1
|
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
|
2
|
Zhou S, Liu Y, Huang X, Wu C, Pórszász R. Omecamtiv Mecarbil in the treatment of heart failure: the past, the present, and the future. Front Cardiovasc Med 2024; 11:1337154. [PMID: 38566963 PMCID: PMC10985333 DOI: 10.3389/fcvm.2024.1337154] [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: 11/12/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Heart failure, a prevailing global health issue, imposes a substantial burden on both healthcare systems and patients worldwide. With an escalating prevalence of heart failure, prolonged survival rates, and an aging demographic, an increasing number of individuals are progressing to more advanced phases of this incapacitating ailment. Against this backdrop, the quest for pharmacological agents capable of addressing the diverse subtypes of heart failure becomes a paramount pursuit. From this viewpoint, the present article focuses on Omecamtiv Mecarbil (OM), an emerging chemical compound said to exert inotropic effects without altering calcium homeostasis. For the first time, as a review, the present article uniquely started from the very basic pathophysiology of heart failure, its classification, and the strategies underpinning drug design, to on-going debates of OM's underlying mechanism of action and the latest large-scale clinical trials. Furthermore, we not only saw the advantages of OM, but also exhaustively summarized the concerns in sense of its effects. These of no doubt make the present article the most systemic and informative one among the existing literature. Overall, by offering new mechanistic insights and therapeutic possibilities, OM has carved a significant niche in the treatment of heart failure, making it a compelling subject of study.
Collapse
Affiliation(s)
- Shujing Zhou
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ying Liu
- Department of Cardiology, Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xufeng Huang
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Chuhan Wu
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Róbert Pórszász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
3
|
Aggarwal K, Boyapati SP, Valecha J, Noor A, Kanwal F, Jain R, Kanagala SG. Arrhythmias and Hypertrophic Cardiomyopathy: Unravelling the Connection. Curr Cardiol Rev 2024; 20:e240124226139. [PMID: 38279754 PMCID: PMC11327833 DOI: 10.2174/011573403x279223231227111737] [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: 08/26/2023] [Revised: 11/15/2023] [Accepted: 11/30/2023] [Indexed: 01/28/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) results from gene mutations affecting cardiac sarcomeres and is inherited in an autosomal dominant manner. With a prevalence of 1:200-1:500 in the general population, HCM is characterised by a hypertrophied and non-dilated left ventricle with predominant involvement of the interventricular septum. The myocardium's structural and intracellular factors, combined with triggers such as physical exertion, autonomic dysfunction, and ischemia, can lead to reentry events, and atrial and ventricular arrhythmias, including atrial fibrillation (AF) which is common among HCM patients. To manage the increased risk of mortality arising from congestive heart failure and thromboembolism, in patients with AF long-term anticoagulation and antiarrhythmic drugs are employed. HCM patients may also encounter supraventricular and ventricular arrhythmias, such as nonsustained ventricular tachycardia and ventricular premature beats, which can potentially lead to sudden cardiac death and necessitate treatment with implanted defibrillators. Physicians must comprehensively analyse clinical, anatomical, hemodynamic, rhythmic, functional, and genetic characteristics to identify HCM patients at high risk of sudden death. This article aims to discuss the pathophysiology of arrhythmia in HCM and clinical recommendations for various ventricular and atrial fibrillation including catheter ablation and implantable cardioverter-defibrillator (ICD).
Collapse
Affiliation(s)
| | | | | | - Amna Noor
- Services Hospital Center, Lahore, Pakistan
| | - Fnu Kanwal
- Chandka Medical College, Larkana, Sindh, Pakistan
| | - Rohit Jain
- Penn State Milton S Hershey Medical Center, Hershey, Pennsylvania, USA
| | | |
Collapse
|
4
|
Orgil BO, Purevjav E. Molecular Pathways and Animal Models of Cardiomyopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:991-1019. [PMID: 38884766 DOI: 10.1007/978-3-031-44087-8_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
5
|
Eisner D, Neher E, Taschenberger H, Smith G. Physiology of intracellular calcium buffering. Physiol Rev 2023; 103:2767-2845. [PMID: 37326298 PMCID: PMC11550887 DOI: 10.1152/physrev.00042.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/08/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023] Open
Abstract
Calcium signaling underlies much of physiology. Almost all the Ca2+ in the cytoplasm is bound to buffers, with typically only ∼1% being freely ionized at resting levels in most cells. Physiological Ca2+ buffers include small molecules and proteins, and experimentally Ca2+ indicators will also buffer calcium. The chemistry of interactions between Ca2+ and buffers determines the extent and speed of Ca2+ binding. The physiological effects of Ca2+ buffers are determined by the kinetics with which they bind Ca2+ and their mobility within the cell. The degree of buffering depends on factors such as the affinity for Ca2+, the Ca2+ concentration, and whether Ca2+ ions bind cooperatively. Buffering affects both the amplitude and time course of cytoplasmic Ca2+ signals as well as changes of Ca2+ concentration in organelles. It can also facilitate Ca2+ diffusion inside the cell. Ca2+ buffering affects synaptic transmission, muscle contraction, Ca2+ transport across epithelia, and the killing of bacteria. Saturation of buffers leads to synaptic facilitation and tetanic contraction in skeletal muscle and may play a role in inotropy in the heart. This review focuses on the link between buffer chemistry and function and how Ca2+ buffering affects normal physiology and the consequences of changes in disease. As well as summarizing what is known, we point out the many areas where further work is required.
Collapse
Affiliation(s)
- David Eisner
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Erwin Neher
- Membrane Biophysics Laboratory, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Godfrey Smith
- School of Cardiovascular and Metabolic Health, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
6
|
Sharma AK, Singh S, Bhat M, Gill K, Zaid M, Kumar S, Shakya A, Tantray J, Jose D, Gupta R, Yangzom T, Sharma RK, Sahu SK, Rathore G, Chandolia P, Singh M, Mishra A, Raj S, Gupta A, Agarwal M, Kifayat S, Gupta A, Gupta P, Vashist A, Vaibhav P, Kathuria N, Yadav V, Singh RP, Garg A. New drug discovery of cardiac anti-arrhythmic drugs: insights in animal models. Sci Rep 2023; 13:16420. [PMID: 37775650 PMCID: PMC10541452 DOI: 10.1038/s41598-023-41942-4] [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: 04/24/2023] [Accepted: 09/04/2023] [Indexed: 10/01/2023] Open
Abstract
Cardiac rhythm regulated by micro-macroscopic structures of heart. Pacemaker abnormalities or disruptions in electrical conduction, lead to arrhythmic disorders may be benign, typical, threatening, ultimately fatal, occurs in clinical practice, patients on digitalis, anaesthesia or acute myocardial infarction. Both traditional and genetic animal models are: In-vitro: Isolated ventricular Myocytes, Guinea pig papillary muscles, Patch-Clamp Experiments, Porcine Atrial Myocytes, Guinea pig ventricular myocytes, Guinea pig papillary muscle: action potential and refractory period, Langendorff technique, Arrhythmia by acetylcholine or potassium. Acquired arrhythmia disorders: Transverse Aortic Constriction, Myocardial Ischemia, Complete Heart Block and AV Node Ablation, Chronic Tachypacing, Inflammation, Metabolic and Drug-Induced Arrhythmia. In-Vivo: Chemically induced arrhythmia: Aconitine antagonism, Digoxin-induced arrhythmia, Strophanthin/ouabain-induced arrhythmia, Adrenaline-induced arrhythmia, and Calcium-induced arrhythmia. Electrically induced arrhythmia: Ventricular fibrillation electrical threshold, Arrhythmia through programmed electrical stimulation, sudden coronary death in dogs, Exercise ventricular fibrillation. Genetic Arrhythmia: Channelopathies, Calcium Release Deficiency Syndrome, Long QT Syndrome, Short QT Syndrome, Brugada Syndrome. Genetic with Structural Heart Disease: Arrhythmogenic Right Ventricular Cardiomyopathy/Dysplasia, Dilated Cardiomyopathy, Hypertrophic Cardiomyopathy, Atrial Fibrillation, Sick Sinus Syndrome, Atrioventricular Block, Preexcitation Syndrome. Arrhythmia in Pluripotent Stem Cell Cardiomyocytes. Conclusion: Both traditional and genetic, experimental models of cardiac arrhythmias' characteristics and significance help in development of new antiarrhythmic drugs.
Collapse
Affiliation(s)
- Ashish Kumar Sharma
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India.
| | - Shivam Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mehvish Bhat
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Kartik Gill
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mohammad Zaid
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anjali Shakya
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Junaid Tantray
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Divyamol Jose
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Rashmi Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Tsering Yangzom
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Rajesh Kumar Sharma
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | | | - Gulshan Rathore
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Priyanka Chandolia
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mithilesh Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anurag Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Shobhit Raj
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Archita Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Mohit Agarwal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Sumaiya Kifayat
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Anamika Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Prashant Gupta
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Ankit Vashist
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Parth Vaibhav
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Nancy Kathuria
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Vipin Yadav
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Ravindra Pal Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, 303121, India
| | - Arun Garg
- MVN University, Palwal, Haryana, India
| |
Collapse
|
7
|
Shafaattalab S, Li AY, Jayousi F, Maaref Y, Dababneh S, Hamledari H, Baygi DH, Barszczewski T, Ruprai B, Jannati S, Nagalingam R, Cool AM, Langa P, Chiao M, Roston T, Solaro RJ, Sanatani S, Toepfer C, Lindert S, Lange P, Tibbits GF. Mechanisms of Pathogenicity of Hypertrophic Cardiomyopathy-Associated Troponin T (TNNT2) Variant R278C +/- During Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.542948. [PMID: 37609317 PMCID: PMC10441323 DOI: 10.1101/2023.06.06.542948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is one of the most common heritable cardiovascular diseases and variants of TNNT2 (cardiac troponin T) are linked to increased risk of sudden cardiac arrest despite causing limited hypertrophy. In this study, a TNNT2 variant, R278C+/-, was generated in both human cardiac recombinant/reconstituted thin filaments (hcRTF) and human- induced pluripotent stem cells (hiPSCs) to investigate the mechanisms by which the R278C+/- variant affects cardiomyocytes at the proteomic and functional levels. The results of proteomics analysis showed a significant upregulation of markers of cardiac hypertrophy and remodeling in R278C+/- vs. the isogenic control. Functional measurements showed that R278C+/- variant enhances the myofilament sensitivity to Ca2+, increases the kinetics of contraction, and causes arrhythmia at frequencies >75 bpm. This study uniquely shows the profound impact of the TNNT2 R278C+/- variant on the cardiomyocyte proteomic profile, cardiac electrical and contractile function in the early stages of cardiac development.
Collapse
Affiliation(s)
- Sanam Shafaattalab
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Alison Y Li
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Farah Jayousi
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Yasaman Maaref
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Homa Hamledari
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Dina Hosseini Baygi
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Tiffany Barszczewski
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Balwinder Ruprai
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Shayan Jannati
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Mechanical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Raghu Nagalingam
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Austin M Cool
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, USA
| | - Paulina Langa
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mu Chiao
- Mechanical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Thomas Roston
- Division of Cardiology and Centre for Cardiovascular Innovation, The University of British Columbia 1081 Burrard Street, Level 4 Cardiology Vancouver, BC, V6Z 1Y6, Canada
| | - R John Solaro
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shubhayan Sanatani
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
| | | | - Steffen Lindert
- Mechanical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Philipp Lange
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
| | - Glen F Tibbits
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
8
|
Claassen WJ, Baelde RJ, Galli RA, de Winter JM, Ottenheijm CAC. Small molecule drugs to improve sarcomere function in those with acquired and inherited myopathies. Am J Physiol Cell Physiol 2023; 325:C60-C68. [PMID: 37212548 PMCID: PMC10281779 DOI: 10.1152/ajpcell.00047.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Muscle weakness is a hallmark of inherited or acquired myopathies. It is a major cause of functional impairment and can advance to life-threatening respiratory insufficiency. During the past decade, several small-molecule drugs that improve the contractility of skeletal muscle fibers have been developed. In this review, we provide an overview of the available literature and the mechanisms of action of small-molecule drugs that modulate the contractility of sarcomeres, the smallest contractile units in striated muscle, by acting on myosin and troponin. We also discuss their use in the treatment of skeletal myopathies. The first of three classes of drugs discussed here increase contractility by decreasing the dissociation rate of calcium from troponin and thereby sensitizing the muscle to calcium. The second two classes of drugs directly act on myosin and stimulate or inhibit the kinetics of myosin-actin interactions, which may be useful in patients with muscle weakness or stiffness.NEW & NOTEWORTHY During the past decade, several small molecule drugs that improve the contractility of skeletal muscle fibers have been developed. In this review, we provide an overview of the available literature and the mechanisms of action of small molecule drugs that modulate the contractility of sarcomeres, the smallest contractile units in striated muscle, by acting on myosin and troponin.
