1
|
Zhou S, Liu Y, Huang X, Wu C, Pórszász R. Omecamtiv Mecarbil in the treatment of heart failure: the past, the present, and the future. Front Cardiovasc Med 2024; 11:1337154. [PMID: 38566963 PMCID: PMC10985333 DOI: 10.3389/fcvm.2024.1337154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Heart failure, a prevailing global health issue, imposes a substantial burden on both healthcare systems and patients worldwide. With an escalating prevalence of heart failure, prolonged survival rates, and an aging demographic, an increasing number of individuals are progressing to more advanced phases of this incapacitating ailment. Against this backdrop, the quest for pharmacological agents capable of addressing the diverse subtypes of heart failure becomes a paramount pursuit. From this viewpoint, the present article focuses on Omecamtiv Mecarbil (OM), an emerging chemical compound said to exert inotropic effects without altering calcium homeostasis. For the first time, as a review, the present article uniquely started from the very basic pathophysiology of heart failure, its classification, and the strategies underpinning drug design, to on-going debates of OM's underlying mechanism of action and the latest large-scale clinical trials. Furthermore, we not only saw the advantages of OM, but also exhaustively summarized the concerns in sense of its effects. These of no doubt make the present article the most systemic and informative one among the existing literature. Overall, by offering new mechanistic insights and therapeutic possibilities, OM has carved a significant niche in the treatment of heart failure, making it a compelling subject of study.
Collapse
Affiliation(s)
- Shujing Zhou
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ying Liu
- Department of Cardiology, Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xufeng Huang
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Chuhan Wu
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Róbert Pórszász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Janssens JV, Raaijmakers AJA, Weeks KL, Bell JR, Mellor KM, Curl CL, Delbridge LMD. The cardiomyocyte origins of diastolic dysfunction: cellular components of myocardial "stiffness". Am J Physiol Heart Circ Physiol 2024; 326:H584-H598. [PMID: 38180448 DOI: 10.1152/ajpheart.00334.2023] [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: 06/08/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The impaired ability of the heart to relax and stretch to accommodate venous return is generally understood to represent a state of "diastolic dysfunction" and often described using the all-purpose noun "stiffness." Despite the now common qualitative usage of this term in fields of cardiac patho/physiology, the specific quantitative concept of stiffness as a molecular and biophysical entity with real practical interpretation in healthy and diseased hearts is sometimes obscure. The focus of this review is to characterize the concept of cardiomyocyte stiffness and to develop interpretation of "stiffness" attributes at the cellular and molecular levels. Here, we consider "stiffness"-related terminology interpretation and make links between cardiomyocyte stiffness and aspects of functional and structural cardiac performance. We discuss cross bridge-derived stiffness sources, considering the contributions of diastolic myofilament activation and impaired relaxation. This includes commentary relating to the role of cardiomyocyte Ca2+ flux and Ca2+ levels in diastole, the troponin-tropomyosin complex role as a Ca2+ effector in diastole, the myosin ADP dissociation rate as a modulator of cross bridge attachment and regulation of cross-bridge attachment by myosin binding protein C. We also discuss non-cross bridge-derived stiffness sources, including the titin sarcomeric spring protein, microtubule and intermediate filaments, and cytoskeletal extracellular matrix interactions. As the prevalence of conditions involving diastolic heart failure has escalated, a more sophisticated understanding of the molecular, cellular, and tissue determinants of cardiomyocyte stiffness offers potential to develop imaging and molecular intervention tools.
Collapse
Affiliation(s)
- Johannes V Janssens
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Antonia J A Raaijmakers
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Kate L Weeks
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
- Department of Diabetes, Monash University, Parkville, Victoria, Australia
| | - James R Bell
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Kimberley M Mellor
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Claire L Curl
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Gao B, Abi-Gerges N, Truong K, Stafford A, Nguyen W, Sutherland W, Vargas HM, Qu Y. Assessment of sarcomere shortening and calcium transient in primary human and dog ventricular myocytes. J Pharmacol Toxicol Methods 2023; 123:107278. [PMID: 37268094 DOI: 10.1016/j.vascn.2023.107278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 06/04/2023]
Abstract
Understanding translation from preclinical observations to clinical findings is important for evaluating the efficacy and safety of novel compounds. Of relevance to cardiac safety is profiling drug effects on cardiomyocyte (CM) sarcomere shortening and intracellular Ca2+ dynamics. Although CM from different animal species have been used to assess such effects, primary human CM isolated from human organ donor heart represent an ideal non-animal alternative approach. We performed a study to evaluate primary human CM and have them compared to freshly isolated dog cardiomyocytes for their basic function and responses to positive inotropes with well-known mechanisms. Our data showed that simultaneous assessment of sarcomere shortening and Ca2+-transient can be performed with both myocytes using the IonOptix system. Amplitude of sarcomere shortening and Ca2+-transient (CaT) were significantly higher in dog compared to human CM in the basic condition (absence of treatment), while longer duration of sarcomere shortening and CaT were observed in human cells. We observed that human and dog CMs have similar pharmacological responses to five inotropes with different mechanisms, including dobutamine and isoproterenol (β-adrenergic stimulation), milrinone (PDE3 inhibition), pimobendan and levosimendan (increase of Ca2+sensitization as well as PDE3 inhibition). In conclusion, our study suggests that myocytes obtained from both human donor hearts and dog hearts can be used to simultaneously assess drug-induced effects on sarcomere shortening and CaT using the IonOptix platform.
Collapse
Affiliation(s)
- BaoXi Gao
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - Ky Truong
- AnaBios Corporation, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - Alexa Stafford
- AnaBios Corporation, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - William Nguyen
- AnaBios Corporation, 3030 Bunker Hill St, San Diego, CA 92109, USA
| | - Weston Sutherland
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Hugo M Vargas
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Yusheng Qu
- Amgen Research, Translational Safety & Bioanalytical Sciences, Amgen Inc, 1 Amgen Center Drive, Thousand Oaks, CA 91320, USA
| |
Collapse
|
4
|
Ting CY, Shih CL, Yu MC, Wu CL, Wu SN. Characterization of Stimulatory Action on Voltage-Gated Na + Currents Caused by Omecamtiv Mecarbil, Known to Be a Myosin Activator. Biomedicines 2023; 11:biomedicines11051351. [PMID: 37239022 DOI: 10.3390/biomedicines11051351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Omecamtiv mecarbil (OM, CK-1827452) is recognized as an activator of myosin and has been demonstrated to be beneficial for the treatment of systolic heart failure. However, the mechanisms by which this compound interacts with ionic currents in electrically excitable cells remain largely unknown. The objective of this study was to investigate the effects of OM on ionic currents in GH3 pituitary cells and Neuro-2a neuroblastoma cells. In GH3 cells, whole-cell current recordings showed that the addition of OM had different potencies in stimulating the transient (INa(T)) and late components (INa(L)) of the voltage-gated Na+ current (INa) with different potencies in GH3 cells. The EC50 value required to observe the stimulatory effect of this compound on INa(T) or INa(L) in GH3 cells was found to be 15.8 and 2.3 µM, respectively. Exposure to OM did not affect the current versus voltage relationship of INa(T). However, the steady-state inactivation curve of the current was observed to shift towards a depolarized potential of approximately 11 mV, with no changes in the slope factor of the curve. The addition of OM resulted in an increase in the decaying time constant during the cumulative inhibition of INa(T) in response to pulse-train depolarizing stimuli. Furthermore, the presence of OM led to a shortening of the recovery time constant in the slow inactivation of INa(T). Adding OM also resulted in an augmentation of the strength of the window Na+ current, which was evoked by a short ascending ramp voltage. However, the OM exposure had little to no effect on the magnitude of L-type Ca2+ currents in GH3 cells. On the other hand, the delayed-rectifier K+ currents in GH3 cells were observed to be mildly suppressed in its presence. Neuro-2a cells also showed a susceptibility to the differential stimulation of INa(T) or INa(L) upon the addition of OM. Molecular analysis revealed potential interactions between the OM molecule and hNaV1.7 channels. Overall, the direct stimulation of INa(T) and INa(L) by OM is assumed to not be mediated by an interaction with myosin, and this has potential implications for its pharmacological or therapeutic actions occurring in vivo.
