1
|
Scott B, Greenberg L, Squarci C, Campbell KS, Greenberg MJ. Danicamtiv reduces myosin's working stroke but enhances contraction by activating the thin filament. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617269. [PMID: 39416013 PMCID: PMC11482770 DOI: 10.1101/2024.10.09.617269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Heart failure is a leading cause of death worldwide, and even with current treatments, the 5-year transplant-free survival rate is only ~50-70%. As such, there is a need to develop new treatments for patients that improve survival and quality of life. Recently, there have been efforts to develop small molecules for heart failure that directly target components of the sarcomere, including cardiac myosin. One such molecule, danicamtiv, recently entered phase II clinical trials; however, its mechanism of action and direct effects on myosin's mechanics and kinetics are not well understood. Using optical trapping techniques, stopped flow transient kinetics, and in vitro reconstitution assays, we found that danicamtiv reduces the size of cardiac myosin's working stroke, and in contrast to studies in muscle fibers, we found that it does not affect actomyosin detachment kinetics at the level of individual crossbridges. We demonstrate that danicamtiv accelerates actomyosin association kinetics, leading to increased recruitment of myosin crossbridges and subsequent thin filament activation at physiologically-relevant calcium concentrations. Finally, we computationally model how the observed changes in mechanics and kinetics at the level of single crossbridges contribute to increased cardiac contraction and improved diastolic function compared to the related myotrope, omecamtiv mecarbil. Taken together, our results have important implications for the design of new sarcomeric-targeting compounds for heart failure.
Collapse
Affiliation(s)
- Brent Scott
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Caterina Squarci
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Kenneth S. Campbell
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
2
|
Childers MC, Regnier M. Dynamics of the Pre-Powerstroke Myosin Lever Arm and the Effects of Omecamtiv Mecarbil. Int J Mol Sci 2024; 25:10425. [PMID: 39408754 PMCID: PMC11477208 DOI: 10.3390/ijms251910425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
The binding of small molecules to sarcomeric myosin can elicit powerful effects on the chemomechanical cycle, making them effective therapeutics in the clinic and research tools at the benchtop. However, these myotropes can have complex effects that act on different phases of the crossbridge cycle and which depend on structural, dynamic, and environmental variables. While small molecule binding sites have been identified crystallographically and their effects on contraction studied extensively, small molecule-induced dynamic changes that link structure-function are less studied. Here, we use molecular dynamics simulations to explore how omecamtiv mecarbil (OM), a cardiac myosin-specific myotrope, alters the coordinated dynamics of the lever arm and the motor domain in the pre-powerstroke state. We show that the lever arm adopts a range of orientations and find that different lever arm orientations are accompanied by changes in the hydrogen bonding patterns near the converter. We find that the binding of OM to myosin reduces the conformational heterogeneity of the lever arm orientation and also adjusts the average lever arm orientation. Finally, we map out the distinct conformations and ligand-protein interactions adopted by OM. These results uncover some structural factors that govern the motor domain-tail orientations and the mechanisms by which OM primes the pre-powerstroke myosin heads.
Collapse
Affiliation(s)
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| |
Collapse
|
3
|
Bodt SML, Ge J, Ma W, Rasicci DV, Desetty R, McCammon JA, Yengo CM. Dilated cardiomyopathy mutation in beta-cardiac myosin enhances actin activation of the power stroke and phosphate release. PNAS NEXUS 2024; 3:pgae279. [PMID: 39108304 PMCID: PMC11302452 DOI: 10.1093/pnasnexus/pgae279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/25/2024] [Indexed: 08/13/2024]
Abstract
Inherited mutations in human beta-cardiac myosin (M2β) can lead to severe forms of heart failure. The E525K mutation in M2β is associated with dilated cardiomyopathy (DCM) and was found to stabilize the interacting heads motif (IHM) and autoinhibited super-relaxed (SRX) state in dimeric heavy meromyosin. However, in monomeric M2β subfragment 1 (S1) we found that E525K enhances (threefold) the maximum steady-state actin-activated ATPase activity (k cat) and decreases (eightfold) the actin concentration at which ATPase is one-half maximal (K ATPase). We also found a twofold to fourfold increase in the actin-activated power stroke and phosphate release rate constants at 30 μM actin, which overall enhanced the duty ratio threefold. Loaded motility assays revealed that the enhanced intrinsic motor activity translates to increased ensemble force in M2β S1. Glutamate 525, located near the actin binding region in the so-called activation loop, is highly conserved and predicted to form a salt bridge with another conserved residue (lysine 484) in the relay helix. Enhanced sampling molecular dynamics simulations predict that the charge reversal mutation disrupts the E525-K484 salt bridge, inducing conformations with a more flexible relay helix and a wide phosphate release tunnel. Our results highlight a highly conserved allosteric pathway associated with actin activation of the power stroke and phosphate release and suggest an important feature of the autoinhibited IHM is to prevent this region of myosin from interacting with actin. The ability of the E525K mutation to stabilize the IHM likely overrides the enhanced intrinsic motor properties, which may be key to triggering DCM pathogenesis.
Collapse
Affiliation(s)
- Skylar M L Bodt
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - Jinghua Ge
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - Wen Ma
- Department of Physics, University of Vermont, 149 Beaumont Avenue, Burlington, VT 05405, USA
| | - David V Rasicci
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University School of Medicine, 64 Medical Center Dr, Morgantown, WV 26506, USA
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - J Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| |
Collapse
|
4
|
Wang Y, Lei C, Wang Q, Zhang X, Zhi L, Liu X. Design and synthesis of 7-azaindole derivatives as potent CDK8 inhibitors for the treatment of acute myeloid leukemia. RSC Med Chem 2024:d4md00465e. [PMID: 39157854 PMCID: PMC11325196 DOI: 10.1039/d4md00465e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
It is of great significance to design and synthesize novel structural inhibitors with good antitumor activity. In this study, based on rational design, a total of 42 7-azaindole derivatives as novel CDK8 inhibitors were designed and synthesized. All compounds were screened with antitumor activity and compound 6 (1-(3-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)phenyl)-3-(m-tolyl)urea) exhibited the best activity, especially in acute myeloid leukemia (GI50 MV4-11 = 1.97 ± 1.24 μM). This compound also exhibited excellent inhibitory activity against CDK8 (IC50 = 51.3 ± 4.6 nM). Further mechanism studies shown that it could inhibit STAT5 phosphorylation and induce cell cycle arrest in the G1 phase, leading to apoptosis in acute myeloid leukemia cells. In addition, acute toxicity at a dose of 1000 mg kg-1 indicated the low toxicity of this compound.
Collapse
Affiliation(s)
- Yumeng Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University Hefei P. R. China
| | - Cencen Lei
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University Hefei P. R. China
| | - Quan Wang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University Hefei P. R. China
| | - Xingxing Zhang
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University Hefei P. R. China
| | - Liping Zhi
- School of Health Management, Anhui Medical University Hefei 230032 PR China
| | - Xinhua Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University Hefei P. R. China
| |
Collapse
|
5
|
Li Y, Ma K, Dong Z, Gao S, Zhang J, Huang S, Yang J, Fang G, Li Y, Li X, Welch C, Griffin EL, Ramaswamy P, Valivullah Z, Liu X, Dong J, Wang DW, Du, Chung WK, Li Y. Frameshift variants in C10orf71 cause dilated cardiomyopathy in human, mouse, and organoid models. J Clin Invest 2024; 134:e177172. [PMID: 38950288 PMCID: PMC11178530 DOI: 10.1172/jci177172] [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: 11/01/2023] [Accepted: 04/25/2024] [Indexed: 07/03/2024] Open
Abstract
Research advances over the past 30 years have confirmed a critical role for genetics in the etiology of dilated cardiomyopathies (DCMs). However, full knowledge of the genetic architecture of DCM remains incomplete. We identified candidate DCM causal gene, C10orf71, in a large family with 8 patients with DCM by whole-exome sequencing. Four loss-of-function variants of C10orf71 were subsequently identified in an additional group of492 patients with sporadic DCM from 2 independent cohorts. C10orf71 was found to be an intrinsically disordered protein specifically expressed in cardiomyocytes. C10orf71-KO mice had abnormal heart morphogenesis during embryonic development and cardiac dysfunction as adults with altered expression and splicing of contractile cardiac genes. C10orf71-null cardiomyocytes exhibited impaired contractile function with unaffected sarcomere structure. Cardiomyocytes and heart organoids derived from human induced pluripotent stem cells with C10orf71 frameshift variants also had contractile defects with normal electrophysiological activity. A rescue study using a cardiac myosin activator, omecamtiv mecarbil, restored contractile function in C10orf71-KO mice. These data support C10orf71 as a causal gene for DCM by contributing to the contractile function of cardiomyocytes. Mutation-specific pathophysiology may suggest therapeutic targets and more individualized therapy.
