1
|
Chakraborty P, Bokhari MM, Massé S, Azam MA, Lai PFH, Liang T, Si D, Bhaskaran A, Riazi S, Billia F, Nanthakumar K. Acute effects of dantrolene on the mechanical performance of myopathic human hearts. Heart Rhythm 2024; 21:2055-2057. [PMID: 38574788 DOI: 10.1016/j.hrthm.2024.03.1815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024]
Affiliation(s)
- Praloy Chakraborty
- Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mahmoud M Bokhari
- Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, University Health Network, Toronto, Ontario, Canada
| | - Mohammed A Azam
- The Hull Family Cardiac Fibrillation Management Laboratory, University Health Network, Toronto, Ontario, Canada
| | - Patrick F H Lai
- The Hull Family Cardiac Fibrillation Management Laboratory, University Health Network, Toronto, Ontario, Canada
| | - Timothy Liang
- The Hull Family Cardiac Fibrillation Management Laboratory, University Health Network, Toronto, Ontario, Canada
| | - Daoyuan Si
- The Hull Family Cardiac Fibrillation Management Laboratory, University Health Network, Toronto, Ontario, Canada
| | - Abhishek Bhaskaran
- Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Sheila Riazi
- Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Filio Billia
- Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Kumaraswamy Nanthakumar
- Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada; The Hull Family Cardiac Fibrillation Management Laboratory, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Pandit SV, Lampe JW, Silver AE. Recurrence of ventricular fibrillation in out-of-hospital cardiac arrest: Clinical evidence and underlying ionic mechanisms. J Physiol 2024; 602:4649-4667. [PMID: 38661672 DOI: 10.1113/jp284621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/08/2024] [Indexed: 04/26/2024] Open
Abstract
Defibrillation remains the optimal therapy for terminating ventricular fibrillation (VF) in out-of-hospital cardiac arrest (OHCA) patients, with reported shock success rates of ∼90%. A key persistent challenge, however, is the high rate of VF recurrence (∼50-80%) seen during post-shock cardiopulmonary resuscitation (CPR). Studies have shown that the incidence and time spent in recurrent VF are negatively associated with neurologically-intact survival. Recurrent VF also results in the administration of extra shocks at escalating energy levels, which can cause cardiac dysfunction. Unfortunately, the mechanisms underlying recurrent VF remain poorly understood. In particular, the role of chest-compressions (CC) administered during CPR in mediating recurrent VF remains controversial. In this review, we first summarize the available clinical evidence for refibrillation occurring during CPR in OHCA patients, including the postulated contribution of CC and non-CC related pathways. Next, we examine experimental studies highlighting how CC can re-induce VF via direct mechano-electric feedback. We postulate the ionic mechanisms involved by comparison with similar phenomena seen in commotio cordis. Subsequently, the hypothesized contribution of partial cardiac reperfusion (either as a result of CC or CC independent organized rhythm) in re-initiating VF in a globally ischaemic heart is examined. An overview of the proposed ionic mechanisms contributing to VF recurrence in OHCA during CPR from a cellular level to the whole heart is outlined. Possible therapeutic implications of the proposed mechanistic theories for VF recurrence in OHCA are briefly discussed.
Collapse
|
3
|
Zhao Y, Du B, Chakraborty P, Denham N, Massé S, Lai PF, Azam MA, Billia F, Thavendiranathan P, Abdel‐Qadir H, Lopaschuk GD, Nanthakumar K. Impaired Cardiac AMPK (5'-Adenosine Monophosphate-Activated Protein Kinase) and Ca 2+-Handling, and Action Potential Duration Heterogeneity in Ibrutinib-Induced Ventricular Arrhythmia Vulnerability. J Am Heart Assoc 2024; 13:e032357. [PMID: 38842296 PMCID: PMC11255774 DOI: 10.1161/jaha.123.032357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/03/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND We recently demonstrated that acute administration of ibrutinib, a Bruton's tyrosine kinase inhibitor used in chemotherapy for blood malignancies, increases ventricular arrhythmia (VA) vulnerability. A pathway of ibrutinib-induced vulnerability to VA that can be modulated for cardioprotection remains unclear. METHODS AND RESULTS The effects of ibrutinib on cardiac electrical activity and Ca2+ dynamics were investigated in Langendorff-perfused hearts using optical mapping. We also conducted Western blotting analysis to evaluate the impact of ibrutinib on various regulatory and Ca2+-handling proteins in rat cardiac tissues. Treatment with ibrutinib (10 mg/kg per day) for 4 weeks was associated with an increased VA inducibility (72.2%±6.3% versus 38.9±7.0% in controls, P<0.002) and shorter action potential durations during pacing at various frequencies (P<0.05). Ibrutinib also decreased heart rate thresholds for beat-to-beat duration alternans of the cardiac action potential (P<0.05). Significant changes in myocardial Ca2+ transients included lower amplitude alternans ratios (P<0.05), longer times-to-peak (P<0.05), and greater spontaneous intracellular Ca2+ elevations (P<0.01). We also found lower abundance and phosphorylation of myocardial AMPK (5'-adenosine monophosphate-activated protein kinase), indicating reduced AMPK activity in hearts after ibrutinib treatment. An acute treatment with the AMPK activator 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside ameliorated abnormalities in action potential and Ca2+ dynamics, and significantly reduced VA inducibility (37.1%±13.4% versus 72.2%±6.3% in the absence of 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside, P<0.05) in hearts from ibrutinib-treated rats. CONCLUSIONS VA vulnerability inflicted by ibrutinib may be mediated in part by an impairment of myocardial AMPK activity. Pharmacological activation of AMPK may be a protective strategy against ibrutinib-induced cardiotoxicity.
Collapse
MESH Headings
- Animals
- Adenine/analogs & derivatives
- Adenine/pharmacology
- Piperidines/pharmacology
- Action Potentials/drug effects
- Pyrimidines/pharmacology
- AMP-Activated Protein Kinases/metabolism
- Pyrazoles/pharmacology
- Male
- Arrhythmias, Cardiac/chemically induced
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/prevention & control
- Protein Kinase Inhibitors/pharmacology
- Heart Rate/drug effects
- Isolated Heart Preparation
- Calcium/metabolism
- Rats
- Disease Models, Animal
- Rats, Sprague-Dawley
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Calcium Signaling/drug effects
- Time Factors
Collapse
Affiliation(s)
- Yanan Zhao
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
- China‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Beibei Du
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
- China‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Nathan Denham
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Patrick F.H. Lai
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Mohammed Ali Azam
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| | - Filio Billia
- Toronto General Hospital Research InstituteTorontoCanada
- Ted Rogers Centre for Heart ResearchTorontoCanada
| | | | - Husam Abdel‐Qadir
- Toronto General Hospital Research InstituteTorontoCanada
- Ted Rogers Centre for Heart ResearchTorontoCanada
| | | | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General HospitalTorontoCanada
- Toronto General Hospital Research InstituteTorontoCanada
| |
Collapse
|
4
|
Zhao Y, Chakraborty P, Tomassetti J, Subha T, Massé S, Thavendiranathan P, Billia F, Lai PFH, Abdel-Qadir H, Nanthakumar K. Arrhythmogenic Ventricular Remodeling by Next-Generation Bruton's Tyrosine Kinase Inhibitor Acalabrutinib. Int J Mol Sci 2024; 25:6207. [PMID: 38892396 PMCID: PMC11173147 DOI: 10.3390/ijms25116207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiac arrhythmias remain a significant concern with Ibrutinib (IBR), a first-generation Bruton's tyrosine kinase inhibitor (BTKi). Acalabrutinib (ABR), a next-generation BTKi, is associated with reduced atrial arrhythmia events. However, the role of ABR in ventricular arrhythmia (VA) has not been adequately evaluated. Our study aimed to investigate VA vulnerability and ventricular electrophysiology following chronic ABR therapy in male Sprague-Dawley rats utilizing epicardial optical mapping for ventricular voltage and Ca2+ dynamics and VA induction by electrical stimulation in ex-vivo perfused hearts. Ventricular tissues were snap-frozen for protein analysis for sarcoplasmic Ca2+ and metabolic regulatory proteins. The results show that both ABR and IBR treatments increased VA vulnerability, with ABR showing higher VA regularity index (RI). IBR, but not ABR, is associated with the abbreviation of action potential duration (APD) and APD alternans. Both IBR and ABR increased diastolic Ca2+ leak and Ca2+ alternans, reduced conduction velocity (CV), and increased CV dispersion. Decreased SERCA2a expression and AMPK phosphorylation were observed with both treatments. Our results suggest that ABR treatment also increases the risk of VA by inducing proarrhythmic changes in Ca2+ signaling and membrane electrophysiology, as seen with IBR. However, the different impacts of these two BTKi on ventricular electrophysiology may contribute to differences in VA vulnerability and distinct VA characteristics.
Collapse
Affiliation(s)
- Yanan Zhao
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Praloy Chakraborty
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Julianna Tomassetti
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Tasnia Subha
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Stéphane Massé
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Paaladinesh Thavendiranathan
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
- Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Filio Billia
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
- Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Patrick F. H. Lai
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| | - Husam Abdel-Qadir
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
- Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Women’s College Hospital, Toronto, ON M5S 1B2, Canada
| | - Kumaraswamy Nanthakumar
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2M1, Canada; (Y.Z.); (P.C.); (J.T.); (T.S.); (S.M.); (P.T.); (F.B.); (P.F.H.L.); (H.A.-Q.)
| |
Collapse
|
5
|
Gao Y, Li S, Liu X, Si D, Chen W, Yang F, Sun H, Yang P. RyR2 Stabilizer Attenuates Cardiac Hypertrophy by Downregulating TNF-α/NF-κB/NLRP3 Signaling Pathway through Inhibiting Calcineurin. J Cardiovasc Transl Res 2024; 17:481-495. [PMID: 38652413 DOI: 10.1007/s12265-023-10376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/13/2023] [Indexed: 04/25/2024]
Abstract
The effect of Ryanodine receptor2 (RyR2) and its stabilizer on cardiac hypertrophy is not well known. C57/BL6 mice underwent transverse aortic contraction (TAC) or sham surgery were administered dantrolene, the RyR2 stabilizer, or control drug. Dantrolene significantly alleviated TAC-induced cardiac hypertrophy in mice, and RNA sequencing was performed implying calcineurin/NFAT3 and TNF-α/NF-κB/NLRP3 as critical signaling pathways. Further expression analysis and Western blot with heart tissue as well as neonatal rat cardiomyocyte (NRCM) model confirmed dantrolene decreases the activation of calcineurin/NFAT3 signaling pathway and TNF-α/NF-κB/NLRP3 signaling pathway, which was similar to FK506 and might be attenuated by calcineurin overexpression. The present study shows for the first time that RyR2 stabilizer dantrolene attenuates cardiac hypertrophy by inhibiting the calcineurin, therefore downregulating the TNF-α/NF-κB/NLRP3 pathway.
Collapse
MESH Headings
- Animals
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/metabolism
- Ryanodine Receptor Calcium Release Channel/metabolism
- Ryanodine Receptor Calcium Release Channel/genetics
- Ryanodine Receptor Calcium Release Channel/drug effects
- Calcineurin/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Mice, Inbred C57BL
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Disease Models, Animal
- NF-kappa B/metabolism
- Down-Regulation
- Dantrolene/pharmacology
- Male
- Calcineurin Inhibitors/pharmacology
- NFATC Transcription Factors/metabolism
- Cells, Cultured
- Cardiomegaly/metabolism
- Cardiomegaly/prevention & control
- Cardiomegaly/pathology
- Cardiomegaly/drug therapy
- Rats, Sprague-Dawley
- Rats
- Hypertrophy, Left Ventricular/prevention & control
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
Collapse
Affiliation(s)
- Yi Gao
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China
| | - Shuai Li
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xueyan Liu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China
| | - Daoyuan Si
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China
| | - Weiwei Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China
| | - Fenghua Yang
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Huan Sun
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China.