Collapse
Affiliation(s)
- Chih-Yu Ting
- Department of Emergency Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Chia-Lung Shih
- Clinical Research Center, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Meng-Cheng Yu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Chao-Liang Wu
- Clinical Research Center, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
- School of Medicine, National Sun Yat-Sen University College of Medicine, Kaohsiung 80424, Taiwan
| |
Collapse
|
5
|
Ráduly AP, Sárkány F, Kovács MB, Bernát B, Juhász B, Szilvássy Z, Porszász R, Horváth B, Szentandrássy N, Nánási P, Csanádi Z, Édes I, Tóth A, Papp Z, Priksz D, Borbély A. The Novel Cardiac Myosin Activator Danicamtiv Improves Cardiac Systolic Function at the Expense of Diastolic Dysfunction In Vitro and In Vivo: Implications for Clinical Applications. Int J Mol Sci 2022; 24:ijms24010446. [PMID: 36613900 PMCID: PMC9820393 DOI: 10.3390/ijms24010446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Recent cardiotropic drug developments have focused on cardiac myofilaments. Danicamtiv, the second direct myosin activator, has achieved encouraging results in preclinical and clinical studies, thus implicating its potential applicability in the treatment of heart failure with reduced ejection fraction (HFrEF). Here, we analyzed the inotropic effects of danicamtiv in detail. To this end, changes in sarcomere length and intracellular Ca2+ levels were monitored in parallel, in enzymatically isolated canine cardiomyocytes, and detailed echocardiographic examinations were performed in anesthetized rats in the absence or presence of danicamtiv. The systolic and diastolic sarcomere lengths decreased; contraction and relaxation kinetics slowed down with increasing danicamtiv concentrations without changes in intracellular Ca2+ transients in vitro. Danicamtiv evoked remarkable increases in left ventricular ejection fraction and fractional shortening, also reflected by changes in systolic strain. Nevertheless, the systolic ejection time was significantly prolonged, the ratio of diastolic to systolic duration was reduced, and signs of diastolic dysfunction were also observed upon danicamtiv treatment in vivo. Taken together, danicamtiv improves cardiac systolic function, but it can also limit diastolic performance, especially at high drug concentrations.
Collapse
Affiliation(s)
- Arnold Péter Ráduly
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Kálmán Laki Doctoral School, University of Debrecen, 4032 Debrecen, Hungary
| | - Fruzsina Sárkány
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Kálmán Laki Doctoral School, University of Debrecen, 4032 Debrecen, Hungary
| | - Máté Balázs Kovács
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Brigitta Bernát
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Róbert Porszász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Norbert Szentandrássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - Péter Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - Zoltán Csanádi
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - István Édes
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Kálmán Laki Doctoral School, University of Debrecen, 4032 Debrecen, Hungary
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, 4032 Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Kálmán Laki Doctoral School, University of Debrecen, 4032 Debrecen, Hungary
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, 4032 Debrecen, Hungary
- Correspondence: ; Tel.: +36-52-255-978/54329
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Attila Borbély
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Kálmán Laki Doctoral School, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
6
|
Nagy N, Tóth N, Nánási PP. Antiarrhythmic and Inotropic Effects of Selective Na +/Ca 2+ Exchanger Inhibition: What Can We Learn from the Pharmacological Studies? Int J Mol Sci 2022; 23:ijms232314651. [PMID: 36498977 PMCID: PMC9736231 DOI: 10.3390/ijms232314651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
Life-long stable heart function requires a critical balance of intracellular Ca2+. Several ion channels and pumps cooperate in a complex machinery that controls the influx, release, and efflux of Ca2+. Probably one of the most interesting and most complex players of this crosstalk is the Na+/Ca2+ exchanger, which represents the main Ca2+ efflux mechanism; however, under some circumstances, it can also bring Ca2+ into the cell. Therefore, the inhibition of the Na+/Ca2+ exchanger has emerged as one of the most promising possible pharmacological targets to increase Ca2+ levels, to decrease arrhythmogenic depolarizations, and to reduce excessive Ca2+ influx. In line with this, as a response to increasing demand, several more or less selective Na+/Ca2+ exchanger inhibitor compounds have been developed. In the past 20 years, several results have been published regarding the effect of Na+/Ca2+ exchanger inhibition under various circumstances, e.g., species, inhibitor compounds, and experimental conditions; however, the results are often controversial. Does selective Na+/Ca2+ exchanger inhibition have any future in clinical pharmacological practice? In this review, the experimental results of Na+/Ca2+ exchanger inhibition are summarized focusing on the data obtained by novel highly selective inhibitors.
Collapse
Affiliation(s)
- Norbert Nagy
- ELKH-SZTE Research Group of Cardiovascular Pharmacology, 6720 Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-682; Fax: +36-62-545-680
| | - Noémi Tóth
- ELKH-SZTE Research Group of Cardiovascular Pharmacology, 6720 Szeged, Hungary
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
7
|
Streiff ME, Sachse FB. Effects of Sarcolemmal Background Ca2+ Entry and Sarcoplasmic Ca2+ Leak Currents on Electrophysiology and Ca2+ Transients in Human Ventricular Cardiomyocytes: A Computational Comparison. Front Physiol 2022; 13:916278. [PMID: 35784869 PMCID: PMC9243544 DOI: 10.3389/fphys.2022.916278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022] Open
Abstract
The intricate regulation of the compartmental Ca2+ concentrations in cardiomyocytes is critical for electrophysiology, excitation-contraction coupling, and other signaling pathways. Research into the complex signaling pathways is motivated by cardiac pathologies including arrhythmia and maladaptive myocyte remodeling, which result from Ca2+ dysregulation. Of interest to this investigation are two types of Ca2+ currents in cardiomyocytes: 1) background Ca2+ entry, i.e., Ca2+ transport across the sarcolemma from the extracellular space into the cytosol, and 2) Ca2+ leak from the sarcoplasmic reticulum (SR) across the SR membrane into the cytosol. Candidates for the ion channels underlying background Ca2+ entry and SR Ca2+ leak channels include members of the mechano-modulated transient receptor potential (TRP) family. We used a mathematical model of a human ventricular myocyte to analyze the individual contributions of background Ca2+ entry and SR Ca2+ leak to the modulation of Ca2+ transients and SR Ca2+ load at rest and during action potentials. Background Ca2+ entry exhibited a positive relationship with both [Ca2+]i and [Ca2+]SR. Modulating SR Ca2+ leak had opposite effects of background Ca2+ entry. Effects of SR Ca2+ leak on Ca2+ were particularly pronounced at lower pacing frequency. In contrast to the pronounced effects of background and leak Ca2+ currents on Ca2+ concentrations, the effects on cellular electrophysiology were marginal. Our studies provide quantitative insights into the differential modulation of compartmental Ca2+ concentrations by the background and leak Ca2+ currents. Furthermore, our studies support the hypothesis that TRP channels play a role in strain-modulation of cardiac contractility. In summary, our investigations shed light on the physiological effects of the background and leak Ca2+ currents and their contribution to the development of disease caused by Ca2+ dysregulation.