Collapse
Affiliation(s)
- Yang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Ke Ma
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Zhujun Dong
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jing Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shan Huang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jie Yang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Guangming Fang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Yujie Li
- Novogene Co. Ltd., Beijing, China
| | - Xiaowei Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Carrie Welch
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Emily L. Griffin
- Department of Pediatrics, Columbia University, New York, New York, USA
| | | | | | | | - Jianzeng Dong
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Wendy K. Chung
- Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung & Blood Vessel Disease, Beijing, China
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| |
Collapse
|
6
|
Romero A, Ashcraft L, Chandra A, DiMassa V, Cremin P, Collibee SE, Chuang C, Hartman J, Hwee DT, St. Jean D, Malinowski J, DeBenedetto M, Moebius D, Payette J, Vargas R, Yeoman J, Motani A, Reagan J, Malik FI, Morgan BP. Discovery of Nelutroctiv (CK-136), a Selective Cardiac Troponin Activator for the Treatment of Cardiovascular Diseases Associated with Reduced Cardiac Contractility. J Med Chem 2024; 67:7825-7835. [PMID: 38729623 PMCID: PMC11129190 DOI: 10.1021/acs.jmedchem.3c02413] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/11/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024]
Abstract
Cardiac myosin activation has been shown to be a viable approach for the treatment of heart failure with reduced ejection fraction. Here, we report the discovery of nelutroctiv (CK-136), a selective cardiac troponin activator intended for patients with cardiovascular conditions where cardiac contractility is reduced. Discovery of nelutroctiv began with a high-throughput screen that identified compound 1R, a muscle selective cardiac sarcomere activator devoid of phosphodiesterase-3 activity. Optimization of druglike properties for 1R led to the replacement of the sulfonamide and aniline substituents which resulted in improved pharmacokinetic (PK) profiles and a reduced potential for human drug-drug interactions. In vivo echocardiography assessment of the optimized leads showed concentration dependent increases in fractional shortening and an improved pharmacodynamic window compared to myosin activator CK-138. Overall, nelutroctiv was found to possess the desired selectivity, a favorable pharmacodynamic window relative to myosin activators, and a preclinical PK profile to support clinical development.
Collapse
Affiliation(s)
- Antonio Romero
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Luke Ashcraft
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Aroop Chandra
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Vincent DiMassa
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Peadar Cremin
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Scott E. Collibee
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Chihyuan Chuang
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - James Hartman
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Darren T. Hwee
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - David St. Jean
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Justin Malinowski
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Mikkel DeBenedetto
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - David Moebius
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Joshua Payette
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Richard Vargas
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - John Yeoman
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Alykhan Motani
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Jeffrey Reagan
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Fady I. Malik
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Bradley P. Morgan
- Cytokinetics, Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
7
|
Collibee SE, Romero A, Muci AR, Hwee DT, Chuang C, Hartman JJ, Motani AS, Ashcraft L, DeRosier A, Grillo M, Lu Q, Malik FI, Morgan BP. Cardiac Troponin Activator CK-963 Increases Cardiac Contractility in Rats. J Med Chem 2024; 67:7859-7869. [PMID: 38451215 PMCID: PMC11129196 DOI: 10.1021/acs.jmedchem.3c02412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
Novel cardiac troponin activators were identified using a high throughput cardiac myofibril ATPase assay and confirmed using a series of biochemical and biophysical assays. HTS hit 2 increased rat cardiomyocyte fractional shortening without increasing intracellular calcium concentrations, and the biological target of 1 and 2 was determined to be the cardiac thin filament. Subsequent optimization to increase solubility and remove PDE-3 inhibition led to the discovery of CK-963 and enabled pharmacological evaluation of cardiac troponin activation without the competing effects of PDE-3 inhibition. Rat echocardiography studies using CK-963 demonstrated concentration-dependent increases in cardiac fractional shortening up to 95%. Isothermal calorimetry studies confirmed a direct interaction between CK-963 and a cardiac troponin chimera with a dissociation constant of 11.5 ± 3.2 μM. These results provide evidence that direct activation of cardiac troponin without the confounding effects of PDE-3 inhibition may provide benefit for patients with cardiovascular conditions where contractility is reduced.
Collapse
Affiliation(s)
- Scott E. Collibee
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Antonio Romero
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Alexander R. Muci
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Darren T. Hwee
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Chihyuan Chuang
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - James J. Hartman
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Alykhan S. Motani
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Luke Ashcraft
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Andre DeRosier
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Mark Grillo
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Qing Lu
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Fady I. Malik
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Bradley P. Morgan
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
8
|
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
|
9
|
Garg A, Lavine KJ, Greenberg MJ. Assessing Cardiac Contractility From Single Molecules to Whole Hearts. JACC Basic Transl Sci 2024; 9:414-439. [PMID: 38559627 PMCID: PMC10978360 DOI: 10.1016/j.jacbts.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 04/04/2024]
Abstract
Fundamentally, the heart needs to generate sufficient force and power output to dynamically meet the needs of the body. Cardiomyocytes contain specialized structures referred to as sarcomeres that power and regulate contraction. Disruption of sarcomeric function or regulation impairs contractility and leads to cardiomyopathies and heart failure. Basic, translational, and clinical studies have adapted numerous methods to assess cardiac contraction in a variety of pathophysiological contexts. These tools measure aspects of cardiac contraction at different scales ranging from single molecules to whole organisms. Moreover, these studies have revealed new pathogenic mechanisms of heart disease leading to the development of novel therapies targeting contractility. In this review, the authors explore the breadth of tools available for studying cardiac contractile function across scales, discuss their strengths and limitations, highlight new insights into cardiac physiology and pathophysiology, and describe how these insights can be harnessed for therapeutic candidate development and translational.
Collapse
Affiliation(s)
- Ankit Garg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kory J. Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Jani VP, Song T, Gao C, Gong H, Sadayappan S, Kass DA, Irving TC, Ma W. The structural OFF and ON states of myosin can be decoupled from the biochemical super- and disordered-relaxed states. PNAS NEXUS 2024; 3:pgae039. [PMID: 38328779 PMCID: PMC10849796 DOI: 10.1093/pnasnexus/pgae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
There is a growing awareness that both thick-filament and classical thin-filament regulations play central roles in modulating muscle contraction. Myosin ATPase assays have demonstrated that under relaxed conditions, myosin may reside either in a high-energy-consuming disordered-relaxed (DRX) state available for binding actin to generate force or in an energy-sparing super-relaxed (SRX) state unavailable for actin binding. X-ray diffraction studies have shown that the majority of myosin heads are in a quasi-helically ordered OFF state in a resting muscle and that this helical ordering is lost when myosin heads are turned ON for contraction. It has been assumed that myosin heads in SRX and DRX states are equivalent to the OFF and ON states, respectively, and the terms have been used interchangeably. In this study, we use X-ray diffraction and ATP turnover assays to track the structural and biochemical transitions of myosin heads, respectively, induced with either omecamtiv mecarbil (OM) or piperine in relaxed porcine myocardium. We find that while OM and piperine induce dramatic shifts of myosin heads from the OFF to the ON state, there are no appreciable changes in the population of myosin heads in the SRX and DRX states in both unloaded and loaded preparations. Our results show that biochemically defined SRX and DRX can be decoupled from structurally defined OFF and ON states. In summary, while SRX/DRX and OFF/ON transitions can be correlated in some cases, these two phenomena are measured using different approaches, reflect different properties of the thick filament, and should be investigated and interpreted separately.
Collapse
Affiliation(s)
- Vivek P Jani
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Chengqian Gao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - David A Kass
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thomas C Irving
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Weikang Ma
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
11
|
Parijat P, Attili S, Hoare Z, Shattock M, Kenyon V, Kampourakis T. Discovery of a novel cardiac-specific myosin modulator using artificial intelligence-based virtual screening. Nat Commun 2023; 14:7692. [PMID: 38001148 PMCID: PMC10673995 DOI: 10.1038/s41467-023-43538-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Direct modulation of cardiac myosin function has emerged as a therapeutic target for both heart disease and heart failure. However, the development of myosin-based therapeutics has been hampered by the lack of targeted in vitro screening assays. In this study we use Artificial Intelligence-based virtual high throughput screening (vHTS) to identify novel small molecule effectors of human β-cardiac myosin. We test the top scoring compounds from vHTS in biochemical counter-screens and identify a novel chemical scaffold called 'F10' as a cardiac-specific low-micromolar myosin inhibitor. Biochemical and biophysical characterization in both isolated proteins and muscle fibers show that F10 stabilizes both the biochemical (i.e. super-relaxed state) and structural (i.e. interacting heads motif) OFF state of cardiac myosin, and reduces force and left ventricular pressure development in isolated myofilaments and Langendorff-perfused hearts, respectively. F10 is a tunable scaffold for the further development of a novel class of myosin modulators.