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China.
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China.
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China.
- Jilin Provincial International Joint Research Center of Cardiovascular Disease Precision Medicine, Changchun, China.
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Changchun, China.
| |
Collapse
|
6
|
Joshi P, Estes S, DeMazumder D, Knollmann BC, Dey S. Ryanodine receptor 2 inhibition reduces dispersion of cardiac repolarization, improves contractile function, and prevents sudden arrhythmic death in failing hearts. eLife 2023; 12:RP88638. [PMID: 38078905 PMCID: PMC10712946 DOI: 10.7554/elife.88638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Sudden cardiac death (SCD) from ventricular tachycardia/fibrillation (VT/VF) is a leading cause of death, but current therapies are limited. Despite extensive research on drugs targeting sarcolemmal ion channels, none have proven sufficiently effective for preventing SCD. Sarcoplasmic ryanodine receptor 2 (RyR2) Ca2+ release channels, the downstream effectors of sarcolemmal ion channels, are underexplored in this context. Recent evidence implicates reactive oxygen species (ROS)-mediated oxidation and hyperactivity of RyR2s in the pathophysiology of SCD. We tested the hypothesis that RyR2 inhibition of failing arrhythmogenic hearts reduces sarcoplasmic Ca2+ leak and repolarization lability, mitigates VT/VF/SCD and improves contractile function. We used a guinea pig model that replicates key clinical aspects of human nonischemic HF, such as a prolonged QT interval, a high prevalence of spontaneous arrhythmic SCD, and profound Ca2+ leak via a hyperactive RyR2. HF animals were randomized to receive dantrolene (DS) or placebo in early or chronic HF. We assessed the incidence of VT/VF and SCD (primary outcome), ECG heart rate and QT variability, echocardiographic left ventricular (LV) structure and function, immunohistochemical LV fibrosis, and sarcoplasmic RyR2 oxidation. DS treatment prevented VT/VF and SCD by decreasing dispersion of repolarization and ventricular arrhythmias. Compared to placebo, DS lowered resting heart rate, preserved chronotropic competency during transient β-adrenergic challenge, and improved heart rate variability and cardiac function. Inhibition of RyR2 hyperactivity with dantrolene mitigates the vicious cycle of sarcoplasmic Ca2+ leak-induced increases in diastolic Ca2+ and ROS-mediated RyR2 oxidation, thereby reducing repolarization lability and protecting against VT/VF/SCD. Moreover, the consequent increase in sarcoplasmic Ca2+ load improves contractile function. These potentially life-saving effects of RyR2 inhibition warrant further investigation, such as clinical studies of repurposing dantrolene as a potential new therapy for heart failure and/or SCD.
Collapse
Affiliation(s)
- Pooja Joshi
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| | - Shanea Estes
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| | - Deeptankar DeMazumder
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Internal Medicine, Veterans Affairs Pittsburgh Health SystemPittsburghUnited States
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Internal Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghUnited States
- Department of Surgery, University of Pittsburgh School of MedicinePittsburghUnited States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| | - Swati Dey
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| |
Collapse
|
7
|
Chakraborty P, Aggarwal AK, Nair MKK, Massé S, Riazi S, Nanthakumar K. Restoration of calcium release synchrony: A novel target for heart failure and ventricular arrhythmia. Heart Rhythm 2023; 20:1773-1781. [PMID: 37678492 DOI: 10.1016/j.hrthm.2023.08.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/13/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Abstract
Myocardial calcium (Ca2+) signaling plays a crucial role in contractile function and membrane electrophysiology. An abnormal myocardial Ca2+ transient is linked to heart failure and ventricular arrhythmias. At the subcellular level, the synchronous release of Ca2+ sparks from sarcoplasmic Ca2+ release units determines the configuration and amplitude of the global Ca2+ transient. This narrative review evaluates the role of aberrant Ca2+ release synchrony in the pathophysiology of cardiomyopathies and ventricular arrhythmias. The potential therapeutic benefits of restoration of Ca2+ release synchrony in heart failure and ventricular arrhythmias are also discussed.
Collapse
Affiliation(s)
- Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Heart Rhythm Institute, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Arjun K Aggarwal
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Madhav Krishna Kumar Nair
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia and Pain Management, University Health Network, Toronto, Ontario, Canada
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
George SA, Brennan-McLean JA, Trampel KA, Rytkin E, Faye NR, Knollmann BC, Efimov IR. Ryanodine receptor inhibition with acute dantrolene treatment reduces arrhythmia susceptibility in human hearts. Am J Physiol Heart Circ Physiol 2023; 325:H720-H728. [PMID: 37566110 DOI: 10.1152/ajpheart.00103.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Ryanodine receptor 2 (RyR2) hyperactivity is observed in structural heart diseases that are a result of ischemia or heart failure. It causes abnormal calcium handling and calcium leaks that cause metabolic, electrical, and mechanical dysfunction, which can trigger arrhythmias. Here, we tested the antiarrhythmic potential of dantrolene (RyR inhibitor) in human hearts. Human hearts not used in transplantation were obtained, and right ventricular outflow tract (RVOT) wedges and left ventricular (LV) slices were prepared. Pseudo-ECGs were recorded to determine premature ventricular contraction (PVC) incidences. Optical mapping was performed to determine arrhythmogenic substrates. After baseline optical recordings, tissues were treated with 1) isoproterenol (250 nM), 2) caffeine (200 mM), and 3) dantrolene (2 or 10 mM). Optical recordings were obtained after each treatment. Isoproterenol and caffeine treatment increased PVC incidence, whereas dantrolene reduced the PVC burden. Isoproterenol shortened action potential duration (APD) in the RV, RVOT, and LV regions and shortened calcium transient duration (CaTD) in the LV. Caffeine further shortened APD in the RV, did not modulate APD in the RVOT, and prolonged APD in the LV. In addition, in the LV, CaTD prolongation was also observed. More importantly, adding dantrolene did not alter APD in the RV or RVOT regions but produced a trend toward APD prolongation and significant CaTD prolongation in the LV, restoring these parameters to baseline values. In conclusions, dantrolene treatment suppresses triggers and reverses arrhythmogenic substrates in the human heart and could be a novel antiarrhythmic therapy in patients with structural heart disease.NEW & NOTEWORTHY Ryanodine receptor 2 hyperactivity is observed in structural heart diseases caused by ischemia or heart failure. It causes abnormal calcium leaks, which can trigger arrhythmias. To prevent arrhythmias, we applied dantrolene in human hearts ex vivo. Isoproterenol and caffeine treatment increased PVC incidence, whereas dantrolene reduced the PVC burden. Dantrolene treatment suppresses triggers and reverses arrhythmogenic substrates and could be a novel antiarrhythmic therapy in patients with structural heart disease.
Collapse
Affiliation(s)
- Sharon A George
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - Jaclyn A Brennan-McLean
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
| | - Katy A Trampel
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - Eric Rytkin
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| | - N Rokhaya Faye
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Igor R Efimov
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia, United States
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
9
|
Joshi P, Estes S, DeMazumder D, Knollmann BC, Dey S. Ryanodine receptor 2 inhibition reduces dispersion of cardiac repolarization, improves contractile function and prevents sudden arrhythmic death in failing hearts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.29.526151. [PMID: 37662391 PMCID: PMC10473608 DOI: 10.1101/2023.01.29.526151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Introduction Sudden cardiac death (SCD) from ventricular tachycardia/fibrillation (VT/VF) are a leading cause of death, but current therapies are limited. Despite extensive research on drugs targeting sarcolemmal ion channels, none have proven sufficiently effective for preventing SCD. Sarcoplasmic ryanodine receptor 2 (RyR2) Ca 2+ release channels, the downstream effectors of sarcolemmal ion channels, are underexplored in this context. Recent evidence implicates reactive oxygen species (ROS)- mediated oxidation and hyperactivity of RyR2s in the pathophysiology of SCD. Objective To test the hypothesis that RyR2 inhibition of failing arrhythmogenic hearts reduces sarcoplasmic Ca 2+ leak and repolarization lability, mitigates VT/VF/SCD and improves contractile function. Methods We used a guinea pig model that replicates key clinical aspects of human nonischemic HF, such as a prolonged QT interval, a high prevalence of spontaneous arrhythmic SCD, and profound Ca 2+ leak via a hyperactive RyR2. HF animals were randomized to receive dantrolene (DS) or placebo in early or chronic HF. We assessed the incidence of VT/VF and SCD (primary outcome), ECG heart rate and QT variability, echocardiographic left ventricular (LV) structure and function, immunohistochemical LV fibrosis, and sarcoplasmic RyR2 oxidation. Results DS treatment prevented VT/VF and SCD by decreasing dispersion of repolarization and ventricular arrhythmias. Compared to placebo, DS lowered resting heart rate, preserved chronotropic competency during transient β-adrenergic challenge, and improved heart rate variability and cardiac function. Conclusion Inhibition of RyR2 hyperactivity with dantrolene mitigates the vicious cycle of sarcoplasmic Ca 2+ leak-induced increases in diastolic Ca 2+ and ROS-mediated RyR2 oxidation, thereby increasing repolarization lability and protecting against VT/VF/SCD. Moreover, the consequent increase in sarcoplasmic Ca 2+ load improves contractile function. These potentially life-saving effects of RyR2 inhibition warrant further investigation, such as clinical studies of repurposing dantrolene as a potential new therapy for heart failure and/or SCD.
Collapse
|
10
|
Schmeckpeper J, Kim K, George SA, Blackwell DJ, Brennan JA, Efimov IR, Knollmann BC. RyR2 inhibition with dantrolene is antiarrhythmic, prevents further pathological remodeling, and improves cardiac function in chronic ischemic heart disease. J Mol Cell Cardiol 2023; 181:67-78. [PMID: 37285929 PMCID: PMC10526741 DOI: 10.1016/j.yjmcc.2023.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/30/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Diastolic Ca2+ leak due to cardiac ryanodine receptor (RyR2) hyperactivity has been widely documented in chronic ischemic heart disease (CIHD) and may contribute to ventricular tachycardia (VT) risk and progressive left-ventricular (LV) remodeling. Here we test the hypothesis that targeting RyR2 hyperactivity can suppress VT inducibility and progressive heart failure in CIHD by the RyR2 inhibitor dantrolene. METHODS AND RESULTS: CIHD was induced in C57BL/6 J mice by left coronary artery ligation. Four weeks later, mice were randomized to either acute or chronic (6 weeks via implanted osmotic pump) treatment with dantrolene or vehicle. VT inducibility was assessed by programmed stimulation in vivo and in isolated hearts. Electrical substrate remodeling was assessed by optical mapping. Ca2+ sparks and spontaneous Ca2+ releases were measured in isolated cardiomyocytes. Cardiac remodeling was quantified by histology and qRT-PCR. Cardiac function and contractility were measured using echocardiography. Compared to vehicle, acute dantrolene treatment reduced VT inducibility. Optical mapping demonstrated reentrant VT prevention by dantrolene, which normalized the shortened refractory period (VERP) and prolonged action potential duration (APD), preventing APD alternans. In single CIHD cardiomyocytes, dantrolene normalized RyR2 hyperactivity and prevented spontaneous intracellular Ca2+ release. Chronic dantrolene treatment not only reduced VT inducibility but also reduced peri-infarct fibrosis and prevented further progression of LV dysfunction in CIHD mice. CONCLUSIONS: RyR2 hyperactivity plays a mechanistic role for VT risk, post-infarct remodeling, and contractile dysfunction in CIHD mice. Our data provide proof of concept for the anti-arrhythmic and anti-remodeling efficacy of dantrolene in CIHD.