Collapse
Affiliation(s)
- Molly E. Streiff
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Frank B. Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- *Correspondence: Frank B. Sachse,
| |
Collapse
|
8
|
Day SM, Tardiff JC, Ostap EM. Myosin modulators: emerging approaches for the treatment of cardiomyopathies and heart failure. J Clin Invest 2022; 132:148557. [PMID: 35229734 PMCID: PMC8884898 DOI: 10.1172/jci148557] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Myosin modulators are a novel class of pharmaceutical agents that are being developed to treat patients with a range of cardiomyopathies. The therapeutic goal of these drugs is to target cardiac myosins directly to modulate contractility and cardiac power output to alleviate symptoms that lead to heart failure and arrhythmias, without altering calcium signaling. In this Review, we discuss two classes of drugs that have been developed to either activate (omecamtiv mecarbil) or inhibit (mavacamten) cardiac contractility by binding to β-cardiac myosin (MYH7). We discuss progress in understanding the mechanisms by which the drugs alter myosin mechanochemistry, and we provide an appraisal of the results from clinical trials of these drugs, with consideration for the importance of disease heterogeneity and genetic etiology for predicting treatment benefit.
Collapse
Affiliation(s)
- Sharlene M Day
- Division of Cardiovascular Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jil C Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | - E Michael Ostap
- Pennsylvania Muscle Institute and Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Rhoden A, Schulze T, Pietsch N, Christ T, Hansen A, Eschenhagen T. Comprehensive analyses of the inotropic compound omecamtiv mecarbil in rat and human cardiac preparations. Am J Physiol Heart Circ Physiol 2022; 322:H373-H385. [PMID: 35030072 DOI: 10.1152/ajpheart.00534.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Omecamtiv mecarbil (OM), a myosin activator, was reported to induce complex concentration- and species-dependent effects on contractile function and clinical studies indicated a low therapeutic index with diastolic dysfunction at concentrations above 1 µM. To further characterize effects of OM in a human context and under different preload conditions, we constructed a setup that allows isometric contractility analyses of human induced pluripotent stem cell (hiPSC)-derived engineered heart tissues (EHTs). The results were compared to effects of OM on the very same EHTs measured under auxotonic conditions. OM induced a sustained, concentration-dependent increase in time-to-peak under all conditions (maximally 2-3 fold). Peak force, in contrast, was increased by OM only in human, but not rat EHTs and only under isometric conditions, varied between hiPSC lines and showed a biphasic concentration-dependency with maximal effects at 1 µM. Relaxation time tended to fall under auxotonic and strongly increase under isometric conditions, again with biphasic concentration-dependency. Diastolic tension concentration-dependently increased under all conditions. The latter was reduced by an inhibitor of the mitochondrial sodium calcium exchanger (CGP-37157). OM induced increases in mitochondrial oxidation in isolated cardiomyocytes, indicating that OM, an inotrope that does not increase intracellular and mitochondrial Ca2+, can induce mismatch between an increase in ATP and ROS production and unstimulated mitochondrial redox capacity. Taken together, we developed a novel setup well suitable for isometric measurements of EHTs. The effects of OM on contractility and diastolic tension are complex with concentration-, time-, species- and loading-dependent differences. Effects on mitochondrial function require further studies.
Collapse
Affiliation(s)
- Alexandra Rhoden
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Schulze
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Niels Pietsch
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
10
|
Kiss D, Horváth B, Hézső T, Dienes C, Kovács Z, Topal L, Szentandrássy N, Almássy J, Prorok J, Virág L, Bányász T, Varró A, Nánási PP, Magyar J. Late Na + Current Is [Ca 2+] i-Dependent in Canine Ventricular Myocytes. Pharmaceuticals (Basel) 2021; 14:ph14111142. [PMID: 34832924 PMCID: PMC8623624 DOI: 10.3390/ph14111142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Enhancement of the late sodium current (INaL) increases arrhythmia propensity in the heart, whereas suppression of the current is antiarrhythmic. In the present study, we investigated INaL in canine ventricular cardiomyocytes under action potential voltage-clamp conditions using the selective Na+ channel inhibitors GS967 and tetrodotoxin. Both 1 µM GS967 and 10 µM tetrodotoxin dissected largely similar inward currents. The amplitude and integral of the GS967-sensitive current was significantly smaller after the reduction of intracellular Ca2+ concentration ([Ca2+]i) either by superfusion of the cells with 1 µM nisoldipine or by intracellular application of 10 mM BAPTA. Inhibiting calcium/calmodulin-dependent protein kinase II (CaMKII) by KN-93 or the autocamtide-2-related inhibitor peptide similarly reduced the amplitude and integral of INaL. Action potential duration was shortened in a reverse rate-dependent manner and the plateau potential was depressed by GS967. This GS967-induced depression of plateau was reduced by pretreatment of the cells with BAPTA-AM. We conclude that (1) INaL depends on the magnitude of [Ca2+]i in canine ventricular cells, (2) this [Ca2+]i-dependence of INaL is mediated by the Ca2+-dependent activation of CaMKII, and (3) INaL is augmented by the baseline CaMKII activity.
Collapse
Affiliation(s)
- Dénes Kiss
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
- Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Hézső
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
| | - Csaba Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
| | - Zsigmond Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
| | - Leila Topal
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary; (L.T.); (J.P.); (L.V.); (A.V.)
| | - Norbert Szentandrássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
| | - János Prorok
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary; (L.T.); (J.P.); (L.V.); (A.V.)
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6725 Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary; (L.T.); (J.P.); (L.V.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6725 Szeged, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary; (L.T.); (J.P.); (L.V.); (A.V.)
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6725 Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6725 Szeged, Hungary
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: ; Tel.: +36-52255575; Fax: +36-52255116
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (D.K.); (B.H.); (T.H.); (C.D.); (Z.K.); (N.S.); (J.A.); (T.B.); (J.M.)
- Division of Sport Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
11
|
Parikh J, Rumbell T, Butova X, Myachina T, Acero JC, Khamzin S, Solovyova O, Kozloski J, Khokhlova A, Gurev V. Generative adversarial networks for construction of virtual populations of mechanistic models: simulations to study Omecamtiv Mecarbil action. J Pharmacokinet Pharmacodyn 2021; 49:51-64. [PMID: 34716531 PMCID: PMC8837558 DOI: 10.1007/s10928-021-09787-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/23/2021] [Indexed: 11/30/2022]
Abstract
Biophysical models are increasingly used to gain mechanistic insights by fitting and reproducing experimental and clinical data. The inherent variability in the recorded datasets, however, presents a key challenge. In this study, we present a novel approach, which integrates mechanistic modeling and machine learning to analyze in vitro cardiac mechanics data and solve the inverse problem of model parameter inference. We designed a novel generative adversarial network (GAN) and employed it to construct virtual populations of cardiac ventricular myocyte models in order to study the action of Omecamtiv Mecarbil (OM), a positive cardiac inotrope. Populations of models were calibrated from mechanically unloaded myocyte shortening recordings obtained in experiments on rat myocytes in the presence and absence of OM. The GAN was able to infer model parameters while incorporating prior information about which model parameters OM targets. The generated populations of models reproduced variations in myocyte contraction recorded during in vitro experiments and provided improved understanding of OM’s mechanism of action. Inverse mapping of the experimental data using our approach suggests a novel action of OM, whereby it modifies interactions between myosin and tropomyosin proteins. To validate our approach, the inferred model parameters were used to replicate other in vitro experimental protocols, such as skinned preparations demonstrating an increase in calcium sensitivity and a decrease in the Hill coefficient of the force–calcium (F–Ca) curve under OM action. Our approach thereby facilitated the identification of the mechanistic underpinnings of experimental observations and the exploration of different hypotheses regarding variability in this complex biological system.