Collapse
Affiliation(s)
- Priyanka Parijat
- Randall Centre for Cell and Molecular Biophysics; and British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, United Kingdom
| | - Seetharamaiah Attili
- Randall Centre for Cell and Molecular Biophysics; and British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, United Kingdom
| | - Zoe Hoare
- School of Cardiovascular and Metabolic Medicine and Sciences; Rayne Institute and British Heart Foundation Centre of Research Excellence, King's College London, London, SE5 9NU, United Kingdom
| | - Michael Shattock
- School of Cardiovascular and Metabolic Medicine and Sciences; Rayne Institute and British Heart Foundation Centre of Research Excellence, King's College London, London, SE5 9NU, United Kingdom
| | | | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics; and British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, United Kingdom.
| |
Collapse
|
12
|
Bodt SML, Ge J, Ma W, Rasicci DV, Desetty R, McCammon JA, Yengo CM. Dilated cardiomyopathy mutation in beta-cardiac myosin enhances actin activation of the power stroke and phosphate release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566646. [PMID: 38014187 PMCID: PMC10680644 DOI: 10.1101/2023.11.10.566646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Inherited mutations in human beta-cardiac myosin (M2β) can lead to severe forms of heart failure. The E525K mutation in M2β is associated with dilated cardiomyopathy (DCM) and was found to stabilize the interacting heads motif (IHM) and autoinhibited super-relaxed (SRX) state in dimeric heavy meromyosin. However, in monomeric M2β subfragment 1 (S1) we found that E525K enhances (3-fold) the maximum steady-state actin-activated ATPase activity (kcat) and decreases (6-fold) the actin concentration at which ATPase is one-half maximal (KATPase). We also found a 3 to 4-fold increase in the actin-activated power stroke and phosphate release rate constants at 30 μM actin, which overall enhanced the duty ratio 3-fold. Loaded motility assays revealed that the enhanced intrinsic motor activity translates to increased ensemble force in M2β S1. Glutamate 525, located near the actin binding region in the so-called activation loop, is highly conserved and predicted to form a salt-bridge with another conserved residue (lysine 484) in the relay helix. Enhanced sampling molecular dynamics simulations predict that the charge reversal mutation disrupts the E525-K484 salt-bridge, inducing conformations with a more flexible relay helix and a wide phosphate release tunnel. Our results highlight a highly conserved allosteric pathway associated with actin activation of the power stroke and phosphate release and suggest an important feature of the autoinhibited IHM is to prevent this region of myosin from interacting with actin. The ability of the E525K mutation to stabilize the IHM likely overrides the enhanced intrinsic motor properties, which may be key to triggering DCM pathogenesis.
Collapse
Affiliation(s)
- Skylar M. L. Bodt
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jinghua Ge
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Wen Ma
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California
| | - David V. Rasicci
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
13
|
Ma W, Jani VP, Song T, Gao C, Gong H, Sadayappan S, Kass DA, Irving TC. The structural OFF and ON states of myosin can be decoupled from the biochemical super-relaxed and disordered-relaxed states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562891. [PMID: 37904972 PMCID: PMC10614925 DOI: 10.1101/2023.10.18.562891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
There is a growing awareness that both thick filament and classical thin filament regulation play central roles in modulating muscle contraction. Myosin ATPase assays have demonstrated that under relaxed conditions, myosin may reside in either a high energy-consuming disordered-relaxed (DRX) state available for binding actin to generate force, or in an energy-sparing super-relaxed (SRX) state unavailable for actin binding. X-ray diffraction studies have shown the majority of myosin heads are in a quasi-helically ordered OFF state in a resting muscle and that this helical ordering is lost when myosin heads are turned ON for contraction. It has been assumed that myosin heads in SRX and DRX states are equivalent to the OFF and ON state respectively and the terms have been used interchangeably. Here, we use X-ray diffraction and ATP turnover assays to track the structural and biochemical transitions of myosin heads respectively induced with either omecamtiv mecarbil (OM) or piperine in relaxed porcine myocardium. We find that while OM and piperine induce dramatic shifts of myosin heads from the OFF to ON states, there are no appreciable changes in the population of myosin heads in the SRX and DRX states in both unloaded and loaded preparations. Our results show that biochemically defined SRX and DRX can be decoupled from structurally-defined OFF and ON states. In summary, while SRX/DRX and OFF/ON transitions can be correlated in some cases, these two phenomena are measured using different approaches, do not necessarily reflect the same properties of the thick filament and should be investigated and interpreted separately.
Collapse
Affiliation(s)
- Weikang Ma
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Vivek P. Jani
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, OH, USA
| | - Chengqian Gao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, OH, USA
| | - David A. Kass
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Thomas C. Irving
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
14
|
Claassen WJ, Baelde RJ, Galli RA, de Winter JM, Ottenheijm CAC. Small molecule drugs to improve sarcomere function in those with acquired and inherited myopathies. Am J Physiol Cell Physiol 2023; 325:C60-C68. [PMID: 37212548 PMCID: PMC10281779 DOI: 10.1152/ajpcell.00047.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Muscle weakness is a hallmark of inherited or acquired myopathies. It is a major cause of functional impairment and can advance to life-threatening respiratory insufficiency. During the past decade, several small-molecule drugs that improve the contractility of skeletal muscle fibers have been developed. In this review, we provide an overview of the available literature and the mechanisms of action of small-molecule drugs that modulate the contractility of sarcomeres, the smallest contractile units in striated muscle, by acting on myosin and troponin. We also discuss their use in the treatment of skeletal myopathies. The first of three classes of drugs discussed here increase contractility by decreasing the dissociation rate of calcium from troponin and thereby sensitizing the muscle to calcium. The second two classes of drugs directly act on myosin and stimulate or inhibit the kinetics of myosin-actin interactions, which may be useful in patients with muscle weakness or stiffness.NEW & NOTEWORTHY During the past decade, several small molecule drugs that improve the contractility of skeletal muscle fibers have been developed. In this review, we provide an overview of the available literature and the mechanisms of action of small molecule drugs that modulate the contractility of sarcomeres, the smallest contractile units in striated muscle, by acting on myosin and troponin.
Collapse
Affiliation(s)
- Wout J Claassen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Rianne J Baelde
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Ricardo A Galli
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Josine M de Winter
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Coen A C Ottenheijm
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| |
Collapse
|
15
|
Chakraborti A, Tardiff JC, Schwartz SD. Insights into the Mechanism of the Cardiac Drug Omecamtiv Mecarbil─A Computational Study. J Phys Chem B 2022; 126:10069-10082. [PMID: 36448224 PMCID: PMC9830884 DOI: 10.1021/acs.jpcb.2c06679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Omecamtiv mecarbil (OM) is a positive inotrope that is thought to bind directly to an allosteric site of the β-cardiac myosin. The drug is under investigation for the treatment of systolic heart failure. The drug is classified as a cardiac myosin modulator and has been observed to affect multiple vital steps of the cross-bridge cycle including the recovery stroke and the chemical step. We explored the free-energy surface of the recovery stroke of the human cardiac β-myosin in the presence of OM to determine its influence on this process. We also investigated the effects of OM on the recovery stroke in the presence of genetic cardiomyopathic mutations R712L, F764L, and P710R using metadynamics. We also utilized the method of transition path sampling to generate an unbiased ensemble of reactive trajectories for the ATP hydrolysis step in the presence of OM that were able to provide insight into the differences observed due to OM in the dynamics and mechanism of the decomposition of ATP to ADP and HPO42-, a central part of the power generation in cardiac muscle. We studied chemistry in the presence of the same three mutations to further elucidate the effect of OM, and its use in the treatment of cardiac disease.
Collapse
Affiliation(s)
- Ananya Chakraborti
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| | - Jil C. Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85724, United States
| | - Steven D. Schwartz
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
16
|
Langa P, Wolska BM, Solaro RJ. The Hippo Signaling Pathway as a Drug Target in Familial Dilated Cardiomyopathy. INTERNATIONAL JOURNAL OF DRUG DISCOVERY AND PHARMACOLOGY 2022; 1:4. [PMID: 38818406 PMCID: PMC11139043 DOI: 10.53941/ijddp.v1i1.189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
We focus here on the Hippo pathway in the hierarchical sensing and modulation of the mechanical state of the adult heart in health and disease. The Hippo pathway interrogates the micro-environment of cardiac myocytes providing surveillance of the mechanical state with engagement of signaling pathways critical to homeostasis of cardiac development, remodeling, and function and vulnerable to pathologies. Our discussion centers on Hippo signaling in the altered mechanical state instigated by variants of genes expressing mutant sarcomere proteins that trigger a progression to dilated cardiomyopathy (familial DCM). There is an unmet need for therapies in DCM. Recent progress in the discovery of small molecules that target Hippo signaling and are intended for use in cardiac disorders provides leads for modifying Hippo in DCM. As we emphasize, identifying useful targets in DCM requires in depth understanding of cell specific Hippo signaling in the cardiac micro-environment.
