a novel ACM model with spontaneous AF by cardiac-specifically overexpressing CREM-IbΔC-X in mice, providing valuable insights into the mechanisms and therapeutic targets of ACM.
Collapse
Affiliation(s)
- Jiuzhou Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Huiyuan Qin
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Jingzhe Hao
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Qing Wang
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Shaojie Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Gang Yang
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Mingfang Li
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| | - Xiyu Zhu
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Dongjin Wang
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hongwu Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China.
| | - Chang Cui
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China.
| | - Minglong Chen
- Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
5
|
Zhao S, Hulsurkar MM, Lahiri SK, Aguilar-Sanchez Y, Munivez E, Müller FU, Jain A, Malovannaya A, Yiu CHK, Reilly S, Wehrens XHT. Atrial proteomic profiling reveals a switch towards profibrotic gene expression program in CREM-IbΔC-X mice with persistent atrial fibrillation. J Mol Cell Cardiol 2024; 190:1-12. [PMID: 38514002 DOI: 10.1016/j.yjmcc.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/19/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Overexpression of the CREM (cAMP response element-binding modulator) isoform CREM-IbΔC-X in transgenic mice (CREM-Tg) causes the age-dependent development of spontaneous AF. PURPOSE To identify key proteome signatures and biological processes accompanying the development of persistent AF through integrated proteomics and bioinformatics analysis. METHODS Atrial tissue samples from three CREM-Tg mice and three wild-type littermates were subjected to unbiased mass spectrometry-based quantitative proteomics, differential expression and pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. RESULTS A total of 98 differentially expressed proteins were identified. Gene ontology analysis revealed enrichment for biological processes regulating actin cytoskeleton organization and extracellular matrix (ECM) dynamics. Changes in ITGAV, FBLN5, and LCP1 were identified as being relevant to atrial fibrosis and structural based on expression changes, co-expression patterns, and PPI network analysis. Comparative analysis with previously published datasets revealed a shift in protein expression patterns from ion-channel and metabolic regulators in young CREM-Tg mice to profibrotic remodeling factors in older CREM-Tg mice. Furthermore, older CREM-Tg mice exhibited protein expression patterns reminiscent of those seen in humans with persistent AF. CONCLUSIONS This study uncovered distinct temporal changes in atrial protein expression patterns with age in CREM-Tg mice consistent with the progressive evolution of AF. Future studies into the role of the key differentially abundant proteins identified in this study in AF progression may open new therapeutic avenues to control atrial fibrosis and substrate development in AF.
Collapse
Affiliation(s)
- Shuai Zhao
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohit M Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Satadru K Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuriana Aguilar-Sanchez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elda Munivez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA; Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chi Him Kendrick Yiu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA; Center for Space Medicine, Baylor College of Medicine, Houston, USA.
| |
Collapse
|
6
|
Mehdizadeh M, Naud P, Abu-Taha IH, Hiram R, Xiong F, Xiao J, Saljic A, Kamler M, Vuong-Robillard N, Thorin E, Ferbeyre G, Tardif JC, Sirois MG, Tanguay JF, Dobrev D, Nattel S. The role of cellular senescence in profibrillatory atrial remodelling associated with cardiac pathology. Cardiovasc Res 2024; 120:506-518. [PMID: 38181429 PMCID: PMC11060482 DOI: 10.1093/cvr/cvae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/21/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
AIMS Cellular senescence is a stress-related or aging response believed to contribute to many cardiac conditions; however, its role in atrial fibrillation (AF) is unknown. Age is the single most important determinant of the risk of AF. The present study was designed to (i) evaluate AF susceptibility and senescence marker expression in rat models of aging and myocardial infarction (MI), (ii) study the effect of reducing senescent-cell burden with senolytic therapy on the atrial substrate in MI rats, and (iii) assess senescence markers in human atrial tissue as a function of age and the presence of AF. METHODS AND RESULTS AF susceptibility was studied with programmed electrical stimulation. Gene and protein expression was evaluated by immunoblot or immunofluorescence (protein) and digital polymerase chain reaction (PCR) or reverse transcriptase quantitative PCR (messenger RNA). A previously validated senolytic combination, dasatinib and quercetin, (D+Q; or corresponding vehicle) was administered from the time of sham or MI surgery through 28 days later. Experiments were performed blinded to treatment assignment. Burst pacing-induced AF was seen in 100% of aged (18-month old) rats, 87.5% of young MI rats, and 10% of young control (3-month old) rats (P ≤ 0.001 vs. each). Conduction velocity was slower in aged [both left atrium (LA) and right atrium (RA)] and young MI (LA) rats vs. young control rats (P ≤ 0.001 vs. each). Atrial fibrosis was greater in aged (LA and RA) and young MI (LA) vs. young control rats (P < 0.05 for each). Senolytic therapy reduced AF inducibility in MI rats (from 8/9 rats, 89% in MI vehicle, to 0/9 rats, 0% in MI D + Q, P < 0.001) and attenuated LA fibrosis. Double staining suggested that D + Q acts by clearing senescent myofibroblasts and endothelial cells. In human atria, senescence markers were upregulated in older (≥70 years) and long-standing AF patients vs. individuals ≤60 and sinus rhythm controls, respectively. CONCLUSION Our results point to a potentially significant role of cellular senescence in AF pathophysiology. Modulating cell senescence might provide a basis for novel therapeutic approaches to AF.
Collapse
Affiliation(s)
- Mozhdeh Mehdizadeh
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, Quebec H3G 1Y6, Canada
| | - Patrice Naud
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada
| | - Issam H Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, Essen 45122, Germany
| | - Roddy Hiram
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada
| | - Feng Xiong
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
| | - Jiening Xiao
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
| | - Arnela Saljic
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, Essen 45122, Germany
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Norregade 10, P.O. Box 2177, Copenhagen, Denmark
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, University Hospital Essen, Hufelandstrasse 55, Essen 45122, Germany
| | - Nhung Vuong-Robillard
- Department of Biochemistry, Université de Montréal, CRCHUM, 900 Saint Denis St, Montreal, Quebec H2X 0A9, Canada
| | - Eric Thorin
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
- Department of Surgery, Université de Montréal, Pavillon Roger-Gaudry, Montreal, Quebec H3C 3J7, Canada
| | - Gerardo Ferbeyre
- Department of Biochemistry, Université de Montréal, CRCHUM, 900 Saint Denis St, Montreal, Quebec H2X 0A9, Canada
| | - Jean-Claude Tardif
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
| | - Martin G Sirois
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Pavillon Roger-GaudryOffice S-436, 2900 boulevard Édouard-Montpetit, Montreal, Quebec H3T 1J4, Canada
| | - Jean Francois Tanguay
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
| | - Dobromir Dobrev
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, Essen 45122, Germany
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Stanley Nattel
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, Quebec H1T 1C8, Canada
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, Quebec H3G 1Y6, Canada
- Department of Medicine, Université de Montréal, Pavillon Roger-Gaudry, 2900 Edouard Montpetit Blvd, Montreal, Quebec H3T 1J4, Canada
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Hufelandstrasse 55, Essen 45122, Germany
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Pavillon Roger-GaudryOffice S-436, 2900 boulevard Édouard-Montpetit, Montreal, Quebec H3T 1J4, Canada
- IHU Liryc and Fondation Bordeaux Université, 166 cours de l’Argonne, Bordeaux 33000, France
| |
Collapse
|
7
|
Arabia G, Bellicini MG, Cersosimo A, Memo M, Mazzarotto F, Inciardi RM, Cerini M, Chen LY, Aboelhassan M, Benzoni P, Mitacchione G, Bontempi L, Curnis A. Ion channel dysfunction and fibrosis in atrial fibrillation: Two sides of the same coin. Pacing Clin Electrophysiol 2024; 47:417-428. [PMID: 38375940 DOI: 10.1111/pace.14944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/10/2024] [Accepted: 01/23/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) is a common heart rhythm disorder that is associated with an increased risk of stroke and heart failure (HF). Initially, an association between AF and ion channel dysfunction was identified, classifying the pathology as a predominantly electrical disease. More recently it has been recognized that fibrosis and structural atrial remodeling play a driving role in the development of this arrhythmia also in these cases. PURPOSE Understanding the role of fibrosis in genetic determined AF could be important to better comprise the pathophysiology of this arrhythmia and to refine its management also in nongenetic forms. In this review we analyze genetic and epigenetic mechanisms responsible for AF and their link with atrial fibrosis, then we will consider analogies with the pathophysiological mechanism in nongenetic AF, and discuss consequent therapeutic options.
Collapse
Affiliation(s)
- Gianmarco Arabia
- Cardiology Department, Spedali Civili Hospital, University of Brescia, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Angelica Cersosimo
- Cardiology Department, Spedali Civili Hospital, University of Brescia, Brescia, Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- National Heart and Lung Institute, Imperial College London (F.M., J. Ware), London, UK
| | | | - Manuel Cerini
- Cardiology Department, Spedali Civili Hospital, University of Brescia, Brescia, Italy
| | - Lin Yee Chen
- University of Minnesota (L.Y.C.), Minneapolis, USA
| | | | - Patrizia Benzoni
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | | | - Luca Bontempi
- Unit of Cardiology, Cardiac Electrophysiology and, Electrostimulation Laboratory, "Bolognini" Hospital of Seriate - ASST Bergamo Est, Bergamo, Italy
| | - Antonio Curnis
- Cardiology Department, Spedali Civili Hospital, University of Brescia, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
8
|
Gong Q, LE X, Yu P, Zhuang L. Therapeutic advances in atrial fibrillation based on animal models. J Zhejiang Univ Sci B 2024; 25:135-152. [PMID: 38303497 PMCID: PMC10835209 DOI: 10.1631/jzus.b2300285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/14/2023] [Indexed: 02/03/2024]
Abstract
Atrial fibrillation (AF) is the most prevalent sustained cardiac arrhythmia among humans, with its incidence increasing significantly with age. Despite the high frequency of AF in clinical practice, its etiology and management remain elusive. To develop effective treatment strategies, it is imperative to comprehend the underlying mechanisms of AF; therefore, the establishment of animal models of AF is vital to explore its pathogenesis. While spontaneous AF is rare in most animal species, several large animal models, particularly those of pigs, dogs, and horses, have proven as invaluable in recent years in advancing our knowledge of AF pathogenesis and developing novel therapeutic options. This review aims to provide a comprehensive discussion of various animal models of AF, with an emphasis on the unique features of each model and its utility in AF research and treatment. The data summarized in this review provide valuable insights into the mechanisms of AF and can be used to evaluate the efficacy and safety of novel therapeutic interventions.
Collapse
Affiliation(s)
- Qian Gong
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Xuan LE
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Pengcheng Yu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Lenan Zhuang
- Institute of Genetics and Reproduction, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
9
|
Navarro-Garcia JA, Bruns F, Moore OM, Tekook MA, Dobrev D, Miyake CY, Wehrens XH. In Vivo Cardiac Electrophysiology in Mice: Determination of Atrial and Ventricular Arrhythmic Substrates. Curr Protoc 2024; 4:e994. [PMID: 38372479 PMCID: PMC10883620 DOI: 10.1002/cpz1.994] [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: 02/20/2024]
Abstract
Cardiac arrhythmias are a common cardiac condition that might lead to fatal outcomes. A better understanding of the molecular and cellular basis of arrhythmia mechanisms is necessary for the development of better treatment modalities. To aid these efforts, various mouse models have been developed for studying cardiac arrhythmias. Both genetic and surgical mouse models are commonly used to assess the incidence and mechanisms of arrhythmias. Since spontaneous arrhythmias are uncommon in healthy young mice, intracardiac programmed electrical stimulation (PES) can be performed to assess the susceptibility to pacing-induced arrhythmias and uncover the possible presence of a proarrhythmogenic substrate. This procedure is performed by positioning an octopolar catheter inside the right atrium and ventricle of the heart through the right jugular vein. PES can provide insights into atrial and ventricular electrical activity and reveal whether atrial and/or ventricular arrhythmias are present or can be induced. Here, we explain detailed procedures used to perform this technique, possible troubleshooting scenarios, and methods to interpret the results obtained. © 2024 Wiley Periodicals LLC. Basic Protocol: Programmed electrical stimulation in mice.