Collapse
Affiliation(s)
| | | | - Xenia Butova
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences (UB RAS), Yekaterinburg, Russia
| | - Tatiana Myachina
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences (UB RAS), Yekaterinburg, Russia
| | - Jorge Corral Acero
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Svyatoslav Khamzin
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences (UB RAS), Yekaterinburg, Russia
| | - Olga Solovyova
- Ural Federal University, Yekaterinburg, Russia.,Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences (UB RAS), Yekaterinburg, Russia
| | | | - Anastasia Khokhlova
- Ural Federal University, Yekaterinburg, Russia.,Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences (UB RAS), Yekaterinburg, Russia
| | | |
Collapse
|
12
|
Lookin O, Kuznetsov D, Protsenko Y. Omecamtiv mecarbil attenuates length-tension relationship in healthy rat myocardium and preserves it in monocrotaline-induced pulmonary heart failure. Clin Exp Pharmacol Physiol 2021; 49:84-93. [PMID: 34459025 DOI: 10.1111/1440-1681.13584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023]
Abstract
The cardiac-specific myosin activator, omecamtiv mecarbil (OM), is an effective inotrope for treating heart failure but its effects on active force and Ca2+ kinetics in healthy and diseased myocardium remain poorly studied. We tested the effect of two concentrations of OM (0.2 and 1 µmol/L in saline) on isometric contraction and Ca-transient (CaT) in right ventricular trabeculae of healthy rats (CONT, n = 8) and rats with monocrotaline-induced pulmonary heart failure (MCT, n = 8). The contractions were obtained under preload of 75%-100% of optimal length (tension-length relationship). The 0.2 µmol/L OM did not affect the diastolic level, amplitude, or kinetics of isometric contraction and CaT, irrespective of the group of rats or preload. The 1 µmol/L OM significantly suppressed active tension-length relationships in CONT but not in MCT, while leading in both groups to a significantly prolonged relaxation. CaT time-to-peak was unaffected in CONT and MCT, but CaT decay was slightly accelerated in its early phase and considerably prolonged in its late phase to a similar extent in both groups. We conclude that the substantial prolongation of CaT decay is due to enhanced Ca2+ utilisation by troponin C mediated by the direct effect of OM on the cooperative activation of myofilaments. The lack of beneficial effect of OM in the healthy rat myocardium may be due to a relatively high level of activating Ca2+ in cells with normal Ca2+ handling, whereas the preservation of the tension-length relationship in the failing heart may relate to the diminished Ca2+ levels of sarcoplasmic reticulum.
Collapse
Affiliation(s)
- Oleg Lookin
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Daniil Kuznetsov
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Yuri Protsenko
- Institute of Immunology and Physiology, Ural Branch of Russian Academy of Sciences, Yekaterinburg, Russian Federation
| |
Collapse
|
13
|
Tóth N, Szlovák J, Kohajda Z, Bitay G, Veress R, Horváth B, Papp JG, Varró A, Nagy N. The development of L-type Ca 2+ current mediated alternans does not depend on the restitution slope in canine ventricular myocardium. Sci Rep 2021; 11:16652. [PMID: 34404848 PMCID: PMC8371021 DOI: 10.1038/s41598-021-95299-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/23/2021] [Indexed: 11/25/2022] Open
Abstract
Cardiac alternans have crucial importance in the onset of ventricular fibrillation. The early explanation for alternans development was the voltage-driven mechanism, where the action potential (AP) restitution steepness was considered as crucial determining factor. Recent results suggest that restitution slope is an inadequate predictor for alternans development, but several studies still claim the role of membrane potential as underlying mechanism of alternans. These controversial data indicate that the relationship of restitution and alternans development is not completely understood. APs were measured by conventional microelectrode technique from canine right ventricular papillary muscles. Ionic currents combined with fluorescent measurements were recorded by patch-clamp technique. APs combined with fluorescent measurements were monitored by sharp microelectrodes. Rapid pacing evoked restitution-independent AP duration (APD) alternans. When non-alternating AP voltage command was used, Ca2+i-transient (CaT) alternans were not observed. When alternating rectangular voltage pulses were applied, CaT alternans were proportional to ICaL amplitude alternans. Selective ICaL inhibition did not influence the fast phase of APD restitution. In this study we found that ICaL has minor contribution in shaping the fast phase of restitution curve suggesting that ICaL—if it plays important role in the alternans mechanism—could be an additional factor that attenuates the reliability of APD restitution slope to predict alternans.
Collapse
Affiliation(s)
- Noémi Tóth
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, P.O. Box 427, 6720, Szeged, Hungary
| | - Jozefina Szlovák
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, P.O. Box 427, 6720, Szeged, Hungary
| | - Zsófia Kohajda
- ELKH-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Gergő Bitay
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, P.O. Box 427, 6720, Szeged, Hungary
| | - Roland Veress
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Julius Gy Papp
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, P.O. Box 427, 6720, Szeged, Hungary.,ELKH-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, P.O. Box 427, 6720, Szeged, Hungary.,ELKH-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dóm tér 12, P.O. Box 427, 6720, Szeged, Hungary. .,ELKH-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary.
| |
Collapse
|
14
|
Ion current profiles in canine ventricular myocytes obtained by the "onion peeling" technique. J Mol Cell Cardiol 2021; 158:153-162. [PMID: 34089737 DOI: 10.1016/j.yjmcc.2021.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/05/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023]
Abstract
The profiles of ion currents during the cardiac action potential can be visualized by the action potential voltage clamp technique. To obtain multiple ion current data from the same cell, the "onion peeling" technique, based on sequential pharmacological dissection of ion currents, has to be applied. Combination of the two methods allows recording of several ion current profiles from the same myocyte under largely physiological conditions. Using this approach, we have studied the densities and integrals of the major cardiac inward (ICa, INCX, INa-late) and outward (IKr, IKs, IK1) currents in canine ventricular cells and studied the correlation between them. For this purpose, canine ventricular cardiomyocytes were chosen because their electrophysiological properties are similar to those of human ones. Significant positive correlation was observed between the density and integral of ICa and IKr, and positive correlation was found also between the integral of ICa and INCX. No further correlations were detected. The Ca2+-sensitivity of K+ currents was studied by comparing their parameters in the case of normal calcium homeostasis and following blockade of ICa. Out of the three K+ currents studied, only IKs was Ca2+-sensitive. The density and integral of IKs was significantly greater, while its time-to-peak value was shorter at normal Ca2+ cycling than following ICa blockade. No differences were detected for IKr or IK1 in this regard. Present results indicate that the positive correlation between ICa and IKr prominently contribute to the balance between inward and outward fluxes during the action potential plateau in canine myocytes. The results also suggest that the profiles of cardiac ion currents have to be studied under physiological conditions, since their behavior may strongly be influenced by the intracellular Ca2+ homeostasis and the applied membrane potential protocol.