Collapse
Affiliation(s)
- Paulina Langa
- Department of Physiology and Biophysics and the Center for Cardiovascular Research,University of Illinois at Chicago, Chicago, IL,USA
| | - Beata M. Wolska
- Department of Physiology and Biophysics and the Center for Cardiovascular Research,University of Illinois at Chicago, Chicago, IL,USA
- Department of Medicine, Division of Cardiology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - R. John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research,University of Illinois at Chicago, Chicago, IL,USA
| |
Collapse
|
17
|
Singh RR, Slater RE, Wang J, Wang C, Guo Q, Motani AS, Hartman JJ, Sadayappan S, Ason BL. Distinct Mechanisms for Increased Cardiac Contraction Through Selective Alteration of Either Myosin or Troponin Activity. JACC Basic Transl Sci 2022; 7:1021-1037. [DOI: 10.1016/j.jacbts.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 10/14/2022]
|
18
|
Solaro RJ. Widely cited publications of Michael Bárány in 1964 and 1967 as tipping points in understanding myosin molecular motors. Arch Biochem Biophys 2022; 727:109319. [PMID: 35709967 DOI: 10.1016/j.abb.2022.109319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 11/02/2022]
Abstract
In 1964 Michael Bárány and colleagues published a paper ((M. Bárány, E. Gaetjens, K. Bárány, Karp E. Arch Biochem Biophys 106(1964)280-93. http://10.1016/0003-9861(64)90,189-4)) that has been one of the most cited papers in Archives of Biochemistry and Biophysics. This was followed in 1967 by another most cited paper (M. Bárány. J Gen Physiol 50(1967)197-218. https://doi.org/10.1085/jgp.50.6.197). I have commemorated these achievements as tipping points in the understanding of myosin motors in muscle function. Tipping points are generally defined as a temporal point in which a series of progressive advances (in this case the understanding of the relations between myosin ATP hydrolysis and muscle function) inspire more expansive, wide-ranging, significant changes. I first concisely summarize the background against which the papers came to publication as well as the unimaginable personal challenges faced by Michael and Kate Bárány. A final section summarizes the impact of these publications as key steps in the progression of contemporary understanding of diverse control of myosin ATPase activity with focus on the thick filaments in cardiac homeostasis, disorders, and as targets for therapeutic applications in translational investigations.
Collapse
Affiliation(s)
- R John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, 835 South Wolcott Avenue, Chicago, IL, USA.
| |
Collapse
|
19
|
Solaro RJ. Commentary on the 1964 Bárány Publication in Archives of Biochemistry and Biophysics as an advance in understanding myosin function. Arch Biochem Biophys 2022; 726:109300. [PMID: 35709968 DOI: 10.1016/j.abb.2022.109300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/13/2022] [Accepted: 05/20/2022] [Indexed: 11/15/2022]
Affiliation(s)
- R John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
20
|
Abstract
Variants in >12 genes encoding sarcomeric proteins can cause various cardiomyopathies. The two most common are hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Current therapeutics do not target the root causes of these diseases, but attempt to prevent disease progression and/or to manage symptoms. Accordingly, novel approaches are being developed to treat the cardiac muscle dysfunction directly. Challenges to developing therapeutics for these diseases include the diverse mechanisms of pathogenesis, some of which are still being debated and defined. Four small molecules that modulate the myosin motor protein in the cardiac sarcomere have shown great promise in the settings of HCM and DCM, regardless of the underlying genetic pathogenesis, and similar approaches are being developed to target other components of the sarcomere. In the setting of HCM, mavacamten and aficamten bind to the myosin motor and decrease the ATPase activity of myosin. In the setting of DCM, omecamtiv mecarbil and danicamtiv increase myosin activity in cardiac muscle (but omecamtiv mecarbil decreases myosin activity in vitro). In this Review, we discuss the therapeutic strategies to alter sarcomere contractile activity and summarize the data indicating that targeting one protein in the sarcomere can be effective in treating patients with genetic variants in other sarcomeric proteins, as well as in patients with non-sarcomere-based disease.
Collapse
Affiliation(s)
- Sarah J Lehman
- BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, USA
| | - Claudia Crocini
- BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, USA
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Leslie A Leinwand
- BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, USA.
- Molecular, Cellular, and Developmental Biology, University of Colorado at Boulder, Boulder, CO, USA.
| |
Collapse
|
21
|
Halas M, Langa P, Warren CM, Goldspink PH, Wolska BM, Solaro RJ. Effects of Sarcomere Activators and Inhibitors Targeting Myosin Cross-Bridges on Ca2+-Activation of Mature and Immature Mouse Cardiac Myofilaments. Mol Pharmacol 2022; 101:286-299. [PMID: 35236770 PMCID: PMC9092471 DOI: 10.1124/molpharm.121.000420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/16/2022] [Indexed: 11/22/2022] Open
Abstract
We tested the hypothesis that isoform shifts in sarcomeres of the immature heart modify the effect of cardiac myosin-directed sarcomere inhibitors and activators. Omecamtiv mecarbil (OM) activates tension and is in clinical trials for the treatment of adult acute and chronic heart failure. Mavacamten (Mava) inhibits tension and is in clinical trials to relieve hyper-contractility and outflow obstruction in advanced genetic hypertrophic cardiomyopathy (HCM) linked commonly to mutations in sarcomeric proteins. To address the effect of these agents in developing sarcomeres we isolated heart fiber bundles, extracted membranes with Triton X-100, and measured tension developed over a range of Ca2+ concentrations with and without OM or Mava treatment. We made measurements in fiber bundles from hearts of adult non-transgenic controls (NTG) expressing cardiac troponin I (cTnI), and from hearts of transgenic mice (TG-ssTnI) expressing the fetal/neonatal form, slow skeletal troponin I (ssTnI). We also compared fibers from 7+14-day-old NTG mice expressing ssTnI and cTnI. These studies were repeated with 7+14-day old transgenic mice (TG-cTnT-R92Q) expressing a mutant form of cardiac TnT (cTnT) linked to HCM. OM increased Ca2+-sensitivity and decreased cooperative activation in both ssTnI- and cTnI- regulated myofilaments with a similar effect reducing sub-maximal tension in immature and mature myofilaments. Although Mava decreased tension similarly in cTnI- and ssTnI-regulated myofilaments controlled either by cTnT or cTnT-R92Q, its effect involved a depressed Ca2+-sensitivity in the mature cTnT-R92-myofilaments. Our data demonstrate an influence of myosin and thin filament-associated proteins on the actions of myosin-directed agents such as OM and Mava. Significance Statement The effects of myosin-targeted activators and inhibitors on Ca2+-activated tension in developing cardiac sarcomeres presented here provide novel, ex-vivo evidence as to their actions in early-stage cardiac disorders. These studies advance understanding of the molecular mechanisms of these agents that is important in pre-clinical studies employing sarcomere Ca2+-response as a screening approach. The data also inform the use of commonly immature cardiac myocytes generated from human inducible pluripotent stem cells in screening for sarcomere activators and inhibitors.
Collapse
Affiliation(s)
- Monika Halas
- Physiology and Biophysics, University of Illinois at Chicago, United States
| | - Paulina Langa
- Physiology and Biophysics, University of Illinois at Chicago, United States
| | - Chad M Warren
- Physiology and Biophysics, University of Illinois at Chicago, United States
| | - Paul H Goldspink
- Physiology and Biophysics, University of Illinois at Chicago, United States
| | - Beata M Wolska
- Department of Medicine, University of Illinois at Chicago, United States
| | - R John Solaro
- Physiology and Biophysics, University of Illinois at Chicago, United States
| |
Collapse
|
22
|
Weisert M, Su JA, Menteer J, Shaddy RE, Kantor PF. Drug Treatment of Heart Failure in Children: Gaps and Opportunities. Paediatr Drugs 2022; 24:121-136. [PMID: 35084696 DOI: 10.1007/s40272-021-00485-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2021] [Indexed: 12/11/2022]
Abstract
Medical therapy for pediatric heart failure is based on a detailed mechanistic understanding of the underlying causes, which are diverse and unlike those encountered in most adult patients. Diuresis and improved perfusion are the immediate goals of care in the child with acute decompensated heart failure. Conversion to maintenance oral therapy for heart failure is based on the results of landmark studies in adults, as well as recent pediatric clinical trials and heart failure guidelines. There will continue to be an important role for newer drugs, some of which are in active trials in adults, and some of which are already approved for use in children. The need to plan for clinical trials in children during drug development for heart failure is emphasized.