Collapse
Affiliation(s)
- Jeffrey Schmeckpeper
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kyungsoo Kim
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sharon A George
- Department of Biomedical Engineering, the George Washington University, Washington DC, USA; Department of Biomedical Engineering, Northwestern University, Chicago, IL, USA
| | - Daniel J Blackwell
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jaclyn A Brennan
- Department of Biomedical Engineering, the George Washington University, Washington DC, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, the George Washington University, Washington DC, USA; Department of Biomedical Engineering, Northwestern University, Chicago, IL, USA
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
11
|
Molecular Aspects Implicated in Dantrolene Selectivity with Respect to Ryanodine Receptor Isoforms. Int J Mol Sci 2023; 24:ijms24065409. [PMID: 36982484 PMCID: PMC10049336 DOI: 10.3390/ijms24065409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/24/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
Dantrolene is an intra-cellularly acting skeletal muscle relaxant used for the treatment of the rare genetic disorder, malignant hyperthermia (MH). In most cases, MH susceptibility is caused by dysfunction of the skeletal ryanodine receptor (RyR1) harboring one of nearly 230 single-point MH mutations. The therapeutic effect of dantrolene is the result of a direct inhibitory action on the RyR1 channel, thus suppressing aberrant Ca2+ release from the sarcoplasmic reticulum. Despite the almost identical dantrolene-binding sequence exits in all three mammalian RyR isoforms, dantrolene appears to be an isoform-selective inhibitor. Whereas RyR1 and RyR3 channels are competent to bind dantrolene, the RyR2 channel, predominantly expressed in the heart, is unresponsive. However, a large body of evidence suggests that the RyR2 channel becomes sensitive to dantrolene-mediated inhibition under certain pathological conditions. Although a consistent picture of the dantrolene effect emerges from in vivo studies, in vitro results are often contradictory. Hence, our goal in this perspective is to provide the best possible clues to the molecular mechanism of dantrolene’s action on RyR isoforms by identifying and discussing potential sources of conflicting results, mainly coming from cell-free experiments. Moreover, we propose that, specifically in the case of the RyR2 channel, its phosphorylation could be implicated in acquiring the channel responsiveness to dantrolene inhibition, interpreting functional findings in the structural context.
Collapse
|
12
|
Si D, Chakraborty P, Azam MA, Nair MKK, Massé S, Lai PF, Labos C, Riazi S, Nanthakumar K. Synchronizing systolic calcium release with azumolene in an experimental model. Heart Rhythm O2 2022; 3:568-576. [PMID: 36340488 PMCID: PMC9626747 DOI: 10.1016/j.hroo.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background Post-defibrillation myocardial contractile dysfunction adversely affects the survival of patients after cardiac arrest. Attenuation of diastolic calcium (Ca2+) overload by stabilization of the cardiac ryanodine receptor (RyR2) is found to reduce refibrillation after long-duration ventricular fibrillation (LDVF). Objective In the present study, we explored the effects of RyR2 stabilization by azumolene on systolic Ca2+ release synchrony and myocardial contractility. Methods After completion of baseline optical mapping, Langendorff-perfused rabbit hearts were subjected to global ischemia followed by reperfusion with azumolene or deionized distilled water (vehicle). Following reperfusion, LDVF was induced with burst pacing. In the first series of experiments (n = 16), epicardial Ca2+ transient was analyzed for Ca2+ transient amplitude alternans and dispersion of Ca2+ transient amplitude alternans index (CAAI). In the second series of experiments following the same protocol (n = 12), ventricular contractility was assessed by measuring the left ventricular pressure. Results Ischemic LDVF led to greater CAAI (0.06 ± 0.02 at baseline vs 0.12 ± 0.02 post-LDVF, P < .01) and magnitude of dispersion of CAAI (0.04 ± 0.01 vs 0.09 ± 0.01, P < .01) in control hearts. In azumolene-treated hearts, no significant changes in CAAI (0.05 ± 0.01 vs 0.05 ± 0.01, P = .84) and dispersion of CAAI (0.04 ± 0.01 vs 0.04 ± 0.01, P = .99) were noted following ischemic LDVF. Ischemic LDVF was associated with reduction in left ventricular developed pressure (100% vs 36.8% ± 6.1%, P = .002) and dP/dtmax (100% vs 45.3% ± 6.5%, P = .003) in control hearts, but these reductions were mitigated (left ventricular developed pressure: 100% vs 74.0% ± 8.1%, P = .052, dP/dtmax: 100% vs 80.8% ± 7.9%, P = .09) in azumolene-treated hearts. Conclusion Treatment with azumolene is associated with improvement of systolic Ca2+ release synchrony and myocardial contractility following ischemic LDVF.
Collapse
Affiliation(s)
- Daoyuan Si
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Mohammed Ali Azam
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Madhav Krishna Kumar Nair
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Patrick F.H. Lai
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | | | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia and Pain Management, University Health Network, Toronto, Canada
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| |
Collapse
|
13
|
Mahmood A, Ahmed K, Zhang Y. β-Adrenergic Receptor Desensitization/Down-Regulation in Heart Failure: A Friend or Foe? Front Cardiovasc Med 2022; 9:925692. [PMID: 35845057 PMCID: PMC9283919 DOI: 10.3389/fcvm.2022.925692] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/06/2022] [Indexed: 11/22/2022] Open
Abstract
Cardiac sympathetic activation, mediated by β-adrenergic receptors (β-ARs), normally increases cardiac contraction and relaxation. Accomplishing this task requires a physiological, concerted Ca2+ signaling, being able to increase Ca2+ release from sarcoplasmic reticulum (SR) in systole and speed up Ca2+ re-uptake in diastole. In heart failure (HF) myocardial β-ARs undergo desensitization/down-regulation due to sustained sympathetic adrenergic activation. β-AR desensitization/down-regulation diminishes adrenergic signaling and cardiac contractile reserve, and is conventionally considered to be detrimental in HF progression. Abnormal Ca2+ handling, manifested as cardiac ryanodine receptor (RyR2) dysfunction and diastolic Ca2+ leak (due to sustained adrenergic activation) also occur in HF. RyR2 dysfunction and Ca2+ leak deplete SR Ca2+ store, diminish Ca2+ release in systole and elevate Ca2+ levels in diastole, impairing both systolic and diastolic ventricular function. Moreover, elevated Ca2+ levels in diastole promote triggered activity and arrhythmogenesis. In the presence of RyR2 dysfunction and Ca2+ leak, further activation of the β-AR signaling in HF would worsen the existing abnormal Ca2+ handling, exacerbating not only cardiac dysfunction, but also ventricular arrhythmogenesis and sudden cardiac death. Thus, we conclude that β-AR desensitization/down-regulation may be a self-preserving, adaptive process (acting like an intrinsic β-AR blocker) protecting the failing heart from developing lethal ventricular arrhythmias under conditions of elevated sympathetic drive and catecholamine levels in HF, rather than a conventionally considered detrimental process. This also implies that medications simply enhancing β-AR signaling (like β-AR agonists) may not be so beneficial unless they can also correct dysfunctional Ca2+ handling in HF.
Collapse
Affiliation(s)
- Abrahim Mahmood
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Kinza Ahmed
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Youhua Zhang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| |
Collapse
|
14
|
Therapeutic Approaches of Ryanodine Receptor-Associated Heart Diseases. Int J Mol Sci 2022; 23:ijms23084435. [PMID: 35457253 PMCID: PMC9031589 DOI: 10.3390/ijms23084435] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 01/08/2023] Open
Abstract
Cardiac diseases are the leading causes of death, with a growing number of cases worldwide, posing a challenge for both healthcare and research. Therefore, the most relevant aim of cardiac research is to unravel the molecular pathomechanisms and identify new therapeutic targets. Cardiac ryanodine receptor (RyR2), the Ca2+ release channel of the sarcoplasmic reticulum, is believed to be a good therapeutic target in a group of certain heart diseases, collectively called cardiac ryanopathies. Ryanopathies are associated with the impaired function of the RyR, leading to heart diseases such as congestive heart failure (CHF), catecholaminergic polymorphic ventricular tachycardia (CPVT), arrhythmogenic right ventricular dysplasia type 2 (ARVD2), and calcium release deficiency syndrome (CRDS). The aim of the current review is to provide a short insight into the pathological mechanisms of ryanopathies and discuss the pharmacological approaches targeting RyR2.
Collapse
|
15
|
Emerging Antiarrhythmic Drugs for Atrial Fibrillation. Int J Mol Sci 2022; 23:ijms23084096. [PMID: 35456912 PMCID: PMC9029767 DOI: 10.3390/ijms23084096] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia worldwide, is driven by complex mechanisms that differ between subgroups of patients. This complexity is apparent from the different forms in which AF presents itself (post-operative, paroxysmal and persistent), each with heterogeneous patterns and variable progression. Our current understanding of the mechanisms responsible for initiation, maintenance and progression of the different forms of AF has increased significantly in recent years. Nevertheless, antiarrhythmic drugs for the management of AF have not been developed based on the underlying arrhythmia mechanisms and none of the currently used drugs were specifically developed to target AF. With the increased knowledge on the mechanisms underlying different forms of AF, new opportunities for developing more effective and safer AF therapies are emerging. In this review, we provide an overview of potential novel antiarrhythmic approaches based on the underlying mechanisms of AF, focusing both on the development of novel antiarrhythmic agents and on the possibility of repurposing already marketed drugs. In addition, we discuss the opportunity of targeting some of the key players involved in the underlying AF mechanisms, such as ryanodine receptor type-2 (RyR2) channels and atrial-selective K+-currents (IK2P and ISK) for antiarrhythmic therapy. In addition, we highlight the opportunities for targeting components of inflammatory signaling (e.g., the NLRP3-inflammasome) and upstream mechanisms targeting fibroblast function to prevent structural remodeling and progression of AF. Finally, we critically appraise emerging antiarrhythmic drug principles and future directions for antiarrhythmic drug development, as well as their potential for improving AF management.
Collapse
|
16
|
Chakraborty P, Massé S, Azam MA, Thollon C, Niri A, Lai PFH, Bouly M, Riazi S, Nanthakumar K. Effects of azumolene on arrhythmia substrate in a model of recurrent long-duration ventricular fibrillation. Biochem Biophys Res Commun 2022; 600:123-129. [PMID: 35219100 DOI: 10.1016/j.bbrc.2022.02.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Proarrhythmic risk of conventional anti-arrhythmic agents is linked to unintended modulation of membrane voltage dynamics. We have demonstrated that the anti-fibrillatory effect of azumolene is mediated via stabilization of the hyperphosphorylated ryanodine receptor (RyR2), leading to attenuation of diastolic calcium leak. However, the concomitant effects on membrane voltage dynamics have not been evaluated yet. METHODS After baseline optical mapping, Langendorff-perfused rabbit hearts treated with azumolene, or vehicle, were subjected to global ischemia-reperfusion (I/R) followed by two episodes of long-duration ventricular fibrillation (LDVF). Simultaneous dual epicardial calcium transient (CaT) and voltage dynamics were studied optically. RESULTS Pre-treatment with azumolene was associated with higher CaT amplitude alternans ratios (0.94 ± 0.02 vs. 0.78 ± 0.03 in control hearts, at 6 Hz; p = 0.005; and action potential amplitude alternans ratio (0.95 ± 0.02 vs. 0.78 ± 0.04 at 6.0 Hz; p = 0.02), and reduction of action potential duration (APD80) dispersion (9.0 ± 4.8 msec vs. 19.3 ± 6.6 msec at 6.0 Hz p = 0.02) and optical action potential upstroke rise time (26.3 ± 2.6 msec in control vs. 13.8 ± 0.6 msec at 6.0 Hz, p = 0.02) after LDVF. No change in action potential duration (APD) was noted with azumolene treatment. CONCLUSION In a model of ischemic recurrent LDVF, treatment with azumolene led to reduction of cardiac alternans, i.e., calcium and voltage alternans. Unlike conventional anti-arrhythmic agents, reduction of action potential upstroke rise time and preservation of action potential duration following azumolene treatment may reduce the proarrhythmia risk.