Collapse
Affiliation(s)
- Wout J Claassen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Rianne J Baelde
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Ricardo A Galli
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Josine M de Winter
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Coen A C Ottenheijm
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| |
Collapse
|
9
|
Langa P, Shafaattalab S, Goldspink PH, Wolska BM, Fernandes AA, Tibbits GF, Solaro RJ. A perspective on Notch signalling in progression and arrhythmogenesis in familial hypertrophic and dilated cardiomyopathies. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220176. [PMID: 37122209 PMCID: PMC10150215 DOI: 10.1098/rstb.2022.0176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/13/2022] [Indexed: 05/02/2023] Open
Abstract
In this perspective, we discussed emerging data indicating a role for Notch signalling in inherited disorders of the heart failure with focus on hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) linked to variants of genes encoding mutant proteins of the sarcomere. We recently reported an upregulation of elements in the Notch signalling cascade in cardiomyocytes derived from human inducible pluripotent stem cells expressing a TNNT2 variant encoding cardiac troponin T (cTnT-I79N+/-), which induces hypertrophy, remodelling, abnormalities in excitation-contraction coupling and electrical instabilities (Shafaattalab S et al. 2021 Front. Cell Dev. Biol. 9, 787581. (doi:10.3389/fcell.2021.787581)). Our search of the literature revealed the novelty of this finding and stimulated us to discuss potential connections between the Notch signalling pathway and familial cardiomyopathies. Our considerations focused on the potential role of these interactions in arrhythmias, microvascular ischaemia, and fibrosis. This finding underscored a need to consider the role of Notch signalling in familial cardiomyopathies which are trigged by sarcomere mutations engaging mechano-signalling pathways for which there is evidence of a role for Notch signalling with crosstalk with Hippo signalling. Our discussion included a role for both cardiac myocytes and non-cardiac myocytes in progression of HCM and DCM. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Paulina Langa
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Sanam Shafaattalab
- Molecular Biology and Biochemistry; BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4; Simon Fraser University Burnaby, British Columbia, V5A 4H4, Canada
| | - Paul H. Goldspink
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Beata M. Wolska
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
- Department of Medicine, Division of Cardiology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Aurelia A. Fernandes
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Glen F. Tibbits
- Molecular Biology and Biochemistry; BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4; Simon Fraser University Burnaby, British Columbia, V5A 4H4, Canada
| | - R. John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| |
Collapse
|
10
|
Sequeira V, Waddingham MT, Tsuchimochi H, Maack C, Pearson JT. Mechano-energetic uncoupling in hypertrophic cardiomyopathy: Pathophysiological mechanisms and therapeutic opportunities. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 4:100036. [PMID: 39801694 PMCID: PMC11708264 DOI: 10.1016/j.jmccpl.2023.100036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 01/16/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a frequent inherited form of heart failure. The underlying cause of HCM is generally attributed to mutations in genes that encode for sarcomeric proteins, but the pathogenesis of the disease is also influenced by non-genetic factors, which can contribute to diastolic dysfunction and hypertrophic remodeling. Central to the pathogenesis of HCM is hypercontractility, a state that is an antecedent to several key derangements, including increased mitochondrial workload and oxidative stress. As a result, energy depletion and mechano-energetic uncoupling drive cardiac growth through signaling pathways such as ERK and/or potentially AMPK downregulation. Metabolic remodeling also occurs in HCM, characterized by decreased fatty acid oxidation and increased glucose uptake. In some instances, ketones may also feed the heart with energy and act as signaling molecules to reduce oxidative stress and hypertrophic signaling. In addition, arrhythmias are frequently triggered in HCM, resulting from the high Ca2+-buffering of the myofilaments and changes in the ATP/ADP ratio. Understanding the mechanisms driving the progression of HCM is critical to the development of effective therapeutic strategies. This paper presents evidence from both experimental and clinical studies that support the role of hypercontractility and cellular energy alterations in the progression of HCM towards heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Vasco Sequeira
- DZHI, Department of Translational Science Universitätsklinikum, Würzburg, Germany
| | - Mark T. Waddingham
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Christoph Maack
- DZHI, Department of Translational Science Universitätsklinikum, Würzburg, Germany
| | - James T. Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
- Department of Physiology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
11
|
Tamargo J, Tamargo M, Caballero R. Hypertrophic cardiomyopathy: an up-to-date snapshot of the clinical drug development pipeline. Expert Opin Investig Drugs 2022; 31:1027-1052. [PMID: 36062808 DOI: 10.1080/13543784.2022.2113374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM) is a complex cardiac disease with highly variable phenotypic expression and clinical course most often caused by sarcomeric gene mutations resulting in left ventricular hypertrophy, fibrosis, hypercontractility, and diastolic dysfunction. For almost 60 years, HCM has remained an orphan disease and still lacks a disease-specific treatment. AREAS COVERED This review summarizes recent preclinical and clinical trials with repurposed drugs and new emerging pharmacological and gene-based therapies for the treatment of HCM. EXPERT OPINION The off-label drugs routinely used alleviate symptoms but do not target the core pathophysiology of HCM or prevent or revert the phenotype. Recent advances in the genetics and pathophysiology of HCM led to the development of cardiac myosin adenosine triphosphatase inhibitors specifically directed to counteract the hypercontractility associated with HCM-causing mutations. Mavacamten, the first drug specifically developed for HCM successfully tested in a phase 3 trial, represents the major advance for the treatment of HCM. This opens new horizons for the development of novel drugs targeting HCM molecular substrates which hopefully modify the natural history of the disease. The role of current drugs in development and genetic-based approaches for the treatment of HCM are also discussed.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain
| | - María Tamargo
- Department of Cardiology, Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, Doctor Esquerdo, 46, 28007 Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain
| |
Collapse
|
12
|
Shen H, Dong SY, Ren MS, Wang R. Ventricular arrhythmia and sudden cardiac death in hypertrophic cardiomyopathy: From bench to bedside. Front Cardiovasc Med 2022; 9:949294. [PMID: 36061538 PMCID: PMC9433716 DOI: 10.3389/fcvm.2022.949294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with hypertrophic cardiomyopathy (HCM) mostly experience minimal symptoms throughout their lifetime, and some individuals have an increased risk of ventricular arrhythmias and sudden cardiac death (SCD). How to identify patients with a higher risk of ventricular arrythmias and SCD is the priority in HCM research. The American College of Cardiology/American Heart Association (ACC/AHA) and the European Society of Cardiology (ESC) both recommend the use of risk algorithms to identify patients at high risk of ventricular arrhythmias, to be selected for implantation of implantable cardioverters/defibrillators (ICDs) for primary prevention of SCD, although major discrepancies exist. The present SCD risk scoring systems cannot accurately identify early-stage HCM patients with modest structural remodeling and mild disease manifestations. Unfortunately, SCD events could occur in young asymptomatic HCM patients and even as initial symptoms, prompting the determination of new risk factors for SCD. This review summarizes the studies based on patients' surgical specimens, transgenic animals, and patient-derived induced pluripotent stem cells (hiPSCs) to explore the possible molecular mechanism of ventricular arrhythmia and SCD. Ion channel remodeling, Ca2+ homeostasis abnormalities, and increased myofilament Ca2+ sensitivity may contribute to changes in action potential duration (APD), reentry circuit formation, and trigger activities, such as early aferdepolarization (EAD) or delayed afterdepolarization (DAD), leading to ventricular arrhythmia in HCM. Besides the ICD implantation, novel drugs represented by the late sodium current channel inhibitor and myosin inhibitor also shed light on the prevention of HCM-related arrhythmias. The ideal prevention strategy of SCD in early-stage HCM patients needs to be combined with gene screening, hiPSC-CM testing, machine learning, and advanced ECG studies, thus achieving individualized SCD prevention.
Collapse
Affiliation(s)
- Hua Shen
- Division of Adult Cardiac Surgery, Department of Cardiovascular Medicine, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shi-Yong Dong
- Department of Cardiovascular Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ming-Shi Ren
- Division of Adult Cardiac Surgery, Department of Cardiovascular Medicine, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
- Graduate School, Chinese PLA General Hospital & Chinese PLA Medical School, Beijing, China
| | - Rong Wang
- Division of Adult Cardiac Surgery, Department of Cardiovascular Medicine, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Cardiovascular Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Rong Wang
| |
Collapse
|
13
|
Coscarella IL, Landim-Vieira M, Pinto JR, Chelko SP. Arrhythmogenic Cardiomyopathy: Exercise Pitfalls, Role of Connexin-43, and Moving beyond Antiarrhythmics. Int J Mol Sci 2022; 23:ijms23158753. [PMID: 35955883 PMCID: PMC9369094 DOI: 10.3390/ijms23158753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 12/11/2022] Open
Abstract
Arrhythmogenic Cardiomyopathy (ACM), a Mendelian disorder that can affect both left and right ventricles, is most often associated with pathogenic desmosomal variants that can lead to fibrofatty replacement of the myocardium, a pathological hallmark of this disease. Current therapies are aimed to prevent the worsening of disease phenotypes and sudden cardiac death (SCD). Despite the use of implantable cardioverter defibrillators (ICDs) there is no present therapy that would mitigate the loss in electrical signal and propagation by these fibrofatty barriers. Recent studies have shown the influence of forced vs. voluntary exercise in a variety of healthy and diseased mice; more specifically, that exercised mice show increased Connexin-43 (Cx43) expression levels. Fascinatingly, increased Cx43 expression ameliorated the abnormal electrical signal conduction in the myocardium of diseased mice. These findings point to a major translational pitfall in current therapeutics for ACM patients, who are advised to completely cease exercising and already demonstrate reduced Cx43 levels at the myocyte intercalated disc. Considering cardiac dysfunction in ACM arises from the loss of cardiomyocytes and electrical signal conduction abnormalities, an increase in Cx43 expression-promoted by low to moderate intensity exercise and/or gene therapy-could very well improve cardiac function in ACM patients.
Collapse
Affiliation(s)
- Isabella Leite Coscarella
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
| | - José Renato Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
| | - Stephen P. Chelko
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32303, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21215, USA
- Correspondence: ; Tel.: +1-850-644-2215
| |
Collapse
|
14
|
Jeon YK, Kwon JW, Jang J, Choi SW, Woo J, Cho SH, Yu BI, Chun YS, Youm JB, Zhang YH, Kim SJ. Lower troponin expression in the right ventricle of rats explains interventricular differences in E-C coupling. J Gen Physiol 2022; 154:212990. [PMID: 35099502 PMCID: PMC8823606 DOI: 10.1085/jgp.202112949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Despite distinctive functional and anatomic differences, a precise understanding of the cardiac interventricular differences in excitation–contraction (E–C) coupling mechanisms is still lacking. Here, we directly compared rat right and left cardiomyocytes (RVCM and LVCM). Whole-cell patch clamp, the IonOptix system, and fura-2 fluorimetry were used to measure electrical properties (action potential and ionic currents), single-cell contractility, and cytosolic Ca2+ ([Ca2+]i), respectively. Myofilament proteins were analyzed by immunoblotting. RVCM showed significantly shorter action potential duration (APD) and higher density of transient outward K+ current (Ito). However, the triggered [Ca2+]i change (Ca2+ transient) was not different, while the decay rate of the Ca2+ transient was slower in RVCM. Although the relaxation speed was also slower, the sarcomere shortening amplitude (ΔSL) was smaller in RVCM. SERCA activity was ∼60% lower in RVCM, which is partly responsible for the slower decay of the Ca2+ transient. Immunoblot analysis revealed lower expression of the cardiac troponin complex (cTn) in RVCM, implying a smaller Ca2+ buffering capacity (κS), which was proved by in situ analysis. The introduction of these new levels of cTn, Ito, and SERCA into a mathematical model of rat LVCM reproduced the similar Ca2+ transient, slower Ca2+ decay, shorter APD, and smaller ΔSL of RVCM. Taken together, these data show reduced expression of cTn proteins in the RVCM, which provides an explanation for the interventricular difference in the E–C coupling kinetics.