Collapse
Affiliation(s)
- Jose Alberto Navarro-Garcia
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, Houston, USA
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, USA
| | - Florian Bruns
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Oliver M. Moore
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, Houston, USA
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, USA
| | - Marcel A. Tekook
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, USA
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal
| | - Christina Y. Miyake
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, Houston, USA
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, USA
- Department of Pediatrics, Division of Cardiology, Baylor College of Medicine, Houston, US
| | - Xander H.T. Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, Houston, USA
- Department of Integrative Physiology, Baylor College of Medicine, One Baylor Plaza, Houston, USA
- Department of Medicine, Division of Cardiology, Baylor College of Medicine, Houston, US
- Department of Neuroscience, Baylor College of Medicine, Houston, US
- Department of Pediatrics, Division of Cardiology, Baylor College of Medicine, Houston, US
- Center for Space Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
10
|
Zhao S, Hulsurkar MM, Lahiri SK, Aguilar-Sanchez Y, Munivez E, Müller FU, Jain A, Malovannaya A, Yiu K, Reilly S, Wehrens XH. Atrial Proteomic Profiling Reveals a Switch Towards Profibrotic Gene Expression Program in CREM-IbΔC-X Mice with Persistent Atrial Fibrillation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575097. [PMID: 38260363 PMCID: PMC10802622 DOI: 10.1101/2024.01.10.575097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Overexpression of the CREM (cAMP response element-binding modulator) isoform CREM-IbΔC-X in transgenic mice (CREM-Tg) causes the age-dependent development of spontaneous AF. Purpose To identify key proteome signatures and biological processes accompanying the development of persistent AF through integrated proteomics and bioinformatics analysis. Methods Atrial tissue samples from three CREM-Tg mice and three wild-type littermates were subjected to unbiased mass spectrometry-based quantitative proteomics, differential expression and pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. Results A total of 98 differentially expressed proteins were identified. Gene ontology analysis revealed enrichment for biological processes regulating actin cytoskeleton organization and extracellular matrix (ECM) dynamics. Changes in ITGAV, FBLN5, and LCP1 were identified as being relevant to atrial fibrosis and remodeling based on expression changes, co-expression patterns, and PPI network analysis. Comparative analysis with previously published datasets revealed a shift in protein expression patterns from ion-channel and metabolic regulators in young CREM-Tg mice to profibrotic remodeling factors in older CREM-Tg mice. Furthermore, older CREM-Tg mice exhibited protein expression patterns that resembled those of humans with persistent AF. Conclusions This study uncovered distinct temporal changes in atrial protein expression patterns with age in CREM-Tg mice consistent with the progressive evolution of AF. Future studies into the role of the key differentially abundant proteins identified in this study in AF progression may open new therapeutic avenues to control atrial fibrosis and substrate development in AF.
Collapse
Affiliation(s)
- Shuai Zhao
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mohit M. Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Satadru K. Lahiri
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuriana Aguilar-Sanchez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elda Munivez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
- Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kendrick Yiu
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, NIHR Oxford BRC, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Xander H.T. Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics (in Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, USA
| |
Collapse
|
11
|
Yang H, Zhu J, Fu H, Shuai W. Dapansutrile Ameliorates Atrial Inflammation and Vulnerability to Atrial Fibrillation in HFpEF Rats. Heart Lung Circ 2024; 33:65-77. [PMID: 38040503 DOI: 10.1016/j.hlc.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND Numerous studies have demonstrated that NLRP3 inflammasomes are key players in the progression of atrial fibrillation (AF) in heart failure with preserved ejection fraction (HFpEF). This study aimed to analyse the effect of pharmacological inhibition of NLRP3 inflammasomes using dapansutrile (DAPA), an oral NLRP3-specific inhibitor. METHODS Dahl salt-sensitive rats were fed a high-salt diet (HSD, 8% NaCl) to induce HFpEF. Either DAPA (200 mg/kg/day) or saline was administered daily via gavage for 4 weeks. Electrophysiological studies were performed to assess the AF inducibility. Confocal fluorescence microscopy and western blot analysis were used to study calcium handling. RESULTS The DAPA-treated HFpEF rats were less prone to AF induction by programmed electrical stimulation. Atrial fibrosis and inflammation were attenuated in DAPA-treated HFpEF hearts. Dapansutrile treatment showed an increase in the Ca2+ transient sarcoplasmic reticulum-Ca2+ load, and protein expression of SERCA2; NCX1 and phosphorylation of PLB at Thr17 were decreased following DAPA treatment. The increased frequency of spontaneous Ca2+ spark in the HFpEF rats was related to the hyperphosphorylation of RyR2 at Ser2814, which was blunted in DAPA treatment. Dapansutrile treatment also decreased the phosphorylation of CaMKII expression in the HFpEF rats. Mechanistically, DAPA exerts an anti-arrhythmic effect, mainly by inhibiting activation of the NLRP3 inflammasome. CONCLUSION These data provide evidence that the beneficial cardiac effects of DAPA are associated with reduced atrial inflammation and improved CaMKII-dependent Ca2+-handling abnormalities via blunting activation of the NLRP3 inflammasome, and DAPA may be beneficial in a rat model of HFpEF-induced AF.
Collapse
Affiliation(s)
- Hongjie Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hui Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
12
|
Ni H, Morotti S, Zhang X, Dobrev D, Grandi E. Integrative human atrial modelling unravels interactive protein kinase A and Ca2+/calmodulin-dependent protein kinase II signalling as key determinants of atrial arrhythmogenesis. Cardiovasc Res 2023; 119:2294-2311. [PMID: 37523735 PMCID: PMC11318383 DOI: 10.1093/cvr/cvad118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/18/2023] [Accepted: 06/05/2023] [Indexed: 08/02/2023] Open
Abstract
AIMS Atrial fibrillation (AF), the most prevalent clinical arrhythmia, is associated with atrial remodelling manifesting as acute and chronic alterations in expression, function, and regulation of atrial electrophysiological and Ca2+-handling processes. These AF-induced modifications crosstalk and propagate across spatial scales creating a complex pathophysiological network, which renders AF resistant to existing pharmacotherapies that predominantly target transmembrane ion channels. Developing innovative therapeutic strategies requires a systems approach to disentangle quantitatively the pro-arrhythmic contributions of individual AF-induced alterations. METHODS AND RESULTS Here, we built a novel computational framework for simulating electrophysiology and Ca2+-handling in human atrial cardiomyocytes and tissues, and their regulation by key upstream signalling pathways [i.e. protein kinase A (PKA), and Ca2+/calmodulin-dependent protein kinase II (CaMKII)] involved in AF-pathogenesis. Populations of atrial cardiomyocyte models were constructed to determine the influence of subcellular ionic processes, signalling components, and regulatory networks on atrial arrhythmogenesis. Our results reveal a novel synergistic crosstalk between PKA and CaMKII that promotes atrial cardiomyocyte electrical instability and arrhythmogenic triggered activity. Simulations of heterogeneous tissue demonstrate that this cellular triggered activity is further amplified by CaMKII- and PKA-dependent alterations of tissue properties, further exacerbating atrial arrhythmogenesis. CONCLUSIONS Our analysis reveals potential mechanisms by which the stress-associated adaptive changes turn into maladaptive pro-arrhythmic triggers at the cellular and tissue levels and identifies potential anti-AF targets. Collectively, our integrative approach is powerful and instrumental to assemble and reconcile existing knowledge into a systems network for identifying novel anti-AF targets and innovative approaches moving beyond the traditional ion channel-based strategy.
Collapse
Affiliation(s)
- Haibo Ni
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Xianwei Zhang
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, Faculty of Medicine, University
Duisburg-Essen, Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and
Université de Montréal, Montréal, Canada
- Department of Molecular Physiology and Biophysics, Baylor College of
Medicine, Houston, TX, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| |
Collapse
|
13
|
Ramos-Mondragón R, Lozhkin A, Vendrov AE, Runge MS, Isom LL, Madamanchi NR. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1833. [PMID: 37891912 PMCID: PMC10604902 DOI: 10.3390/antiox12101833] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and its prevalence increases with age. The irregular and rapid contraction of the atria can lead to ineffective blood pumping, local blood stasis, blood clots, ischemic stroke, and heart failure. NADPH oxidases (NOX) and mitochondria are the main sources of reactive oxygen species in the heart, and dysregulated activation of NOX and mitochondrial dysfunction are associated with AF pathogenesis. NOX- and mitochondria-derived oxidative stress contribute to the onset of paroxysmal AF by inducing electrophysiological changes in atrial myocytes and structural remodeling in the atria. Because high atrial activity causes cardiac myocytes to expend extremely high energy to maintain excitation-contraction coupling during persistent AF, mitochondria, the primary energy source, undergo metabolic stress, affecting their morphology, Ca2+ handling, and ATP generation. In this review, we discuss the role of oxidative stress in activating AF-triggered activities, regulating intracellular Ca2+ handling, and functional and anatomical reentry mechanisms, all of which are associated with AF initiation, perpetuation, and progression. Changes in the extracellular matrix, inflammation, ion channel expression and function, myofibril structure, and mitochondrial function occur during the early transitional stages of AF, opening a window of opportunity to target NOX and mitochondria-derived oxidative stress using isoform-specific NOX inhibitors and mitochondrial ROS scavengers, as well as drugs that improve mitochondrial dynamics and metabolism to treat persistent AF and its transition to permanent AF.
Collapse
Affiliation(s)
- Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Aleksandr E. Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Marschall S. Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| |
Collapse
|
14
|
George SA, Brennan-McLean JA, Trampel KA, Rytkin E, Faye NR, Knollmann BC, Efimov IR. Ryanodine receptor inhibition with acute dantrolene treatment reduces arrhythmia susceptibility in human hearts. Am J Physiol Heart Circ Physiol 2023; 325:H720-H728. [PMID: 37566110 DOI: 10.1152/ajpheart.00103.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Ryanodine receptor 2 (RyR2) hyperactivity is observed in structural heart diseases that are a result of ischemia or heart failure. It causes abnormal calcium handling and calcium leaks that cause metabolic, electrical, and mechanical dysfunction, which can trigger arrhythmias. Here, we tested the antiarrhythmic potential of dantrolene (RyR inhibitor) in human hearts. Human hearts not used in transplantation were obtained, and right ventricular outflow tract (RVOT) wedges and left ventricular (LV) slices were prepared. Pseudo-ECGs were recorded to determine premature ventricular contraction (PVC) incidences. Optical mapping was performed to determine arrhythmogenic substrates. After baseline optical recordings, tissues were treated with 1) isoproterenol (250 nM), 2) caffeine (200 mM), and 3) dantrolene (2 or 10 mM). Optical recordings were obtained after each treatment. Isoproterenol and caffeine treatment increased PVC incidence, whereas dantrolene reduced the PVC burden. Isoproterenol shortened action potential duration (APD) in the RV, RVOT, and LV regions and shortened calcium transient duration (CaTD) in the LV. Caffeine further shortened APD in the RV, did not modulate APD in the RVOT, and prolonged APD in the LV. In addition, in the LV, CaTD prolongation was also observed. More importantly, adding dantrolene did not alter APD in the RV or RVOT regions but produced a trend toward APD prolongation and significant CaTD prolongation in the LV, restoring these parameters to baseline values. In conclusions, dantrolene treatment suppresses triggers and reverses arrhythmogenic substrates in the human heart and could be a novel antiarrhythmic therapy in patients with structural heart disease.NEW & NOTEWORTHY Ryanodine receptor 2 hyperactivity is observed in structural heart diseases caused by ischemia or heart failure. It causes abnormal calcium leaks, which can trigger arrhythmias. To prevent arrhythmias, we applied dantrolene in human hearts ex vivo. Isoproterenol and caffeine treatment increased PVC incidence, whereas dantrolene reduced the PVC burden. Dantrolene treatment suppresses triggers and reverses arrhythmogenic substrates and could be a novel antiarrhythmic therapy in patients with structural heart disease.