Collapse
Affiliation(s)
- Molly Weisert
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Cardiology, Heart Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Jennifer A Su
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Cardiology, Heart Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Jondavid Menteer
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Cardiology, Heart Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Robert E Shaddy
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Cardiology, Heart Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Paul F Kantor
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Division of Cardiology, Heart Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
23
|
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
|
24
|
Barrick SK, Greenberg MJ. Cardiac myosin contraction and mechanotransduction in health and disease. J Biol Chem 2021; 297:101297. [PMID: 34634306 PMCID: PMC8559575 DOI: 10.1016/j.jbc.2021.101297] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
Cardiac myosin is the molecular motor that powers heart contraction by converting chemical energy from ATP hydrolysis into mechanical force. The power output of the heart is tightly regulated to meet the physiological needs of the body. Recent multiscale studies spanning from molecules to tissues have revealed complex regulatory mechanisms that fine-tune cardiac contraction, in which myosin not only generates power output but also plays an active role in its regulation. Thus, myosin is both shaped by and actively involved in shaping its mechanical environment. Moreover, these studies have shown that cardiac myosin-generated tension affects physiological processes beyond muscle contraction. Here, we review these novel regulatory mechanisms, as well as the roles that myosin-based force generation and mechanotransduction play in development and disease. We describe how key intra- and intermolecular interactions contribute to the regulation of myosin-based contractility and the role of mechanical forces in tuning myosin function. We also discuss the emergence of cardiac myosin as a drug target for diseases including heart failure, leading to the discovery of therapeutics that directly tune myosin contractility. Finally, we highlight some of the outstanding questions that must be addressed to better understand myosin's functions and regulation, and we discuss prospects for translating these discoveries into precision medicine therapeutics targeting contractility and mechanotransduction.
Collapse
Affiliation(s)
- Samantha K Barrick
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
25
|
Fucili A, Cimaglia P, Severi P, Giannini F, Boccadoro A, Micillo M, Rapezzi C, Tavazzi L, Ferrari R. Looking for a Tailored Therapy for Heart Failure: Are We Capable of Treating the Patient Instead of the Disease? J Clin Med 2021; 10:4325. [PMID: 34640341 PMCID: PMC8509308 DOI: 10.3390/jcm10194325] [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: 07/28/2021] [Revised: 08/23/2021] [Accepted: 09/16/2021] [Indexed: 12/11/2022] Open
Abstract
After almost a decade of stagnation in clinical research for HF treatment, five large randomized trials recently published have supported the use of four new classes of drugs, namely: angiotensin receptor/neprilysin inhibitor, sodium-glucose co-transporters 2 inhibitors, soluble guanylate cyclase modulators, and myosin activators. Each treatment has proved to be beneficial for both long-term outcomes and quality of life. Beside their clinical relevance, all these novel treatments have a different mechanism of action beyond the usual neuro-hormonal blockage. These different pathways, together with the unquestionable clinical evidence, advocate a re-thinking of HF treatment and of the appropriate drug to integrate with the existing standard therapy, according to different characteristics of HFrEF patients. This study aimed to offer a synthetic overview of the mechanisms of action of the new drugs and to propose a more personalized approach, considering patients' characteristics and safety profiles. To this end, we have identified seven profiles for patients with chronic heart failure with reduced ejection fraction and two for pre-discharge patients.
Collapse
Affiliation(s)
- Alessandro Fucili
- Cardiovascular Research Centre, Ferrara University, Via Aldo Moro 8, 44124 Ferrara, Italy; (A.F.); (P.S.); (A.B.); (M.M.); (C.R.)
| | - Paolo Cimaglia
- Cardiovascular Department, GVM Care and Research, Maria Cecilia Hospital, Via Corriera 1, 48033 Cotignola, Italy; (P.C.); (F.G.); (L.T.)
| | - Paolo Severi
- Cardiovascular Research Centre, Ferrara University, Via Aldo Moro 8, 44124 Ferrara, Italy; (A.F.); (P.S.); (A.B.); (M.M.); (C.R.)
| | - Francesco Giannini
- Cardiovascular Department, GVM Care and Research, Maria Cecilia Hospital, Via Corriera 1, 48033 Cotignola, Italy; (P.C.); (F.G.); (L.T.)
| | - Alberto Boccadoro
- Cardiovascular Research Centre, Ferrara University, Via Aldo Moro 8, 44124 Ferrara, Italy; (A.F.); (P.S.); (A.B.); (M.M.); (C.R.)
| | - Marco Micillo
- Cardiovascular Research Centre, Ferrara University, Via Aldo Moro 8, 44124 Ferrara, Italy; (A.F.); (P.S.); (A.B.); (M.M.); (C.R.)
| | - Claudio Rapezzi
- Cardiovascular Research Centre, Ferrara University, Via Aldo Moro 8, 44124 Ferrara, Italy; (A.F.); (P.S.); (A.B.); (M.M.); (C.R.)
- Cardiovascular Department, GVM Care and Research, Maria Cecilia Hospital, Via Corriera 1, 48033 Cotignola, Italy; (P.C.); (F.G.); (L.T.)
| | - Luigi Tavazzi
- Cardiovascular Department, GVM Care and Research, Maria Cecilia Hospital, Via Corriera 1, 48033 Cotignola, Italy; (P.C.); (F.G.); (L.T.)
| | - Roberto Ferrari
- Cardiovascular Research Centre, Ferrara University, Via Aldo Moro 8, 44124 Ferrara, Italy; (A.F.); (P.S.); (A.B.); (M.M.); (C.R.)
- Cardiovascular Department, GVM Care and Research, Maria Cecilia Hospital, Via Corriera 1, 48033 Cotignola, Italy; (P.C.); (F.G.); (L.T.)
- Via Ercole I° D’Este 32, 44124 Ferrara, Italy
| |
Collapse
|
26
|
Synthesis, characterization, and biological evaluations of substituted phenethylamine-based urea as anticancer and antioxidant agents. MONATSHEFTE FUR CHEMIE 2021. [DOI: 10.1007/s00706-021-02830-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
27
|
Sequeira V. When fat meets the engine: implications of dietary rumenic acid on myosin-targeting therapies in heart failure. J Physiol 2021; 599:3635-3636. [PMID: 34114231 DOI: 10.1113/jp281846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/09/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Vasco Sequeira
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, Würzburg, 97078, Germany
| |
Collapse
|
28
|
Greenberg MJ, Tardiff JC. Complexity in genetic cardiomyopathies and new approaches for mechanism-based precision medicine. J Gen Physiol 2021; 153:e202012662. [PMID: 33512404 PMCID: PMC7852459 DOI: 10.1085/jgp.202012662] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Genetic cardiomyopathies have been studied for decades, and it has become increasingly clear that these progressive diseases are more complex than originally thought. These complexities can be seen both in the molecular etiologies of these disorders and in the clinical phenotypes observed in patients. While these disorders can be caused by mutations in cardiac genes, including ones encoding sarcomeric proteins, the disease presentation varies depending on the patient mutation, where mutations even within the same gene can cause divergent phenotypes. Moreover, it is challenging to connect the mutation-induced molecular insult that drives the disease pathogenesis with the various compensatory and maladaptive pathways that are activated during the course of the subsequent progressive, pathogenic cardiac remodeling. These inherent complexities have frustrated our ability to understand and develop broadly effective treatments for these disorders. It has been proposed that it might be possible to improve patient outcomes by adopting a precision medicine approach. Here, we lay out a practical framework for such an approach, where patient subpopulations are binned based on common underlying biophysical mechanisms that drive the molecular disease pathogenesis, and we propose that this function-based approach will enable the development of targeted therapeutics that ameliorate these effects. We highlight several mutations to illustrate the need for mechanistic molecular experiments that span organizational and temporal scales, and we describe recent advances in the development of novel therapeutics based on functional targets. Finally, we describe many of the outstanding questions for the field and how fundamental mechanistic studies, informed by our more nuanced understanding of the clinical disorders, will play a central role in realizing the potential of precision medicine for genetic cardiomyopathies.
Collapse
Affiliation(s)
- Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
| | - Jil C. Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ
- Department of Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
29
|
Chaudhary AG, Alreefi FM, Aziz MA. Emerging Pharmacologic Therapies for Heart Failure With Reduced Ejection Fraction. CJC Open 2021; 3:646-657. [PMID: 34027369 PMCID: PMC8134937 DOI: 10.1016/j.cjco.2021.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/17/2021] [Indexed: 10/27/2022] Open
Abstract
The global burden of heart failure has reached epidemic proportions with tremendous health and economic consequences. Sodium glucose cotransporter 2 inhibitors, vericiguat, and omecamtiv mecarbil are novel agents that promise to blunt the high residual risk of heart failure with reduced ejection fraction. We review the vast knowledge base that has rapidly materialized for these agents and is poised to shape the current and future trends and recommendations in heart failure pharmacotherapy.