Collapse
Affiliation(s)
- Young Keul Jeon
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae Won Kwon
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jihyun Jang
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Center for Vascular and Inflammatory Disease, University of Maryland School of Medicine, Baltimore, MD
| | - Seong Woo Choi
- Department of Physiology and Ion Channel Disease Research Center, Dongguk University College of Medicine, Seoul, Republic of Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joohan Woo
- Department of Physiology and Ion Channel Disease Research Center, Dongguk University College of Medicine, Seoul, Republic of Korea
| | - Su Han Cho
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Byeong Il Yu
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yang Sook Chun
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae Boum Youm
- Cardiovascular and Metabolic Disease Center, Department of Physiology, College of Medicine, Inje University, Busan, Republic of Korea
| | - Yin Hua Zhang
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Yanbian University Hospital, Yanji, China.,Institute of Cardiovascular Sciences, Faculty of Biology, Medicine and Health Sciences, University of Manchester, Manchester, UK
| | - Sung Joon Kim
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Republic of Korea
| |
Collapse
|
15
|
Monasky MM, Micaglio E, D'Imperio S, Pappone C. The Mechanism of Ajmaline and Thus Brugada Syndrome: Not Only the Sodium Channel! Front Cardiovasc Med 2022; 8:782596. [PMID: 35004896 PMCID: PMC8733296 DOI: 10.3389/fcvm.2021.782596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Ajmaline is an anti-arrhythmic drug that is used to unmask the type-1 Brugada syndrome (BrS) electrocardiogram pattern to diagnose the syndrome. Thus, the disease is defined at its core as a particular response to this or other drugs. Ajmaline is usually described as a sodium-channel blocker, and most research into the mechanism of BrS has centered around this idea that the sodium channel is somehow impaired in BrS, and thus the genetics research has placed much emphasis on sodium channel gene mutations, especially the gene SCN5A, to the point that it has even been suggested that only the SCN5A gene should be screened in BrS patients. However, pathogenic rare variants in SCN5A are identified in only 20–30% of cases, and recent data indicates that SCN5A variants are actually, in many cases, prognostic rather than diagnostic, resulting in a more severe phenotype. Furthermore, the misconception by some that ajmaline only influences the sodium current is flawed, in that ajmaline actually acts additionally on potassium and calcium currents, as well as mitochondria and metabolic pathways. Clinical studies have implicated several candidate genes in BrS, encoding not only for sodium, potassium, and calcium channel proteins, but also for signaling-related, scaffolding-related, sarcomeric, and mitochondrial proteins. Thus, these proteins, as well as any proteins that act upon them, could prove absolutely relevant in the mechanism of BrS.
Collapse
Affiliation(s)
- Michelle M Monasky
- Arrhythmology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Emanuele Micaglio
- Arrhythmology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Sara D'Imperio
- Arrhythmology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
16
|
Shafaattalab S, Li AY, Gunawan MG, Kim B, Jayousi F, Maaref Y, Song Z, Weiss JN, Solaro RJ, Qu Z, Tibbits GF. Mechanisms of Arrhythmogenicity of Hypertrophic Cardiomyopathy-Associated Troponin T ( TNNT2) Variant I79N. Front Cell Dev Biol 2022; 9:787581. [PMID: 34977031 PMCID: PMC8718794 DOI: 10.3389/fcell.2021.787581] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heritable cardiovascular disease and often results in cardiac remodeling and an increased incidence of sudden cardiac arrest (SCA) and death, especially in youth and young adults. Among thousands of different variants found in HCM patients, variants of TNNT2 (cardiac troponin T—TNNT2) are linked to increased risk of ventricular arrhythmogenesis and sudden death despite causing little to no cardiac hypertrophy. Therefore, studying the effect of TNNT2 variants on cardiac propensity for arrhythmogenesis can pave the way for characterizing HCM in susceptible patients before sudden cardiac arrest occurs. In this study, a TNNT2 variant, I79N, was generated in human cardiac recombinant/reconstituted thin filaments (hcRTF) to investigate the effect of the mutation on myofilament Ca2+ sensitivity and Ca2+ dissociation rate using steady-state and stopped-flow fluorescence techniques. The results revealed that the I79N variant significantly increases myofilament Ca2+ sensitivity and decreases the Ca2+ off-rate constant (koff). To investigate further, a heterozygous I79N+/−TNNT2 variant was introduced into human-induced pluripotent stem cells using CRISPR/Cas9 and subsequently differentiated into ventricular cardiomyocytes (hiPSC-CMs). To study the arrhythmogenic properties, monolayers of I79N+/− hiPSC-CMs were studied in comparison to their isogenic controls. Arrhythmogenesis was investigated by measuring voltage (Vm) and cytosolic Ca2+ transients over a range of stimulation frequencies. An increasing stimulation frequency was applied to the cells, from 55 to 75 bpm. The results of this protocol showed that the TnT-I79N cells had reduced intracellular Ca2+ transients due to the enhanced cytosolic Ca2+ buffering. These changes in Ca2+ handling resulted in beat-to-beat instability and triangulation of the cardiac action potential, which are predictors of arrhythmia risk. While wild-type (WT) hiPSC-CMs were accurately entrained to frequencies of at least 150 bpm, the I79N hiPSC-CMs demonstrated clear patterns of alternans for both Vm and Ca2+ transients at frequencies >75 bpm. Lastly, a transcriptomic analysis was conducted on WT vs. I79N+/−TNNT2 hiPSC-CMs using a custom NanoString codeset. The results showed a significant upregulation of NPPA (atrial natriuretic peptide), NPPB (brain natriuretic peptide), Notch signaling pathway components, and other extracellular matrix (ECM) remodeling components in I79N+/− vs. the isogenic control. This significant shift demonstrates that this missense in the TNNT2 transcript likely causes a biophysical trigger, which initiates this significant alteration in the transcriptome. This TnT-I79N hiPSC-CM model not only reproduces key cellular features of HCM-linked mutations but also suggests that this variant causes uncharted pro-arrhythmic changes to the human action potential and gene expression.
Collapse
Affiliation(s)
- Sanam Shafaattalab
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.,Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Alison Y Li
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Marvin G Gunawan
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.,Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - BaRun Kim
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Farah Jayousi
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.,Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Yasaman Maaref
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.,Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Zhen Song
- UCLA Cardiac Computation Lab, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - James N Weiss
- UCLA Cardiac Computation Lab, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - R John Solaro
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States
| | - Zhilin Qu
- UCLA Cardiac Computation Lab, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Glen F Tibbits
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada.,Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada.,Department of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Increased cytosolic calcium buffering contributes to a cellular arrhythmogenic substrate in iPSC-cardiomyocytes from patients with dilated cardiomyopathy. Basic Res Cardiol 2022; 117:5. [PMID: 35499658 PMCID: PMC9061684 DOI: 10.1007/s00395-022-00912-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 01/31/2023]
Abstract
Dilated cardiomyopathy (DCM) is a major risk factor for heart failure and is associated with the development of life-threatening cardiac arrhythmias. Using a patient-specific induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) model harbouring a mutation in cardiac troponin T (R173W), we aim to examine the cellular basis of arrhythmogenesis in DCM patients with this mutation. iPSC from control (Ctrl) and DCM-TnT-R173W donors from the same family were differentiated into iPSC-CM and analysed through optical action potential (AP) recordings, simultaneous measurement of cytosolic calcium concentration ([Ca2+]i) and membrane currents and separately assayed using field stimulation to detect the threshold for AP- and [Ca2+]i-alternans development. AP duration was unaltered in TnT-R173W iPSC-CM. Nevertheless, TnT-R173W iPSC-CM showed a strikingly low stimulation threshold for AP- and [Ca2+]i-alternans. Myofilaments are known to play a role as intracellular Ca2+ buffers and here we show increased Ca2+ affinity of intracellular buffers in TnT-R173W cells, indicating increased myofilament sensitivity to Ca2+. Similarly, EMD57033, a myofilament Ca2+ sensitiser, replicated the abnormal [Ca2+]i dynamics observed in TnT-R173W samples and lowered the threshold for alternans development. In contrast, application of a Ca2+ desensitiser (blebbistatin) to TnT-R173W iPSC-CM was able to phenotypically rescue Ca2+ dynamics, normalising Ca2+ transient profile and minimising the occurrence of Ca2+ alternans at physiological frequencies. This finding suggests that increased Ca2+ buffering likely plays a major arrhythmogenic role in patients with DCM, specifically in those with mutations in cardiac troponin T. In addition, we propose that modulation of myofilament Ca2+ sensitivity could be an effective anti-arrhythmic target for pharmacological management of this disease.
Collapse
|
18
|
Pollmann K, Kaltenecker E, Schleihauf J, Ewert P, Görlach A, Wolf CM. Compound Mutation in Cardiac Sarcomere Proteins Is Associated with Increased Risk for Major Arrhythmic Events in Pediatric Onset Hypertrophic Cardiomyopathy. J Clin Med 2021; 10:jcm10225256. [PMID: 34830538 PMCID: PMC8617951 DOI: 10.3390/jcm10225256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/02/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is associated with adverse left ventricular (LV) remodeling causing dysfunction and malignant arrhythmias. Severely affected patients present with disease onset during childhood and sudden cardiac death risk (SCD) stratification is of the highest importance in this cohort. This study aimed to investigate genotype–phenotype association regarding clinical outcome and disease progression in pediatric onset HCM. Medical charts from forty-nine patients with pediatric HCM who had undergone genetic testing were reviewed for retrospective analysis. Demographic, clinical, transthoracic echocardiographic, electrocardiographic, long-term electrocardiogram, cardiopulmonary exercise test, cardiac magnetic resonance, and medication data were recorded. Childhood onset HCM was diagnosed in 29 males and 20 females. Median age at last follow-up was 18.7 years (range 2.6–51.7 years) with a median follow-up time since diagnosis of 8.5 years (range 0.2–38.0 years). Comparison of patients carrying mutations in distinct genes and comparison of genotype-negative with genotype-positive individuals, revealed no differences in functional classification, LV morphology, hypertrophy, systolic and diastolic function, fibrosis and cardiac medication. Patients with compound mutations had a significantly higher risk for major arrhythmic events than a single-mutation carrier. No association between affected genes and disease severity or progression was identified in this cohort.
Collapse
Affiliation(s)
- Kathrin Pollmann
- German Heart Center Munich, Department of Congenital Heart Disease and Pediatric Cardiology, School of Medicine & Health, Technical University of Munich, 80636 Munich, Germany; (K.P.); (E.K.); (J.S.); (P.E.); (A.G.)
| | - Emanuel Kaltenecker
- German Heart Center Munich, Department of Congenital Heart Disease and Pediatric Cardiology, School of Medicine & Health, Technical University of Munich, 80636 Munich, Germany; (K.P.); (E.K.); (J.S.); (P.E.); (A.G.)
| | - Julia Schleihauf
- German Heart Center Munich, Department of Congenital Heart Disease and Pediatric Cardiology, School of Medicine & Health, Technical University of Munich, 80636 Munich, Germany; (K.P.); (E.K.); (J.S.); (P.E.); (A.G.)
| | - Peter Ewert
- German Heart Center Munich, Department of Congenital Heart Disease and Pediatric Cardiology, School of Medicine & Health, Technical University of Munich, 80636 Munich, Germany; (K.P.); (E.K.); (J.S.); (P.E.); (A.G.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802 Munich, Germany
| | - Agnes Görlach
- German Heart Center Munich, Department of Congenital Heart Disease and Pediatric Cardiology, School of Medicine & Health, Technical University of Munich, 80636 Munich, Germany; (K.P.); (E.K.); (J.S.); (P.E.); (A.G.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802 Munich, Germany
- Experimental and Molecular Pediatric Cardiology, Technical University of Munich, 80636 Munich, Germany
| | - Cordula M. Wolf
- German Heart Center Munich, Department of Congenital Heart Disease and Pediatric Cardiology, School of Medicine & Health, Technical University of Munich, 80636 Munich, Germany; (K.P.); (E.K.); (J.S.); (P.E.); (A.G.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80802 Munich, Germany
- Correspondence:
| |
Collapse
|
19
|
Cheng Z, Fang T, Huang J, Guo Y, Alam M, Qian H. Hypertrophic Cardiomyopathy: From Phenotype and Pathogenesis to Treatment. Front Cardiovasc Med 2021; 8:722340. [PMID: 34760939 PMCID: PMC8572854 DOI: 10.3389/fcvm.2021.722340] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/17/2021] [Indexed: 02/05/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a very common inherited cardiovascular disease (CAD) and the incidence is about 1/500 of the common population. It is caused by more than 1,400 mutations in 11 or more genes encoding the proteins of the cardiac sarcomere. HCM presents a heterogeneous clinical profile and complex pathophysiology and HCM is the most important cause of sudden cardiac death (SCD) in young people. HCM also contributes to functional disability from heart failure and stroke (caused by atrial fibrillation). Current treatments for HCM (medication, myectomy, and alcohol septal ablation) are geared toward slowing down the disease progression and symptom relief and implanted cardiac defibrillator (ICD) to prevent SCD. HCM is, however, entering a period of tight translational research that holds promise for the major advances in disease-specific therapy. Main insights into the genetic landscape of HCM have improved our understanding of molecular pathogenesis and pointed the potential targets for the development of therapeutic agents. We reviewed the critical discoveries about the treatments, mechanism of HCM, and their implications for future research.