Collapse
|
15
|
Dashwood A, Cheesman E, Wong YW, Haqqani H, Beard N, Hay K, Spratt M, Chan W, Molenaar P. Effects of omecamtiv mecarbil on failing human ventricular trabeculae and interaction with (-)-noradrenaline. Pharmacol Res Perspect 2021; 9:e00760. [PMID: 33929079 PMCID: PMC8085933 DOI: 10.1002/prp2.760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 01/10/2023] Open
Abstract
Omecamtiv mecarbil (OM) is a novel medicine for systolic heart failure, targeting myosin to enhance cardiomyocyte performance. To assist translation to clinical practice we investigated OMs effect on explanted human failing hearts, specifically; contractile dynamics, interaction with the β1–adrenoceptor (AR) agonist (−)‐noradrenaline and spontaneous contractions. Left and right ventricular trabeculae from 13 explanted failing hearts, and trabeculae from 58 right atrial appendages of non‐failing hearts, were incubated with or without a single concentration of OM for 120 min. Time to peak force (TPF) and 50% relaxation (t50%) were recorded. In other experiments, trabeculae were observed for spontaneous contractions and cumulative concentration‐effect curves were established to (−)‐noradrenaline at β1‐ARs in the absence or presence of OM. OM prolonged TPF and t50% in ventricular trabeculae (600 nM, 2 µM, p < .001). OM had no significant inotropic effect but reduced time dependent deterioration in contractile strength compared to control (p < .001). OM did not affect the generation of spontaneous contractions. The potency of (−)‐noradrenaline (pEC50 6.05 ± 0.10), for inotropic effect, was unchanged in the presence of OM 600 nM or 2 µM. Co‐incubation with (−)‐noradrenaline reduced TPF and t50%, reversing the negative diastolic effects of OM. OM, at both 600 nM and 2 µM, preserved contractile force in left ventricular trabeculae, but imparted negative diastolic effects in trabeculae from human failing heart. (−)‐Noradrenaline reversed the negative diastolic effects, co‐administration may limit the titration of inotropes by reducing the threshold for ischemic side effects.
Collapse
Affiliation(s)
- Alexander Dashwood
- Heart Lung Institute, The Prince Charles Hospital, Chermside, QLD, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, University of Queensland, Brisbane, QLD, Australia
| | - Elizabeth Cheesman
- Cardio-Vascular Molecular & Therapeutics Translational Research Group, University of Queensland, Brisbane, QLD, Australia
| | - Yee Weng Wong
- Heart Lung Institute, The Prince Charles Hospital, Chermside, QLD, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, University of Queensland, Brisbane, QLD, Australia
| | - Haris Haqqani
- Heart Lung Institute, The Prince Charles Hospital, Chermside, QLD, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, University of Queensland, Brisbane, QLD, Australia
| | - Nicole Beard
- Queensland University of Technology, Brisbane, Australia.,Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Karen Hay
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Melanie Spratt
- Heart Lung Institute, The Prince Charles Hospital, Chermside, QLD, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, University of Queensland, Brisbane, QLD, Australia.,Queensland University of Technology, Brisbane, Australia
| | - Wandy Chan
- Heart Lung Institute, The Prince Charles Hospital, Chermside, QLD, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, University of Queensland, Brisbane, QLD, Australia
| | - Peter Molenaar
- Heart Lung Institute, The Prince Charles Hospital, Chermside, QLD, Australia.,Cardio-Vascular Molecular & Therapeutics Translational Research Group, University of Queensland, Brisbane, QLD, Australia.,Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
16
|
Fülöp GÁ, Oláh A, Csipo T, Kovács Á, Pórszász R, Veress R, Horváth B, Nagy L, Bódi B, Fagyas M, Helgadottir SL, Bánhegyi V, Juhász B, Bombicz M, Priksz D, Nanasi P, Merkely B, Édes I, Csanádi Z, Papp Z, Radovits T, Tóth A. Omecamtiv mecarbil evokes diastolic dysfunction and leads to periodic electromechanical alternans. Basic Res Cardiol 2021; 116:24. [PMID: 33844095 PMCID: PMC8041714 DOI: 10.1007/s00395-021-00866-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 03/31/2021] [Indexed: 01/31/2023]
Abstract
Omecamtiv mecarbil (OM) is a promising novel drug for improving cardiac contractility. We tested the therapeutic range of OM and identified previously unrecognized side effects. The Ca2+ sensitivity of isometric force production (pCa50) and force at low Ca2+ levels increased with OM concentration in human permeabilized cardiomyocytes. OM (1 µM) slowed the kinetics of contractions and relaxations and evoked an oscillation between normal and reduced intracellular Ca2+ transients, action potential lengths and contractions in isolated canine cardiomyocytes. Echocardiographic studies and left ventricular pressure-volume analyses demonstrated concentration-dependent improvements in cardiac systolic function at OM concentrations of 600-1200 µg/kg in rats. Administration of OM at a concentration of 1200 µg/kg was associated with hypotension, while doses of 600-1200 µg/kg were associated with the following aspects of diastolic dysfunction: decreases in E/A ratio and the maximal rate of diastolic pressure decrement (dP/dtmin) and increases in isovolumic relaxation time, left atrial diameter, the isovolumic relaxation constant Tau, left ventricular end-diastolic pressure and the slope of the end-diastolic pressure-volume relationship. Moreover, OM 1200 µg/kg frequently evoked transient electromechanical alternans in the rat in vivo in which normal systoles were followed by smaller contractions (and T-wave amplitudes) without major differences on the QRS complexes. Besides improving systolic function, OM evoked diastolic dysfunction and pulsus alternans. The narrow therapeutic window for OM may necessitate the monitoring of additional clinical safety parameters in clinical application.
Collapse
MESH Headings
- Action Potentials/drug effects
- Adult
- Animals
- Arrhythmias, Cardiac/chemically induced
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Blood Pressure/drug effects
- Calcium Signaling/drug effects
- Cardiotonic Agents/toxicity
- Diastole
- Dogs
- Dose-Response Relationship, Drug
- Female
- Heart Rate/drug effects
- Humans
- Hypotension/chemically induced
- Hypotension/metabolism
- Hypotension/physiopathology
- Kinetics
- Male
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Rats, Inbred WKY
- Systole
- Urea/analogs & derivatives
- Urea/toxicity
- Ventricular Dysfunction, Left/chemically induced
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left/drug effects
- Rats
Collapse
Grants
- GINOP-2.3.2-15-2016-00043 Ministry for National Economy of Hungary, co-financed by the European Union and the European Regional Development Fund
- ÚNKP-18-3-III-DE-209 Ministry of Human Capacities of Hungary, co-financed by the European Union and the European Regional Development Fund
- ED_18-1-2019-0028, TKP2020-IKA-04 and TKP2020-NKA-04 The Thematic Excellence Programme of the Ministry for Innovation and Technology, also supported from the National Research, Development and Innovation Fund of Hungary
- FK 128809 National Research, Development and Innovation Fund of Hungary
- FK 128116 National Research, Development and Innovation Fund of Hungary
- K 134939 National Research, Development and Innovation Fund of Hungary.
- K 116940 and K 132623 National Research, Development and Innovation Fund of Hungary.
- Therapeutic Development thematic programme of the Semmelweis University Higher Education Institutional Excellence Programme of the Ministry for Innovation and Technology in Hungary
- 2020-4.1.1.-TKP2020, Therapeutic Development and Bioimaging thematic programme of the Semmelweis University The Thematic Excellence Programme of the Ministry for Innovation and Technology was also supported from the National Research, Development and Innovation Fund of Hungary
- The Thematic Excellence Programme of the Ministry for Innovation and Technology, also supported from the National Research, Development and Innovation Fund of Hungary
- The Thematic Excellence Programme of the Ministry for Innovation and Technology was also supported from the National Research, Development and Innovation Fund of Hungary
- University of Debrecen
Collapse
Affiliation(s)
- Gábor Á Fülöp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Doctoral School of Kálmán Laki, University of Debrecen, Debrecen, Hungary
| | - Attila Oláh
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Tamas Csipo
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Doctoral School of Kálmán Laki, University of Debrecen, Debrecen, Hungary
| | - Árpád Kovács
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Róbert Pórszász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Roland Veress
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Nagy
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Beáta Bódi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
| | - Miklós Fagyas
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Solveig Lind Helgadottir
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
| | - Viktor Bánhegyi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- Doctoral School of Kálmán Laki, University of Debrecen, Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Nanasi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - István Édes
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Csanádi
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, 22 Móricz Zsigmond Street, 4032, Debrecen, Hungary.