Collapse
Affiliation(s)
- Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Mohammed Ali Azam
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | | | - Ahmed Niri
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Patrick F H Lai
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Muriel Bouly
- Institut de Recherches Internationales Servier (IRIS), Suresnes, France
| | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia and Pain Management, University Health Network, Toronto, Ontario, Canada
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
17
|
Azam MA, Chakraborty P, Bokhari MM, Dadson K, Du B, Massé S, Si D, Niri A, Aggarwal AK, Lai PF, Riazi S, Billia F, Nanthakumar K. Cardioprotective effects of dantrolene in doxorubicin-induced cardiomyopathy in mice. Heart Rhythm O2 2021; 2:733-741. [PMID: 34988524 PMCID: PMC8710625 DOI: 10.1016/j.hroo.2021.08.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Doxorubicin (Dox) is a potent chemotherapeutic agent, but its usage is limited by dose-dependent cardiotoxicity. Intracellular calcium dysregulation has been reported to be involved in doxorubicin-induced cardiomyopathy (DICM). The cardioprotective role of RyR stabilizer dantrolene (Dan) on the calcium dynamics of DICM has not yet been explored. OBJECTIVE To evaluate the effects of dantrolene on intracellular calcium dysregulation and cardiac contractile function in a DICM model. METHODS Adult male C57BL/6 mice were randomized into 4 groups: (1) Control, (2) Dox Only, (3) Dan Only, and (4) Dan + Dox. Fractional shortening (FS) and left ventricular ejection fraction (LVEF) were assessed by echocardiography. In addition, mice were sacrificed 2 weeks after doxorubicin injection for optical mapping of the heart in a Langendorff setup. RESULTS Treatment with Dox was associated with a reduction in both FS and LVEF at 2 weeks (P < .0001) and 4 weeks (P < .006). Dox treatment was also associated with prolongation of calcium transient durations CaTD50 (P = .0005) and CaTD80 (P < .0001) and reduction of calcium amplitude alternans ratio (P < .0001). Concomitant treatment with Dan prevented the Dox-induced decline in FS and LVEF (P < .002 at both 2 and 4 weeks). Dan also prevented Dox-induced prolongation of CaTD50 and CaTD80 and improved the CaT alternans ratio (P < .0001). Finally, calcium transient rise time was increased in the doxorubicin-treated group, indicating RyR2 dyssynchrony, and dantrolene prevented this prolongation (P = .02). CONCLUSION Dantrolene prevents cardiac contractile dysfunction following doxorubicin treatment by mitigating dysregulation of calcium dynamics.
Collapse
Affiliation(s)
- Mohammed Ali Azam
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Mahmoud M. Bokhari
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Keith Dadson
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Beibei Du
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Daoyuan Si
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Ahmed Niri
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Arjun K. Aggarwal
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Patrick F.H. Lai
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Filio Billia
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| |
Collapse
|
18
|
Gao L, Zhang L, He F, Chen J, Zhao M, Li S, Wu H, Liu Y, Zhang Y, Ping Q, Hu L, Qiao H. Surfactant Assisted Rapid-Release Liposomal Strategies Enhance the Antitumor Efficiency of Bufalin Derivative and Reduce Cardiotoxicity. Int J Nanomedicine 2021; 16:3581-3598. [PMID: 34079251 PMCID: PMC8165102 DOI: 10.2147/ijn.s313153] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background BF211, a derivative of bufalin (BF), shows significantly improved solubility and potent antitumor efficiency compared to BF. Unfortunately, the unwanted toxicity such as cardiotoxicity caused by unspecific distribution has hindered its clinical use. Methods PEGylated BF211 liposomes (BF211@Lipo) were designed and optimizely prepared based on the pre-prescription research. In vitro and in vivo cardiotoxicity was evaluated. In vivo pharmacokinetics and biodistribution of BF211@Lipo were investigated. In vivo antitumor activity and toxicity were evaluated in HepG2 cell xenograft models. The rapid-release triggered by Poloxamer 188 (P188) was assessed in vitro and in vivo. Results The optimized BF211@Lipo displayed a spherical morphology with a size of (164.6 ± 10.3) nm and a high encapsulation efficiency of (93.24 ± 2.15) %. The in vivo concentration–time curves of BF211 loaded in liposomes showed a prolonged half-life in plasma and increased tumor accumulation. No obvious abnormality in electrocardiograms was observed in guinea pigs even at 9 mg/kg. Moreover, to improve the efficient release of BF211@Lipo, a surfactant-assisted rapid-release strategy was developed, and the release-promoting mechanism was revealed by the fluorescence resonance energy transfer (FRET) and fluorescence nanoparticle tracking analysis (fl-NTA) technology. Sequential injection of BF211@Lipo and P188 could ignite the “cold” liposomes locally in tumor regions, facilitating the burst release of BF211 and enhancing the therapeutic index. Conclusion Our progressive efforts that begin with preparation technology and dosage regimen enable BF211 to like a drug, providing a promising nano platform to deliver the cardiac glycosides and alleviate the side effects by decreasing unspecific biodistribution.
Collapse
Affiliation(s)
- Lina Gao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Lei Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Fengjun He
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Jing Chen
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Meng Zhao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Simin Li
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Hao Wu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yumeng Liu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yinan Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Qineng Ping
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Hongzhi Qiao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.,Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| |
Collapse
|
19
|
Azam MA, Chakraborty P, Si D, Du B, Massé S, Lai PFH, Ha ACT, Nanthakumar K. Anti-arrhythmic and inotropic effects of empagliflozin following myocardial ischemia. Life Sci 2021; 276:119440. [PMID: 33781832 DOI: 10.1016/j.lfs.2021.119440] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Empagliflozin (EMPA) reduces heart failure hospitalization and mortality. The benefit in terms of ventricular arrhythmia and contractility has not been explored. OBJECTIVE To determine the direct effects of EMPA on ventricular arrhythmia and cardiac contractility in an ex-vivo model of global ischemia-reperfusion (I/R). METHODS Langendorff-perfused rabbit hearts were subjected to 30 min of complete perfusion arrest and reperfusion. Either EMPA (1 μM) or normal saline (controls) was then infused into the perfusate in a randomized fashion. Ten minutes following drug infusion, calcium imaging was performed. At the end of each experiment, the heart was electrically stimulated 5 times to assess the inducibility of ventricular fibrillation (VF). In a separate series of experiments, left ventricular (LV) pressure and epicardial NADH fluorescence were simultaneously recorded. LV specimens were then collected for western blotting. RESULTS Post-ischemia, EMPA treatment was associated with reduction in the induction of VF >10s (rate of induction: 16.7 ± 3.3% vs. 60 ± 8.7% in control hearts, p = 0.003), improvement of LV developed pressure (LVDP; 68.10 ± 9.02% vs. 47.61 ± 5.15% in controls, p = 0.03) and reduction of NADH fluorescence (87.42 ± 2.79% vs. 112.88 ± 2.27% in control hearts, p = 0.04) along with an increase in NAD+/NADH ratio (2.75 ± 0.55 vs. 1.09 ± 0.32 in the control group, p = 0.04) A higher calcium amplitude alternans threshold was also observed with EMPA-treatment (5.42 ± 0.1 Hz vs. 4.75 ± 0.1 Hz in controls, p = 0.006). Sodium-glucose co-transporter-2 (SGLT2) expression was not detected in LV tissues. CONCLUSIONS EMPA treatment reduced ventricular arrhythmia vulnerability and mitigated contractile dysfunction in the global I/R model while improving calcium cycling and mitochondrial redox by SGLT2-independent mechanisms.
Collapse
Affiliation(s)
- Mohammed Ali Azam
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Canada
| | - Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Canada
| | - Daoyuan Si
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Canada
| | - BeiBei Du
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Canada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Canada
| | - Patrick F H Lai
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Canada
| | - Andrew C T Ha
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Canada; Toronto General Hospital, Canada
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, Canada.
| |
Collapse
|
20
|
Anthracycline-induced cardiomyopathy: cellular and molecular mechanisms. Clin Sci (Lond) 2021; 134:1859-1885. [PMID: 32677679 DOI: 10.1042/cs20190653] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023]
Abstract
Despite the known risk of cardiotoxicity, anthracyclines are widely prescribed chemotherapeutic agents. They are broadly characterized as being a robust effector of cellular apoptosis in rapidly proliferating cells through its actions in the nucleus and formation of reactive oxygen species (ROS). And, despite the early use of dexrazoxane, no effective treatment strategy has emerged to prevent the development of cardiomyopathy, despite decades of study, suggesting that much more insight into the underlying mechanism of the development of cardiomyopathy is needed. In this review, we detail the specific intracellular activities of anthracyclines, from the cell membrane to the sarcoplasmic reticulum, and highlight potential therapeutic windows that represent the forefront of research into the underlying causes of anthracycline-induced cardiomyopathy.
Collapse
|
21
|
Samiotis I, Papakonstantinou NA, Dedeilias P, Vasileiadis I, Papalois A, Deftereos S, Kotanidou A. Dantrolene Induces Mitigation of Myocardial Ischemia-Reperfusion Injury by Ryanodine Receptor Inhibition. Semin Thorac Cardiovasc Surg 2021; 34:123-132. [PMID: 33460764 DOI: 10.1053/j.semtcvs.2021.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 11/11/2022]
Abstract
The impairment of intracellular calcium homeostasis plays an essential role during ischemia-reperfusion injury. Calcium release from sarcoplasmic reticulum which is triggered by myocardial ischemia is mainly mediated by ryanodine receptors. Dantrolene sodium is a ryanodine receptor antagonist. The objective of the present study was to evaluate the in-vivo impact of dantrolene sodium on myocardial ischemia-reperfusion injury in swine models. An in vivo, experimental trial comparing 10 experimental animals which received dantrolene sodium with 9 control swine models was conducted. Their left anterior descending coronary artery was temporarily occluded for 75 minutes via a vessel tourniquet, which was then released. Myocardial reperfusion was allowed for 24 hours. Dantrolene was administered at the onset of the reperfusion period and levels of troponin, creatine phosphokinase and creatine kinase myocardial band between the two groups were compared. Additionally, various other hemodynamic parameters and left ventricular morphology and function were examined. There were significantly lower values of troponin, creatine phosphokinase and creatine kinase myocardial band in the dantrolene group indicating less ischemia-reperfusion injury. Moreover, the postischemic cardiac index was also greater in the dantrolene group, whereas viable myocardium was also better preserved. In conclusion, the in vivo cardioprotective role of dantrolene sodium against ischemia-reperfusion injury in swine models was indicated in this study. Therefore, dantrolene sodium administration could be a promising treatment against ischemia-reperfusion injury in humans. However, large randomized clinical studies should be firstly carried out to prove this hypothesis.