Collapse
Affiliation(s)
- Sharon A George
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - Jaclyn A Brennan-McLean
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
| | - Katy A Trampel
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - Eric Rytkin
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - N Rokhaya Faye
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Igor R Efimov
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
15
|
Shi S, Mao X, Lv J, Wang Y, Zhang X, Shou X, Zhang B, Li Y, Wu H, Song Q, Hu Y. Qi-Po-Sheng-Mai granule ameliorates Ach-CaCl 2 -induced atrial fibrillation by regulating calcium homeostasis in cardiomyocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:155017. [PMID: 37597360 DOI: 10.1016/j.phymed.2023.155017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/15/2023] [Accepted: 08/06/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is one of the most common arrhythmias encountered in clinical settings. Currently, the pathophysiology of AF remains unclear, which severely limits the effectiveness and safety of medical therapies. The Chinese herbal formula Qi-Po-Sheng-Mai Granule (QPSM) has been widely used in China to treat AF. However, its pharmacological and molecular mechanisms remain unknown. PURPOSE The purpose of this study was to investigate the molecular mechanisms and potential targets of QPSM for AF. STUDY DESIGN AND METHODS The AF model was induced by Ach (66 μg/ml) and CaCl2 (10 mg/kg), and the dose of 0.1 ml/100 g was injected into the tail vein for 5 weeks. QPSM was administered daily at doses of 4.42 and 8.84 g/kg, and amiodarone (0.18 g/kg) was used as the positive control. The effect of QPSM on AF was assessed by electrocardiogram, echocardiography, and histopathological analysis. Then, we employed network pharmacology with single nucleus RNA sequencing (snRNA-Seq) to investigate the molecular mechanisms and potential targets of QPSM for AF. Furthermore, high performance liquid chromatography (HPLC) method was used for component analysis of QPSM, and molecular docking was used to verify the potential targets. Using the IonOptix single cell contraction and ion synchronization test equipment, single myocyte length and calcium ion variations were observed in real time. The expression levels of calcium Transporter-related proteins were detected by western blot and immunohistochemistry. RESULTS Based on an Ach-CaCl2-induced AF model, we found that QPSM treatment significantly reduced atrial electrical remodeling-related markers, such as AF inducibility and duration, and attenuated atrial dilation and fibrosis. Network pharmacology identified 52 active ingredients and 119 potential targets for QPSM in the treatment of AF, and 45 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were enriched, among which calcium pathway had the greatest impact. Using single nucleus sequencing (snRNA-seq), we identified cardiomyocytes as the most differentially expressed in response to drug treatment, with nine differentially expressed genes enriched in calcium signaling pathways. High performance liquid chromatography and molecular docking confirmed that the core components of QPSM strongly bind to the key factors in the calcium signaling pathway. Additional experiments have shown that QPSM increases calcium transients (CaT) and contractility in the individual cardiomyocyte. This was accomplished by increasing the expression of CACNA1C and SERCA2a and decreasing the expression of CAMK2B and NCX1. CONCLUSION The present study has systematically elucidated the role of QPSM in maintaining calcium homeostasis in cardiomyocytes through the regulation of calcium transporters, which could lead to new drug development ideas for AF.
Collapse
Affiliation(s)
- Shuqing Shi
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Xinxin Mao
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Jiayu Lv
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Yajiao Wang
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Xuesong Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xintian Shou
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Yumeng Li
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Huaqin Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingqiao Song
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China.
| | - Yuanhui Hu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
16
|
Schmeckpeper J, Kim K, George SA, Blackwell DJ, Brennan JA, Efimov IR, Knollmann BC. RyR2 inhibition with dantrolene is antiarrhythmic, prevents further pathological remodeling, and improves cardiac function in chronic ischemic heart disease. J Mol Cell Cardiol 2023; 181:67-78. [PMID: 37285929 PMCID: PMC10526741 DOI: 10.1016/j.yjmcc.2023.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/30/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Diastolic Ca2+ leak due to cardiac ryanodine receptor (RyR2) hyperactivity has been widely documented in chronic ischemic heart disease (CIHD) and may contribute to ventricular tachycardia (VT) risk and progressive left-ventricular (LV) remodeling. Here we test the hypothesis that targeting RyR2 hyperactivity can suppress VT inducibility and progressive heart failure in CIHD by the RyR2 inhibitor dantrolene. METHODS AND RESULTS: CIHD was induced in C57BL/6 J mice by left coronary artery ligation. Four weeks later, mice were randomized to either acute or chronic (6 weeks via implanted osmotic pump) treatment with dantrolene or vehicle. VT inducibility was assessed by programmed stimulation in vivo and in isolated hearts. Electrical substrate remodeling was assessed by optical mapping. Ca2+ sparks and spontaneous Ca2+ releases were measured in isolated cardiomyocytes. Cardiac remodeling was quantified by histology and qRT-PCR. Cardiac function and contractility were measured using echocardiography. Compared to vehicle, acute dantrolene treatment reduced VT inducibility. Optical mapping demonstrated reentrant VT prevention by dantrolene, which normalized the shortened refractory period (VERP) and prolonged action potential duration (APD), preventing APD alternans. In single CIHD cardiomyocytes, dantrolene normalized RyR2 hyperactivity and prevented spontaneous intracellular Ca2+ release. Chronic dantrolene treatment not only reduced VT inducibility but also reduced peri-infarct fibrosis and prevented further progression of LV dysfunction in CIHD mice. CONCLUSIONS: RyR2 hyperactivity plays a mechanistic role for VT risk, post-infarct remodeling, and contractile dysfunction in CIHD mice. Our data provide proof of concept for the anti-arrhythmic and anti-remodeling efficacy of dantrolene in CIHD.
Collapse
Affiliation(s)
- Jeffrey Schmeckpeper
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kyungsoo Kim
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sharon A George
- Department of Biomedical Engineering, the George Washington University, Washington DC, USA; Department of Biomedical Engineering, Northwestern University, Chicago, IL, USA
| | - Daniel J Blackwell
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jaclyn A Brennan
- Department of Biomedical Engineering, the George Washington University, Washington DC, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, the George Washington University, Washington DC, USA; Department of Biomedical Engineering, Northwestern University, Chicago, IL, USA
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
17
|
Voigt N, Seibertz F, Fakuade FE. Vicious Twins: RyR2 Dysfunction and Structural Remodeling. Circ Res 2023; 133:193-195. [PMID: 37410856 DOI: 10.1161/circresaha.123.323144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Affiliation(s)
- Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Germany (N.V., F.S., F.E.F.)
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (N.V., F.S., F.E.F.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (N.V., F.S., F.E.F.)
| | - Fitzwilliam Seibertz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Germany (N.V., F.S., F.E.F.)
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (N.V., F.S., F.E.F.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (N.V., F.S., F.E.F.)
| | - Funsho E Fakuade
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Germany (N.V., F.S., F.E.F.)
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany (N.V., F.S., F.E.F.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany (N.V., F.S., F.E.F.)
| |
Collapse
|
18
|
Gaido OER, Pavlaki N, Granger JM, Mesubi OO, Liu B, Lin BL, Long A, Walker D, Mayourian J, Schole KL, Terrillion CE, Nkashama LJ, Hulsurkar MM, Dorn LE, Ferrero KM, Huganir RL, Müller FU, Wehrens XHT, Liu JO, Luczak ED, Bezzerides VJ, Anderson ME. An improved reporter identifies ruxolitinib as a potent and cardioprotective CaMKII inhibitor. Sci Transl Med 2023; 15:eabq7839. [PMID: 37343080 PMCID: PMC11022683 DOI: 10.1126/scitranslmed.abq7839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 05/31/2023] [Indexed: 06/23/2023]
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) hyperactivity causes cardiac arrhythmias, a major source of morbidity and mortality worldwide. Despite proven benefits of CaMKII inhibition in numerous preclinical models of heart disease, translation of CaMKII antagonists into humans has been stymied by low potency, toxicity, and an enduring concern for adverse effects on cognition due to an established role of CaMKII in learning and memory. To address these challenges, we asked whether any clinically approved drugs, developed for other purposes, were potent CaMKII inhibitors. For this, we engineered an improved fluorescent reporter, CaMKAR (CaMKII activity reporter), which features superior sensitivity, kinetics, and tractability for high-throughput screening. Using this tool, we carried out a drug repurposing screen (4475 compounds in clinical use) in human cells expressing constitutively active CaMKII. This yielded five previously unrecognized CaMKII inhibitors with clinically relevant potency: ruxolitinib, baricitinib, silmitasertib, crenolanib, and abemaciclib. We found that ruxolitinib, an orally bioavailable and U.S. Food and Drug Administration-approved medication, inhibited CaMKII in cultured cardiomyocytes and in mice. Ruxolitinib abolished arrhythmogenesis in mouse and patient-derived models of CaMKII-driven arrhythmias. A 10-min pretreatment in vivo was sufficient to prevent catecholaminergic polymorphic ventricular tachycardia, a congenital source of pediatric cardiac arrest, and rescue atrial fibrillation, the most common clinical arrhythmia. At cardioprotective doses, ruxolitinib-treated mice did not show any adverse effects in established cognitive assays. Our results support further clinical investigation of ruxolitinib as a potential treatment for cardiac indications.