Collapse
Affiliation(s)
- Zeyi Cheng
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tingting Fang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinglei Huang
- School of Medicine, Lanzhou University, Lanzhou, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Mahboob Alam
- Division of Cardiovascular Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Hong Qian
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Ion Channel Impairment and Myofilament Ca 2+ Sensitization: Two Parallel Mechanisms Underlying Arrhythmogenesis in Hypertrophic Cardiomyopathy. Cells 2021; 10:cells10102789. [PMID: 34685769 PMCID: PMC8534456 DOI: 10.3390/cells10102789] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
Life-threatening ventricular arrhythmias are the main clinical burden in patients with hypertrophic cardiomyopathy (HCM), and frequently occur in young patients with mild structural disease. While massive hypertrophy, fibrosis and microvascular ischemia are the main mechanisms underlying sustained reentry-based ventricular arrhythmias in advanced HCM, cardiomyocyte-based functional arrhythmogenic mechanisms are likely prevalent at earlier stages of the disease. In this review, we will describe studies conducted in human surgical samples from HCM patients, transgenic animal models and human cultured cell lines derived from induced pluripotent stem cells. Current pieces of evidence concur to attribute the increased risk of ventricular arrhythmias in early HCM to different cellular mechanisms. The increase of late sodium current and L-type calcium current is an early observation in HCM, which follows post-translation channel modifications and increases the occurrence of early and delayed afterdepolarizations. Increased myofilament Ca2+ sensitivity, commonly observed in HCM, may promote afterdepolarizations and reentry arrhythmias with direct mechanisms. Decrease of K+-currents due to transcriptional regulation occurs in the advanced disease and contributes to reducing the repolarization-reserve and increasing the early afterdepolarizations (EADs). The presented evidence supports the idea that patients with early-stage HCM should be considered and managed as subjects with an acquired channelopathy rather than with a structural cardiac disease.
Collapse
|
21
|
Hassoun R, Budde H, Mügge A, Hamdani N. Cardiomyocyte Dysfunction in Inherited Cardiomyopathies. Int J Mol Sci 2021; 22:11154. [PMID: 34681814 PMCID: PMC8541428 DOI: 10.3390/ijms222011154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 01/10/2023] Open
Abstract
Inherited cardiomyopathies form a heterogenous group of disorders that affect the structure and function of the heart. Defects in the genes encoding sarcomeric proteins are associated with various perturbations that induce contractile dysfunction and promote disease development. In this review we aimed to outline the functional consequences of the major inherited cardiomyopathies in terms of myocardial contraction and kinetics, and to highlight the structural and functional alterations in some sarcomeric variants that have been demonstrated to be involved in the pathogenesis of the inherited cardiomyopathies. A particular focus was made on mutation-induced alterations in cardiomyocyte mechanics. Since no disease-specific treatments for familial cardiomyopathies exist, several novel agents have been developed to modulate sarcomere contractility. Understanding the molecular basis of the disease opens new avenues for the development of new therapies. Furthermore, the earlier the awareness of the genetic defect, the better the clinical prognostication would be for patients and the better the prevention of development of the disease.
Collapse
Affiliation(s)
- Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
22
|
Mutations in MYBPC3 and MYH7 in Association with Brugada Type 1 ECG Pattern: Overlap between Brugada Syndrome and Hypertrophic Cardiomyopathy? CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11030016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Brugada syndrome (BrS) is an inherited disorder with high allelic and genetic heterogeneity clinically characterized by typical coved-type ST segment elevation at the electrocardiogram (ECG), which may occur either spontaneously or after provocative drug testing. BrS is classically described as an arrhythmic condition occurring in a structurally normal heart and is associated with the risk of ventricular fibrillation and sudden cardiac death (SCD). We studied five patients with spontaneous or drug-induced type 1 ECG pattern, variably associated with symptoms and a positive family history through a Next Generation Sequencing panels approach, which includes genes of both channelopathies and cardiomyopathies. We identified variants in MYBPC3 and in MYH7, hypertrophic cardiomyopathy (HCM) genes (MYBPC3: p.Lys1065Glnfs*12 and c.1458-1G > A, MYH7: p.Arg783His, p.Val1213Met, p.Lys744Thr). Our data propose that Brugada type 1 ECG may be an early electrocardiographic marker of a concealed structural heart disease, possibly enlarging the genotypic overlap between Brugada syndrome and cardiomyopathies.
Collapse
|
23
|
Stătescu C, Enachi Ș, Ureche C, Țăpoi L, Anghel L, Șalaru D, Pleșoianu C, Bostan M, Marcu D, Ovanez Balasanian M, Sascău RA. Pushing the Limits of Medical Management in HCM: A Review of Current Pharmacological Therapy Options. Int J Mol Sci 2021; 22:ijms22137218. [PMID: 34281272 PMCID: PMC8268685 DOI: 10.3390/ijms22137218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common monogenic cardiac disease with a highly variable phenotypic expression, ranging from asymptomatic to drug refractory heart failure (HF) presentation. Pharmacological therapy is the first line of treatment, but options are currently limited to nonspecific medication like betablockers or calcium channel inhibitors, with frequent suboptimal results. While being the gold standard practice for the management of drug refractory HCM patients, septal reduction therapy (SRT) remains an invasive procedure with associated surgical risks and it requires the expertise of the operating centre, thus limiting its accessibility. It is therefore with high interest that researchers look for pharmacological alternatives that could provide higher rates of success. With new data gathering these past years as well as the development of a new drug class showing promising results, this review provides an up-to-date focused synthesis of existing medical treatment options and future directions for HCM pharmacological treatment.
Collapse
Affiliation(s)
- Cristian Stătescu
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ștefana Enachi
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Correspondence: ; Tel.: +40-749-630-641
| | - Carina Ureche
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laura Țăpoi
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
| | - Larisa Anghel
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Delia Șalaru
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Carmen Pleșoianu
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mădălina Bostan
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Dragoș Marcu
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mircea Ovanez Balasanian
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Radu Andy Sascău
- Cardiology Department, Cardiovascular Diseases Institute “Prof. Dr. George I.M. Georgescu”, Carol I Boulevard No. 50, 700503 Iași, Romania; (C.S.); (C.U.); (L.Ț.); (L.A.); (D.Ș.); (C.P.); (M.B.); (D.M.); (M.O.B.); (R.A.S.)
- Internal Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
24
|
Fülöp GÁ, Oláh A, Csipo T, Kovács Á, Pórszász R, Veress R, Horváth B, Nagy L, Bódi B, Fagyas M, Helgadottir SL, Bánhegyi V, Juhász B, Bombicz M, Priksz D, Nanasi P, Merkely B, Édes I, Csanádi Z, Papp Z, Radovits T, Tóth A. Omecamtiv mecarbil evokes diastolic dysfunction and leads to periodic electromechanical alternans. Basic Res Cardiol 2021; 116:24. [PMID: 33844095 PMCID: PMC8041714 DOI: 10.1007/s00395-021-00866-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 03/31/2021] [Indexed: 01/31/2023]
Abstract
Omecamtiv mecarbil (OM) is a promising novel drug for improving cardiac contractility. We tested the therapeutic range of OM and identified previously unrecognized side effects. The Ca2+ sensitivity of isometric force production (pCa50) and force at low Ca2+ levels increased with OM concentration in human permeabilized cardiomyocytes. OM (1 µM) slowed the kinetics of contractions and relaxations and evoked an oscillation between normal and reduced intracellular Ca2+ transients, action potential lengths and contractions in isolated canine cardiomyocytes. Echocardiographic studies and left ventricular pressure-volume analyses demonstrated concentration-dependent improvements in cardiac systolic function at OM concentrations of 600-1200 µg/kg in rats. Administration of OM at a concentration of 1200 µg/kg was associated with hypotension, while doses of 600-1200 µg/kg were associated with the following aspects of diastolic dysfunction: decreases in E/A ratio and the maximal rate of diastolic pressure decrement (dP/dtmin) and increases in isovolumic relaxation time, left atrial diameter, the isovolumic relaxation constant Tau, left ventricular end-diastolic pressure and the slope of the end-diastolic pressure-volume relationship. Moreover, OM 1200 µg/kg frequently evoked transient electromechanical alternans in the rat in vivo in which normal systoles were followed by smaller contractions (and T-wave amplitudes) without major differences on the QRS complexes. Besides improving systolic function, OM evoked diastolic dysfunction and pulsus alternans. The narrow therapeutic window for OM may necessitate the monitoring of additional clinical safety parameters in clinical application.
Collapse
MESH Headings
- Action Potentials/drug effects
- Adult
- Animals
- Arrhythmias, Cardiac/chemically induced
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Blood Pressure/drug effects
- Calcium Signaling/drug effects
- Cardiotonic Agents/toxicity
- Diastole
- Dogs
- Dose-Response Relationship, Drug
- Female
- Heart Rate/drug effects
- Humans
- Hypotension/chemically induced
- Hypotension/metabolism
- Hypotension/physiopathology
- Kinetics
- Male
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Rats, Inbred WKY
- Systole
- Urea/analogs & derivatives
- Urea/toxicity
- Ventricular Dysfunction, Left/chemically induced
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left/drug effects
- Rats
Collapse
Grants
- GINOP-2.3.2-15-2016-00043 Ministry for National Economy of Hungary, co-financed by the European Union and the European Regional Development Fund
- ÚNKP-18-3-III-DE-209 Ministry of Human Capacities of Hungary, co-financed by the European Union and the European Regional Development Fund
- ED_18-1-2019-0028, TKP2020-IKA-04 and TKP2020-NKA-04 The Thematic Excellence Programme of the Ministry for Innovation and Technology, also supported from the National Research, Development and Innovation Fund of Hungary
- FK 128809 National Research, Development and Innovation Fund of Hungary
- FK 128116 National Research, Development and Innovation Fund of Hungary
- K 134939 National Research, Development and Innovation Fund of Hungary.
- K 116940 and K 132623 National Research, Development and Innovation Fund of Hungary.
- Therapeutic Development thematic programme of the Semmelweis University Higher Education Institutional Excellence Programme of the Ministry for Innovation and Technology in Hungary
- 2020-4.1.1.-TKP2020, Therapeutic Development and Bioimaging thematic programme of the Semmelweis University The Thematic Excellence Programme of the Ministry for Innovation and Technology was also supported from the National Research, Development and Innovation Fund of Hungary
- The Thematic Excellence Programme of the Ministry for Innovation and Technology, also supported from the National Research, Development and Innovation Fund of Hungary
- The Thematic Excellence Programme of the Ministry for Innovation and Technology was also supported from the National Research, Development and Innovation Fund of Hungary
- University of Debrecen
Collapse
Affiliation(s)
- Gábor Á Fülöp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Doctoral School of Kálmán Laki, University of Debrecen, Debrecen, Hungary
| | - Attila Oláh
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Tamas Csipo
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Doctoral School of Kálmán Laki, University of Debrecen, Debrecen, Hungary
| | - Árpád Kovács
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Róbert Pórszász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Roland Veress
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Nagy
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beáta Bódi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
| | - Miklós Fagyas
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Solveig Lind Helgadottir
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
| | - Viktor Bánhegyi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Doctoral School of Kálmán Laki, University of Debrecen, Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Nanasi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - István Édes
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Csanádi
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary.
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
25
|
Assessment of dynamic cardiac repolarization and contractility in patients with hypertrophic cardiomyopathy. PLoS One 2021; 16:e0246768. [PMID: 33571287 PMCID: PMC7877626 DOI: 10.1371/journal.pone.0246768] [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: 10/09/2020] [Accepted: 01/25/2021] [Indexed: 11/24/2022] Open
Abstract
Aims Arrhythmia mechanisms in hypertrophic cardiomyopathy remain uncertain. Preclinical models suggest hypertrophic cardiomyopathy-linked mutations perturb sarcomere length-dependent activation, alter cardiac repolarization in rate-dependent fashion and potentiate triggered electrical activity. This study was designed to assess rate-dependence of clinical surrogates of contractility and repolarization in humans with hypertrophic cardiomyopathy. Methods All participants had a cardiac implantable device capable of atrial pacing. Cases had clinical diagnosis of hypertrophic cardiomyopathy, controls were age-matched. Continuous electrocardiogram and blood pressure were recorded during and immediately after 30 second pacing trains delivered at increasing rates. Results Nine hypertrophic cardiomyopathy patients and 10 controls were enrolled (47% female, median 55 years), with similar baseline QRS duration, QT interval and blood pressure. Median septal thickness in hypertrophic cardiomyopathy patients was 18mm; 33% of hypertrophic cardiomyopathy patients had peak sub-aortic velocity >50mmHg. Ventricular ectopy occurred during or immediately after pacing trains in 4/9 hypertrophic cardiomyopathy patients and 0/10 controls (P = 0.03). During delivery of steady rate pacing across a range of cycle lengths, the QT-RR relationship was not statistically different between HCM and control groups; no differences were seen in subgroup analysis of patients with or without intact AV node conduction. Similarly, there was no difference between groups in the QT interval of the first post-pause recovery beat after pacing trains. No statistically significant differences were seen in surrogate measures for cardiac contractility. Conclusion Rapid pacing trains triggered ventricular ectopy in hypertrophic cardiomyopathy patients, but not controls. This finding aligns with pre-clinical descriptions of excessive cardiomyocyte calcium loading during rapid pacing, increased post-pause sarcoplasmic reticulum calcium release, and subsequent calcium-triggered activity. Normal contractility at all diastolic intervals argues against clinical significance of altered length-dependent myofilament activation.