Collapse
Affiliation(s)
- Ammar G Chaudhary
- Cardiovascular Diseases Department, King Faisal Specialist Hospital and Research Centre (Gen. Org.), Jeddah, Saudi Arabia
| | - Fadi M Alreefi
- Cardiovascular Diseases Department, King Faisal Specialist Hospital and Research Centre (Gen. Org.), Jeddah, Saudi Arabia
| | - Mohammad A Aziz
- Cardiovascular Diseases Department, King Faisal Specialist Hospital and Research Centre (Gen. Org.), Jeddah, Saudi Arabia
| |
Collapse
|
30
|
Parijat P, Kondacs L, Alexandrovich A, Gautel M, Cobb AJA, Kampourakis T. High Throughput Screen Identifies Small Molecule Effectors That Modulate Thin Filament Activation in Cardiac Muscle. ACS Chem Biol 2021; 16:225-235. [PMID: 33315370 DOI: 10.1021/acschembio.0c00908] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Current therapeutic interventions for both heart disease and heart failure are largely insufficient and associated with undesired side effects. Biomedical research has emphasized the role of sarcomeric protein function for the normal performance and energy efficiency of the heart, suggesting that directly targeting the contractile myofilaments themselves using small molecule effectors has therapeutic potential and will likely result in greater drug efficacy and selectivity. In this study, we developed a robust and highly reproducible fluorescence polarization-based high throughput screening (HTS) assay that directly targets the calcium-dependent interaction between cardiac troponin C (cTnC) and the switch region of cardiac troponin I (cTnISP), with the aim of identifying small molecule effectors of the cardiac thin filament activation pathway. We screened a commercially available small molecule library and identified several hit compounds with both inhibitory and activating effects. We used a range of biophysical and biochemical methods to characterize hit compounds and identified fingolimod, a sphingosin-1-phosphate receptor modulator, as a new troponin-based small molecule effector. Fingolimod decreased the ATPase activity and calcium sensitivity of demembranated cardiac muscle fibers in a dose-dependent manner, suggesting that the compound acts as a calcium desensitizer. We investigated fingolimod's mechanism of action using a combination of computational studies, biophysical methods, and synthetic chemistry, showing that fingolimod bound to cTnC repels cTnISP via mainly electrostatic repulsion of its positively charged tail. These results suggest that fingolimod is a potential new lead compound/scaffold for the development of troponin-directed heart failure therapeutics.
Collapse
Affiliation(s)
- Priyanka Parijat
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Laszlo Kondacs
- Department of Chemistry, King’s College London, 7 Trinity Street, London, SE1 1DB, United Kingdom
| | - Alexander Alexandrovich
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Alexander J. A. Cobb
- Department of Chemistry, King’s College London, 7 Trinity Street, London, SE1 1DB, United Kingdom
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| |
Collapse
|
31
|
Patel PH, Nguyen M, Rodriguez R, Surani S, Udeani G. Omecamtiv Mecarbil: A Novel Mechanistic and Therapeutic Approach to Chronic Heart Failure Management. Cureus 2021; 13:e12419. [PMID: 33542867 PMCID: PMC7847774 DOI: 10.7759/cureus.12419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a major public health problem in the United States as well as worldwide. Chronic heart failure is a syndrome of reduced cardiac output resulting from impaired ventricular function, impaired filling, or a combination of both. Associated symptoms include dyspnea, fatigue, and decreased exercise tolerance. HF has a marked effect on morbidity and mortality, given limited therapeutic choices. The first line of therapeutic agents indicated in heart failure are beta-blockers. Other drugs and therapeutic modalities employed in HF treatment include angiotensin-receptor blockers (ARBs), sacubitril (neprilysin inhibitor) combination with the ARB, valsartan, small doses of aldosterone receptor antagonists (ARAs) in the setting of angiotensin-converting enzyme (ACE) inhibitors, and beta-blockers. Additionally, the sodium-glucose transporter-2 inhibitor, dapagliflozin in the setting of ACE inhibitors, ARBs, or sacubitril-valsartan plus beta-blocker have been employed. Other therapeutic modalities have included loop diuretics, digoxin, the hydralazine-isosorbide dinitrate combination, ivabradine, the inotropes, dobutamine, milrinone, and dopamine. Decreased cardiac contractility is central to the systolic HF. Therapeutic agents employed to increase cardiac contractility in HF are limited because of their mechanistic-related adverse effect profiles. Omecamtiv mecarbil (OM) is a first of its class cardiac myosin activator that increases the cardiac contractility by specifically binding to the catalytic S1 domain of cardiac myosin, to be employed in heart failure treatment. This agent has demonstrated benefit in reducing heart rate, peripheral vascular resistance, mean left arterial pressure, and left ventricular end-diastolic pressure in the animal models. Additionally, OM is known to improve systolic wall thickening, stroke volume (SV), and cardiac output (CO). OM increases systolic ejection time (SET), cardiac myocyte fractional shortening without significant increase of LV dP/dtmax, myocardial oxygen consumption, and myocyte intracellular calcium. The benefits of OM have been demonstrated through key trials, as (i) The Acute Treatment with Omecamtiv mecarbil to Increase Contractility in Acute Heart Failure (ATOMIC-AHF), and (ii) The Chronic Oral Study of Myosin Activation to Increase Contractility in Heart Failure (COSMIC-HF). The Global Approach to Lowering Adverse Cardiac Outcomes Through Improving Contractility in Heart Failure (GALACTIC-HF) trial is ongoing and can help provide further clinical data. OM provides a novel mechanism and therapeutic approach to managing patients with HF. Preclinical and clinical data suggest that OM capability can improve cardiac function, decrease ventricular wall stress, reverse ventricular remodeling, and promote sympathetic withdrawal.
Collapse
Affiliation(s)
- Pooja H Patel
- College of Pharmacy, Texas A&M University, Kingsville, USA
| | | | - Rubi Rodriguez
- College of Pharmacy, Texas A&M University, Kingsville, USA
| | - Salim Surani
- Internal Medicine, Corpus Christi Medical Center, Corpus Christi, USA.,Internal Medicine, University of North Texas, Dallas, USA
| | - George Udeani
- College of Pharmacy, Texas A&M University, Kingsville, USA
| |
Collapse
|
32
|
Loss KL, Shaddy RE, Kantor PF. Recent and Upcoming Drug Therapies for Pediatric Heart Failure. Front Pediatr 2021; 9:681224. [PMID: 34858897 PMCID: PMC8632454 DOI: 10.3389/fped.2021.681224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/01/2021] [Indexed: 12/23/2022] Open
Abstract
Pediatric heart failure (HF) is an important clinical condition with high morbidity, mortality, and costs. Due to the heterogeneity in clinical presentation and etiologies, the development of therapeutic strategies is more challenging in children than adults. Most guidelines recommending drug therapy for pediatric HF are extrapolated from studies in adults. Unfortunately, even using all available treatment, progression to cardiac transplantation is common. The development of prospective clinical trials in the pediatric population has significant obstacles, including small sample sizes, slow recruitment rates, challenging endpoints, and high costs. However, progress is being made as evidenced by the recent introduction of ivabradine and of sacubitril/valsartan. In the last 5 years, new drugs have also been developed for HF with reduced ejection fraction (HFrEF) in adults. The use of well-designed prospective clinical trials will be fundamental in the evaluation of safety and efficacy of these new drugs on the pediatric population. The aim of this article is to review the clinical presentation and management of acute and chronic pediatric heart failure, focusing on systolic dysfunction in patients with biventricular circulation and a systemic left ventricle. We discuss the drugs recently approved for children and those emerging, or in use for adults with HFrEF.