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
17
|
Gao B, Sutherland W, Vargas HM, Qu Y. Effects of omecamtiv mecarbil on calcium-transients and contractility in a translational canine myocyte model. Pharmacol Res Perspect 2020; 8:e00656. [PMID: 32969560 PMCID: PMC7512116 DOI: 10.1002/prp2.656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 11/25/2022] Open
Abstract
Omecamtiv mecarbil (OM) is a selective cardiac myosin activator (myotrope), currently in Phase 3 clinical investigation as a novel treatment for heart failure with reduced ejection fraction. OM increases cardiac contractility by enhancing interaction between myosin and actin in a calcium-independent fashion. This study aims to characterize the mechanism of action by evaluating its simultaneous effect on myocyte contractility and calcium-transients (CTs) in healthy canine ventricular myocytes. Left ventricular myocytes were isolated from canines and loaded with Fura-2 AM. With an IonOptix system, contractility parameters including amplitude and duration of sarcomere shortening, contraction and relaxation velocity, and resting sarcomere length were measured. CT parameters including amplitude at systole and diastole, velocity at systole and diastole, and duration at 50% from peak were simultaneously measured. OM was tested at 0.03, 0.1, 0.3, 1, and 3 µmol\L concentrations to simulate therapeutic human plasma exposure levels. OM and isoproterenol (ISO) demonstrated differential effects on CTs and myocyte contractility. OM increased contractility mainly by prolonging duration of contraction while ISO increased contractility mainly by augmenting the amplitude of contraction. ISO increased the amplitude and velocity of CT, shortened duration of CT concurrent with increasing myocyte contraction, while OM did not change the amplitude, velocity, and duration of CT up to 1 µmol\L. Decreases in relaxation velocity and increases in duration were present only at 3 µmol\L. In this translational myocyte model study, therapeutically relevant concentrations of OM increased contractility but did not alter intracellular CTs, a mechanism of action distinct from traditional calcitropes.
Collapse
Affiliation(s)
- BaoXi Gao
- Translational Safety & Bioanalytical SciencesAmgen Inc.Thousand OaksCAUSA
| | - Weston Sutherland
- Translational Safety & Bioanalytical SciencesAmgen Inc.Thousand OaksCAUSA
| | - Hugo M. Vargas
- Translational Safety & Bioanalytical SciencesAmgen Inc.Thousand OaksCAUSA
| | - Yusheng Qu
- Translational Safety & Bioanalytical SciencesAmgen Inc.Thousand OaksCAUSA
| |
Collapse
|
18
|
Qu Y, Feric N, Pallotta I, Singh R, Sobbi R, Vargas HM. Inotropic assessment in engineered 3D cardiac tissues using human induced pluripotent stem cell-derived cardiomyocytes in the Biowire TM II platform. J Pharmacol Toxicol Methods 2020; 105:106886. [PMID: 32629159 DOI: 10.1016/j.vascn.2020.106886] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/22/2020] [Accepted: 06/08/2020] [Indexed: 11/28/2022]
Abstract
To develop therapeutics for cardiovascular disease, especially heart failure, translational models for assessing cardiac contractility are necessary for preclinical target validation and lead optimization. The availability of stem cell-derived cardiomyocytes (SC-CM) has generated a great opportunity in developing new in-vitro models for assessing cardiac contractility. However, the immature phenotype of SC-CM is a well-recognized limitation in inotropic evaluation, especially regarding the lack of or diminished positive inotropic response to β-adrenergic agonists. Recent development of 3D engineered cardiac tissues (ECTs) using human induced pluripotent stem cell derived-cardiomyocytes (hiPSC-CM) in the BiowireTM II platform has shown improved maturation. To evaluate their suitability to detect drug-induced changes in cardiac contractility, positive inotropes with diverse mechanisms, including β-adrenergic agonists, PDE3 inhibitors, Ca2+-sensitizers, myosin and troponin activators, and an apelin receptor agonist, were tested blindly. A total of 8 compounds were evaluated, including dobutamine, milrinone, pimobendan, levosimendan, omecamtiv mecarbil, AMG1, AMG2, and pyr-apelin-13. Contractility was evaluated by analyzing the amplitude, velocity and duration of contraction and relaxation. All tested agents, except pyr-apelin-13, increased contractility by increasing the amplitude of contraction and velocity. In addition, myosin and troponin activators increase contraction duration. These results indicate that ECTs generated in the BiowireTM II platform can identify inotropes with different mechanisms and provides a human-based in-vitro model for evaluating potential therapeutics.
Collapse
Affiliation(s)
- Yusheng Qu
- Amgen Research, Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA, USA.
| | | | | | | | | | - Hugo M Vargas
- Amgen Research, Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA, USA
| |
Collapse
|
19
|
Eisner DA, Caldwell JL, Trafford AW, Hutchings DC. The Control of Diastolic Calcium in the Heart: Basic Mechanisms and Functional Implications. Circ Res 2020; 126:395-412. [PMID: 31999537 PMCID: PMC7004450 DOI: 10.1161/circresaha.119.315891] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Normal cardiac function requires that intracellular Ca2+ concentration be reduced to low levels in diastole so that the ventricle can relax and refill with blood. Heart failure is often associated with impaired cardiac relaxation. Little, however, is known about how diastolic intracellular Ca2+ concentration is regulated. This article first discusses the reasons for this ignorance before reviewing the basic mechanisms that control diastolic intracellular Ca2+ concentration. It then considers how the control of systolic and diastolic intracellular Ca2+ concentration is intimately connected. Finally, it discusses the changes that occur in heart failure and how these may result in heart failure with preserved versus reduced ejection fraction.
Collapse
Affiliation(s)
- David A Eisner
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Jessica L Caldwell
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Andrew W Trafford
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - David C Hutchings
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| |
Collapse
|
20
|
Saleem U, Mannhardt I, Braren I, Denning C, Eschenhagen T, Hansen A. Force and Calcium Transients Analysis in Human Engineered Heart Tissues Reveals Positive Force-Frequency Relation at Physiological Frequency. Stem Cell Reports 2020; 14:312-324. [PMID: 31956082 PMCID: PMC7013237 DOI: 10.1016/j.stemcr.2019.12.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 01/29/2023] Open
Abstract
Force measurements in ex vivo and engineered heart tissues are well established. Analysis of calcium transients (CaT) is complementary to force, and the combined analysis is meaningful to the study of cardiomyocyte biology and disease. This article describes a model of human induced pluripotent stem cell cardiomyocyte-derived engineered heart tissues (hiPSC-CM EHTs) transduced with the calcium sensor GCaMP6f followed by sequential analysis of force and CaT. Average peak analysis demonstrated the temporal sequence of the CaT preceding the contraction twitch. The pharmacological relevance of the test system was demonstrated with inotropic indicator compounds. Force-frequency relationship was analyzed in the presence of ivabradine (300 nM), which reduced spontaneous frequency and unmasked a positive correlation of force and CaT at physiological human heart beating frequency with stimulation frequency between 0.75 and 2.5 Hz (force +96%; CaT +102%). This work demonstrates the usefulness of combined force/CaT analysis and demonstrates a positive force-frequency relationship in hiPSC-CM EHTs. Analysis of calcium transients and force in engineered heart tissues Accurate replications of drug effects on calcium transients and force analysis Positive force- and calcium transients-frequency relationship Reverse correlation between omecamtiv mecarbil's inotropic effect and frequency
Collapse
Affiliation(s)
- Umber Saleem
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Ingke Braren
- Vector Facility, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Chris Denning
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany.