Collapse
|
26
|
Alsulami K, Marston S. Small Molecules acting on Myofilaments as Treatments for Heart and Skeletal Muscle Diseases. Int J Mol Sci 2020; 21:E9599. [PMID: 33339418 PMCID: PMC7767104 DOI: 10.3390/ijms21249599] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are the most prevalent forms of the chronic and progressive pathological condition known as cardiomyopathy. These diseases have different aetiologies; however, they share the feature of haemodynamic abnormalities, which is mainly due to dysfunction in the contractile proteins that make up the contractile unit known as the sarcomere. To date, pharmacological treatment options are not disease-specific and rather focus on managing the symptoms, without addressing the disease mechanism. Earliest attempts at improving cardiac contractility by modulating the sarcomere indirectly (inotropes) resulted in unwanted effects. In contrast, targeting the sarcomere directly, aided by high-throughput screening systems, could identify small molecules with a superior therapeutic value in cardiac muscle disorders. Herein, an extensive literature review of 21 small molecules directed to five different targets was conducted. A simple scoring system was created to assess the suitability of small molecules for therapy by evaluating them in eight different criteria. Most of the compounds failed due to lack of target specificity or poor physicochemical properties. Six compounds stood out, showing a potential therapeutic value in HCM, DCM or heart failure (HF). Omecamtiv Mecarbil and Danicamtiv (myosin activators), Mavacamten, CK-274 and MYK-581 (myosin inhibitors) and AMG 594 (Ca2+-sensitiser) are all small molecules that allosterically modulate troponin or myosin. Omecamtiv Mecarbil showed limited efficacy in phase III GALACTIC-HF trial, while, results from phase III EXPLORER-HCM trial were recently published, indicating that Mavacamten reduced left ventricular outflow tract (LVOT) obstruction and diastolic dysfunction and improved the health status of patients with HCM. A novel category of small molecules known as "recouplers" was reported to target a phenomenon termed uncoupling commonly found in familial cardiomyopathies but has not progressed beyond preclinical work. In conclusion, the contractile apparatus is a promising target for new drug development.
Collapse
Affiliation(s)
- Khulud Alsulami
- Imperial Centre for Translational and Experimental Medicine, Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology, Riyadh 11461, Saudi Arabia
| | - Steven Marston
- Imperial Centre for Translational and Experimental Medicine, Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
27
|
Awinda PO, Bishaw Y, Watanabe M, Guglin MA, Campbell KS, Tanner BCW. Effects of mavacamten on Ca 2+ sensitivity of contraction as sarcomere length varied in human myocardium. Br J Pharmacol 2020; 177:5609-5621. [PMID: 32960449 PMCID: PMC7707091 DOI: 10.1111/bph.15271] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Heart failure can reflect impaired contractile function at the myofilament level. In healthy hearts, myofilaments become more sensitive to Ca2+ as cells are stretched. This represents a fundamental property of the myocardium that contributes to the Frank-Starling response, although the molecular mechanisms underlying the effect remain unclear. Mavacamten, which binds to myosin, is under investigation as a potential therapy for heart disease. We investigated how mavacamten affects the sarcomere-length dependence of Ca2+ -sensitive isometric contraction to determine how mavacamten might modulate the Frank-Starling mechanism. EXPERIMENTAL APPROACH Multicellular preparations from the left ventricular-free wall of hearts from organ donors were chemically permeabilized and Ca2+ activated in the presence or absence of 0.5-μM mavacamten at 1.9 or 2.3-μm sarcomere length (37°C). Isometric force and frequency-dependent viscoelastic myocardial stiffness measurements were made. KEY RESULTS At both sarcomere lengths, mavacamten reduced maximal force and Ca2+ sensitivity of contraction. In the presence and absence of mavacamten, Ca2+ sensitivity of force increased as sarcomere length increased. This suggests that the length-dependent activation response was maintained in human myocardium, even though mavacamten reduced Ca2+ sensitivity. There were subtle effects of mavacamten reducing force values under relaxed conditions (pCa 8.0), as well as slowing myosin cross-bridge recruitment and speeding cross-bridge detachment under maximally activated conditions (pCa 4.5). CONCLUSION AND IMPLICATIONS Mavacamten did not eliminate sarcomere length-dependent increases in the Ca2+ sensitivity of contraction in myocardial strips from organ donors at physiological temperature. Drugs that modulate myofilament function may be useful therapies for cardiomyopathies.
Collapse
Affiliation(s)
- Peter O. Awinda
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Yemeserach Bishaw
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Marissa Watanabe
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| | - Maya A. Guglin
- Division of Cardiovascular MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Kenneth S. Campbell
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- Division of Cardiovascular MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Bertrand C. W. Tanner
- Department of Integrative Physiology and NeuroscienceWashington State UniversityPullmanWashingtonUSA
| |
Collapse
|
28
|
Maack C, Eschenhagen T, Hamdani N, Heinzel FR, Lyon AR, Manstein DJ, Metzger J, Papp Z, Tocchetti CG, Yilmaz MB, Anker SD, Balligand JL, Bauersachs J, Brutsaert D, Carrier L, Chlopicki S, Cleland JG, de Boer RA, Dietl A, Fischmeister R, Harjola VP, Heymans S, Hilfiker-Kleiner D, Holzmeister J, de Keulenaer G, Limongelli G, Linke WA, Lund LH, Masip J, Metra M, Mueller C, Pieske B, Ponikowski P, Ristić A, Ruschitzka F, Seferović PM, Skouri H, Zimmermann WH, Mebazaa A. Treatments targeting inotropy. Eur Heart J 2020; 40:3626-3644. [PMID: 30295807 DOI: 10.1093/eurheartj/ehy600] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/06/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Acute heart failure (HF) and in particular, cardiogenic shock are associated with high morbidity and mortality. A therapeutic dilemma is that the use of positive inotropic agents, such as catecholamines or phosphodiesterase-inhibitors, is associated with increased mortality. Newer drugs, such as levosimendan or omecamtiv mecarbil, target sarcomeres to improve systolic function putatively without elevating intracellular Ca2+. Although meta-analyses of smaller trials suggested that levosimendan is associated with a better outcome than dobutamine, larger comparative trials failed to confirm this observation. For omecamtiv mecarbil, Phase II clinical trials suggest a favourable haemodynamic profile in patients with acute and chronic HF, and a Phase III morbidity/mortality trial in patients with chronic HF has recently begun. Here, we review the pathophysiological basis of systolic dysfunction in patients with HF and the mechanisms through which different inotropic agents improve cardiac function. Since adenosine triphosphate and reactive oxygen species production in mitochondria are intimately linked to the processes of excitation-contraction coupling, we also discuss the impact of inotropic agents on mitochondrial bioenergetics and redox regulation. Therefore, this position paper should help identify novel targets for treatments that could not only safely improve systolic and diastolic function acutely, but potentially also myocardial structure and function over a longer-term.
Collapse
Affiliation(s)
- Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, Würzburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Partner site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Frank R Heinzel
- Department of Cardiology, Charité University Medicine, Berlin, Germany
| | - Alexander R Lyon
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany.,Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Joseph Metzger
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - M Birhan Yilmaz
- Department of Cardiology, Cumhuriyet University, Sivas, Turkey
| | - Stefan D Anker
- Department of Cardiology and Pneumology, University Medical Center Göttingen and DZHK (German Center for Cardiovascular Research), Göttingen, Germany.,Division of Cardiology and Metabolism - Heart Failure, Cachexia and Sarcopenia, Department of Internal Medicine and Cardiology, Berlin-Brandenburg Center for Regenerative Therapies (BCRT) at Charité University Medicine, Berlin, Germany
| | - Jean-Luc Balligand
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Universite Catholique de Louvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover D-30625, Germany
| | | | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Partner site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Stefan Chlopicki
- Department of Pharmacology, Medical College, Jagiellonian University, Krakow, Poland
| | - John G Cleland
- University of Hull, Kingston upon Hull, UK.,National Heart and Lung Institute, Royal Brompton and Harefield Hospitals NHS Trust, Imperial College, London, UK
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexander Dietl
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Rodolphe Fischmeister
- Inserm UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | | | | | | | | | - Gilles de Keulenaer
- Laboratory of Physiopharmacology (University of Antwerp) and Department of Cardiology, ZNA Hospital, Antwerp, Belgium
| | - Giuseppe Limongelli
- Department of Cardiothoracic Sciences, Second University of Naples, Naples, Italy
| | | | - Lars H Lund
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josep Masip
- Intensive Care Department, Consorci Sanitari Integral, University of Barcelona, Spain
| | - Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Italy
| | - Christian Mueller
- Department of Cardiology and Cardiovascular Research Institute Basel (CRIB), University Hospital Basel, University of Basel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center Berlin, and German Centre for Cardiovascular Research (DZHK), Partner site Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Piotr Ponikowski
- Department of Cardiology, Medical University, Clinical Military Hospital, Wroclaw, Poland
| | - Arsen Ristić
- Department of Cardiology of the Clinical Center of Serbia and Belgrade University School of Medicine, Belgrade, Serbia
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Switzerland
| | | | - Hadi Skouri
- Division of Cardiology, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner siteGöttingen, Göttingen, Germany
| | - Alexandre Mebazaa
- Hôpital Lariboisière, Université Paris Diderot, Inserm U 942, Paris, France
| |
Collapse
|
29
|
Sarcomere integrated biosensor detects myofilament-activating ligands in real time during twitch contractions in live cardiac muscle. J Mol Cell Cardiol 2020; 147:49-61. [PMID: 32791214 DOI: 10.1016/j.yjmcc.2020.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 11/24/2022]
Abstract
The sarcomere is the functional unit of cardiac muscle, essential for normal heart function. To date, it has not been possible to study, in real time, thin filament-based activation dynamics in live cardiac muscle. We report here results from a cardiac troponin C (TnC) FRET-based biosensor integrated into the cardiac sarcomere via stoichiometric replacement of endogenous TnC. The TnC biosensor provides, for the first time, evidence of multiple thin filament activating ligands, including troponin I interfacing with TnC and cycling myosin, during a cardiac twitch. Results show that the TnC FRET biosensor transient significantly precedes that of peak twitch force. Using small molecules and genetic modifiers known to alter sarcomere activation, independently of the intracellular Ca2+ transient, the data show that the TnC biosensor detects significant effects of the troponin I switch domain as a sarcomere-activating ligand. Interestingly, the TnC biosensor also detected the effects of load-dependent altered myosin cycling, as shown by a significant delay in TnC biosensor transient inactivation during the isometric twitch. In addition, the TnC biosensor detected the effects of myosin as an activating ligand during the twitch by using a small molecule that directly alters cross-bridge cycling, independently of the intracellular Ca2+ transient. Collectively, these results aid in illuminating the basis of cardiac muscle contractile activation with implications for gene, protein, and small molecule-based strategies designed to target the sarcomere in regulating beat-to-beat heart performance in health and disease.
Collapse
|
30
|
Tadros HJ, Life CS, Garcia G, Pirozzi E, Jones EG, Datta S, Parvatiyar MS, Chase PB, Allen HD, Kim JJ, Pinto JR, Landstrom AP. Meta-analysis of cardiomyopathy-associated variants in troponin genes identifies loci and intragenic hot spots that are associated with worse clinical outcomes. J Mol Cell Cardiol 2020; 142:118-125. [PMID: 32278834 PMCID: PMC7275889 DOI: 10.1016/j.yjmcc.2020.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/01/2020] [Accepted: 04/05/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Troponin (TNN)-encoded cardiac troponins (Tn) are critical for sensing calcium and triggering myofilament contraction. TNN variants are associated with development of cardiomyopathy; however, recent advances in genetic analysis have identified rare population variants. It is unclear how certain variants are associated with disease while others are tolerated. OBJECTIVE To compare probands with TNNT2, TNNI3, and TNNC1 variants and utilize high-resolution variant comparison mapping of pathologic and rare population variants to identify loci associated with disease pathogenesis. METHODS Cardiomyopathy-associated TNN variants were identified in the literature and topology mapping conducted. Clinical features were compiled and compared. Rare population variants were obtained from the gnomAD database. Signal-to-noise (S:N) normalized pathologic variant frequency against population variant frequency. Abstract review of clinical phenotypes was applied to "significant" hot spots. RESULTS Probands were compiled (N = 70 studies, 224 probands) as were rare variants (N = 125,748 exomes; 15,708 genomes, MAF <0.001). TNNC1-positive probands demonstrated the youngest age of presentation (20.0 years; P = .016 vs TNNT2; P = .004 vs TNNI3) and the highest death, transplant, or ventricular fibrillation events (P = .093 vs TNNT2; P = .024 vs TNNI3; Kaplan Meir: P = .025). S:N analysis yielded hot spots of diagnostic significance within the tropomyosin-binding domains, α-helix 1, and the N-Terminus in TNNT2 with increased sudden cardiac death and ventricular fibrillation (P = .004). The inhibitory region and C-terminal region in TNNI3 exhibited increased restrictive cardiomyopathy (P =.008). HCM and RCM models tended to have increased calcium sensitivity and DCM decreased sensitivity (P < .001). DCM and HCM studies typically showed no differences in Hill coefficient which was decreased in RCM models (P < .001). CM models typically demonstrated no changes to Fmax (P = .239). CONCLUSION TNNC1-positive probands had younger ages of diagnosis and poorer clinical outcomes. Mapping of TNN variants identified locations in TNNT2 and TNNI3 associated with heightened pathogenicity, RCM diagnosis, and increased risk of sudden death.