Collapse
Affiliation(s)
- Oscar E. Reyes Gaido
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nikoleta Pavlaki
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan M. Granger
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Olurotimi O. Mesubi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bian Liu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Brian L. Lin
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alan Long
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David Walker
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Joshua Mayourian
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kate L. Schole
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chantelle E. Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lubika J. Nkashama
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mohit M. Hulsurkar
- Cardiovascular Research Institute and Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lauren E. Dorn
- Cardiovascular Research Institute and Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kimberly M. Ferrero
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L. Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Frank U. Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster 48149, Germany
| | - Xander H. T. Wehrens
- Cardiovascular Research Institute and Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Medicine, Neuroscience, and Pediatrics, Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth D. Luczak
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Vassilios J. Bezzerides
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mark E. Anderson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Biological Sciences and the Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
19
|
Liu X, Ren L, Yu S, Li G, He P, Yang Q, Wei X, Thai PN, Wu L, Huo Y. Late sodium current in synergism with Ca 2+/calmodulin-dependent protein kinase II contributes to β-adrenergic activation-induced atrial fibrillation. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220163. [PMID: 37122215 PMCID: PMC10150221 DOI: 10.1098/rstb.2022.0163] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Atrial fibrillation (AF) is frequently associated with β-adrenergic stimulation, especially in patients with structural heart diseases. The objective of this study was to determine the synergism of late sodium current (late INa) and Ca2+/calmodulin-dependent protein kinase (CaMKII)-mediated arrhythmogenic activities in β-adrenergic overactivation-associated AF. Monophasic action potential, conduction properties, protein phosphorylation, ion currents and cellular trigger activities were measured from rabbit-isolated hearts, atrial tissue and atrial myocytes, respectively. Isoproterenol (ISO, 1-15 nM) increased atrial conduction inhomogeneity index, phospho-Nav1.5 and phospho-CaMKII protein levels and late INa by 108%, 65%, 135% and 87%, respectively, and induced triggered activities and episodes of AF in all hearts studied (p < 0.05). Sea anemone toxin II (ATX-II, 2 nM) was insufficient to induce any atrial arrhythmias, whereas the propensities of AF were greater in hearts treated with a combination of ATX-II and ISO. Ranolazine, eleclazine and KN-93 abolished ISO-induced AF, attenuated the phosphorylation of Nav1.5 and CaMKII, and reversed the increase of late INa (p < 0.05) in a synergistic mode. Overall, late INa in association with the activation of CaMKII potentiates β-adrenergic stimulation-induced AF and the inhibition of both late INa and CaMKII exerted synergistic anti-arrhythmic effects to suppress atrial arrhythmic activities associated with catecholaminergic activation. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Lu Ren
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Shandong Yu
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Gang Li
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
| | - Pengkang He
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
| | - Qiaomei Yang
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
| | - Xiaohong Wei
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Lin Wu
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, 8, Xishiku Street, West District, Beijing 100034, People's Republic of China
| |
Collapse
|
20
|
Keefe JA, Hulsurkar MM, Reilly S, Wehrens XHT. Mouse models of spontaneous atrial fibrillation. Mamm Genome 2023; 34:298-311. [PMID: 36173465 PMCID: PMC10898345 DOI: 10.1007/s00335-022-09964-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in adults, with a prevalence increasing with age. Current clinical management of AF is focused on tertiary prevention (i.e., treating the symptoms and sequelae) rather than addressing the underlying molecular pathophysiology. Robust animal models of AF, particularly those that do not require supraphysiologic stimuli to induce AF (i.e., showing spontaneous AF), enable studies that can uncover the underlying mechanisms of AF. Several mouse models of AF have been described to exhibit spontaneous AF, but pathophysiologic drivers of AF differ among models. Here, we describe relevant AF mechanisms and provide an overview of large and small animal models of AF. We then provide an in-depth review of the spontaneous mouse models of AF, highlighting the relevant AF mechanisms for each model.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mohit M Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
Ezeani M, Prabhu S. PI3K(p110α) as a determinant and gene therapy for atrial enlargement in atrial fibrillation. Mol Cell Biochem 2023; 478:471-490. [PMID: 35900667 PMCID: PMC9938077 DOI: 10.1007/s11010-022-04526-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/05/2022] [Indexed: 11/28/2022]
Abstract
Atrial fibrillation (AF) is an irregular heart rhythm, characterised by chaotic atrial activation, which is promoted by remodelling. Once initiated, AF can also propagate the progression of itself in the so-called ''AF begets AF''. Several lines of investigation have shown that signalling molecules, including reactive oxygen species, angiotensin II, and phosphoinositide 3-kinases (PI3Ks), in presence or absence of cardiovascular disease risk factors, stabilise and promote AF maintenance. In particular, reduced cardiac-specific PI3K activity that is not associated with oncology is cardiotoxic and increases susceptibility to AF. Atrial-specific PI3K(p110α) transgene can cause pathological atrial enlargement. Highlighting the crucial importance of the p110α protein in a clinical problem that currently challenges the professional health care practice, in over forty (40) transgenic mouse models of AF (Table1), currently existing, of which some of the models are models of human genetic disorders, including PI3K(p110α) transgenic mouse model, over 70% of them reporting atrial size showed enlarged, greater atrial size. Individuals with minimal to severely dilated atria develop AF more likely. Left atrial diameter and volume stratification are an assessment for follow-up surveillance to detect AF. Gene therapy to reduce atrial size will be associated with a reduction in AF burden. In this overview, PI3K(p110α), a master regulator of organ size, was investigated in atrial enlargement and in physiological determinants that promote AF.
Collapse
Affiliation(s)
- Martin Ezeani
- NanoBiotechnology Laboratory, Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, 3004, Australia.
| | - Sandeep Prabhu
- The Alfred, and Baker Heart and Diabetes Institute, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
| |
Collapse
|
22
|
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
|
23
|
Zhu M, Yan T, Zhu S, Weng F, Zhu K, Wang C, Guo C. Identification and verification of FN1, P4HA1 and CREBBP as potential biomarkers in human atrial fibrillation. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:6947-6965. [PMID: 37161136 DOI: 10.3934/mbe.2023300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is a common arrhythmia that can lead to cardiac complications. The mechanisms involved in AF remain elusive. We aimed to explore the potential biomarkers and mechanisms underpinning AF. METHODS An independent dataset, GSE2240, was obtained from the Gene Expression Omnibus database. The R package, "limma", was used to screen for differentially expressed genes (DEGs) in individuals with AF and normal sinus rhythm (SR). Weighted gene co-expression network analysis (WGCNA) was applied to cluster DEGs into different modules based on functional disparities. Enrichment analyses were performed using the Database for Annotation, Visualization and Integrated Discovery. A protein-protein interaction network was constructed, and hub genes were identified using cytoHubba. Quantitative reverse-transcription PCR was used to validate mRNA expression in individuals with AF and SR. RESULTS We identified 2, 589 DEGs clustered into 10 modules using WGCNA. Gene Ontology analysis showed specific clustered genes significantly enriched in pathways associated with the extracellular matrix and collagen organization. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that the target genes were mainly enriched for proteoglycans in cancer, extracellular matrix-receptor interaction, focal adhesion, and the PI3K-Akt signaling pathway. Three hub genes, FN1, P4HA1 and CREBBP, were identified, which were highly correlated with AF endogenesis. mRNA expression of hub genes in patients with AF were higher than in individuals with normal SR, consistent with the results of bioinformatics analysis. CONCLUSIONS FN1, P4HA1, and CREBBP may play critical roles in AF. Using bioinformatics, we found that expression of these genes was significantly elevated in patients with AF than in individuals with normal SR. Furthermore, these genes were elevated at core positions in the mRNA interaction network. These genes should be further explored as novel biomarkers and target candidates for AF therapy.
Collapse
Affiliation(s)
- Miao Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Tao Yan
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Shijie Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Fan Weng
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Kai Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Chunsheng Wang
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Changfa Guo
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
24
|
Potter AS, Hulsukar MM, Wu L, Narasimhan B, Karimzad K, Koutroumpakis E, Palaskas N, Deswal A, Kantharia BK, Wehrens XH. Kinase Inhibitors and Atrial Fibrillation. JACC Clin Electrophysiol 2023; 9:591-602. [PMID: 37100538 DOI: 10.1016/j.jacep.2022.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/13/2022] [Accepted: 11/30/2022] [Indexed: 02/24/2023]
Abstract
Recent advances have significantly expanded the options of available therapeutics for cancer treatment, including novel targeted cancer therapies. Within this broad category of targeted therapies is the class of kinase inhibitors (KIs), which target kinases that have undergone aberrant activation in cancerous cells. Although KIs have shown a benefit in treating various forms of malignancy, they have also been shown to cause a wide array of cardiovascular toxicities, with cardiac arrhythmias, in particular atrial fibrillation (AF), being 1 of the predominant side effects. The occurrence of AF in patients undergoing cancer treatment can complicate the treatment approach and poses unique clinical challenges. The association of KIs and AF has led to new research aimed at trying to elucidate the underlying mechanisms. Furthermore, there are unique considerations to treating KI-induced AF because of the anticoagulant properties of some KIs as well as drug-drug interactions with KIs and some cardiovascular medications. Here, we review the current literature pertaining to KI-induced AF.
Collapse
|
25
|
Jiménez-Sábado V, Casabella-Ramón S, Llach A, Gich I, Casellas S, Ciruela F, Chen SRW, Guerra JM, Ginel A, Benítez R, Cinca J, Tarifa C, Hove-Madsen L. Beta-blocker treatment of patients with atrial fibrillation attenuates spontaneous calcium release-induced electrical activity. Biomed Pharmacother 2023; 158:114169. [PMID: 36592495 DOI: 10.1016/j.biopha.2022.114169] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
AIMS Atrial fibrillation (AF) has been associated with excessive spontaneous calcium release, linked to cyclic AMP (cAMP)-dependent phosphorylation of calcium regulatory proteins. Because β-blockers are expected to attenuate cAMP-dependent signaling, we aimed to examine whether the treatment of patients with β-blockers affected the incidence of spontaneous calcium release events or transient inward currents (ITI). METHODS The impact of treatment with commonly used β-blockers was analyzed in human atrial myocytes from 371 patients using patch-clamp technique, confocal calcium imaging or immunofluorescent labeling. Data were analyzed using multivariate regression analysis taking into account potentially confounding effects of relevant clinical factors RESULTS: The L-type calcium current (ICa) density was diminished significantly in patients with chronic but not paroxysmal AF and the treatment of patients with β-blockers did not affect ICa density in any group. By contrast, the ITI frequency was elevated in patients with either paroxysmal or chronic AF that did not receive treatment, and β-blocker treatment reduced the frequency to levels observed in patients without AF. Confocal calcium imaging showed that β-blocker treatment also reduced the calcium spark frequency in patients with AF to levels observed in those without AF. Furthermore, phosphorylation of the ryanodine receptor (RyR2) at Ser-2808 and phospholamban at Ser-16 was significantly lower in patients with AF that received β-blockers. CONCLUSION Together, our findings demonstrate that β-blocker treatment may be of therapeutic utility to prevent spontaneous calcium release-induced atrial electrical activity; especially in patients with a history of paroxysmal AF displaying preserved ICa density.
Collapse
Affiliation(s)
- Verónica Jiménez-Sábado
- CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Sergi Casabella-Ramón
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain; Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Llach
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ignasi Gich
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Casellas
- Servicio de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Dept. Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, The Libin Cardiovascular Institute, University of Calgary, Canada
| | - José M Guerra
- CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Servicio de Cardiología and Univ. Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Antonino Ginel
- Servicio de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Raúl Benítez
- Dept. d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Univ. Politècnica de Catalunya, Barcelona, Spain
| | - Juan Cinca
- Servicio de Cardiología and Univ. Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Carmen Tarifa
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - Leif Hove-Madsen
- CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain.
| |
Collapse
|
26
|
Guillot B, Boileve A, Walton R, Harfoush A, Conte C, Sainte-Marie Y, Charron S, Bernus O, Recalde A, Sallé L, Brette F, Lezoualc'h F. Inhibition of EPAC1 signaling pathway alters atrial electrophysiology and prevents atrial fibrillation. Front Physiol 2023; 14:1120336. [PMID: 36909224 PMCID: PMC9992743 DOI: 10.3389/fphys.2023.1120336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Introduction: Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and is associated with increased mortality and morbidity. The Exchange Protein directly Activated by cAMP (EPAC), has been implicated in pro-arrhythmic signaling pathways in the atria, but the underlying mechanisms remain unknown. Methods: In this study, we investigated the involvement of EPAC1 and EPAC2 isoforms in the genesis of AF in wild type (WT) mice and knockout (KO) mice for EPAC1 or EPAC2. We also employed EPAC pharmacological modulators to selectively activate EPAC proteins (8-CPT-AM; 10 μM), or inhibit either EPAC1 (AM-001; 20 μM) or EPAC2 (ESI-05; 25 μM). Transesophageal stimulation was used to characterize the induction of AF in vivo in mice. Optical mapping experiments were performed on isolated mouse atria and cellular electrophysiology was examined by whole-cell patch-clamp technique. Results: In wild type mice, we found 8-CPT-AM slightly increased AF susceptibility and that this was blocked by the EPAC1 inhibitor AM-001 but not the EPAC2 inhibitor ESI-05. Consistent with this, in EPAC1 KO mice, occurrence of AF was observed in 3/12 (vs. 4/10 WT littermates) and 4/10 in EPAC2 KO (vs. 5/10 WT littermates). In wild type animals, optical mapping experiments revealed that 8-CPT-AM perfusion increased action potential duration even in the presence of AM-001 or ESI-05. Interestingly, 8-CPT-AM perfusion decreased conduction velocity, an effect blunted by AM-001 but not ESI-05. Patch-clamp experiments demonstrated action potential prolongation after 8-CPT-AM perfusion in both wild type and EPAC1 KO mice and this effect was partially prevented by AM-001 in WT. Conclusion: Together, these results indicate that EPAC1 and EPAC2 signaling pathways differentially alter atrial electrophysiology but only the EPAC1 isoform is involved in the genesis of AF. Selective blockade of EPAC1 with AM-001 prevents AF in mice.