Collapse
Affiliation(s)
- Ilias Samiotis
- Cardiovascular and Thoracic Surgery Department, General Hospital of Athens "Evangelismos'', Greece
| | | | - Panagiotis Dedeilias
- Cardiovascular and Thoracic Surgery Department, General Hospital of Athens "Evangelismos'', Greece
| | - Ioannis Vasileiadis
- 1st Respiratory Medicine Department, Hospital for Diseases of the Chest "Sotiria", National and Kapodistrian University of Athens, Greece
| | - Apostolos Papalois
- Experimental, Educational and Research Center, ELPEN Pharmaceuticals, Athens, Greece; School of Medicine European University of Cyprus, Nicosia, Cyprus
| | - Spyridon Deftereos
- 2nd Department of Cardiology, "Attikon" Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine, General Hospital of Athens "Evangelismos'', School of Medicine, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
22
|
Jiang B, Liang S, Liang G, Wei H. Could dantrolene be explored as a repurposed drug to treat COVID-19 patients by restoring intracellular calcium homeostasis? EUROPEAN REVIEW FOR MEDICAL AND PHARMACOLOGICAL SCIENCES 2020; 24:10228-10238. [PMID: 33090434 DOI: 10.26355/eurrev_202010_23247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dantrolene, an FDA approved drug to treat malignant hyperthermia and muscle spasm, has been demonstrated to inhibit severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mediated toxicity of host cells. Ryanodine receptor overactivation and associated disruption of intracellular Ca2+ homeostasis play important roles in SARS-CoV-2 infection and replication of host cells. Dantrolene, as an inhibitor of RyRs, is expected to ameliorate these detrimental effects of SARS-CoV-2 in host cells. Additionally, dantrolene has also been shown to inhibit multiple cell or organ damage induced by hypoxia/ischemia, mitochondria damage, oxidative stresses, inflammation, impairment of autophagy and apoptosis, etc., which are often the causes of severity and mortality of COVID-19 patients. We have repurposed that dantrolene has a high potential at treating COVID-19 patients and reducing its morbidity and mortality.
Collapse
Affiliation(s)
- B Jiang
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA.
| | | | | | | |
Collapse
|
23
|
Orini M, Yanni J, Taggart P, Hanson B, Hayward M, Smith A, Zhang H, Colman M, Jones G, Jie X, Dobrzynski H, Boyett MR, Lambiase PD. Mechanistic insights from targeted molecular profiling of repolarization alternans in the intact human heart. Europace 2020; 21:981-989. [PMID: 30753421 PMCID: PMC6545501 DOI: 10.1093/europace/euz007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/17/2018] [Accepted: 01/21/2019] [Indexed: 02/05/2023] Open
Abstract
AIMS Action potential duration (APD) alternans is an established precursor or arrhythmia and sudden cardiac death. Important differences in fundamental electrophysiological properties relevant to arrhythmia exist between experimental models and the diseased in vivo human heart. To investigate mechanisms of APD alternans using a novel approach combining intact heart and cellular cardiac electrophysiology in human in vivo. METHODS AND RESULTS We developed a novel approach combining intact heart electrophysiological mapping during cardiac surgery with rapid on-site data analysis to guide myocardial biopsies for laboratory analysis, thereby linking repolarization dynamics observed at the organ level with underlying ion channel expression. Alternans-susceptible and alternans-resistant regions were identified by an incremental pacing protocol. Biopsies from these sites (n = 13) demonstrated greater RNA expression in Calsequestrin (CSQN) and Ryanodine (RyR) and ion channels underlying IK1 and Ito at alternans-susceptible sites. Electrical restitution properties (n = 7) showed no difference between alternans-susceptible and resistant sites, whereas spatial gradients of repolarization were greater in alternans-susceptible than in alternans-resistant sites (P = 0.001). The degree of histological fibrosis between alternans-susceptible and resistant sites was equivalent. Mathematical modelling of these changes indicated that both CSQN and RyR up-regulation are key determinants of APD alternans. CONCLUSION Combined intact heart and cellular electrophysiology show that regions of myocardium in the in vivo human heart exhibiting APD alternans are associated with greater expression of CSQN and RyR and show no difference in restitution properties compared to non-alternans regions. In silico modelling identifies up-regulation and interaction of CSQN with RyR as a major mechanism underlying APD alternans.
Collapse
Affiliation(s)
- Michele Orini
- Institute of Cardiovascular Science, University College London, London, UK.,Department of Electrophysiology, Barts Heart Centre at St Bartholomew's Hospital, London, UK
| | - Joseph Yanni
- Division of Cardiovascular Science, University of Manchester, Manchester, UK
| | - Peter Taggart
- Institute of Cardiovascular Science, University College London, London, UK
| | - Ben Hanson
- Department of Mechanical Engineering, University College London, UK
| | - Martin Hayward
- Department of Cardiothoracic Surgery, The Heart Hospital, University College London Hospitals, London, UK
| | - Andrew Smith
- Department of Electrophysiology, Barts Heart Centre at St Bartholomew's Hospital, London, UK
| | - Henggui Zhang
- Division of Cardiovascular Science, University of Manchester, Manchester, UK.,School of Physics and Astronomy, University of Manchester, Manchester, UK
| | | | - Gareth Jones
- School of Physics and Astronomy, University of Manchester, Manchester, UK
| | - Xiao Jie
- Institute of Cardiovascular Science, University College London, London, UK
| | - Halina Dobrzynski
- Division of Cardiovascular Science, University of Manchester, Manchester, UK
| | - Mark R Boyett
- Division of Cardiovascular Science, University of Manchester, Manchester, UK
| | - Pier D Lambiase
- Institute of Cardiovascular Science, University College London, London, UK.,Department of Electrophysiology, Barts Heart Centre at St Bartholomew's Hospital, London, UK
| |
Collapse
|
24
|
Nofi C, Zhang K, Tang YD, Li Y, Migirov A, Ojamaa K, Gerdes AM, Zhang Y. Chronic dantrolene treatment attenuates cardiac dysfunction and reduces atrial fibrillation inducibility in a rat myocardial infarction heart failure model. Heart Rhythm O2 2020; 1:126-135. [PMID: 34113867 PMCID: PMC8183840 DOI: 10.1016/j.hroo.2020.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Cardiac ryanodine receptor 2 (RyR2) dysfunction and elevated diastolic Ca2+ leak have been linked to arrhythmogenesis not only in inherited arrhythmia syndromes but also in acquired forms of heart disease including heart failure (HF) and atrial fibrillation (AF). Thus, stabilizing RyR2 may exert therapeutic effects in these conditions. Objective The purpose of this study was to investigate the effects of stabilizing RyR2 with chronic dantrolene treatment on HF development and AF inducibility in a myocardial infarction (MI)-induced HF model in rats. Methods MI was induced in adult Sprague-Dawley rats by ligation of the left anterior descending coronary artery. Two weeks after MI surgery, rats with large MI (≥40%) were randomly assigned to MI-vehicle (n = 14) or MI-dantrolene (10 mg/kg/d; n = 13) groups. Sham-surgery rats (n = 7) served as controls. Results Compared to the MI-vehicle group, 4-week dantrolene treatment significantly improved cardiac function, with increased left ventricular (LV) fractional shortening (19.48% ± 3.61% vs 15.43% ± 2.65%; P <.01), and decreased LV end-diastolic pressure (12.58 ± 8.52 mm Hg vs 21.91 ± 7.25 mm Hg; P <.01), left atrial diameter (4.97 ± 0.75 mm vs 6.09 ± 1.53 mm; P <.05), and fibrosis content (6.42% ± 0.78% vs 9.76% ± 2.25%; P <.001). Dantrolene significantly decreased AF inducibility (69% in MI-vehicle vs 23% in MI-dantrolene; P <.05). Dantrolene treatment was associated with reduced RyR2 phosphorylation and favorably altered gene expression involving ion channels, sympathetic signaling, oxidative stress, and inflammatory markers. Conclusion Chronic dantrolene treatment attenuated LV dysfunction and reduced AF inducibility, which was associated with decreased RyR2 phosphorylation and normalization of many adverse changes in gene expression. Thus, stabilizing RyR2 with chronic dantrolene treatment is a promising novel strategy for decreasing AF in HF.
Collapse
Affiliation(s)
- Colleen Nofi
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York
| | - Kuo Zhang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York.,Department of Internal Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Da Tang
- Department of Internal Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Li
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York
| | - Allan Migirov
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York
| | - Kaie Ojamaa
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York
| | - A Martin Gerdes
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York
| | - Youhua Zhang
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York
| |
Collapse
|
25
|
Mucke HA. Drug Repurposing Patent Applications April–June 2019. Assay Drug Dev Technol 2020. [DOI: 10.1089/adt.2019.968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
26
|
Avula UMR, Hernandez JJ, Yamazaki M, Valdivia CR, Chu A, Rojas-Pena A, Kaur K, Ramos-Mondragón R, Anumonwo JM, Nattel S, Valdivia HH, Kalifa J. Atrial Infarction-Induced Spontaneous Focal Discharges and Atrial Fibrillation in Sheep: Role of Dantrolene-Sensitive Aberrant Ryanodine Receptor Calcium Release. Circ Arrhythm Electrophysiol 2019. [PMID: 29540372 DOI: 10.1161/circep.117.005659] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mechanisms underlying spontaneous atrial fibrillation (AF) associated with atrial ischemia/infarction are incompletely elucidated. Here, we investigate the mechanisms underlying spontaneous AF in an ovine model of left atrial myocardial infarction (LAMI). METHODS AND RESULTS LAMI was created by ligating the atrial branch of the left anterior descending coronary artery. ECG loop recorders were implanted to monitor AF episodes. In 7 sheep, dantrolene-a ryanodine receptor blocker-was administered in vivo during the 8-day observation period (LAMI-D, 2.5 mg/kg, IV, BID). LAMI animals experienced numerous spontaneous AF episodes during the 8-day monitoring period that were suppressed by dantrolene (LAMI, 26.1±5.1; sham, 4.3±1.1; LAMI-D, 2.8±0.8; mean±SEM episodes per sheep, P<0.01). Optical mapping showed spontaneous focal discharges (SFDs) originating from the ischemic/normal-zone border. SFDs were calcium driven, rate dependent, and enhanced by isoproterenol (0.03 µmol/L, from 210±87 to 3816±1450, SFDs per sheep) but suppressed by dantrolene (to 55.8±32.8, SFDs per sheep, mean±SEM). SFDs initiated AF-maintaining reentrant rotors anchored by marked conduction delays at the ischemic/normal-zone border. NOS1 (NO synthase-1) protein expression decreased in ischemic zone myocytes, whereas NADPH (nicotinamide adenine dinucleotide phosphate, reduced form) oxidase and xanthine oxidase enzyme activities and reactive oxygen species (DCF [6-carboxy-2',7'-dichlorodihydrofluorescein diacetate]-fluorescence) increased. CaM (calmodulin) aberrantly increased [3H]ryanodine binding to cardiac RyR2 (ryanodine receptors) in the ischemic zone. Dantrolene restored the physiological binding of CaM to RyR2. CONCLUSIONS Atrial ischemia causes spontaneous AF episodes in sheep, caused by SFDs that initiate reentry. Nitroso-redox imbalance in the ischemic zone is associated with intense reactive oxygen species production and altered RyR2 responses to CaM. Dantrolene administration normalizes the CaM response, prevents LAMI-related SFDs, and AF initiation. These findings provide novel insights into the mechanisms underlying ischemia-related atrial arrhythmias.