Collapse
Affiliation(s)
- Karla L Loss
- Division of Cardiology, Department of Pediatrics, Keck School of Medicine at University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Robert E Shaddy
- Division of Cardiology, Department of Pediatrics, Keck School of Medicine at University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Paul F Kantor
- Division of Cardiology, Department of Pediatrics, Keck School of Medicine at University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
33
|
Bernier TD, Buckley LF. Cardiac Myosin Activation for the Treatment of Systolic Heart Failure. J Cardiovasc Pharmacol 2021; 77:4-10. [PMID: 33165138 PMCID: PMC7779665 DOI: 10.1097/fjc.0000000000000929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/24/2020] [Indexed: 01/10/2023]
Abstract
ABSTRACT Left ventricular systolic dysfunction is the hallmark pathology in heart failure with reduced ejection fraction. Increasing left ventricular contractility with beta-adrenergic receptor agonists, phosphodiesterase-3 inhibitors, or levosimendan has failed to improve clinical outcomes and, in some situations, increased the risk of sudden cardiac death. Beta-adrenergic receptor agonists and phosphodiesterase-3 inhibitors retain an important role in advanced heart failure. Thus, there remains an unmet need for safe and effective therapies to improve left ventricular systolic function. Two novel cardiac myotropes, omecamtiv mecarbil and danicamtiv, target cardiac myosin to increase left ventricular systolic performance. Neither omecamtiv mecarbil nor danicamtiv affects cardiomyocyte calcium handling, the proposed mechanism underlying the life-threatening arrhythmias associated with cardiac calcitropes and calcium sensitizers. Phase 2 clinical trials have demonstrated that these cardiac myosin activators prolong left ventricular systolic ejection time and promote left ventricular and atrial reverse remodeling. At higher plasma concentrations, these agents may be associated with myocardial ischemia and impaired diastolic function. An ongoing phase 3 clinical trial will estimate the clinical efficacy and safety of omecamtiv mecarbil. An additional study of these agents, which have minimal hemodynamic and renal effects, is warranted in patients with advanced heart failure refractory to guideline-directed neurohormonal blockers.
Collapse
Affiliation(s)
- Thomas D Bernier
- Department of Pharmacy Services, Brigham and Women's Hospital, Boston, MA
| | | |
Collapse
|
34
|
Alsulami K, Marston S. Small Molecules acting on Myofilaments as Treatments for Heart and Skeletal Muscle Diseases. Int J Mol Sci 2020; 21:E9599. [PMID: 33339418 PMCID: PMC7767104 DOI: 10.3390/ijms21249599] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are the most prevalent forms of the chronic and progressive pathological condition known as cardiomyopathy. These diseases have different aetiologies; however, they share the feature of haemodynamic abnormalities, which is mainly due to dysfunction in the contractile proteins that make up the contractile unit known as the sarcomere. To date, pharmacological treatment options are not disease-specific and rather focus on managing the symptoms, without addressing the disease mechanism. Earliest attempts at improving cardiac contractility by modulating the sarcomere indirectly (inotropes) resulted in unwanted effects. In contrast, targeting the sarcomere directly, aided by high-throughput screening systems, could identify small molecules with a superior therapeutic value in cardiac muscle disorders. Herein, an extensive literature review of 21 small molecules directed to five different targets was conducted. A simple scoring system was created to assess the suitability of small molecules for therapy by evaluating them in eight different criteria. Most of the compounds failed due to lack of target specificity or poor physicochemical properties. Six compounds stood out, showing a potential therapeutic value in HCM, DCM or heart failure (HF). Omecamtiv Mecarbil and Danicamtiv (myosin activators), Mavacamten, CK-274 and MYK-581 (myosin inhibitors) and AMG 594 (Ca2+-sensitiser) are all small molecules that allosterically modulate troponin or myosin. Omecamtiv Mecarbil showed limited efficacy in phase III GALACTIC-HF trial, while, results from phase III EXPLORER-HCM trial were recently published, indicating that Mavacamten reduced left ventricular outflow tract (LVOT) obstruction and diastolic dysfunction and improved the health status of patients with HCM. A novel category of small molecules known as "recouplers" was reported to target a phenomenon termed uncoupling commonly found in familial cardiomyopathies but has not progressed beyond preclinical work. In conclusion, the contractile apparatus is a promising target for new drug development.
Collapse
Affiliation(s)
- Khulud Alsulami
- Imperial Centre for Translational and Experimental Medicine, Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology, Riyadh 11461, Saudi Arabia
| | - Steven Marston
- Imperial Centre for Translational and Experimental Medicine, Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
35
|
Boggu PR, Venkateswararao E, Manickam M, Sharma N, Kang JS, Jung SH. Identification of diphenylalkylisoxazol-5-amine scaffold as novel activator of cardiac myosin. Bioorg Med Chem 2020; 28:115742. [PMID: 33007555 DOI: 10.1016/j.bmc.2020.115742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/26/2020] [Accepted: 08/29/2020] [Indexed: 01/10/2023]
Abstract
To identify novel potent cardiac myosin activator, a series of diphenylalkylisoxazol-5-amine compounds 4-7 have been synthesized and evaluated for cardiac myosin ATPase activation. Among the 37 compounds, 4a (CMA at 10 µM = 81.6%), 4w (CMA at 10 µM = 71.2%) and 6b (CMA at 10 µM = 67.4%) showed potent cardiac myosin activation at a single concentration of 10 µM. These results suggested that the introduction of the amino-isoxazole ring as a bioisostere for urea group is acceptable for the cardiac myosin activation. Additional structure-activity relationship (SAR) studies were conducted. Para substitution (-Cl, -OCH3, -SO2N(CH3)2) to the phenyl rings or replacement of a phenyl ring with a heterocycle (pyridine, piperidine and tetrahydropyran) appeared to attenuate cardiac myosin activation at 10 µM. Additional hydrogen bonding acceptor next to the amino group of the isoxazoles did not enhance the activity. The potent isoxazole compounds showed selectivity for cardiac myosin activation over skeletal and smooth muscle myosin, and therefore these potent and selective isoxazole compounds could be considered as a new series of cardiac myosin ATPase activators for the treatment of systolic heart failure.
Collapse
Affiliation(s)
- Pulla Reddy Boggu
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Eeda Venkateswararao
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Manoj Manickam
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Niti Sharma
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jong Seong Kang
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Sang-Hun Jung
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
36
|
Parajón E, Surcel A, Robinson DN. The mechanobiome: a goldmine for cancer therapeutics. Am J Physiol Cell Physiol 2020; 320:C306-C323. [PMID: 33175572 DOI: 10.1152/ajpcell.00409.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer progression is dependent on heightened mechanical adaptation, both for the cells' ability to change shape and to interact with varying mechanical environments. This type of adaptation is dependent on mechanoresponsive proteins that sense and respond to mechanical stress, as well as their regulators. Mechanoresponsive proteins are part of the mechanobiome, which is the larger network that constitutes the cell's mechanical systems that are also highly integrated with many other cellular systems, such as gene expression, metabolism, and signaling. Despite the altered expression patterns of key mechanobiome proteins across many different cancer types, pharmaceutical targeting of these proteins has been overlooked. Here, we review the biochemistry of key mechanoresponsive proteins, specifically nonmuscle myosin II, α-actinins, and filamins, as well as the partnering proteins 14-3-3 and CLP36. We also examined a wide range of data sets to assess how gene and protein expression levels of these proteins are altered across many different cancer types. Finally, we determined the potential of targeting these proteins to mitigate invasion or metastasis and suggest that the mechanobiome is a goldmine of opportunity for anticancer drug discovery and development.
Collapse
Affiliation(s)
- Eleana Parajón
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexandra Surcel
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
37
|
Maack C, Eschenhagen T, Hamdani N, Heinzel FR, Lyon AR, Manstein DJ, Metzger J, Papp Z, Tocchetti CG, Yilmaz MB, Anker SD, Balligand JL, Bauersachs J, Brutsaert D, Carrier L, Chlopicki S, Cleland JG, de Boer RA, Dietl A, Fischmeister R, Harjola VP, Heymans S, Hilfiker-Kleiner D, Holzmeister J, de Keulenaer G, Limongelli G, Linke WA, Lund LH, Masip J, Metra M, Mueller C, Pieske B, Ponikowski P, Ristić A, Ruschitzka F, Seferović PM, Skouri H, Zimmermann WH, Mebazaa A. Treatments targeting inotropy. Eur Heart J 2020; 40:3626-3644. [PMID: 30295807 DOI: 10.1093/eurheartj/ehy600] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/06/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Acute heart failure (HF) and in particular, cardiogenic shock are associated with high morbidity and mortality. A therapeutic dilemma is that the use of positive inotropic agents, such as catecholamines or phosphodiesterase-inhibitors, is associated with increased mortality. Newer drugs, such as levosimendan or omecamtiv mecarbil, target sarcomeres to improve systolic function putatively without elevating intracellular Ca2+. Although meta-analyses of smaller trials suggested that levosimendan is associated with a better outcome than dobutamine, larger comparative trials failed to confirm this observation. For omecamtiv mecarbil, Phase II clinical trials suggest a favourable haemodynamic profile in patients with acute and chronic HF, and a Phase III morbidity/mortality trial in patients with chronic HF has recently begun. Here, we review the pathophysiological basis of systolic dysfunction in patients with HF and the mechanisms through which different inotropic agents improve cardiac function. Since adenosine triphosphate and reactive oxygen species production in mitochondria are intimately linked to the processes of excitation-contraction coupling, we also discuss the impact of inotropic agents on mitochondrial bioenergetics and redox regulation. Therefore, this position paper should help identify novel targets for treatments that could not only safely improve systolic and diastolic function acutely, but potentially also myocardial structure and function over a longer-term.