Collapse
Affiliation(s)
- Bastien Guillot
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Arthur Boileve
- UR 4650 PSIR, GIP Cyceron, Caen, France.,Université de Caen-Normandie, Caen, France
| | - Richard Walton
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Alexandre Harfoush
- UR 4650 PSIR, GIP Cyceron, Caen, France.,Université de Caen-Normandie, Caen, France
| | - Caroline Conte
- Université de Toulouse-Paul Sabatier, Toulouse, France.,Institut des maladies métaboliques et cardiovasculaires, INSERM UMR-1297, Toulouse, France
| | - Yannis Sainte-Marie
- Université de Toulouse-Paul Sabatier, Toulouse, France.,Institut des maladies métaboliques et cardiovasculaires, INSERM UMR-1297, Toulouse, France
| | - Sabine Charron
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Olivier Bernus
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Alice Recalde
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Laurent Sallé
- UR 4650 PSIR, GIP Cyceron, Caen, France.,Université de Caen-Normandie, Caen, France
| | - Fabien Brette
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France.,PhyMedExp, INSERM U1046, CNRS 9412, Université de Montpellier, Montpellier, France
| | - Frank Lezoualc'h
- Université de Toulouse-Paul Sabatier, Toulouse, France.,Institut des maladies métaboliques et cardiovasculaires, INSERM UMR-1297, Toulouse, France
| |
Collapse
|
27
|
Fan W, Sun X, Yang C, Wan J, Luo H, Liao B. Pacemaker activity and ion channels in the sinoatrial node cells: MicroRNAs and arrhythmia. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:151-167. [PMID: 36450332 DOI: 10.1016/j.pbiomolbio.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/13/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
The primary pacemaking activity of the heart is determined by a spontaneous action potential (AP) within sinoatrial node (SAN) cells. This unique AP generation relies on two mechanisms: membrane clocks and calcium clocks. Nonhomologous arrhythmias are caused by several functional and structural changes in the myocardium. MicroRNAs (miRNAs) are essential regulators of gene expression in cardiomyocytes. These miRNAs play a vital role in regulating the stability of cardiac conduction and in the remodeling process that leads to arrhythmias. Although it remains unclear how miRNAs regulate the expression and function of ion channels in the heart, these regulatory mechanisms may support the development of emerging therapies. This study discusses the spread and generation of AP in the SAN as well as the regulation of miRNAs and individual ion channels. Arrhythmogenicity studies on ion channels will provide a research basis for miRNA modulation as a new therapeutic target.
Collapse
Affiliation(s)
- Wei Fan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Xuemei Sun
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Chao Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| | - Hongli Luo
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| | - Bin Liao
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| |
Collapse
|
28
|
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
|
29
|
RCAN1 deficiency aggravates sepsis-induced cardiac remodeling and dysfunction by accelerating mitochondrial pathological fission. Inflamm Res 2022; 71:1589-1602. [DOI: 10.1007/s00011-022-01628-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/05/2022] Open
|
30
|
Gillis AM, Dobrev D. Targeting the RyR2 to Prevent Atrial Fibrillation. Circ Arrhythm Electrophysiol 2022; 15:e011514. [PMID: 36178743 PMCID: PMC9592734 DOI: 10.1161/circep.122.011514] [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)
- Anne M. Gillis
- Department of Cardiac Sciences, University of Calgary and Libin Cardiovascular Institute, Calgary, Alberta, Canada
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
31
|
Shoemaker MB, Yoneda ZT, Crawford DM, Akers WS, Richardson T, Montgomery JA, Phillips S, Shyr Y, Saavedra P, Estrada J, Kanagasundram A, Shen ST, Michaud G, Crossley G, Ellis CR, Knollmann BC. A Mechanistic Clinical Trial Using ( R)- Versus (S)-Propafenone to Test RyR2 (Ryanodine Receptor) Inhibition for the Prevention of Atrial Fibrillation Induction. Circ Arrhythm Electrophysiol 2022; 15:e010713. [PMID: 36166682 PMCID: PMC9588733 DOI: 10.1161/circep.121.010713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 08/16/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Experimental data suggest ryanodine receptor-mediated intracellular calcium leak is a mechanism for atrial fibrillation (AF), but evidence in humans is still needed. Propafenone is composed of two enantiomers that are equally potent sodium-channel blockers; however, (R)-propafenone is an ryanodine receptor inhibitor whereas (S)-propafenone is not. This study tested the hypothesis that ryanodine receptor inhibition with (R)-propafenone prevents induction of AF compared to (S)-propafenone or placebo in patients referred for AF ablation. METHODS Participants were randomized 4:4:1 to a one-time intravenous dose of (R)-propafenone, (S)-propafenone, or placebo. The study drug was given at the start of the procedure and an AF induction protocol using rapid atrial pacing was performed before ablation. The primary endpoint was 30 s of AF or atrial flutter. RESULTS A total of 193 participants were enrolled and 165 (85%) completed the study protocol (median age: 63 years, 58% male, 95% paroxysmal AF). Sustained AF and/or atrial flutter was induced in 60 participants (84.5%) receiving (R)-propafenone, 60 (80.0%) receiving (S)-propafenone group, and 12 (63.2%) receiving placebo. Atrial flutter occurred significantly more often in the (R)-propafenone (N=23, 32.4%) and (S)-propafenone (N=26, 34.7%) groups compared to placebo (N=1, 5.3%, P=0.029). There was no significant difference between (R)-propafenone and (S)-propafenone for the primary outcome of AF and/or atrial flutter induction in univariable (P=0.522) or multivariable analysis (P=0.199, adjusted for age and serum drug level). CONCLUSIONS There is no difference in AF inducibility between (R)-propafenone and (S)-propafenone at clinically relevant concentrations. These results are confounded by a high rate of inducible atrial flutter due to sodium-channel blockade. REGISTRATION https://clinicaltrials.gov; Unique Identifier: NCT02710669.
Collapse
Affiliation(s)
- M. Benjamin Shoemaker
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Zachary T. Yoneda
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Diane M. Crawford
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Wendell S. Akers
- Department of Pharmacology, Vanderbilt University School of Medicine
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, TN
| | - Travis Richardson
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Jay A. Montgomery
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Sharon Phillips
- Department of Biostatistics, Vanderbilt University School of Medicine
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University School of Medicine
| | - Pablo Saavedra
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - J.C. Estrada
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Arvindh Kanagasundram
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Sharon T. Shen
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Greg Michaud
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - George Crossley
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | - Christopher R. Ellis
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center
| | | |
Collapse
|
32
|
Xie D, Xiong K, Su X, Wang G, Wang L, Zou Q, Zhang C, Cao Y, Liu Y, Chen YH. Memantine targets glutamate receptors in atrial cardiomyocytes to prevent and treat atrial fibrillation. Cell Discov 2022; 8:76. [PMID: 35918317 PMCID: PMC9345967 DOI: 10.1038/s41421-022-00429-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/02/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Duanyang Xie
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Ke Xiong
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Xuling Su
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Guanghua Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Luxin Wang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Qicheng Zou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Caihong Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yuting Cao
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yi Liu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China
- Institute of Medical Genetics, Tongji University, Shanghai, China
| | - Yi-Han Chen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China.
- Institute of Medical Genetics, Tongji University, Shanghai, China.
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, Shanghai, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
33
|
Ziprasidone Induces Rabbit Atrium Arrhythmogenesis via Modification of Oxidative Stress and Sodium/Calcium Homeostasis. Biomedicines 2022; 10:biomedicines10050976. [PMID: 35625713 PMCID: PMC9138982 DOI: 10.3390/biomedicines10050976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Atypical antipsychotics increase the risk of atrial arrhythmias and sudden cardiac death. This study investigated whether ziprasidone, a second-generation antipsychotic, affected intracellular Ca2+ and Na+ regulation and oxidative stress, providing proarrhythmogenic substrates in atriums. Methods: Electromechanical analyses of rabbit atrial tissues were conducted. Intracellular Ca2+ monitoring using Fluo-3, the patch-clamp method for ionic current recordings, and a fluorescence study for the detection of reactive oxygen species and intracellular Na+ levels were conducted in enzymatically dissociated atrial myocytes. Results: Ziprasidone-treated atriums showed sustained triggered activities after rapid pacing, which were inhibited by KN-93 and ranolazine. A reduced peak L-type Ca2+ channel current and enhanced late Na+ current were observed in ziprasidone-treated atrial myocytes, together with an increased cytosolic Na+ level. KN-93 suppressed the enhanced late Na+ current in ziprasidone-treated atrial myocytes. Atrial myocytes treated with ziprasidone showed reduced Ca2+ transient amplitudes and sarcoplasmic reticulum (SR) Ca2+ stores, and increased SR Ca2+ leakage. Cytosolic and mitochondrial reactive oxygen species production was increased in atrial myocytes treated with ziprasidone. TNF-α and NLRP3 were upregulated in ziprasidone-treated myocytes, and the level of phosphorylated calcium/calmodulin-dependent protein kinase II protein was increased. Conclusions: Our results suggest that ziprasidone increases the occurrence of atrial triggered activity and causes intracellular Ca2+ and Na+ dysregulation, which may result from enhanced oxidative stress and activation of the TNF-α/NLRP3 inflammasome pathway in ziprasidone-treated myocytes.
Collapse
|
34
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia despite substantial efforts to understand the pathophysiology of the condition and develop improved treatments. Identifying the underlying causative mechanisms of AF in individual patients is difficult and the efficacy of current therapies is suboptimal. Consequently, the incidence of AF is steadily rising and there is a pressing need for novel therapies. Research has revealed that defects in specific molecular pathways underlie AF pathogenesis, resulting in electrical conduction disorders that drive AF. The severity of this so-called electropathology correlates with the stage of AF disease progression and determines the response to AF treatment. Therefore, unravelling the molecular mechanisms underlying electropathology is expected to fuel the development of innovative personalized diagnostic tools and mechanism-based therapies. Moreover, the co-creation of AF studies with patients to implement novel diagnostic tools and therapies is a prerequisite for successful personalized AF management. Currently, various treatment modalities targeting AF-related electropathology, including lifestyle changes, pharmaceutical and nutraceutical therapy, substrate-based ablative therapy, and neuromodulation, are available to maintain sinus rhythm and might offer a novel holistic strategy to treat AF.