| |
Collapse
|
21
|
van Meer BJ, Krotenberg A, Sala L, Davis RP, Eschenhagen T, Denning C, Tertoolen LGJ, Mummery CL. Simultaneous measurement of excitation-contraction coupling parameters identifies mechanisms underlying contractile responses of hiPSC-derived cardiomyocytes. Nat Commun 2019; 10:4325. [PMID: 31541103 PMCID: PMC6754438 DOI: 10.1038/s41467-019-12354-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 09/04/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiomyocytes from human induced pluripotent stem cells (hiPSC-CMs) are increasingly recognized as valuable for determining the effects of drugs on ion channels but they do not always accurately predict contractile responses of the human heart. This is in part attributable to their immaturity but the sensitivity of measurement tools may also be limiting. Measuring action potential, calcium flux or contraction individually misses critical information that is captured when interrogating the complete excitation-contraction coupling cascade simultaneously. Here, we develop an hypothesis-based statistical algorithm that identifies mechanisms of action. We design and build a high-speed optical system to measure action potential, cytosolic calcium and contraction simultaneously using fluorescent sensors. These measurements are automatically processed, quantified and then assessed by the algorithm. Multiplexing these three critical physical features of hiPSC-CMs allows identification of all major drug classes affecting contractility with detection sensitivities higher than individual measurement of action potential, cytosolic calcium or contraction.
Collapse
Affiliation(s)
- Berend J van Meer
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Ana Krotenberg
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Luca Sala
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands.,Istituto Auxologico Italiano, IRCCS, Center for Cardiac Arrhythmias of Genetic Origin, Laboratory of Cardiovascular Genetics, Via Zucchi 18, 20095, Cusano Milanino, MI, Italy
| | - Richard P Davis
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Thomas Eschenhagen
- Dept. of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Chris Denning
- Dept. of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Leon G J Tertoolen
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Christine L Mummery
- Dept. of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands. .,Dept. of Applied Stem Cell Technologies, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.
| |
Collapse
|
22
|
Clark JA, Weiss JD, Campbell SG. A Microwell Cell Capture Device Reveals Variable Response to Dobutamine in Isolated Cardiomyocytes. Biophys J 2019; 117:1258-1268. [PMID: 31537313 DOI: 10.1016/j.bpj.2019.08.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/14/2019] [Accepted: 08/22/2019] [Indexed: 12/15/2022] Open
Abstract
Isolated ventricular cardiomyocytes exhibit substantial cell-to-cell variability, even when obtained from the same small volume of myocardium. In this study, we investigated the possibility that cardiomyocyte responses to β-adrenergic stimulus are also highly heterogeneous. To achieve the throughput and measurement duration desired for these experiments, we designed and validated a novel microwell system that immobilizes and uniformly orients isolated adult cardiomyocytes. In this configuration, detailed drug responses of dozens of cells can be followed for intervals exceeding 1 h. At the conclusion of an experiment, specific cells can also be harvested via a precision aspirator for single-cell gene expression profiling. Using this system, we followed changes in Ca2+ signaling and contractility of individual cells under sustained application of either dobutamine or omecamtiv mecarbil. Both compounds increased average cardiomyocyte contractility over the course of an hour, but responses of individual cells to dobutamine were significantly more variable. Surprisingly, some dobutamine-treated cardiomyocytes augmented Ca2+ release without increasing contractility. Other cells responded with increased contractility despite unchanged Ca2+ release. Single-cell gene expression analysis revealed significant co-expression of β-adrenergic pathway genes PKA regulatory subunit type I, PKA regulatory subunit type II, and Ca2+/calmodulin-dependent protein kinase II across cardiomyocytes. Other data supported a connection between the effects of dobutamine on relaxation rate and the expression of protein phosphatase 2. These findings suggest that variable drug responses among cells are not merely experimental artifacts. By enabling direct comparison of the functional behavior of an individual cell and the genes it expresses, this new system constitutes a unique tool for interrogating cardiomyocyte drug responses and discovering the genes that modulate them.
Collapse
|
23
|
Obata K, Morita H, Takaki M. The energy-saving effect of a new myosin activator, omecamtiv mecarbil, on LV mechanoenergetics in rat hearts with blood-perfused isovolumic contraction model. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1065-1070. [PMID: 31267148 DOI: 10.1007/s00210-019-01685-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 06/26/2019] [Indexed: 01/10/2023]
Abstract
A novel myosin activator, omecamtiv mecarbil (OM), is a cardiac inotropic agent with a unique new mechanism of action, which is thought to arise from an increase in the transition rate of myosin into the actin-bound force-generating state without increasing calcium (Ca2+) transient. There remains, however, considerable controversy about the effects of OM on cardiac contractility and energy expenditure. In the present study, we investigated the effects of OM on left ventricular (LV) mechanical work and energetics, i.e., mechanoenergetics in rat normal hearts (CTL) and failing hearts induced by chronic administration of isoproterenol (1.2 mg/kg/day) for 4 weeks (ISO-HF). We analyzed the LV end-systolic pressure-volume relation (ESPVR) and the linear relation between the myocardial oxygen consumption per beat (VO2) and systolic pressure-volume area (PVA; a total mechanical energy per beat) in isovolumically contracting rat hearts at 240- or 300-bpm pacing in the absence or presence of OM. OM did not change the ESPVR in CTL and ISO-HF. OM, however, significantly decreased the slope of VO2-PVA relationship in both CTL and ISO-HF, and significantly increased the mean VO2 intercept without changes in basal metabolism in ISO-HF. These results suggested that OM improved the oxygen cost of PVA (contractile efficiency) with the unchanged LV contractility in both CTL and ISO-HF but increased VO2 for Ca2+ handling in excitation-contraction (E-C) coupling in ISO-HF. We concluded that OM improves contractile efficiency in normal and failing hearts but increases O2 consumption of Ca2+ handling in failing hearts in isovolumically contracting rat model.
Collapse
Affiliation(s)
- Koji Obata
- Department of Physiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Hironobu Morita
- Department of Physiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Miyako Takaki
- Department of Physiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
24
|
Kislitsina ON, Rich JD, Wilcox JE, Pham DT, Churyla A, Vorovich EB, Ghafourian K, Yancy CW. Shock - Classification and Pathophysiological Principles of Therapeutics. Curr Cardiol Rev 2019; 15:102-113. [PMID: 30543176 PMCID: PMC6520577 DOI: 10.2174/1573403x15666181212125024] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 10/11/2018] [Accepted: 12/10/2018] [Indexed: 01/10/2023] Open
Abstract
The management of patients with shock is extremely challenging because of the myriad of possible clinical presentations in cardiogenic shock, septic shock and hypovolemic shock and the limitations of contemporary therapeutic options. The treatment of shock includes the administration of endogenous catecholamines (epinephrine, norepinephrine, and dopamine) as well as various vasopressor agents that have shown efficacy in the treatment of the various types of shock. In addition to the endogenous catecholamines, dobutamine, isoproterenol, phenylephrine, and milrinone have served as the mainstays of shock therapy for several decades. Recently, experimental studies have suggested that newer agents such as vasopressin, selepressin, calcium-sensitizing agents like levosimendan, cardiac-specific myosin activators like omecamtiv mecarbil (OM), istaroxime, and natriuretic peptides like nesiritide can enhance shock therapy, especially when shock presents a more complex clinical picture than normal. However, their ability to improve clinical outcomes remains to be proven. It is the purpose of this review to describe the mechanism of action, dosage requirements, advantages and disadvantages, and specific indications and contraindications for the use of each of these catecholamines and vasopressors, as well as to elucidate the most important clinical trials that serve as the basis of contemporary shock therapy.