Collapse
Affiliation(s)
- Uma Mahesh R Avula
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Jonathan J Hernandez
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Masatoshi Yamazaki
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Carmen R Valdivia
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Antony Chu
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Alvaro Rojas-Pena
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Kuljeet Kaur
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Roberto Ramos-Mondragón
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Justus M Anumonwo
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Stanley Nattel
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Héctor H Valdivia
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.)
| | - Jérôme Kalifa
- From the Division of Cardiovascular Medicine, Department of Internal Medicine, Columbia University, New York, NY (U.M.R.A.); Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research (J.J.H., C.R.V., K.K., R.R.-M., J.A., H.H.V.) and Department of Surgery (A.R.-P.), University of Michigan, Ann Arbor; Medical Device Development and Regulation Research Center, The University of Tokyo, Japan (M.Y.); Department of Cardiology, Brown University, Providence, RI (A.C., J.K.); Department of Medicine and Research Center, Montreal Heart Institute, Université de Montréal, Québec (S.N.); Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (S.N.); and Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen (S.N.).
| |
Collapse
|
27
|
Safety of Chronic Cardiac Ryanodine Receptor Modulation: A 10-Year Experience. JACC Clin Electrophysiol 2018; 4:1480-1481. [PMID: 30466856 DOI: 10.1016/j.jacep.2018.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 11/23/2022]
|
28
|
Si D, Azam MA, Lai PFH, Zamiri N, Kichigina G, Asta J, Massé S, Bokhari M, Porta‐Sánchez A, Labos C, Sun H, Yang P, Nanthakumar K. Essential role of ryanodine receptor 2 phosphorylation in the effect of azumolene on ventricular arrhythmia vulnerability in a rabbit heart model. J Cardiovasc Electrophysiol 2018; 29:1707-1715. [DOI: 10.1111/jce.13737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/31/2018] [Accepted: 09/07/2018] [Indexed: 11/28/2022]
Affiliation(s)
- Daoyuan Si
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
- Department of CardiologyChina‐Japan Union Hospital, Jilin UniversityChangchun China
| | - Mohammed Ali Azam
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| | - Patrick F. H. Lai
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| | - Nima Zamiri
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| | - Galina Kichigina
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| | - John Asta
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| | - Mahmoud M. Bokhari
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| | - Andreu Porta‐Sánchez
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| | | | - Huan Sun
- Department of CardiologyChina‐Japan Union Hospital, Jilin UniversityChangchun China
| | - Ping Yang
- Department of CardiologyChina‐Japan Union Hospital, Jilin UniversityChangchun China
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management LaboratoryDivision of Cardiology, Toronto General HospitalToronto Ontario Canada
| |
Collapse
|
29
|
Functional recovery after dantrolene-supplementation of cold stored hearts using an ex vivo isolated working rat heart model. PLoS One 2018; 13:e0205850. [PMID: 30312353 PMCID: PMC6185861 DOI: 10.1371/journal.pone.0205850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/02/2018] [Indexed: 11/19/2022] Open
Abstract
The ryanodine receptor antagonist dantrolene inhibits calcium release from the sarcoplasmic reticulum and reduces cardiac ischaemia-reperfusion injury (IRI) in global warm ischaemia models however the cardioprotective potential of dantrolene under hypothermic conditions is unknown. This study addresses whether the addition of dantrolene during cardioplegia and hypothermic storage of the donor heart can improve functional recovery and reduce IRI. Using an ex vivo isolated working heart model, Wistar rat (3 month and 12 month) hearts were perfused to acquire baseline haemodynamic measurements of aortic flow, coronary flow, cardiac output, pulse pressure and heart rate. Hearts were arrested and stored in Celsior preservation solution supplemented with 0.2–40 μM dantrolene for 6 hours at 4°C, then reperfused (15 min Langendorff, 30 min working mode). In 3-month hearts, supplementation with 1 μM dantrolene significantly improved aortic flow and cardiac output compared to unsupplemented controls however lactate dehydrogenase (LDH) release and contraction bands were comparable. In contrast, 40 μM dantrolene-supplementation yielded poor cardiac recovery, increased post-reperfusion LDH but reduced contraction bands. All 3-month hearts stored in dantrolene displayed significantly reduced cleaved-caspase 3 intensities compared to controls. Analysis of cardioprotective signalling pathways showed no changes in AMPKα however dantrolene increased STAT3 and ERK1/2 signaling in a manner unrelated to functional recovery and AKT activity was reduced in 1 μM dantrolene-stored hearts. In contrast to 3-month hearts, no significant improvements were observed in the functional recovery of 12-month hearts following prolonged storage in 1 μM dantrolene. Conclusions: Dantrolene supplementation at 1 μM during hypothermic heart preservation improved functional recovery of young, but not older (12 month) hearts. Although the molecular mechanisms responsible for dantrolene-mediated cardioprotection are unclear, our studies show no correlation between improved functional recovery and SAFE and RISK pathway activation.
Collapse
|
30
|
Martinez ME, Walton RD, Bayer JD, Haïssaguerre M, Vigmond EJ, Hocini M, Bernus O. Role of the Purkinje-Muscle Junction on the Ventricular Repolarization Heterogeneity in the Healthy and Ischemic Ovine Ventricular Myocardium. Front Physiol 2018; 9:718. [PMID: 29962961 PMCID: PMC6010581 DOI: 10.3389/fphys.2018.00718] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/24/2018] [Indexed: 01/23/2023] Open
Abstract
Alteration of action potential duration (APD) heterogeneity contributes to arrhythmogenesis. Purkinje-muscle junctions (PMJs) present differential electrophysiological properties including longer APD. The goal of this study was to determine if Purkinje-related or myocardial focal activation modulates ventricular repolarization differentially in healthy and ischemic myocardium. Simultaneous epicardial (EPI) and endocardial (ENDO) optical mapping was performed on sheep left ventricular (LV) wedges with intact free-running Purkinje network (N = 7). Preparations were paced on either ENDO or EPI surfaces, or the free-running Purkinje fibers (PFs), mimicking normal activation. EPI and ENDO APDs were assessed for each pacing configuration, before and after (7 min) of the onset of no-flow ischemia. Experiments were supported by simulations. In control conditions, maximal APD was found at endocardial PMJ sites. We observed a significant transmural APD gradient for PF pacing with PMJ APD = 347 ± 41 ms and EPI APD = 273 ± 36 ms (p < 0.001). A similar transmural gradient was observed when pacing ENDO (49 ± 31 ms; p = 0.005). However, the gradient was reduced when pacing EPI (37 ± 20 ms; p = 0.005). Global dispersion of repolarization was the most pronounced for EPI pacing. In ischemia, both ENDO and EPI APD were reduced (p = 0.005) and the transmural APD gradient (109 ± 55 ms) was increased when pacing ENDO compared to control condition or when pacing EPI (p < 0.05). APD maxima remained localized at functional PMJs during ischemia. Local repolarization dispersion was significantly higher at the PMJ than at other sites. The results were consistent with simulations. We found that the activation sequence modulates repolarization heterogeneity in the ischemic sheep LV. PMJs remain active following ischemia and exert significant influence on local repolarization patterns.
Collapse
Affiliation(s)
- Marine E Martinez
- Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,INSERM U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, Bordeaux, France
| | - Richard D Walton
- Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,INSERM U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, Bordeaux, France
| | - Jason D Bayer
- Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,UMR5251, Centre National De La Recherche Scientifique, Institut de Mathématiques de Bordeaux, Bordeaux, France
| | - Michel Haïssaguerre
- Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,INSERM U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, Bordeaux, France.,Centre Hospitalier Universitaire, Bordeaux University Hospital, Hopital Cardiologique du Haut Lévèque, Bordeaux, France
| | - Edward J Vigmond
- Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,UMR5251, Centre National De La Recherche Scientifique, Institut de Mathématiques de Bordeaux, Bordeaux, France
| | - Mélèze Hocini
- Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,INSERM U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, Bordeaux, France.,Centre Hospitalier Universitaire, Bordeaux University Hospital, Hopital Cardiologique du Haut Lévèque, Bordeaux, France
| | - Olivier Bernus
- Centre de Recherche Cardio-Thoracique de Bordeaux, Université de Bordeaux, Bordeaux, France.,INSERM U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France.,IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, Bordeaux, France
| |
Collapse
|
31
|
Reduced threshold for store overload-induced Ca 2+ release is a common defect of RyR1 mutations associated with malignant hyperthermia and central core disease. Biochem J 2017; 474:2749-2761. [PMID: 28687594 DOI: 10.1042/bcj20170282] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/30/2017] [Accepted: 07/06/2017] [Indexed: 12/21/2022]
Abstract
Mutations in the skeletal muscle ryanodine receptor (RyR1) cause malignant hyperthermia (MH) and central core disease (CCD), whereas mutations in the cardiac ryanodine receptor (RyR2) lead to catecholaminergic polymorphic ventricular tachycardia (CPVT). Most disease-associated RyR1 and RyR2 mutations are located in the N-terminal, central, and C-terminal regions of the corresponding ryanodine receptor (RyR) isoform. An increasing body of evidence demonstrates that CPVT-associated RyR2 mutations enhance the propensity for spontaneous Ca2+ release during store Ca2+ overload, a process known as store overload-induced Ca2+ release (SOICR). Considering the similar locations of disease-associated RyR1 and RyR2 mutations in the RyR structure, we hypothesize that like CPVT-associated RyR2 mutations, MH/CCD-associated RyR1 mutations also enhance SOICR. To test this hypothesis, we determined the impact on SOICR of 12 MH/CCD-associated RyR1 mutations E2347-del, R2163H, G2434R, R2435L, R2435H, and R2454H located in the central region, and Y4796C, T4826I, L4838V, A4940T, G4943V, and P4973L located in the C-terminal region of the channel. We found that all these RyR1 mutations reduced the threshold for SOICR. Dantrolene, an acute treatment for MH, suppressed SOICR in HEK293 cells expressing the RyR1 mutants R164C, Y523S, R2136H, R2435H, and Y4796C. Interestingly, carvedilol, a commonly used β-blocker that suppresses RyR2-mediated SOICR, also inhibits SOICR in these RyR1 mutant HEK293 cells. Therefore, these results indicate that a reduced SOICR threshold is a common defect of MH/CCD-associated RyR1 mutations, and that carvedilol, like dantrolene, can suppress RyR1-mediated SOICR. Clinical studies of the effectiveness of carvedilol as a long-term treatment for MH/CCD or other RyR1-associated disorders may be warranted.
Collapse
|
32
|
Azam MA, Zamiri N, Massé S, Kusha M, Lai PFH, Nair GK, Tan NS, Labos C, Nanthakumar K. Effects of Late Sodium Current Blockade on Ventricular Refibrillation in a Rabbit Model. Circ Arrhythm Electrophysiol 2017; 10:CIRCEP.116.004331. [PMID: 28314848 DOI: 10.1161/circep.116.004331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND After defibrillation of initial ventricular fibrillation (VF), it is crucial to prevent refibrillation to ensure successful resuscitation outcomes. Inability of the late Na+ current to inactivate leads to intracellular Ca2+ dysregulation and arrhythmias. Our aim was to determine the effects of ranolazine and GS-967, inhibitors of the late Na+ current, on ventricular refibrillation. METHODS AND RESULTS Long-duration VF was induced electrically in Langendorff-perfused rabbit hearts (n=22) and terminated with a defibrillator after 6 minutes. Fibrillating hearts were randomized into 3 groups: treatment with ranolazine, GS-967, or nontreated controls. In the treated groups, hearts were perfused with ranolazine or GS-967 at 2 minutes of VF. In control experiments, perfusion solution was supplemented with isotonic saline in lieu of a drug. Inducibility of refibrillation was assessed after initial long-duration VF by attempting to reinduce VF. Sustained refibrillation was successful in fewer ranolazine-treated (29.17%; P=0.005) or GS-967-treated (45.83%, P=0.035) hearts compared with that in nontreated control hearts (84.85%). In GS-967-treated hearts, significantly more spontaneous termination of initial long-duration VF was observed (66.67%; P=0.01). Ca2+ transient duration was reduced in ranolazine-treated hearts compared with that in controls (P=0.05) and also Ca2+ alternans (P=0.03). CONCLUSIONS Late Na+ current inhibition during long-duration VF reduces the susceptibility to subsequent refibrillation, partially by mitigating dysregulation of intracellular Ca2+. These results suggest the potential therapeutic use of ranolazine and GS-967 and call for further testing in cardiac arrest models.