Collapse
Affiliation(s)
- Bianca J J M Brundel
- Department of Physiology, Amsterdam University Medical Centers, VU Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.
| | - Xun Ai
- Department of Physiology and Cell Biology, College of Medicine/Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - Myrthe F Kuipers
- AFIPonline.org, Atrial Fibrillation Innovation Platform, Amsterdam, Netherlands
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
35
|
Research Progress on Natural Products’ Therapeutic Effects on Atrial Fibrillation by Regulating Ion Channels. Cardiovasc Ther 2022; 2022:4559809. [PMID: 35387267 PMCID: PMC8964196 DOI: 10.1155/2022/4559809] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 11/18/2022] Open
Abstract
Antiarrhythmic drugs (AADs) have a therapeutic effect on atrial fibrillation (AF) by regulating the function of ion channels. However, several adverse effects and high recurrence rates after drug withdrawal seriously affect patients’ medication compliance and clinical prognosis. Thus, safer and more effective drugs are urgently needed. Active components extracted from natural products are potential choices for AF therapy. Natural products like Panax notoginseng (Burk.) F.H. Chen, Sophora flavescens Ait., Stephania tetrandra S. Moore., Pueraria lobata (Willd.) Ohwi var. thomsonii (Benth.) Vaniot der Maesen., and Coptis chinensis Franch. have a long history in the treatment of arrhythmia, myocardial infarction, stroke, and heart failure in China. Based on the classification of chemical structures, this article discussed the natural product components’ therapeutic effects on atrial fibrillation by regulating ion channels, connexins, and expression of related genes, in order to provide a reference for development of therapeutic drugs for atrial fibrillation.
Collapse
|
36
|
Dixon RE. Nanoscale Organization, Regulation, and Dynamic Reorganization of Cardiac Calcium Channels. Front Physiol 2022; 12:810408. [PMID: 35069264 PMCID: PMC8769284 DOI: 10.3389/fphys.2021.810408] [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: 11/06/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
The architectural specializations and targeted delivery pathways of cardiomyocytes ensure that L-type Ca2+ channels (CaV1.2) are concentrated on the t-tubule sarcolemma within nanometers of their intracellular partners the type 2 ryanodine receptors (RyR2) which cluster on the junctional sarcoplasmic reticulum (jSR). The organization and distribution of these two groups of cardiac calcium channel clusters critically underlies the uniform contraction of the myocardium. Ca2+ signaling between these two sets of adjacent clusters produces Ca2+ sparks that in health, cannot escalate into Ca2+ waves because there is sufficient separation of adjacent clusters so that the release of Ca2+ from one RyR2 cluster or supercluster, cannot activate and sustain the release of Ca2+ from neighboring clusters. Instead, thousands of these Ca2+ release units (CRUs) generate near simultaneous Ca2+ sparks across every cardiomyocyte during the action potential when calcium induced calcium release from RyR2 is stimulated by depolarization induced Ca2+ influx through voltage dependent CaV1.2 channel clusters. These sparks summate to generate a global Ca2+ transient that activates the myofilaments and thus the electrical signal of the action potential is transduced into a functional output, myocardial contraction. To generate more, or less contractile force to match the hemodynamic and metabolic demands of the body, the heart responds to β-adrenergic signaling by altering activity of calcium channels to tune excitation-contraction coupling accordingly. Recent accumulating evidence suggests that this tuning process also involves altered expression, and dynamic reorganization of CaV1.2 and RyR2 channels on their respective membranes to control the amplitude of Ca2+ entry, SR Ca2+ release and myocardial function. In heart failure and aging, altered distribution and reorganization of these key Ca2+ signaling proteins occurs alongside architectural remodeling and is thought to contribute to impaired contractile function. In the present review we discuss these latest developments, their implications, and future questions to be addressed.
Collapse
Affiliation(s)
- Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
37
|
Murphy MB, Kim K, Kannankeril PJ, Subati T, Van Amburg JC, Barnett JV, Murray KT. Optimizing transesophageal atrial pacing in mice to detect atrial fibrillation. Am J Physiol Heart Circ Physiol 2022; 322:H36-H43. [PMID: 34767487 PMCID: PMC8698503 DOI: 10.1152/ajpheart.00434.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 01/03/2023]
Abstract
Mice are routinely used to investigate molecular mechanisms underlying the atrial fibrillation (AF) substrate. We sought to optimize transesophageal rapid atrial pacing (RAP) protocols for the detection of AF susceptibility in mouse models. Hypertensive and control C57Bl/6J mice were subjected to burst RAP at a fixed stimulus amplitude. The role of parasympathetic involvement in pacing-related atrioventricular (AV) block and AF was examined using an intraperitoneal injection of atropine. In a crossover study, burst and decremental RAP at twice diastolic threshold were compared for induction of AV block during pacing. The efficacy of burst and decremental RAP to elicit an AF phenotype was subsequently investigated in mice deficient in the lymphocyte adaptor protein (Lnk-/-) resulting in systemic inflammation, or the paired-like homeodomain-2 transcription factor (Pitx2+/-) as a positive control. When pacing at a fixed stimulus intensity, pacing-induced AV block with AF induction occurred frequently, so that there was no difference in AF burden between hypertensive and control mice. These effects were prevented by atropine administration, implicating parasympathetic activation due to ganglionic stimulation as the etiology. When mice with AV block during pacing were eliminated from the analysis, male Lnk-/- mice displayed an AF phenotype only during burst RAP compared with controls, whereas male Pitx2+/- mice showed AF susceptibility during burst and decremental RAP. Notably, Lnk-/- and Pitx2+/- females exhibited no AF phenotype. Our data support the conclusion that multiple parameters should be used to ascertain AF inducibility and facilitate reproducibility across models and studies.NEW & NOTEWORTHY Methods were developed to optimize transesophageal rapid atrial pacing (RAP) to detect AF susceptibility in new and established mouse models. High stimulus intensity and pacing rates caused parasympathetic stimulation, with pacing-induced AV block and excessive AF induction in normal mice. For a given model, pacing at twice TH enabled improved phenotype discrimination in a pacing mode and sex-specific manner. Transesophageal RAP should be individually optimized when developing a mouse model of AF.
Collapse
Affiliation(s)
- Matthew B Murphy
- Departments of Medicine, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Kyungsoo Kim
- Departments of Medicine, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Prince J Kannankeril
- Departments of Medicine, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Tuerdi Subati
- Departments of Medicine, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Joseph C Van Amburg
- Departments of Medicine, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Joey V Barnett
- Departments of Medicine, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Katherine T Murray
- Departments of Medicine, Pediatrics, and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
38
|
Function and regulation of phosphatase 1 in healthy and diseased heart. Cell Signal 2021; 90:110203. [PMID: 34822978 DOI: 10.1016/j.cellsig.2021.110203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022]
Abstract
Reversible phosphorylation of ion channels and calcium-handling proteins provides precise post-translational regulation of cardiac excitation and contractility. Serine/threonine phosphatases govern dephosphorylation of the majority of cardiac proteins. Accordingly, dysfunction of this regulation contributes to the development and progression of heart failure and atrial fibrillation. On the molecular level, these changes include alterations in the expression level and phosphorylation status of Ca2+ handling and excitation-contraction coupling proteins provoked by dysregulation of phosphatases. The serine/threonine protein phosphatase PP1 is one a major player in the regulation of cardiac excitation-contraction coupling. PP1 essentially impacts on cardiac physiology and pathophysiology via interactions with the cardiac ion channels Cav1.2, NKA, NCX and KCNQ1, sarcoplasmic reticulum-bound Ca2+ handling proteins such as RyR2, SERCA and PLB as well as the contractile proteins MLC2, TnI and MyBP-C. PP1 itself but also PP1-regulatory proteins like inhibitor-1, inhibitor-2 and heat-shock protein 20 are dysregulated in cardiac disease. Therefore, they represent interesting targets to gain more insights in heart pathophysiology and to identify new treatment strategies for patients with heart failure or atrial fibrillation. We describe the genetic and holoenzymatic structure of PP1 and review its role in the heart and cardiac disease. Finally, we highlight the importance of the PP1 regulatory proteins for disease manifestation, provide an overview of genetic models to study the role of PP1 for the development of heart failure and atrial fibrillation and discuss possibilities of pharmacological interventions.
Collapse
|
39
|
Avula UMR, Dridi H, Chen BX, Yuan Q, Katchman AN, Reiken SR, Desai AD, Parsons S, Baksh H, Ma E, Dasrat P, Ji R, Lin Y, Sison C, Lederer WJ, Joca HC, Ward CW, Greiser M, Marks AR, Marx SO, Wan EY. Attenuating persistent sodium current-induced atrial myopathy and fibrillation by preventing mitochondrial oxidative stress. JCI Insight 2021; 6:e147371. [PMID: 34710060 PMCID: PMC8675199 DOI: 10.1172/jci.insight.147371] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/27/2021] [Indexed: 12/19/2022] Open
Abstract
Mechanistically driven therapies for atrial fibrillation (AF), the most common cardiac arrhythmia, are urgently needed, the development of which requires improved understanding of the cellular signaling pathways that facilitate the structural and electrophysiological remodeling that occurs in the atria. Similar to humans, increased persistent Na+ current leads to the development of an atrial myopathy and spontaneous and long-lasting episodes of AF in mice. How increased persistent Na+ current causes both structural and electrophysiological remodeling in the atria is unknown. We crossbred mice expressing human F1759A-NaV1.5 channels with mice expressing human mitochondrial catalase (mCAT). Increased expression of mCAT attenuated mitochondrial and cellular reactive oxygen species (ROS) and the structural remodeling that was induced by persistent F1759A-Na+ current. Despite the heterogeneously prolonged atrial action potential, which was unaffected by the reduction in ROS, the incidences of spontaneous AF, pacing-induced after-depolarizations, and AF were substantially reduced. Expression of mCAT markedly reduced persistent Na+ current-induced ryanodine receptor oxidation and dysfunction. In summary, increased persistent Na+ current in atrial cardiomyocytes, which is observed in patients with AF, induced atrial enlargement, fibrosis, mitochondrial dysmorphology, early after-depolarizations, and AF, all of which can be attenuated by resolving mitochondrial oxidative stress.