Collapse
Affiliation(s)
- Hanna J Tadros
- Department of Pediatrics, Section of Cardiology, Baylor College of Medicine, Houston, TX, United States; Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Chelsea S Life
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, United States
| | - Gustavo Garcia
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, United States
| | - Elisa Pirozzi
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, United States
| | - Edward G Jones
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Michelle S Parvatiyar
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, United States
| | - P Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Hugh D Allen
- Department of Pediatrics, Section of Cardiology, Baylor College of Medicine, Houston, TX, United States
| | - Jeffrey J Kim
- Department of Pediatrics, Section of Cardiology, Baylor College of Medicine, Houston, TX, United States
| | - Jose R Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, United States
| | - Andrew P Landstrom
- Department of Pediatrics, Section of Cardiology, Baylor College of Medicine, Houston, TX, United States; Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
31
|
Abstract
Changes of intracellular Ca2+ concentration regulate many aspects of cardiac myocyte function. About 99% of the cytoplasmic calcium in cardiac myocytes is bound to buffers, and their properties will therefore have a major influence on Ca2+ signaling. This article considers the fundamental properties and identities of the buffers and how to measure them. It reviews the effects of buffering on the systolic Ca2+ transient and how this may change physiologically, and in heart failure and both atrial and ventricular arrhythmias, as well. It is concluded that the consequences of this strong buffering may be more significant than currently appreciated, and a fuller understanding is needed for proper understanding of cardiac calcium cycling and contractility.
Collapse
Affiliation(s)
- Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, UK (G.L.S.)
| | - David A Eisner
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, UK (D.A.E.)
| |
Collapse
|
32
|
Ezekian JE, Clippinger SR, Garcia JM, Yang Q, Denfield S, Jeewa A, Dreyer WJ, Zou W, Fan Y, Allen HD, Kim JJ, Greenberg MJ, Landstrom AP. Variant R94C in TNNT2-Encoded Troponin T Predisposes to Pediatric Restrictive Cardiomyopathy and Sudden Death Through Impaired Thin Filament Relaxation Resulting in Myocardial Diastolic Dysfunction. J Am Heart Assoc 2020; 9:e015111. [PMID: 32098556 PMCID: PMC7335540 DOI: 10.1161/jaha.119.015111] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Pediatric‐onset restrictive cardiomyopathy (RCM) is associated with high mortality, but underlying mechanisms of disease are under investigated. RCM‐associated diastolic dysfunction secondary to variants in TNNT2‐encoded cardiac troponin T (TNNT2) is poorly described. Methods and Results Genetic analysis of a proband and kindred with RCM identified TNNT2‐R94C, which cosegregated in a family with 2 generations of RCM, ventricular arrhythmias, and sudden death. TNNT2‐R94C was absent among large, population‐based cohorts Genome Aggregation Database (gnomAD) and predicted to be pathologic by in silico modeling. Biophysical experiments using recombinant human TNNT2‐R94C demonstrated impaired cardiac regulation at the molecular level attributed to reduced calcium‐dependent blocking of myosin's interaction with the thin filament. Computational modeling predicted a shift in the force‐calcium curve for the R94C mutant toward submaximal calcium activation compared within the wild type, suggesting low levels of muscle activation even at resting calcium concentrations and hypercontractility following activation by calcium. Conclusions The pathogenic TNNT2‐R94C variant activates thin‐filament–mediated sarcomeric contraction at submaximal calcium concentrations, likely resulting in increased muscle tension during diastole and hypercontractility during systole. This describes the proximal biophysical mechanism for development of RCM in this family.
Collapse
Affiliation(s)
- Jordan E Ezekian
- Division of Paediatric Cardiology Department of Pediatrics Duke University School of Medicine Durham NC
| | - Sarah R Clippinger
- Department of Biochemistry and Molecular Biophysics Washington University in St. Louis St. Louis MO
| | - Jaquelin M Garcia
- Department of Biochemistry and Molecular Biophysics Washington University in St. Louis St. Louis MO
| | - Qixin Yang
- Division of Paediatric Cardiology Department of Pediatrics Duke University School of Medicine Durham NC
| | - Susan Denfield
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Aamir Jeewa
- Department of Pediatrics The Hospital for Sick Children Toronto Ontario Canada
| | - William J Dreyer
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Wenxin Zou
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Yuxin Fan
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Hugh D Allen
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Jeffrey J Kim
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics Washington University in St. Louis St. Louis MO
| | - Andrew P Landstrom
- Division of Paediatric Cardiology Department of Pediatrics Duke University School of Medicine Durham NC.,Department of Cell Biology Duke University School of Medicine Durham NC
| |
Collapse
|
33
|
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited heart disease and defined by unexplained isolated progressive myocardial hypertrophy, systolic and diastolic ventricular dysfunction, arrhythmias, sudden cardiac death and histopathologic changes, such as myocyte disarray and myocardial fibrosis. Mutations in genes encoding for proteins of the contractile apparatus of the cardiomyocyte, such as β-myosin heavy chain and myosin binding protein C, have been identified as cause of the disease. Disease is caused by altered biophysical properties of the cardiomyocyte, disturbed calcium handling, and abnormal cellular metabolism. Mutations in sarcomere genes can also activate other signaling pathways via transcriptional activation and can influence non-cardiac cells, such as fibroblasts. Additional environmental, genetic and epigenetic factors result in heterogeneous disease expression. The clinical course of the disease varies greatly with some patients presenting during childhood while others remain asymptomatic until late in life. Patients can present with either heart failure symptoms or the first symptom can be sudden death due to malignant ventricular arrhythmias. The morphological and pathological heterogeneity results in prognosis uncertainty and makes patient management challenging. Current standard therapeutic measures include the prevention of sudden death by prohibition of competitive sport participation and the implantation of cardioverter-defibrillators if indicated, as well as symptomatic heart failure therapies or cardiac transplantation. There exists no causal therapy for this monogenic autosomal-dominant inherited disorder, so that the focus of current management is on early identification of asymptomatic patients at risk through molecular diagnostic and clinical cascade screening of family members, optimal sudden death risk stratification, and timely initiation of preventative therapies to avoid disease progression to the irreversible adverse myocardial remodeling stage. Genetic diagnosis allowing identification of asymptomatic affected patients prior to clinical disease onset, new imaging technologies, and the establishment of international guidelines have optimized treatment and sudden death risk stratification lowering mortality dramatically within the last decade. However, a thorough understanding of underlying disease pathogenesis, regular clinical follow-up, family counseling, and preventative treatment is required to minimize morbidity and mortality of affected patients. This review summarizes current knowledge about molecular genetics and pathogenesis of HCM secondary to mutations in the sarcomere and provides an overview about current evidence and guidelines in clinical patient management. The overview will focus on clinical staging based on disease mechanism allowing timely initiation of preventative measures. An outlook about so far experimental treatments and potential for future therapies will be provided.
Collapse
Affiliation(s)
- Cordula Maria Wolf
- Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Center Munich, Technical University Munich, Munich, Germany
| |
Collapse
|
34
|
Cunningham KS, Spears DA, Care M. Evaluation of cardiac hypertrophy in the setting of sudden cardiac death. Forensic Sci Res 2019; 4:223-240. [PMID: 31489388 PMCID: PMC6713129 DOI: 10.1080/20961790.2019.1633761] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 01/06/2023] Open
Abstract
Ventricular hypertrophy is a common pathological finding at autopsy that can act as a substrate for arrhythmogenesis. Pathologists grapple with the significance of ventricular hypertrophy when assessing the sudden and unexpected deaths of young people and what it could mean for surviving family members. The pathological spectrum of left ventricular hypertrophy (LVH) is reviewed herein. This article is oriented to the practicing autopsy pathologist to help make sense of various patterns of increased heart muscle, particularly those that are not clearly cardiomyopathic, yet present in the setting of sudden cardiac death. The article also reviews factors influencing arrhythmogenesis as well as genetic mutations most commonly associated with ventricular hypertrophy, especially those associated with hypertrophic cardiomyopathy (HCM).
Collapse
Affiliation(s)
- Kristopher S. Cunningham
- Department of Laboratory Medicine and Pathobiology, Ontario Forensic Pathology Service, University of Toronto, Toronto, Canada
| | - Danna A. Spears
- University Health Network, Division of Cardiology – Electrophysiology, University of Toronto, Toronto, Canada
| | - Melanie Care
- Fred A. Litwin Family Centre in Genetic Medicine and Inherited Arrhythmia Clinic, University Health Network & Mount Sinai Hospital, University of Toronto, Toronto, Canada
| |
Collapse
|
35
|
Wijnker PJ, Sequeira V, Kuster DW, van der Velden J. Hypertrophic Cardiomyopathy: A Vicious Cycle Triggered by Sarcomere Mutations and Secondary Disease Hits. Antioxid Redox Signal 2019; 31:318-358. [PMID: 29490477 PMCID: PMC6602117 DOI: 10.1089/ars.2017.7236] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 02/23/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
Abstract
Significance: Hypertrophic cardiomyopathy (HCM) is a cardiac genetic disease characterized by left ventricular hypertrophy, diastolic dysfunction, and myocardial disarray. Disease onset occurs between 20 and 50 years of age, thus affecting patients in the prime of their life. HCM is caused by mutations in sarcomere proteins, the contractile building blocks of the heart. Despite increased knowledge of causal mutations, the exact path from genetic defect leading to cardiomyopathy is complex and involves additional disease hits. Recent Advances: Laboratory-based studies indicate that HCM development not only depends on the primary sarcomere impairment caused by the mutation but also on secondary disease-related alterations in the heart. Here we propose a vicious mutation-induced disease cycle, in which a mutation-induced energy depletion alters cellular metabolism with increased mitochondrial work, which triggers secondary disease modifiers that will worsen disease and ultimately lead to end-stage HCM. Critical Issues: Evidence shows excessive cellular reactive oxygen species (ROS) in HCM patients and HCM animal models. Oxidative stress markers are increased in the heart (oxidized proteins, DNA, and lipids) and serum of HCM patients. In addition, increased mitochondrial ROS production and changes in endogenous antioxidants are reported in HCM. Mutant sarcomeric protein may drive excessive levels of cardiac ROS via changes in cardiac efficiency and metabolism, mitochondrial activation and/or dysfunction, impaired protein quality control, and microvascular dysfunction. Future Directions: Interventions restoring metabolism, mitochondrial function, and improved ROS balance may be promising therapeutic approaches. We discuss the effects of current HCM pharmacological therapies and potential future therapies to prevent and reverse HCM. Antioxid. Redox Signal. 31, 318-358.
Collapse
Affiliation(s)
- Paul J.M. Wijnker
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Vasco Sequeira
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Diederik W.D. Kuster
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
36
|
Marston S. Small molecule studies: the fourth wave of muscle research. J Muscle Res Cell Motil 2019; 40:69-76. [PMID: 31228047 PMCID: PMC6726831 DOI: 10.1007/s10974-019-09526-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/13/2019] [Indexed: 12/28/2022]
Abstract
The study of muscle and contractility is an unusual scientific endeavour since it has from the start been focussed on one problem-What makes muscle work?-and yet has needed a vast range of different approaches and techniques to study it. Its uniqueness lies in the fundamental fascination of a large scale molecular machine that converts chemical energy into mechanical energy at ambient temperature and with high efficiency that is also controlled by an exquisitely intricate yet utterly reliable regulatory system and is an essential component of animal life. The investigation of muscle is as innovative as any other field of research. As soon as one approach appears to be played out another comes along. It is instructive to consider this as a series of waves of novel and heightened activity starting in the 1950s. The thesis of this article is that we are approaching the fourth wave with the recent rise of interest in small molecules as research tools and possible therapies for muscle diseases.