Collapse
Affiliation(s)
- Mohammed Ali Azam
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Nima Zamiri
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Stéphane Massé
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Marjan Kusha
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Patrick F H Lai
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Govind K Nair
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Nigel S Tan
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Christopher Labos
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Kumaraswamy Nanthakumar
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.).
| |
Collapse
|
33
|
Dantrolene versus amiodarone for cardiopulmonary resuscitation: a randomized, double-blinded experimental study. Sci Rep 2017; 7:40875. [PMID: 28098197 PMCID: PMC5241655 DOI: 10.1038/srep40875] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/13/2016] [Indexed: 11/29/2022] Open
Abstract
Dantrolene was introduced for treatment of malignant hyperthermia. It also has antiarrhythmic properties and may thus be an alternative to amiodarone for the treatment of ventricular fibrillation (VF). Aim of this study was to compare the return of spontaneous circulation (ROSC) with dantrolene and amiodarone in a pig model of cardiac arrest. VF was induced in anesthetized pigs. After 8 min of untreated VF, chest compressions and ventilation were started and one of the drugs (amiodarone 5 mg kg−1, dantrolene 2.5 mg kg−1 or saline) was applied. After 4 min of initial CPR, defibrillation was attempted. ROSC rates, hemodynamics and cerebral perfusion measurements were measured. Initial ROSC rates were 7 of 14 animals in the dantrolene group vs. 5 of 14 for amiodarone, and 3 of 10 for saline). ROSC persisted for the 120 min follow-up in 6 animals in the dantrolene group, 4 after amiodarone and 2 in the saline group (n.s.). Hemodynamics were comparable in both dantrolene group amiodarone group after obtaining ROSC. Dantrolene and amiodarone had similar outcomes in our model of prolonged cardiac arrest, However, hemodynamic stability was not significantly improved using dantrolene. Dantrolene might be an alternative drug for resuscitation and should be further investigated.
Collapse
|
34
|
King B, Porta-Sánchez A, Massé S, Zamiri N, Balasundaram K, Kusha M, Jackson N, Haldar S, Umapathy K, Nanthakumar K. Effect of spatial resolution and filtering on mapping cardiac fibrillation. Heart Rhythm 2017; 14:608-615. [PMID: 28104480 DOI: 10.1016/j.hrthm.2017.01.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Endocardial mapping tools use variable interelectrode resolution, whereas body surface mapping tools use narrow bandpass filtering (BPF) to map fibrillatory mechanisms established by high-resolution optical imaging. OBJECTIVE The purpose of this study was to study the effect of resolution and BPF on the underlying mechanism being mapped. METHODS Hearts from 14 healthy New Zealand white rabbits were Langendorff perfused. We studied the effect of spatial resolution and BPF on the location and characterization of rotors by comparing phase singularities detected by high-resolution unfiltered optical maps and of fibrillating myocardium with decimated and filtered maps with simulated electrode spacing of 2, 5, and 8 mm. RESULTS As we decimated the maps with 2-mm, 5-mm, and 8-mm interelectrode spacing, the mean ( ± SD) number of rotors detected decreased from 10.2 ± 9.6, 1.6 ± 3.2, and 0.2 ± 0.5, respectively. Lowering the resolution led to synthesized pseudo-rotors that may be inappropriately identified. Applying a BPF led to fewer mean phase singularities detected (248 ± 207 vs 333 ± 130; P<.01), giving the appearance of pseudo-spatial stability measured as translation index (with BPF 3.6 ± 0.4 mm vs 4.0 ± 0.5 mm without BPF; P<.01) and pseudo-temporal stability with longer duration (70.0 ± 17.6 ms in BPF maps vs 44.1 ± 6.6 ms in unfiltered maps; P<.001) than true underlying fibrillating myocardium mapped. CONCLUSION Electrode resolution and BPF of electrograms can result in distortion of the underlying electrophysiology of fibrillation. Newer mapping techniques need to demonstrate sensitivity analysis to quantify the degree of distortion before clinical use to avoid inaccurate electrophysiologic interpretation.
Collapse
Affiliation(s)
| | | | | | - Nima Zamiri
- University Health Network, Toronto, Ontario, Canada
| | | | - Marjan Kusha
- University Health Network, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
35
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
36
|
Calcium Handling Abnormalities as a Target for Atrial Fibrillation Therapeutics: How Close to Clinical Implementation? J Cardiovasc Pharmacol 2016; 66:515-22. [PMID: 25830486 DOI: 10.1097/fjc.0000000000000253] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia with a substantial impact on morbidity and mortality. Antiarrhythmic drugs play a major role in rhythm-control therapy of AF. However, currently available agents exhibit limited efficacy and pronounced adverse effects, notably drug-induced proarrhythmia. Recent experimental studies have identified that Ca handling abnormalities are critical elements in AF pathophysiology with central roles in atrial ectopic activity, reentry, and atrial remodeling suggesting that Ca handling abnormalities could be promising targets for novel AF therapeutics. Here, we summarize key aspects of AF-related Ca-handling abnormalities, describe currently available compounds targeting atrial Ca handling, and highlight potential novel targets and experimental drugs currently under investigation. Finally, we assess how close AF therapeutics based on Ca-handling abnormalities are to clinical implementation.
Collapse
|
37
|
Hartmann N, Pabel S, Herting J, Schatter F, Renner A, Gummert J, Schotola H, Danner BC, Maier LS, Frey N, Hasenfuss G, Fischer TH, Sossalla S. Antiarrhythmic effects of dantrolene in human diseased cardiomyocytes. Heart Rhythm 2016; 14:412-419. [PMID: 27650424 DOI: 10.1016/j.hrthm.2016.09.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Indexed: 01/27/2023]
Abstract
BACKGROUND Cardiac type 2 ryanodine receptors (RyR2s) play a pivotal role in cellular electrophysiology and contractility. Increased RyR2-mediated diastolic sarcoplasmic reticulum (SR) Ca2+ release is linked to heart failure (HF) and arrhythmias. Dantrolene, a drug used for the treatment of malignant hyperthermia, is known to stabilize RyRs in skeletal muscle. OBJECTIVE The purpose of this study was to investigate the effects of dantrolene on arrhythmogenic triggers and contractile function in human atrial fibrillation (AF) and HF cardiomyocytes (CM). METHODS Human CM were isolated from either patients with HF (ventricular) or patients with AF (atrial), and Ca2+ imaging, patch-clamp, or muscle strip experiments were performed. RESULTS After exposure to dantrolene, human atrial AF and left ventricular HF CM showed significant reductions in proarrhythmic SR Ca2+ spark frequency and diastolic SR Ca2+ leak. Moreover, dantrolene decreased the frequency of Ca2+ waves and spontaneous Ca2+ transients in HF CM. Patch-clamp experiments revealed that dantrolene significantly suppressed delayed afterdepolarizations in HF and AF CM. Importantly, dantrolene had no effect on action potential duration in AF or in HF CM. In addition, dantrolene had neutral effects on contractile force of human isometrically twitching ventricular HF trabeculae. CONCLUSION Our study showed that dantrolene beneficially influenced disrupted SR Ca2+ homeostasis in human HF and AF CM. Cellular arrhythmogenic triggers were potently suppressed by dantrolene, whereas action potential duration and contractility were not affected. As a clinically approved drug for the treatment of malignant hyperthermia, dantrolene may be a potential antiarrhythmic drug for patients with rhythm disorders and merits further clinical investigation.
Collapse
Affiliation(s)
- Nico Hartmann
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - Steffen Pabel
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - Jonas Herting
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - Felix Schatter
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - André Renner
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Jan Gummert
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Hanna Schotola
- Department of Anesthesiology, Emergency and Intensive Care Medicine, Georg-August-University Göttingen, Göttingen, Germany
| | - Bernhard C Danner
- Department of Thoracic and Cardiovascular Surgery, Georg-August-University Göttingen, Göttingen, Germany
| | - Lars S Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Norbert Frey
- Department of Internal Medicine III: Cardiology and Angiology, University of Kiel, Kiel, Germany; DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Thomas H Fischer
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - Samuel Sossalla
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany; Department of Internal Medicine III: Cardiology and Angiology, University of Kiel, Kiel, Germany; DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
38
|
Arevalo HJ, Boyle PM, Trayanova NA. Computational rabbit models to investigate the initiation, perpetuation, and termination of ventricular arrhythmia. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 121:185-94. [PMID: 27334789 DOI: 10.1016/j.pbiomolbio.2016.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/13/2016] [Indexed: 12/29/2022]
Abstract
Current understanding of cardiac electrophysiology has been greatly aided by computational work performed using rabbit ventricular models. This article reviews the contributions of multiscale models of rabbit ventricles in understanding cardiac arrhythmia mechanisms. This review will provide an overview of multiscale modeling of the rabbit ventricles. It will then highlight works that provide insights into the role of the conduction system, complex geometric structures, and heterogeneous cellular electrophysiology in diseased and healthy rabbit hearts to the initiation and maintenance of ventricular arrhythmia. Finally, it will provide an overview on the contributions of rabbit ventricular modeling on understanding the mechanisms underlying shock-induced defibrillation.
Collapse
Affiliation(s)
- Hermenegild J Arevalo
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Simula Research Laboratory, Oslo, Norway
| | - Patrick M Boyle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Natalia A Trayanova
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
39
|
Acsai K, Ördög B, Varró A, Nánási PP. Role of the dysfunctional ryanodine receptor - Na(+)-Ca(2+)exchanger axis in progression of cardiovascular diseases: What we can learn from pharmacological studies? Eur J Pharmacol 2016; 779:91-101. [PMID: 26970182 DOI: 10.1016/j.ejphar.2016.03.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/28/2022]
Abstract
Abnormal Ca(2+)homeostasis is often associated with chronic cardiovascular diseases, such as hypertension, heart failure or cardiac arrhythmias, and typically contributes to the basic ethiology of the disease. Pharmacological targeting of cardiac Ca(2+)handling has great therapeutic potential offering invaluable options for the prevention, slowing down the progression or suppression of the harmful outcomes like life threatening cardiac arrhythmias. In this review we outline the existing knowledge on the involvement of malfunction of the ryanodine receptor and the Na(+)-Ca(2+)exchanger in disturbances of Ca(2+)homeostasis and discuss important proof of concept pharmacological studies targeting these mechanisms in context of hypertension, heart failure, atrial fibrillation and ventricular arrhythmias. We emphasize the promising results of preclinical studies underpinning the potential benefits of the therapeutic strategies based on ryanodine receptor or Na(+)-Ca(2+)exchanger inhibition.