Collapse
Affiliation(s)
| | - Haikel Dridi
- Department of Physiology and Cellular Biophysics and Clyde & Helen Wu Center for Molecular Cardiology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Bi-xing Chen
- Division of Cardiology, Department of Medicine, and
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics and Clyde & Helen Wu Center for Molecular Cardiology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | | - Steven R. Reiken
- Department of Physiology and Cellular Biophysics and Clyde & Helen Wu Center for Molecular Cardiology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | | | | - Haajra Baksh
- Division of Cardiology, Department of Medicine, and
| | - Elaine Ma
- Division of Cardiology, Department of Medicine, and
| | | | - Ruiping Ji
- Division of Cardiology, Department of Medicine, and
| | - Yejun Lin
- Division of Cardiology, Department of Medicine, and
| | | | - W. Jonathan Lederer
- Center for Biomedical Engineering and Technology and Department of Physiology and
| | - Humberto C. Joca
- Center for Biomedical Engineering and Technology and Department of Physiology and
| | - Christopher W. Ward
- Center for Biomedical Engineering and Technology and Department of Physiology and
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Maura Greiser
- Center for Biomedical Engineering and Technology and Department of Physiology and
| | - Andrew R. Marks
- Department of Physiology and Cellular Biophysics and Clyde & Helen Wu Center for Molecular Cardiology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Steven O. Marx
- Division of Cardiology, Department of Medicine, and
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | |
Collapse
|
40
|
Ni L, Lahiri SK, Nie J, Pan X, Abu-Taha I, Reynolds JO, Campbell HM, Wang H, Kamler M, Schmitz W, Müller FU, Li N, Wei X, Wang DW, Dobrev D, Wehrens XHT. Genetic inhibition of Nuclear Factor of Activated T-cell c2 (NFATc2) prevents atrial fibrillation in CREM transgenic mice. Cardiovasc Res 2021; 118:2805-2818. [PMID: 34648001 PMCID: PMC9586567 DOI: 10.1093/cvr/cvab325] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 10/11/2021] [Indexed: 11/14/2022] Open
Abstract
AIMS Abnormal intracellular calcium handling contributes to the progressive nature of atrial fibrillation (AF), the most common sustained cardiac arrhythmia. Evidence in mouse models suggests that activation of the nuclear factor of activated T-cell (NFAT) signaling pathway contributes to atrial remodeling. Our aim was to determine the role of NFATc2 in AF in humans and mouse models. METHODS AND RESULTS Expression levels of NFATc1-c4 isoforms were assessed by quantitative reverse transcription-polymerase chain reaction in right atrial appendages from patients with chronic AF. NFATc1 and NFATc2 mRNA levels were elevated in chronic AF (cAF) patients compared with those in sinus rhythm (SR). Western blotting revealed increased cytosolic and nuclear levels of NFATc2 in AF patients. Similar findings were obtained in CREM-IbΔC-X transgenic (CREM) mice, a model of progressive AF. Telemetry ECG recordings revealed age-dependent spontaneous AF in CREM mice, which was prevented by NFATc2 knockout in CREM: NFATc2-/- mice. Programmed electrical stimulation revealed that CREM: NFATc2-/- mice lacked an AF substrate. Morphometric analysis and histology revealed increased atrial weight and atrial fibrosis in CREM mice compared with WT controls, which was reversed in CREM: NFATc2-/- mice. Confocal microscopy showed an increased Ca2+ spark frequency despite a reduced sarcoplasmic reticulum (SR) Ca2+ load in CREM mice compared with controls, whereas these abnormalities were normalized in CREM: NFATc2-/- mice. Western blotting revealed that genetic inhibition of Ca2+/calmodulin-dependent protein kinase II-mediated phosphorylation of S2814 on RyR2 in CREM: RyR2-S2814A mice suppressed NFATc2 activation observed in CREM mice, suggesting that NFATc2 is activated by excessive SR Ca2+ leak via RyR2. Finally, chromatin immunoprecipitation sequencing from AF patients identified Ras And EF-Hand Domain-Containing Protein (RASEF) as a direct target of NFATc2 mediated transcription. CONCLUSION Our findings reveal activation of the NFAT signaling pathway in patients of Chinese and European descent. NFATc2 knockout prevents the progression of AF in the CREM mouse model. TRANSLATIONAL PERSPECTIVE Atrial fibrillation (AF) is a progressive disease characterized by electrical and structural remodeling which promotes atrial arrhythmias. This study provides evidence for increased 'nuclear factor of activated T-cell' (NFAT) signaling in patients with chronic AF. Studies in the CREM transgenic model of progressive AF revealed that the NFATc2 isoform mediates atrial remodeling associated with AF substrate development. Chromatin immunoprecipitation sequencing of atrial biopsies from AF patients identified 'Ras And EF-Hand Domain-Containing Protein' (RASEF) as a downstream target of NFATc2-mediated transcription, suggesting that targeting these factors might be beneficial for curtailing AF progression.
Collapse
Affiliation(s)
- Li Ni
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Satadru K Lahiri
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xiaolu Pan
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Issam Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Julia O Reynolds
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Hannah M Campbell
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Haihao Wang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Markus Kamler
- Cardiac Surgery II Essen-Huttrop, University Hospital, West German Heart Center, University of Essen, Germany
| | - Wilhelm Schmitz
- Institute of Pharmacology and Toxicology, University of Münster, Germany
| | | | - Na Li
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA.,Institute of Pharmacology and Toxicology, University of Münster, Germany
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, 77030 USA.,Department of Medicine (Cardiology), Baylor College of Medicine, Houston, TX, 77030 USA.,Department of Pediatrics, Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030 USA
| |
Collapse
|
41
|
Yoo S, Geist GE, Pfenniger A, Rottmann M, Arora R. Recent advances in gene therapy for atrial fibrillation. J Cardiovasc Electrophysiol 2021; 32:2854-2864. [PMID: 34053133 PMCID: PMC9281901 DOI: 10.1111/jce.15116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/17/2021] [Accepted: 05/24/2021] [Indexed: 11/28/2022]
Abstract
Atrial fibrillation (AF) is the most common heart rhythm disorder in adults and a major cause of stroke. Unfortunately, current treatments for AF are suboptimal as they are not targeting the molecular mechanisms underlying AF. In this regard, gene therapy is emerging as a promising approach for mechanism-based treatment of AF. In this review, we summarize recent advances and challenges in gene therapy for this important cardiovascular disease.
Collapse
Affiliation(s)
- Shin Yoo
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Gail Elizabeth Geist
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Anna Pfenniger
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Markus Rottmann
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rishi Arora
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
42
|
Szilágyi J, Sághy L. Atrial Remodeling in Atrial Fibrillation. Comorbidities and Markers of Disease Progression Predict Catheter Ablation Outcome. Curr Cardiol Rev 2021; 17:217-229. [PMID: 32693769 PMCID: PMC8226201 DOI: 10.2174/1573403x16666200721153620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 01/19/2023] Open
Abstract
Atrial fibrillation is the most common supraventricular arrhythmia affecting an increasing proportion of the population in which mainstream therapy, i.e. catheter ablation, provides freedom from arrhythmia in only a limited number of patients. Understanding the mechanism is key in order to find more effective therapies and to improve patient selection. In this review, the structural and electrophysiological changes of the atrial musculature that constitute atrial remodeling in atrial fibrillaton and how risk factors and markers of disease progression can predict catheter ablation outcome will be discussed in detail.
Collapse
Affiliation(s)
- Judit Szilágyi
- 2nd Department of Internal Medicine and Cardiology Centre, University of Szeged, Szeged, Hungary
| | - László Sághy
- 2nd Department of Internal Medicine and Cardiology Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
43
|
Scott Jr L, Fender AC, Saljic A, Li L, Chen X, Wang X, Linz D, Lang J, Hohl M, Twomey D, Pham TT, Diaz-Lankenau R, Chelu MG, Kamler M, Entman ML, Taffet GE, Sanders P, Dobrev D, Li N. NLRP3 inflammasome is a key driver of obesity-induced atrial arrhythmias. Cardiovasc Res 2021; 117:1746-1759. [PMID: 33523143 PMCID: PMC8208743 DOI: 10.1093/cvr/cvab024] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/17/2020] [Accepted: 01/18/2021] [Indexed: 12/27/2022] Open
Abstract
AIMS Obesity, an established risk factor of atrial fibrillation (AF), is frequently associated with enhanced inflammatory response. However, whether inflammatory signaling is causally linked to AF pathogenesis in obesity remains elusive. We recently demonstrated that the constitutive activation of the 'NACHT, LRR, and PYD Domains-containing Protein 3' (NLRP3) inflammasome promotes AF susceptibility. In this study, we hypothesized that the NLRP3 inflammasome is a key driver of obesity-induced AF. METHODS AND RESULTS Western blotting was performed to determine the level of NLRP3 inflammasome activation in atrial tissues of obese patients, sheep, and diet-induced obese (DIO) mice. The increased body weight in patients, sheep, and mice was associated with enhanced NLRP3-inflammasome activation. To determine whether NLRP3 contributes to the obesity-induced atrial arrhythmogenesis, wild-type (WT) and NLRP3 homozygous knockout (NLRP3-/-) mice were subjected to high-fat-diet (HFD) or normal chow (NC) for 10 weeks. Relative to NC-fed WT mice, HFD-fed WT mice were more susceptible to pacing-induced AF with longer AF duration. In contrast, HFD-fed NLRP3-/- mice were resistant to pacing-induced AF. Optical mapping in DIO mice revealed an arrhythmogenic substrate characterized by abbreviated refractoriness and action potential duration (APD), two key determinants of reentry-promoting electrical remodeling. Upregulation of ultra-rapid delayed-rectifier K+-channel (Kv1.5) contributed to the shortening of atrial refractoriness. Increased profibrotic signaling and fibrosis along with abnormal Ca2+ release from sarcoplasmic reticulum (SR) accompanied atrial arrhythmogenesis in DIO mice. Conversely, genetic ablation of Nlrp3 (NLRP3-/-) in HFD-fed mice prevented the increases in Kv1.5 and the evolution of electrical remodeling, the upregulation of profibrotic genes, and abnormal SR Ca2+ release in DIO mice. CONCLUSION These results demonstrate that the atrial NLRP3 inflammasome is a key driver of obesity-induced atrial arrhythmogenesis and establishes a mechanistic link between obesity-induced AF and NLRP3-inflammasome activation.
Collapse
Affiliation(s)
- Larry Scott Jr
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Anke C Fender
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Arnela Saljic
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Luge Li
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiaohui Chen
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiaolei Wang
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Dominik Linz
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Jilu Lang
- Department of Cardiac Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Mathias Hohl
- Department of Cardiology/Angiology, University-Clinic of Saarland, Internal Medicine III, Homburg/Saar, Germany
| | | | - Thuy T Pham
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Rodrigo Diaz-Lankenau
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mihail G Chelu
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Markus Kamler
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Mark L Entman
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - George E Taffet
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Na Li
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
44
|
Chiang DY, Lahiri S, Wang G, Karch J, Wang MC, Jung SY, Heck AJR, Scholten A, Wehrens XHT. Phosphorylation-Dependent Interactome of Ryanodine Receptor Type 2 in the Heart. Proteomes 2021; 9:proteomes9020027. [PMID: 34200203 PMCID: PMC8293434 DOI: 10.3390/proteomes9020027] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022] Open
Abstract
Hyperphosphorylation of the calcium release channel/ryanodine receptor type 2 (RyR2) at serine 2814 (S2814) is associated with multiple cardiac diseases including atrial fibrillation and heart failure. Despite recent advances, the molecular mechanisms driving pathological changes associated with RyR2 S2814 phosphorylation are still not well understood. Methods: Using affinity-purification coupled to mass spectrometry (AP-MS), we investigated the RyR2 interactome in ventricles from wild-type (WT) mice and two S2814 knock-in mutants: the unphosphorylated alanine mutant (S2814A) and hyperphosphorylated mimic aspartic acid mutant (S2814D). Western blots were used for validation. Results: In WT mouse ventricular lysates, we identified 22 proteins which were enriched with RyR2 pull-down relative to both IgG control and no antibody (beads-only) pull-downs. Parallel AP-MS using WT, S2814A, and S2814D mouse ventricles identified 72 proteins, with 20 being high confidence RyR2 interactors. Of these, 14 had an increase in their binding to RyR2 S2814A but a decrease in their binding to RyR2 S2814D. We independently validated three protein hits, Idh3b, Aifm1, and Cpt1b, as RyR2 interactors by western blots and showed that Aifm1 and Idh3b had significantly decreased binding to RyR2 S2814D compared to WT and S2814A, consistent with MS findings. Conclusion: By applying state-of-the-art proteomic approaches, we discovered a number of novel RyR2 interactors in the mouse heart. In addition, we found and defined specific alterations in the RyR2 interactome that were dependent on the phosphorylation status of RyR2 at S2814. These findings yield mechanistic insights into RyR2 regulation which may guide future drug designs.