Collapse
Affiliation(s)
- Olga N Kislitsina
- Department of Cardiac Surgery Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States.,Department of Cardiology Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States
| | - Jonathan D Rich
- Department of Cardiology Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States
| | - Jane E Wilcox
- Department of Cardiology Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States
| | - Duc T Pham
- Department of Cardiac Surgery Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States
| | - Andrei Churyla
- Department of Cardiac Surgery Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States
| | - Esther B Vorovich
- Department of Cardiology Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States
| | - Kambiz Ghafourian
- Department of Cardiology Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States
| | - Clyde W Yancy
- Department of Cardiology Bluhm Cardiovascular Institute Feinberg School of Medicine Northwestern University Medical Center, Chicago, Illinois, IL, United States
| |
Collapse
|
25
|
Stroethoff M, Behmenburg F, Meierkord S, Bunte S, Mayer F, Mathes A, Heinen A, Hollmann MW, Huhn R. Cardioprotective Properties of Omecamtiv Mecarbil against Ischemia and Reperfusion Injury. J Clin Med 2019; 8:jcm8030375. [PMID: 30889854 PMCID: PMC6463149 DOI: 10.3390/jcm8030375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/09/2019] [Accepted: 03/13/2019] [Indexed: 01/14/2023] Open
Abstract
Omecamtiv mecarbil (OM) is a first-in-class myosin activator. It was developed as a new inotropic therapy option for heart failure and is currently the object of a phase 3 clinical trial program. OM activates ryanodine receptors, which were shown to be involved in cardioprotection induced by conditioning strategies. We hypothesize that OM exerts a concentration-dependent cardioprotective effect through pre- and postconditioning. Isolated male Wistar rat hearts underwent 33 min of global ischemia and 60 min of reperfusion. OM was administered in various concentrations (1, 3, 10, and 30 µM) over 10 min prior to ischemia. Based on these results, in subsequent experiments 3 and 10 µM OM were given over 10 min after ischemia. Infarct sizes were determined by TTC staining. In controls, the infarct size was 60% ± 10% and 59% ± 12%, respectively. Ten micromolar OM before ischemia reduced the infarct size to 33% ± 8%. The lower concentrations did not initiate cardioprotection, and the next highest concentration did not enhance the protective effect. Even if 10 μM OM was given in the early reperfusion phase, it significantly reduced the infarct size (31% ± 6%), whereas 3 μM OM did not trigger a protective effect (58% ± 15%). This study shows for the first time that OM induces cardioprotection by pre- and postconditioning with a binary phenomenon, which is either ineffective or has a maximal effect.
Collapse
Affiliation(s)
- Martin Stroethoff
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| | - Friederike Behmenburg
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| | - Simon Meierkord
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| | - Sebastian Bunte
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| | - Felix Mayer
- Department of Forensic Medicine, University Hospital Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| | - Alexander Mathes
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany.
| | - André Heinen
- Institute of Cardiovascular Physiology, Heinrich-Heine-University Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany.
| | - Markus W Hollmann
- Department of Anesthesiology, Amsterdam University Medical Center (AUMC), Location AMC, University of Amsterdam, Meiberdreef 9, 1100DD Amsterdam, The Netherlands.
| | - Ragnar Huhn
- Department of Anesthesiology, University Hospital Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| |
Collapse
|
26
|
Woody MS, Greenberg MJ, Barua B, Winkelmann DA, Goldman YE, Ostap EM. Positive cardiac inotrope omecamtiv mecarbil activates muscle despite suppressing the myosin working stroke. Nat Commun 2018; 9:3838. [PMID: 30242219 PMCID: PMC6155018 DOI: 10.1038/s41467-018-06193-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/21/2018] [Indexed: 02/05/2023] Open
Abstract
Omecamtiv mecarbil (OM) is a positive cardiac inotrope in phase-3 clinical trials for treatment of heart failure. Although initially described as a direct myosin activator, subsequent studies are at odds with this description and do not explain OM-mediated increases in cardiac performance. Here we show, via single-molecule, biophysical experiments on cardiac myosin, that OM suppresses myosin's working stroke and prolongs actomyosin attachment 5-fold, which explains inhibitory actions of the drug observed in vitro. OM also causes the actin-detachment rate to become independent of both applied load and ATP concentration. Surprisingly, increased myocardial force output in the presence of OM can be explained by cooperative thin-filament activation by OM-inhibited myosin molecules. Selective suppression of myosin is an unanticipated route to muscle activation that may guide future development of therapeutic drugs.
Collapse
Affiliation(s)
- Michael S Woody
- Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, 700A Clinical Research Building, Philadelphia, PA, 19104-6085, USA
| | - Michael J Greenberg
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, 700A Clinical Research Building, Philadelphia, PA, 19104-6085, USA.,Department of Biochemistry and Molecular Biophysics, Washington University in Saint Louis, St. Louis, 63110, MO, USA
| | - Bipasha Barua
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, 675 Hoes Lane, Piscataway, NJ, 08854, USA
| | - Donald A Winkelmann
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, 675 Hoes Lane, Piscataway, NJ, 08854, USA
| | - Yale E Goldman
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, 700A Clinical Research Building, Philadelphia, PA, 19104-6085, USA.
| | - E Michael Ostap
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, 700A Clinical Research Building, Philadelphia, PA, 19104-6085, USA.
| |
Collapse
|
27
|
Nánási P, Komáromi I, Almássy J. Perspectives of a myosin motor activator agent with increased selectivity. Can J Physiol Pharmacol 2018; 96:676-680. [PMID: 29792814 DOI: 10.1139/cjpp-2017-0741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Clinical treatment of heart failure is still not fully solved. A novel class of agents, the myosin motor activators, acts directly on cardiac myosin resulting in an increased force generation and prolongation of contraction. Omecamtiv mecarbil, the lead molecule of this group, is now in human phase 3 displaying promising clinical performance. However, omecamtiv mecarbil is not selective to myosin, because it readily binds to and activates cardiac ryanodine receptors (RyR-2), an effect that may cause complications in case of overdose. In this study, in silico analysis was performed to investigate the docking of omecamtiv mecarbil and other structural analogues to cardiac myosin heavy chain and RyR-2 to select the structure that has a higher selectivity to myosin over RyR-2. In silico docking studies revealed that omecamtiv mecarbil has comparable affinity to myosin and RyR-2: the respective Kd values are 0.60 and 0.87 μmol/L. Another compound, CK-1032100, has much lower affinity to RyR-2 than omecamtiv mecarbil, while it still has a moderate affinity to myosin. It was concluded that further research starting from the chemical structure of CK-1032100 may result a better myosin activator burdened probably less by the RyR-2 binding side effect. It also is possible, however, that the selectivity of omecamtiv mecarbil to myosin over RyR-2 cannot be substantially improved, because similar moieties seem to be responsible for the high affinity to both myosin and RyR-2.
Collapse
Affiliation(s)
- Péter Nánási
- a Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Komáromi
- b Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Almássy
- c Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|