Collapse
Affiliation(s)
- Károly Acsai
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Balázs Ördög
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - András Varró
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary; Department of Pharmacology and Pharmacotherapy, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Péter P Nánási
- Department of Physiology, University of Debrecen, Debrecen, Hungary; Department of Dentistry, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
40
|
Ríos E, Figueroa L, Manno C, Kraeva N, Riazi S. The couplonopathies: A comparative approach to a class of diseases of skeletal and cardiac muscle. ACTA ACUST UNITED AC 2016; 145:459-74. [PMID: 26009541 PMCID: PMC4442791 DOI: 10.1085/jgp.201411321] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A novel category of diseases of striated muscle is proposed, the couplonopathies, as those that affect components of the couplon and thereby alter its operation. Couplons are the functional units of intracellular calcium release in excitation–contraction coupling. They comprise dihydropyridine receptors, ryanodine receptors (Ca2+ release channels), and a growing list of ancillary proteins whose alteration may lead to disease. Within a generally similar plan, the couplons of skeletal and cardiac muscle show, in a few places, marked structural divergence associated with critical differences in the mechanisms whereby they fulfill their signaling role. Most important among these are the presence of a mechanical or allosteric communication between voltage sensors and Ca2+ release channels, exclusive to the skeletal couplon, and the smaller capacity of the Ca stores in cardiac muscle, which results in greater swings of store concentration during physiological function. Consideration of these structural and functional differences affords insights into the pathogenesis of several couplonopathies. The exclusive mechanical connection of the skeletal couplon explains differences in pathogenesis between malignant hyperthermia (MH) and catecholaminergic polymorphic ventricular tachycardia (CPVT), conditions most commonly caused by mutations in homologous regions of the skeletal and cardiac Ca2+ release channels. Based on mechanistic considerations applicable to both couplons, we identify the plasmalemma as a site of secondary modifications, typically an increase in store-operated calcium entry, that are relevant in MH pathogenesis. Similar considerations help explain the different consequences that mutations in triadin and calsequestrin have in these two tissues. As more information is gathered on the composition of cardiac and skeletal couplons, this comparative and mechanistic approach to couplonopathies should be useful to understand pathogenesis, clarify diagnosis, and propose tissue-specific drug development.
Collapse
Affiliation(s)
- Eduardo Ríos
- Section of Cellular Signaling, Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL 60612
| | - Lourdes Figueroa
- Section of Cellular Signaling, Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL 60612
| | - Carlo Manno
- Section of Cellular Signaling, Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL 60612
| | - Natalia Kraeva
- Malignant Hyperthermia Investigation Unit, University Health Network, Toronto General Hospital, Toronto, Ontario M5G 2C4, Canada
| | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, University Health Network, Toronto General Hospital, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
41
|
|
42
|
Oo YW, Gomez-Hurtado N, Walweel K, van Helden DF, Imtiaz MS, Knollmann BC, Laver DR. Essential Role of Calmodulin in RyR Inhibition by Dantrolene. Mol Pharmacol 2015; 88:57-63. [PMID: 25920678 PMCID: PMC4468648 DOI: 10.1124/mol.115.097691] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 04/28/2015] [Indexed: 12/17/2022] Open
Abstract
Dantrolene is the first line therapy of malignant hyperthermia. Animal studies suggest that dantrolene also protects against heart failure and arrhythmias caused by spontaneous Ca(2+) release. Although dantrolene inhibits Ca(2+) release from the sarcoplasmic reticulum of skeletal and cardiac muscle preparations, its mechanism of action has remained controversial, because dantrolene does not inhibit single ryanodine receptor (RyR) Ca(2+) release channels in lipid bilayers. Here we test the hypothesis that calmodulin (CaM), a physiologic RyR binding partner that is lost during incorporation into lipid bilayers, is required for dantrolene inhibition of RyR channels. In single channel recordings (100 nM cytoplasmic [Ca(2+)] + 2 mM ATP), dantrolene caused inhibition of RyR1 (rabbit skeletal muscle) and RyR2 (sheep) with a maximal inhibition of Po (Emax) to 52 ± 4% of control only after adding physiologic [CaM] = 100 nM. Dantrolene inhibited RyR2 with an IC50 of 0.16 ± 0.03 µM. Mutant N98S-CaM facilitated dantrolene inhibition with an IC50 = 5.9 ± 0.3 nM. In mouse cardiomyocytes, dantrolene had no effect on cardiac Ca(2+) release in the absence of CaM, but reduced Ca(2+) wave frequency (IC50 = 0.42 ± 0.18 µM, Emax = 47 ± 4%) and amplitude (IC50 = 0.19 ± 0.04 µM, Emax = 66 ± 4%) in the presence of 100 nM CaM. We conclude that CaM is essential for dantrolene inhibition of RyR1 and RyR2. Its absence explains why dantrolene inhibition of single RyR channels has not been previously observed.
Collapse
Affiliation(s)
- Ye Win Oo
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia (Y.W.O., K.W., D.F.H., M.S.I., D.R.L.); and Division of Clinical Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee (N.G.-H., B.C.K.)
| | - Nieves Gomez-Hurtado
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia (Y.W.O., K.W., D.F.H., M.S.I., D.R.L.); and Division of Clinical Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee (N.G.-H., B.C.K.)
| | - Kafa Walweel
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia (Y.W.O., K.W., D.F.H., M.S.I., D.R.L.); and Division of Clinical Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee (N.G.-H., B.C.K.)
| | - Dirk F van Helden
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia (Y.W.O., K.W., D.F.H., M.S.I., D.R.L.); and Division of Clinical Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee (N.G.-H., B.C.K.)
| | - Mohammad S Imtiaz
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia (Y.W.O., K.W., D.F.H., M.S.I., D.R.L.); and Division of Clinical Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee (N.G.-H., B.C.K.)
| | - Bjorn C Knollmann
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia (Y.W.O., K.W., D.F.H., M.S.I., D.R.L.); and Division of Clinical Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee (N.G.-H., B.C.K.)
| | - Derek R Laver
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, New South Wales, Australia (Y.W.O., K.W., D.F.H., M.S.I., D.R.L.); and Division of Clinical Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee (N.G.-H., B.C.K.)
| |
Collapse
|
43
|
Domeier TL, Roberts CJ, Gibson AK, Hanft LM, McDonald KS, Segal SS. Dantrolene suppresses spontaneous Ca2+ release without altering excitation-contraction coupling in cardiomyocytes of aged mice. Am J Physiol Heart Circ Physiol 2014; 307:H818-29. [PMID: 25038147 DOI: 10.1152/ajpheart.00287.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac dysfunction in the aged heart reflects abnormalities in cardiomyocyte Ca(2+) homeostasis including altered Ca(2+) cycling through the sarcoplasmic reticulum (SR). The ryanodine receptor antagonist dantrolene exerts antiarrhythmic effects by preventing spontaneous diastolic Ca(2+) release from the SR. We tested the hypothesis that dantrolene prevents spontaneous Ca(2+) release without altering excitation-contraction coupling in aged myocardium. Left ventricular cardiomyocytes isolated from young (3 to 4 mo) and aged (24-26 mo) C57BL/6 mice were loaded with the Ca(2+) indicator fluo-4. Amplitudes of action potential-induced Ca(2+) transients at 1-Hz pacing were similar between young and aged mice, yet cell shortening was impaired in aged mice. Isoproterenol (1 μM) increased Ca(2+) transient amplitude and cell shortening to identical levels in young and aged; dantrolene (1 μM) had no effect on Ca(2+) transients or cell shortening during pacing. Under Ca(2+) overload conditions induced with 10 mM extracellular Ca(2+) concentration, spontaneous Ca(2+) waves were of diminished amplitude and associated with lower SR Ca(2+) content in aged versus young mice. Despite no effect in young mice, dantrolene increased SR Ca(2+) content and Ca(2+) wave amplitude in aged mice. In the presence of isoproterenol following rest from 1-Hz pacing, Ca(2+) spark frequency was elevated in aged mice, yet the time to spontaneous Ca(2+) wave was similar between young and aged mice; dantrolene decreased Ca(2+) spark frequency and prolonged the time to Ca(2+) wave onset in aged mice with no effect in young mice. Thus dantrolene attenuates diastolic Ca(2+) release in the aged murine heart that may prove useful in preventing cardiac dysfunction.
Collapse
Affiliation(s)
- Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Cale J Roberts
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Anne K Gibson
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Laurin M Hanft
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
44
|
Walton RD, Martinez ME, Bishop MJ, Hocini M, Haïssaguerre M, Plank G, Bernus O, Vigmond EJ. Influence of the Purkinje-muscle junction on transmural repolarization heterogeneity. Cardiovasc Res 2014; 103:629-40. [PMID: 24997066 DOI: 10.1093/cvr/cvu165] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS To elucidate the properties of the PMJ and myocardium underlying these effects. Transmural heterogeneity of action potential duration (APD) is known to play an important role in arrhythmogenesis. Regions of non-uniformities of APD gradients often overlap considerably with the location of Purkinje-muscle junctions (PMJs). We therefore hypothesized that such junctions are novel sources of local endocardial and transmural heterogeneity of repolarization, and that remodelling due to heart failure modulates this response. METHODS AND RESULTS Spatial gradients of endocardial APD in left ventricular wedge preparations from healthy sheep (n = 5) were correlated with locations of PMJs identified through Purkinje stimulation under optical mapping. APD prolongation was dependent on proximity of the PMJ to the imaged surface, whereby shallow PMJs significantly modulated local APD when stimulating either Purkinje (P = 0.0116) or endocardium (P = 0.0123). In addition, we model a PMJ in 5 × 5× 10 mm transmural tissue wedges using healthy and novel failing human ventricular and Purkinje ionic models. Short distances of the PMJ to cut surfaces (<0.875 mm) revealed that APD maxima were localized to the PMJ in healthy myocardium, whereas APD minima were observed in failing myocardium. Amplitudes and spatial gradients of APD were prominent at functional PMJs and quiescent PMJs. Furthermore, increasing the extent of Purkinje fibre branching or decreasing tissue conductivity augmented local APD prolongation in both failing and non-failing models. CONCLUSIONS The Purkinje network has the potential to influence myocardial AP morphology and rate-dependent behaviour, and furthermore to underlie enhanced transmural APD heterogeneities and spatial gradients of APD in non-failing and failing myocardium.
Collapse
Affiliation(s)
- Richard D Walton
- Université de Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux U1045, Bordeaux, France Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, CRCTB U1045, PTIB - Campus Xavier Arnozan, Avenue du Haut Lévêque, 33600 Bordeaux, France
| | - Marine E Martinez
- Université de Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux U1045, Bordeaux, France Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, CRCTB U1045, PTIB - Campus Xavier Arnozan, Avenue du Haut Lévêque, 33600 Bordeaux, France
| | - Martin J Bishop
- Biomedical Engineering Department, Division of Imaging Sciences, King's College London, London, UK
| | - Mélèze Hocini
- Université de Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux U1045, Bordeaux, France Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, CRCTB U1045, PTIB - Campus Xavier Arnozan, Avenue du Haut Lévêque, 33600 Bordeaux, France CHU de Bordeaux, Hôpital du Haut lévêque, Pessac, France
| | - Michel Haïssaguerre
- Université de Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux U1045, Bordeaux, France Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, CRCTB U1045, PTIB - Campus Xavier Arnozan, Avenue du Haut Lévêque, 33600 Bordeaux, France CHU de Bordeaux, Hôpital du Haut lévêque, Pessac, France
| | - Gernot Plank
- Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - Olivier Bernus
- Université de Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux U1045, Bordeaux, France Inserm U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Bordeaux, France L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, CRCTB U1045, PTIB - Campus Xavier Arnozan, Avenue du Haut Lévêque, 33600 Bordeaux, France
| | - Edward J Vigmond
- L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Université de Bordeaux, CRCTB U1045, PTIB - Campus Xavier Arnozan, Avenue du Haut Lévêque, 33600 Bordeaux, France L'Institut de Mathématiques de Bordeaux UMR 5251, Université de Bordeaux, Bordeaux, France Department of Electrical and Computer Engineering, University of Calgary, Calgary, Canada
| |
Collapse
|
45
|
Affiliation(s)
- Dan M Roden
- Vanderbilt University School of Medicine, Nashville, TN
| | | |
Collapse
|