Collapse
Affiliation(s)
- David Y. Chiang
- Cardiovascular Division, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA 02115, USA;
| | - Satadru Lahiri
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; (S.L.); (G.W.); (J.K.)
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Guoliang Wang
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; (S.L.); (G.W.); (J.K.)
| | - Jason Karch
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; (S.L.); (G.W.); (J.K.)
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng C. Wang
- Huffington Center on Aging, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sung Y. Jung
- Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, The Netherlands; (A.J.R.H.); (A.S.)
- Netherlands Proteomics Centre, 3584 Utrecht, The Netherlands
| | - Arjen Scholten
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, The Netherlands; (A.J.R.H.); (A.S.)
- Netherlands Proteomics Centre, 3584 Utrecht, The Netherlands
| | - Xander H. T. Wehrens
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; (S.L.); (G.W.); (J.K.)
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine (Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics (Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-798-4261
| |
Collapse
|
45
|
Zhang C, Li Y, Cao J, Yu B, Zhang K, Li K, Xu X, Guo Z, Liang Y, Yang X, Yang Z, Sun Y, Kaartinen V, Ding K, Wang J. Hedgehog signalling controls sinoatrial node development and atrioventricular cushion formation. Open Biol 2021; 11:210020. [PMID: 34062094 PMCID: PMC8169207 DOI: 10.1098/rsob.210020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Smoothened is a key receptor of the hedgehog pathway, but the roles of Smoothened in cardiac development remain incompletely understood. In this study, we found that the conditional knockout of Smoothened from the mesoderm impaired the development of the venous pole of the heart and resulted in hypoplasia of the atrium/inflow tract (IFT) and a low heart rate. The blockage of Smoothened led to reduced expression of genes critical for sinoatrial node (SAN) development in the IFT. In a cardiac cell culture model, we identified a Gli2–Tbx5–Hcn4 pathway that controls SAN development. In the mutant embryos, the endocardial-to-mesenchymal transition (EndMT) in the atrioventricular cushion failed, and Bmp signalling was downregulated. The addition of Bmp2 rescued the EndMT in mutant explant cultures. Furthermore, we analysed Gli2+ scRNAseq and Tbx5−/− RNAseq data and explored the potential genes downstream of hedgehog signalling in posterior second heart field derivatives. In conclusion, our study reveals that Smoothened-mediated hedgehog signalling controls posterior cardiac progenitor commitment, which suggests that the mutation of Smoothened might be involved in the aetiology of congenital heart diseases related to the cardiac conduction system and heart valves.
Collapse
Affiliation(s)
- Chaohui Zhang
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Yuxin Li
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Jiaheng Cao
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Beibei Yu
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Kaiyue Zhang
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Ke Li
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Xinhui Xu
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Zhikun Guo
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Yinming Liang
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Institute of Lifeomics, Beijing 102206, People's Republic of China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing 210061, People's Republic of China
| | - Yunfu Sun
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, Tongji University School of Medicine, Shanghai 200120, People's Republic of China
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Keyue Ding
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou 450003, People's Republic of China
| | - Jikui Wang
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan Province, People's Republic of China
| |
Collapse
|
46
|
Chang SH, Chan YH, Chen WJ, Chang GJ, Lee JL, Yeh YH. Tachypacing-induced CREB/CD44 signaling contributes to the suppression of L-type calcium channel expression and the development of atrial remodeling. Heart Rhythm 2021; 18:1760-1771. [PMID: 34023501 DOI: 10.1016/j.hrthm.2021.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Atrial fibrillation (AF), a common arrhythmia in clinics, is characterized as downregulation of L-type calcium channel (LTCC) and shortening of atrial action potential duration (APD). Our prior studies have shown the association of CD44 with AF genesis. OBJECTIVE The purpose of this study was to explore the potential role of CD44 and its related signaling in tachypacing-induced downregulation of LTCC. METHODS AND RESULTS In vitro, tachypacing in atrium-derived myocytes (HL-1 cell line) induced activation (phosphorylation) of cyclic adenosine monophosphate response element-binding protein (CREB). Furthermore, tachypacing promoted an association between CREB and CD44 in HL-1 myocytes, which was documented in atrial tissues from patients with AF. Deletion and mutational analysis of the LTCC promoter along with chromatin immunoprecipitation revealed that cyclic adenosine monophosphate response element is essential for tachypacing-inhibited LTCC transcription. Tachypacing also hindered the binding of p-CREB to the promoter of LTCC. Blockade of CREB/CD44 signaling in HL-1 cells attenuated tachypacing-triggered downregulation of LTCC and shortening of APD. Atrial myocytes isolated from CD44-/- mice exhibited higher LTCC current and longer APD than did those from wild-type mice. Ex vivo, tachypacing caused less activation of CREB in CD44-/- mice than in wild-type mice. In vivo, burst atrial pacing stimulated less inducibility of AF in CREB inhibitor-treated mice than in controls. CONCLUSION Tachypacing-induced CREB/CD44 signaling contributes to the suppression of LTCC, which provides valuable information about the pathogenesis of atrial modeling and AF.
Collapse
Affiliation(s)
- Shang-Hung Chang
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Yi-Hsin Chan
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Wei-Jan Chen
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan.
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Tao-Yuan, Taiwan
| | - Jia-Lin Lee
- Institute of Molecular and Cellular Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yung-Hsin Yeh
- Division of Cardiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| |
Collapse
|
47
|
Dai W, Kesaraju S, Weber CR. Transcriptional factors in calcium mishandling and atrial fibrillation development. Pflugers Arch 2021; 473:1177-1197. [PMID: 34003377 DOI: 10.1007/s00424-021-02553-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/19/2021] [Accepted: 02/05/2021] [Indexed: 12/19/2022]
Abstract
Healthy cardiac conduction relies on the coordinated electrical activity of distinct populations of cardiomyocytes. Disruption of cell-cell conduction results in cardiac arrhythmias, a leading cause of morbidity and mortality worldwide. Recent genetic studies have highlighted a major heritable component and identified numerous loci associated with risk of atrial fibrillation, including transcription factor genes, particularly those important in cardiac development, microRNAs, and long noncoding RNAs. Identification of such genetic factors has prompted the search to understand the mechanisms that underlie the genetic component of AF. Recent studies have found several mechanisms by which genetic alterations can result in AF formation via disruption of calcium handling. Loss of developmental transcription factors in adult cardiomyocytes can result in disruption of SR calcium ATPase, sodium calcium exchanger, calcium channels, among other ion channels, which underlie action potential abnormalities and triggered activity that can contribute to AF. This review aims to summarize the complex network of transcription factors and their roles in calcium handling.
Collapse
Affiliation(s)
- Wenli Dai
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Sneha Kesaraju
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
48
|
Sun H, Shao Y. Transcriptome analysis reveals key pathways that vary in patients with paroxysmal and persistent atrial fibrillation. Exp Ther Med 2021; 21:571. [PMID: 33850543 PMCID: PMC8027719 DOI: 10.3892/etm.2021.10003] [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: 11/25/2020] [Accepted: 02/25/2021] [Indexed: 12/14/2022] Open
Abstract
The present study evaluated mRNA and long non-coding RNA (lncRNA) expression profiles and the pathways involved in paroxysmal atrial fibrillation (ParoAF) and persistent atrial fibrillation (PersAF). Nine left atrial appendage (LAA) tissues collected from the hearts of patients with AF (patients with ParoAF=3; and patients with PersAF=3) and healthy donors (n=3) were analyzed by RNA sequencing. Differentially expressed (DE) mRNAs and lncRNAs were identified by |Log2 fold change|>2 and P<0.05. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes pathway enrichment, protein-protein interaction network and mRNA-lncRNA interaction network analyses of DE mRNA and mRNA at the upstream/downstream of DE lncRNA were conducted. A total of 285 and 275 DE mRNAs, 575 and 583 DE lncRNAs were detected in ParoAF and PersAF samples compared with controls, respectively. PI3K/Akt and transforming growth factor-β signaling pathways were significantly enriched in the ParoAF_Control and the calcium signaling pathway was significantly enriched in the PersAF_Control. Cis and trans analyses revealed some important interactions in DE mRNAs and lncRNA, including an interaction of GPC-AS2 with dopachrome tautomerase, and phosphodiesterase 4D and cAMP-specific with XLOC_110310 and XLOC_137634. Overall, the present study provides a molecular basis for future clinical studies on ParoAF and PersAF.
Collapse
Affiliation(s)
- Haoliang Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
49
|
Stress-driven cardiac calcium mishandling via a kinase-to-kinase crosstalk. Pflugers Arch 2021; 473:363-375. [PMID: 33590296 PMCID: PMC7940337 DOI: 10.1007/s00424-021-02533-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 01/25/2023]
Abstract
Calcium homeostasis in the cardiomyocyte is critical to the regulation of normal cardiac function. Abnormal calcium dynamics such as altered uptake by the sarcoplasmic reticulum (SR) Ca2+-ATPase and increased diastolic SR calcium leak are involved in the development of maladaptive cardiac remodeling under pathological conditions. Ca2+/calmodulin-dependent protein kinase II-δ (CaMKIIδ) is a well-recognized key molecule in calcium dysregulation in cardiomyocytes. Elevated cellular stress is known as a common feature during pathological remodeling, and c-jun N-terminal kinase (JNK) is an important stress kinase that is activated in response to intrinsic and extrinsic stress stimuli. Our lab recently identified specific actions of JNK isoform 2 (JNK2) in CaMKIIδ expression, activation, and CaMKIIδ-dependent SR Ca2+ mishandling in the stressed heart. This review focuses on the current understanding of cardiac SR calcium handling under physiological and pathological conditions as well as the newly identified contribution of the stress kinase JNK2 in CaMKIIδ-dependent SR Ca2+ abnormal mishandling. The new findings identifying dual roles of JNK2 in CaMKIIδ expression and activation are also discussed in this review.
Collapse
|
50
|
Heijman J, Luermans JGLM, Linz D, van Gelder IC, Crijns HJGM. Risk Factors for Atrial Fibrillation Progression. Card Electrophysiol Clin 2021; 13:201-209. [PMID: 33516398 DOI: 10.1016/j.ccep.2020.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Atrial fibrillation is a chronic, progressive condition that presents a major health burden. This review summarizes recent studies assessing atrial fibrillation progression and its associated risk factors, describes the mechanisms underlying atrial fibrillation progression, and discusses the clinical implications of the progressive nature of atrial fibrillation. Progression of atrial fibrillation burden, and clinical progression from paroxysmal to more advanced (persistent/permanent) forms is common, but progression rates are variable. Atrial fibrillation progression parallels progressive atrial remodeling induced by atrial fibrillation risk factors and atrial fibrillation itself, and is associated with worse clinical outcomes.
Collapse
Affiliation(s)
- Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 5800, Maastricht 6202 AZ, The Netherlands. https://twitter.com/JordiHeijman
| | - Justin G L M Luermans
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 5800, Maastricht 6202 AZ, The Netherlands. https://twitter.com/JLuermans
| | - Dominik Linz
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 5800, Maastricht 6202 AZ, The Netherlands. https://twitter.com/Dominik_Linz
| | - Isabelle C van Gelder
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen 9700 RB, The Netherlands
| | - Harry J G M Crijns
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 5800, Maastricht 6202 AZ, The Netherlands.
| |
Collapse
|