Collapse
Affiliation(s)
- Steven Marston
- Cardiovascular Division, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
37
|
Kresin N, Stücker S, Krämer E, Flenner F, Mearini G, Münch J, Patten M, Redwood C, Carrier L, Friedrich FW. Analysis of Contractile Function of Permeabilized Human Hypertrophic Cardiomyopathy Multicellular Heart Tissue. Front Physiol 2019; 10:239. [PMID: 30984009 PMCID: PMC6447666 DOI: 10.3389/fphys.2019.00239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 02/22/2019] [Indexed: 01/08/2023] Open
Affiliation(s)
- Nico Kresin
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sabrina Stücker
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Elisabeth Krämer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Frederik Flenner
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Giulia Mearini
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Julia Münch
- University Heart Center Hamburg, Hamburg, Germany
| | | | - Charles Redwood
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Felix W Friedrich
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
38
|
In vitro analyses of suspected arrhythmogenic thin filament variants as a cause of sudden cardiac death in infants. Proc Natl Acad Sci U S A 2019; 116:6969-6974. [PMID: 30886088 DOI: 10.1073/pnas.1819023116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Sudden unexpected death of an infant (SUDI) is a devastating occurrence for families. To investigate the genetic pathogenesis of SUDI, we sequenced >70 genes from 191 autopsy-negative SUDI victims. Ten infants sharing a previously unknown variant in troponin I (TnI) were identified. The mutation (TNNI1 R37C+/-) is in the fetal/neonatal paralog of TnI, a gene thought to be expressed in the heart up to the first 24 months of life. Using phylogenetic analysis and molecular dynamics simulations, it was determined that arginine at residue 37 in TNNI1 may play a critical functional role, suggesting that the variant may be pathogenic. We investigated the biophysical properties of the TNNI1 R37C mutation in human reconstituted thin filaments (RTFs) using fluorometry. RTFs reconstituted with the mutant R37C TnI exhibited reduced Ca2+-binding sensitivity due to an increased Ca2+ off-rate constant. Furthermore, we generated TNNI1 R37C+/- mutants in human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) using CRISPR-Cas9. In monolayers of hiPSC-CMs, we simultaneously monitored voltage and Ca2+ transients through optical mapping and compared them to their isogenic controls. We observed normal intrinsic beating patterns under control conditions in TNNI1 R37C+/- at stimulation frequencies of 55 beats/min (bpm), but these cells showed no restitution with increased stimulation frequency to 65 bpm and exhibited alternans at >75 bpm. The WT hiPSC-CMs did not exhibit any sign of arrhythmogenicity even at stimulation frequencies of 120 bpm. The approach used in this study provides critical physiological and mechanistic bases to investigate sarcomeric mutations in the pathogenesis of SUDI.
Collapse
|
39
|
Ramachandra CJ, Mai Ja KPM, Lin YH, Shim W, Boisvert WA, Hausenloy DJ. INDUCED PLURIPOTENT STEM CELLS FOR MODELLING ENERGETIC ALTERATIONS IN HYPERTROPHIC CARDIOMYOPATHY. CONDITIONING MEDICINE 2019; 2:142-151. [PMID: 32457935 PMCID: PMC7250397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is one of the most commonly inherited cardiac disorders that manifests with increased ventricular wall thickening, cardiomyocyte hypertrophy, disarrayed myofibers and interstitial fibrosis. The major pathophysiological features include, diastolic dysfunction, obstruction of the left ventricular outflow tract and cardiac arrhythmias. Mutations in genes that encode mostly for sarcomeric proteins have been associated with HCM but, despite the abundant research conducted to decipher the molecular mechanisms underlying the disease, it remains unclear as to how a primary defect in the sarcomere could lead to secondary phenotypes such as cellular hypertrophy. Mounting evidence suggests energy deficiency could be an important contributor of disease pathogenesis as well. Various animal models of HCM have been generated for gaining deeper insight into disease pathogenesis, however species variation between animals and humans, as well as the limited availability of human myocardial samples, has encouraged researchers to seek alternative 'humanized' models. Using induced pluripotent stem cells (iPSCs), human cardiomyocytes (CMs) have been generated from patients with HCM for investigating disease mechanisms. While these HCM-iPSC models demonstrate most of the phenotypic traits, it is important to ascertain if they recapitulate all pathophysiological features, especially that of energy deficiency. In this review we discuss the currently established HCM-iPSC models with emphasis on altered energetics.
Collapse
Affiliation(s)
- Chrishan J.A. Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - K P Myu Mai Ja
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Ying-Hsi Lin
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Winston Shim
- Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore
| | - William A. Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, USA
| | - Derek J. Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, London, UK
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico
| |
Collapse
|
40
|
Pearman CM, Madders GW, Radcliffe EJ, Kirkwood GJ, Lawless M, Watkins A, Smith CE, Trafford AW, Eisner DA, Dibb KM. Increased Vulnerability to Atrial Fibrillation Is Associated With Increased Susceptibility to Alternans in Old Sheep. J Am Heart Assoc 2018; 7:e009972. [PMID: 30520673 PMCID: PMC6405564 DOI: 10.1161/jaha.118.009972] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/17/2018] [Indexed: 12/17/2022]
Abstract
Background Atrial fibrillation ( AF ) is common in the elderly, but rare in the young; however, the changes that occur with age that promote AF are not fully understood. Action potential ( AP ) alternans may be involved in the initiation of AF . Using a translationally relevant model, we investigated whether age-associated atrial vulnerability to AF was associated with susceptibility to AP alternans. Methods and Results AF was induced in conscious young and old sheep using 50 Hz burst pacing. Old sheep were more vulnerable to AF . Monophasic and cellular AP s were recorded from the right atrium in vivo and from myocytes isolated from the left and right atrial appendages. AP alternans occurred at lower stimulation frequencies in old sheep than young in vivo (old, 3.0±0.1 Hz; young, 3.3±0.1 Hz; P<0.05) and in isolated myocytes (old, 1.6±0.1 Hz; young, 2.0±0.1 Hz; P<0.05). Simultaneous recordings of [Ca2+]i and membrane potential in myocytes showed that alternans of AP s and [Ca2+]i often occurred together. However, at low stimulation rates [Ca2+]i alternans could occur without AP alternans, whereas at high stimulation rates AP alternans could still be observed despite disabling Ca2+ cycling using thapsigargin. Conclusions We have shown, for the first time in a large mammalian model, that aging is associated with increased duration of AF and susceptibility to AP alternans. We suggest that instabilities in Ca2+ handling initiate alternans at low stimulation rates, but that AP restitution alone can sustain alternans at higher rates.
Collapse
|
41
|
Dieseldorff Jones KM, Koh Y, Weller RS, Turna RS, Ahmad F, Huke S, Knollmann BC, Pinto JR, Hwang HS. Pathogenic troponin T mutants with opposing effects on myofilament Ca 2+ sensitivity attenuate cardiomyopathy phenotypes in mice. Arch Biochem Biophys 2018; 661:125-131. [PMID: 30445044 DOI: 10.1016/j.abb.2018.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/28/2018] [Accepted: 11/08/2018] [Indexed: 01/14/2023]
Abstract
Mutations in cardiac troponin T (TnT) associated with hypertrophic cardiomyopathy generally lead to an increase in the Ca2+ sensitivity of contraction and susceptibility to arrhythmias. In contrast, TnT mutations linked to dilated cardiomyopathy decrease the Ca2+ sensitivity of contraction. Here we tested the hypothesis that two TnT disease mutations with opposite effects on myofilament Ca2+ sensitivity can attenuate each other's phenotype. We crossed transgenic mice expressing the HCM TnT-I79N mutation (I79N) with a DCM knock-in mouse model carrying the heterozygous TnT-R141W mutation (HET). The results of the Ca2+ sensitivity in skinned cardiac muscle preparations ranked from highest to lowest were as follow: I79N > I79N/HET > NTg > HET. Echocardiographic measurements revealed an improvement in hemodynamic parameters in I79N/HET compared to I79N and normalization of left ventricular dimensions and volumes compared to both I79N and HET. Ex vivo testing showed that the I79N/HET mouse hearts had reduced arrhythmia susceptibility compared to I79N mice. These results suggest that two disease mutations in TnT that have opposite effects on the myofilament Ca2+ sensitivity can paradoxically ameliorate each other's disease phenotype. Normalizing myofilament Ca2+ sensitivity may be a promising new treatment approach for a variety of diseases.
Collapse
Affiliation(s)
| | - Yeojung Koh
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Rebecca S Weller
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Rajdeep S Turna
- Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Ferhaan Ahmad
- Department of Internal Medicine University of Iowa, Iowa City, IA, USA
| | - Sabine Huke
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA; Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Björn C Knollmann
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA
| | | | - Hyun Seok Hwang
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
42
|
Coppini R, Ferrantini C, Mugelli A, Poggesi C, Cerbai E. Altered Ca 2+ and Na + Homeostasis in Human Hypertrophic Cardiomyopathy: Implications for Arrhythmogenesis. Front Physiol 2018; 9:1391. [PMID: 30420810 PMCID: PMC6215954 DOI: 10.3389/fphys.2018.01391] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/13/2018] [Indexed: 12/12/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common mendelian heart disease, with a prevalence of 1/500. HCM is a primary cause of sudden death, due to an heightened risk of ventricular tachyarrhythmias that often occur in young asymptomatic patients. HCM can slowly progress toward heart failure, either with preserved or reduced ejection fraction, due to worsening of diastolic function. Accumulation of intra-myocardial fibrosis and replacement scars underlies heart failure progression and represents a substrate for sustained arrhythmias in end-stage patients. However, arrhythmias and mechanical abnormalities may occur in hearts with little or no fibrosis, prompting toward functional pathomechanisms. By studying viable cardiomyocytes and trabeculae isolated from inter-ventricular septum samples of non-failing HCM patients with symptomatic obstruction who underwent myectomy operations, we identified that specific abnormalities of intracellular Ca2+ handling are associated with increased cellular arrhytmogenesis and diastolic dysfunction. In HCM cardiomyocytes, diastolic Ca2+ concentration is increased both in the cytosol and in the sarcoplasmic reticulum and the rate of Ca2+ transient decay is slower, while the amplitude of Ca2+-release is preserved. Ca2+ overload is the consequence of an increased Ca2+ entry via L-type Ca2+-current [due to prolongation the action potential (AP) plateau], combined with a reduced rate of Ca2+-extrusion through the Na+/Ca2+ exchanger [due to increased cytosolic (Na+)] and a lower expression of SERCA. Increased late Na+ current (INaL) plays a major role, as it causes both AP prolongation and Na+ overload. Intracellular Ca2+ overload determines an higher frequency of Ca2+ waves leading to delayed-afterdepolarizations (DADs) and premature contractions, but is also linked with the increased diastolic tension and slower relaxation of HCM myocardium. Sustained increase of intracellular [Ca2+] goes hand-in-hand with the increased activation of Ca2+/calmodulin-dependent protein-kinase-II (CaMKII) and augmented phosphorylation of its targets, including Ca2+ handling proteins. In transgenic HCM mouse models, we found that Ca2+ overload, CaMKII and increased INaL drive myocardial remodeling since the earliest stages of disease and underlie the development of hypertrophy, diastolic dysfunction and the arrhythmogenic substrate. In conclusion, diastolic dysfunction and arrhythmogenesis in human HCM myocardium are driven by functional alterations at cellular and molecular level that may be targets of innovative therapies.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department of Neuroscience, Psychology, Drug Sciences and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro Mugelli
- Department of Neuroscience, Psychology, Drug Sciences and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elisabetta Cerbai
- Department of Neuroscience, Psychology, Drug Sciences and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
43
|
Eschenhagen T, Carrier L. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes-a systematic review. Pflugers Arch 2018; 471:755-768. [PMID: 30324321 PMCID: PMC6475632 DOI: 10.1007/s00424-018-2214-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Human-induced pluripotent stem cells (hiPSC) can be differentiated to cardiomyocytes at high efficiency and are increasingly used to study cardiac disease in a human context. This review evaluated 38 studies on hypertrophic (HCM) and dilated cardiomyopathy (DCM) of different genetic causes asking to which extent published data allow the definition of an in vitro HCM/DCM hiPSC-CM phenotype. The data are put in context with the prevailing hypotheses on HCM/DCM dysfunction and pathophysiology. Relatively consistent findings in HCM not reported in DCM were larger cell size (156 ± 85%, n = 15), more nuclear localization of nuclear factor of activated T cells (NFAT; 175 ± 65%, n = 3), and higher β-myosin heavy chain gene expression levels (500 ± 547%, n = 8) than respective controls. Conversely, DCM lines showed consistently less force development than controls (47 ± 23%, n = 9), while HCM forces scattered without clear trend. Both HCM and DCM lines often showed sarcomere disorganization, higher NPPA/NPPB expression levels, and arrhythmic beating behaviour. The data have to be taken with the caveat that reporting frequencies of the various parameters (e.g. cell size, NFAT expression) differ widely between HCM and DCM lines, in which data scatter is large and that only 9/38 studies used isogenic controls. Taken together, the current data provide interesting suggestions for disease-specific phenotypes in HCM/DCM hiPSC-CM but indicate that the field is still in its early days. Systematic, quantitative comparisons and robust, high content assays are warranted to advance the field.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| |
Collapse
|
44
|
Gonzalez-Martinez D, Johnston JR, Landim-Vieira M, Ma W, Antipova O, Awan O, Irving TC, Bryant Chase P, Pinto JR. Structural and functional impact of troponin C-mediated Ca 2+ sensitization on myofilament lattice spacing and cross-bridge mechanics in mouse cardiac muscle. J Mol Cell Cardiol 2018; 123:26-37. [PMID: 30138628 DOI: 10.1016/j.yjmcc.2018.08.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/26/2018] [Accepted: 08/05/2018] [Indexed: 12/25/2022]
Abstract
Acto-myosin cross-bridge kinetics are important for beat-to-beat regulation of cardiac contractility; however, physiological and pathophysiological mechanisms for regulation of contractile kinetics are incompletely understood. Here we explored whether thin filament-mediated Ca2+ sensitization influences cross-bridge kinetics in permeabilized, osmotically compressed cardiac muscle preparations. We used a murine model of hypertrophic cardiomyopathy (HCM) harboring a cardiac troponin C (cTnC) Ca2+-sensitizing mutation, Ala8Val in the regulatory N-domain. We also treated wild-type murine muscle with bepridil, a cTnC-targeting Ca2+ sensitizer. Our findings suggest that both methods of increasing myofilament Ca2+ sensitivity increase cross-bridge cycling rate measured by the rate of tension redevelopment (kTR); force per cross-bridge was also enhanced as measured by sinusoidal stiffness and I1,1/I1,0 ratio from X-ray diffraction. Computational modeling suggests that Ca2+ sensitization through this cTnC mutation or bepridil accelerates kTR primarily by promoting faster cross-bridge detachment. To elucidate if myofilament structural rearrangements are associated with changes in kTR, we used small angle X-ray diffraction to simultaneously measure myofilament lattice spacing and isometric force during steady-state Ca2+ activations. Within in vivo lattice dimensions, lattice spacing and steady-state isometric force increased significantly at submaximal activation. We conclude that the cTnC N-domain controls force by modulating both the number and rate of cycling cross-bridges, and that the both methods of Ca2+ sensitization may act through stabilization of cTnC's D-helix. Furthermore, we propose that the transient expansion of the myofilament lattice during Ca2+ activation may be an additional factor that could increase the rate of cross-bridge cycling in cardiac muscle. These findings may have implications for the pathophysiology of HCM.
Collapse
Affiliation(s)
- David Gonzalez-Martinez
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Jamie R Johnston
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Weikang Ma
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, USA
| | - Olga Antipova
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, USA; X-Ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL, USA
| | - Omar Awan
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Thomas C Irving
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL, USA
| | - P Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - J Renato Pinto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
45
|
Translating emerging molecular genetic insights into clinical practice in inherited cardiomyopathies. J Mol Med (Berl) 2018; 96:993-1024. [PMID: 30128729 DOI: 10.1007/s00109-018-1685-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/22/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022]
Abstract
Cardiomyopathies are primarily genetic disorders of the myocardium associated with higher risk of life-threatening cardiac arrhythmias, heart failure, and sudden cardiac death. The evolving knowledge in genomic medicine during the last decade has reshaped our understanding of cardiomyopathies as diseases of multifactorial nature and complex pathophysiology. Genetic testing in cardiomyopathies has subsequently grown from primarily a research tool into an essential clinical evaluation piece with important clinical implications for patients and their families. The purpose of this review is to provide with a contemporary insight into the implications of genetic testing in diagnosis, therapy, and prognosis of patients with inherited cardiomyopathies. Here, we summarize the contemporary knowledge on genotype-phenotype correlations in inherited cardiomyopathies and highlight the recent significant achievements in the field of translational cardiovascular genetics.
Collapse
|
46
|
Jordà P, García-Álvarez A. Hypertrophic cardiomyopathy: Sudden cardiac death risk stratification in adults. Glob Cardiol Sci Pract 2018; 2018:25. [PMID: 30393637 PMCID: PMC6209451 DOI: 10.21542/gcsp.2018.25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Paloma Jordà
- Cardiology Department, Institut Clínic Cardiovascular, Hospital Clínic, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ana García-Álvarez
- Cardiology Department, Institut Clínic Cardiovascular, Hospital Clínic, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
47
|
Monasky MM, Pappone C, Piccoli M, Ghiroldi A, Micaglio E, Anastasia L. Calcium in Brugada Syndrome: Questions for Future Research. Front Physiol 2018; 9:1088. [PMID: 30147658 PMCID: PMC6095984 DOI: 10.3389/fphys.2018.01088] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022] Open
Abstract
The Brugada syndrome (BrS) is characterized by coved-type ST-segment elevation in the right precordial leads on the electrocardiogram (ECG) and increased risk of sudden cardiac death (SCD). While it is an inheritable disease, determining the true prevalence is a challenge, since patients may report no known family history of the syndrome, present with a normal spontaneous ECG pattern at the time of examination, and test negative for all known BrS-causative genes. In fact, SCD is often the first indication that a person is affected by the syndrome. Men are more likely to be symptomatic than women. Abnormal, low-voltage, fractionated electrograms have been found in the epicardium of the right ventricular outflow tract (RVOT). Ablation of this area abolishes the abnormal electrograms and helps to prevent arrhythmic recurrences. BrS patients are more likely to experience ventricular tachycardia/fibrillation (VT/VF) during fever or during an increase in vagal tone. Isoproterenol helps to reverse the ECG BrS phenotype. In this review, we discuss roles of calcium in various conditions that are relevant to BrS, such as changes in temperature, heart rate, and vagal tone, and the effects of gender and isoproterenol on calcium handling. Studies are warranted to further investigate these mechanisms in models of BrS.
Collapse
Affiliation(s)
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy
| | - Andrea Ghiroldi
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy
| | - Emanuele Micaglio
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Luigi Anastasia
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
48
|
Wen Q, Gandhi K, Capel RA, Hao G, O'Shea C, Neagu G, Pearcey S, Pavlovic D, Terrar DA, Wu J, Faggian G, Camelliti P, Lei M. Transverse cardiac slicing and optical imaging for analysis of transmural gradients in membrane potential and Ca 2+ transients in murine heart. J Physiol 2018; 596:3951-3965. [PMID: 29928770 PMCID: PMC6117587 DOI: 10.1113/jp276239] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/12/2018] [Indexed: 11/18/2022] Open
Abstract
Key points A robust cardiac slicing approach was developed for optical mapping of transmural gradients in transmembrane potential (Vm) and intracellular Ca2+ transient (CaT) of murine heart. Significant transmural gradients in Vm and CaT were observed in the left ventricle. Frequency‐dependent action potentials and CaT alternans were observed in all ventricular regions with rapid pacing, with significantly greater incidence in the endocardium than epicardium. The observations demonstrate the feasibility of our new approach to cardiac slicing for systematic analysis of intrinsic transmural and regional gradients in Vm and CaT.
Abstract Transmural and regional gradients in membrane potential and Ca2+ transient in the murine heart are largely unexplored. Here, we developed and validated a robust approach which combines transverse ultra‐thin cardiac slices and high resolution optical mapping to enable systematic analysis of transmural and regional gradients in transmembrane potential (Vm) and intracellular Ca2+ transient (CaT) across the entire murine ventricles. The voltage dye RH237 or Ca2+ dye Rhod‐2 AM were loaded through the coronary circulation using a Langendorff perfusion system. Short‐axis slices (300 μm thick) were prepared from the entire ventricles (from the apex to the base) by using a high‐precision vibratome. Action potentials (APs) and CaTs were recorded with optical mapping during steady‐state baseline and rapid pacing. Significant transmural gradients in Vm and CaT were observed in the left ventricle, with longer AP duration (APD50 and APD75) and CaT duration (CaTD50 and CaTD75) in the endocardium compared with that in the epicardium. No significant regional gradients were observed along the apico‐basal axis of the left ventricle. Interventricular gradients were detected with significantly shorter APD50, APD75 and CaTD50 in the right ventricle compared with left ventricle and ventricular septum. During rapid pacing, AP and CaT alternans were observed in most ventricular regions, with significantly greater incidence in the endocardium in comparison with epicardium. In conclusion, these observations demonstrate the feasibility of our new approach to cardiac slicing for systematic analysis of intrinsic transmural and regional gradients in Vm and CaT in murine ventricular tissue. A robust cardiac slicing approach was developed for optical mapping of transmural gradients in transmembrane potential (Vm) and intracellular Ca2+ transient (CaT) of murine heart. Significant transmural gradients in Vm and CaT were observed in the left ventricle. Frequency‐dependent action potentials and CaT alternans were observed in all ventricular regions with rapid pacing, with significantly greater incidence in the endocardium than epicardium. The observations demonstrate the feasibility of our new approach to cardiac slicing for systematic analysis of intrinsic transmural and regional gradients in Vm and CaT.
Collapse
Affiliation(s)
- Q Wen
- Institution of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - K Gandhi
- Medical School, University of Verona, Verona, Italy
| | - Rebecca A Capel
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - G Hao
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 6400, China
| | - C O'Shea
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - G Neagu
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - S Pearcey
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - D Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Derek A Terrar
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - J Wu
- Institution of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - G Faggian
- Medical School, University of Verona, Verona, Italy
| | | | - M Lei
- Department of Pharmacology, University of Oxford, Oxford, UK.,Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 6400, China
| |
Collapse
|
49
|
Philipson DJ, DePasquale EC, Yang EH, Baas AS. Emerging pharmacologic and structural therapies for hypertrophic cardiomyopathy. Heart Fail Rev 2018; 22:879-888. [PMID: 28856513 DOI: 10.1007/s10741-017-9648-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Hypertrophic cardiomyopathy is the most common inherited heart disease. Although it was first described over 50 years ago, there has been little in the way of novel disease-specific therapeutic development for these patients. Current treatment practice largely aims at symptomatic control using old drugs made for other diseases and does little to modify the disease course. Septal reduction by surgical myectomy or percutaneous alcohol septal ablation are well-established treatments for pharmacologic-refractory left ventricular outflow tract obstruction in hypertrophic cardiomyopathy patients. In recent years, there has been a relative surge in the development of innovative therapeutics, which aim to target the complex molecular pathophysiology and resulting hemodynamics that underlie hypertrophic cardiomyopathy. Herein, we review the new and emerging therapeutics for hypertrophic cardiomyopathy, which include pharmacologic attenuation of sarcomeric calcium sensitivity, allosteric inhibition of cardiac myosin, myocardial metabolic modulation, and renin-angiotensin-aldosterone system inhibition, as well as structural intervention by percutaneous mitral valve plication and endocardial radiofrequency ablation of septal hypertrophy. In conclusion, while further development of these therapeutic strategies is ongoing, they each mark a significant and promising advancement in treatment for hypertrophic cardiomyopathy patients.
Collapse
Affiliation(s)
- Daniel J Philipson
- Department of Medicine, UCLA, 200 UCLA Medical Plaza Suite 420, Los Angeles, CA, 90095, USA.
| | - Eugene C DePasquale
- Ahmanson-UCLA Cardiomyopathy Center, Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Eric H Yang
- Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Arnold S Baas
- Ahmanson-UCLA Cardiomyopathy Center, Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA, USA
| |
Collapse
|
50
|
Alves ML, Warren CM, Simon JN, Gaffin RD, Montminy EM, Wieczorek DF, Solaro RJ, Wolska BM. Early sensitization of myofilaments to Ca2+ prevents genetically linked dilated cardiomyopathy in mice. Cardiovasc Res 2018; 113:915-925. [PMID: 28379313 DOI: 10.1093/cvr/cvx068] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/31/2017] [Indexed: 12/14/2022] Open
Abstract
Background Dilated cardiomoypathies (DCM) are a heterogeneous group of inherited and acquired diseases characterized by decreased contractility and enlargement of cardiac chambers and a major cause of morbidity and mortality. Mice with Glu54Lys mutation in α-tropomyosin (Tm54) demonstrate typical DCM phenotype with reduced myofilament Ca2+ sensitivity. We tested the hypothesis that early sensitization of the myofilaments to Ca2+ in DCM can prevent the DCM phenotype. Methods and results To sensitize Tm54 myofilaments, we used a genetic approach and crossbred Tm54 mice with mice expressing slow skeletal troponin I (ssTnI) that sensitizes myofilaments to Ca2+. Four groups of mice were used: non-transgenic (NTG), Tm54, ssTnI and Tm54/ssTnI (DTG). Systolic function was significantly reduced in the Tm54 mice compared to NTG, but restored in DTG mice. Tm54 mice also showed increased diastolic LV dimensions and HW/BW ratios, when compared to NTG, which were improved in the DTG group. β-myosin heavy chain expression was increased in the Tm54 animals compared to NTG and was partially restored in DTG group. Analysis by 2D-DIGE indicated a significant decrease in two phosphorylated spots of cardiac troponin I (cTnI) in the DTG animals compared to NTG and Tm54. Analysis by 2D-DIGE also indicated no significant changes in troponin T, regulatory light chain, myosin binding protein C and tropomyosin phosphorylation. Conclusion Our data indicate that decreased myofilament Ca2+ sensitivity is an essential element in the pathophysiology of thin filament linked DCM. Sensitization of myofilaments to Ca2+ in the early stage of DCM may be a useful therapeutic strategy in thin filament linked DCM.
Collapse
Affiliation(s)
- Marco L Alves
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA.,Center for Research in Echocardiography and Cardiology, Heart Institute, University of Sao Paulo, Avenida Dr. Eneas de Carvalho Aguiar 44, 05403-900, Sao Paulo, Brazil
| | - Chad M Warren
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - Jillian N Simon
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - Robert D Gaffin
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - Eric M Montminy
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - David F Wieczorek
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - R John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA
| | - Beata M Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois, 835 S Wolcott Ave. (M/C 901), Chicago, IL 60612, USA.,Department of Medicine, Division of Cardiology, University of Illinois, 840 S Wood St. (M/C 715), Chicago, IL 60612, USA
| |
Collapse
|