Collapse
Affiliation(s)
- Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Am Schwarzenberg 15, Würzburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Partner site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Frank R Heinzel
- Department of Cardiology, Charité University Medicine, Berlin, Germany
| | - Alexander R Lyon
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany.,Division for Structural Biochemistry, Hannover Medical School, Hannover, Germany
| | - Joseph Metzger
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - M Birhan Yilmaz
- Department of Cardiology, Cumhuriyet University, Sivas, Turkey
| | - Stefan D Anker
- Department of Cardiology and Pneumology, University Medical Center Göttingen and DZHK (German Center for Cardiovascular Research), Göttingen, Germany.,Division of Cardiology and Metabolism - Heart Failure, Cachexia and Sarcopenia, Department of Internal Medicine and Cardiology, Berlin-Brandenburg Center for Regenerative Therapies (BCRT) at Charité University Medicine, Berlin, Germany
| | - Jean-Luc Balligand
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Universite Catholique de Louvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover D-30625, Germany
| | | | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Partner site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Stefan Chlopicki
- Department of Pharmacology, Medical College, Jagiellonian University, Krakow, Poland
| | - John G Cleland
- University of Hull, Kingston upon Hull, UK.,National Heart and Lung Institute, Royal Brompton and Harefield Hospitals NHS Trust, Imperial College, London, UK
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexander Dietl
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Rodolphe Fischmeister
- Inserm UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | | | | | | | | | - Gilles de Keulenaer
- Laboratory of Physiopharmacology (University of Antwerp) and Department of Cardiology, ZNA Hospital, Antwerp, Belgium
| | - Giuseppe Limongelli
- Department of Cardiothoracic Sciences, Second University of Naples, Naples, Italy
| | | | - Lars H Lund
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josep Masip
- Intensive Care Department, Consorci Sanitari Integral, University of Barcelona, Spain
| | - Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Italy
| | - Christian Mueller
- Department of Cardiology and Cardiovascular Research Institute Basel (CRIB), University Hospital Basel, University of Basel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center Berlin, and German Centre for Cardiovascular Research (DZHK), Partner site Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Piotr Ponikowski
- Department of Cardiology, Medical University, Clinical Military Hospital, Wroclaw, Poland
| | - Arsen Ristić
- Department of Cardiology of the Clinical Center of Serbia and Belgrade University School of Medicine, Belgrade, Serbia
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Switzerland
| | | | - Hadi Skouri
- Division of Cardiology, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner siteGöttingen, Göttingen, Germany
| | - Alexandre Mebazaa
- Hôpital Lariboisière, Université Paris Diderot, Inserm U 942, Paris, France
| |
Collapse
|
38
|
Shchepkin DV, Nabiev SR, Nikitina LV, Kochurova AM, Berg VY, Bershitsky SY, Kopylova GV. Myosin from the ventricle is more sensitive to omecamtiv mecarbil than myosin from the atrium. Biochem Biophys Res Commun 2020; 528:658-663. [PMID: 32513536 DOI: 10.1016/j.bbrc.2020.05.108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/17/2020] [Indexed: 11/25/2022]
Abstract
Omecamtiv mecarbil (OM), an activator of cardiac myosin, strongly affects contractile characteristics of the ventricles and, to a much lesser extent, the characteristics of atrial contraction. We compared the molecular mechanism of action of OM on the interaction of atrial and ventricular myosin with actin using an optical trap and an in vitro motility assay. In concentrations up to 0.5 μM, OM did not affect the step size of a myosin molecule but reduced it at a higher OM level. OM substantially prolonged the interaction of both isoforms of myosin with actin. However, the interaction characteristics of ventricular myosin with actin were more sensitive to OM than those of atrial myosin. Our results, obtained at the level of isolated proteins, can explain why the impact of OM in therapeutic concentrations on the contractile function of the atrium is less significant as compared to those of the ventricle.
Collapse
Affiliation(s)
- Daniil V Shchepkin
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Salavat R Nabiev
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Larisa V Nikitina
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Anastasia M Kochurova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Valentina Y Berg
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Sergey Y Bershitsky
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Galina V Kopylova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia.
| |
Collapse
|
39
|
Governali S, Caremani M, Gallart C, Pertici I, Stienen G, Piazzesi G, Ottenheijm C, Lombardi V, Linari M. Orthophosphate increases the efficiency of slow muscle-myosin isoform in the presence of omecamtiv mecarbil. Nat Commun 2020; 11:3405. [PMID: 32636378 PMCID: PMC7341760 DOI: 10.1038/s41467-020-17143-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 05/26/2020] [Indexed: 02/08/2023] Open
Abstract
Omecamtiv mecarbil (OM) is a putative positive inotropic tool for treatment of systolic heart dysfunction, based on the finding that in vivo it increases the ejection fraction and in vitro it prolongs the actin-bond life time of the cardiac and slow-skeletal muscle isoforms of myosin. OM action in situ, however, is still poorly understood as the enhanced Ca2+-sensitivity of the myofilaments is at odds with the reduction of force and rate of force development observed at saturating Ca2+. Here we show, by combining fast sarcomere-level mechanics and ATPase measurements in single slow demembranated fibres from rabbit soleus, that the depressant effect of OM on the force per attached motor is reversed, without effect on the ATPase rate, by physiological concentrations of inorganic phosphate (Pi) (1-10 mM). This mechanism could underpin an energetically efficient reduction of systolic tension cost in OM-treated patients, whenever [Pi] increases with heart-beat frequency. Omecamtiv mecarbil is a small molecule effector under clinical trial for the treatment of systolic heart failure. Here the authors define the molecular mechanisms of its inotropic action and find it can increase the efficiency of contraction in muscle fibres when the orthophosphate concentration rises with the beat frequency.
Collapse
Affiliation(s)
- Serena Governali
- PhysioLab, Department of Biology, University of Florence, Sesto Fiorentino, 50019, Italy.,Department of Physiology, Amsterdam UMC (location VUmc), 1081 HZ, Amsterdam, The Netherlands
| | - Marco Caremani
- PhysioLab, Department of Biology, University of Florence, Sesto Fiorentino, 50019, Italy
| | - Cristina Gallart
- PhysioLab, Department of Biology, University of Florence, Sesto Fiorentino, 50019, Italy
| | - Irene Pertici
- PhysioLab, Department of Biology, University of Florence, Sesto Fiorentino, 50019, Italy
| | - Ger Stienen
- Department of Physiology, Amsterdam UMC (location VUmc), 1081 HZ, Amsterdam, The Netherlands.,Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Gabriella Piazzesi
- PhysioLab, Department of Biology, University of Florence, Sesto Fiorentino, 50019, Italy
| | - Coen Ottenheijm
- Department of Physiology, Amsterdam UMC (location VUmc), 1081 HZ, Amsterdam, The Netherlands
| | - Vincenzo Lombardi
- PhysioLab, Department of Biology, University of Florence, Sesto Fiorentino, 50019, Italy.
| | - Marco Linari
- PhysioLab, Department of Biology, University of Florence, Sesto Fiorentino, 50019, Italy
| |
Collapse
|
40
|
Simplified Procedure for General Synthesis of Monosubstituted Piperazines-From a Batch Reaction Vessel to a Flow (Microwave) Reactor. Molecules 2020; 25:molecules25092168. [PMID: 32384633 PMCID: PMC7249161 DOI: 10.3390/molecules25092168] [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: 04/01/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 11/17/2022] Open
Abstract
We reported a novel simplified synthetic procedure for the preparation of monosubstituted piperazine derivatives which can now be easily prepared in a one-pot-one-step way from a protonated piperazine with no need of introduction of a protecting group. Reactions, proceeding either at room or higher temperatures in common solvents, involve heterogeneous catalysis by metal ions supported on commercial polymeric resins. A general synthetic scheme was successfully applied to afford a wide range of monosubstituted piperazines. Furthermore, we picked up a set of piperazine derivatives and studied the possibilities of microwave acceleration of given synthetic reactions to make them even more efficient. Our research proceeded from a simple batch technique to the construction of a flow microwave reactor prototype and resulted in promising findings which are summarized and discussed in the article.
Collapse
|
41
|
Holmes JB, Doh CY, Mamidi R, Li J, Stelzer JE. Strategies for targeting the cardiac sarcomere: avenues for novel drug discovery. Expert Opin Drug Discov 2020; 15:457-469. [PMID: 32067508 PMCID: PMC7065952 DOI: 10.1080/17460441.2020.1722637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/24/2020] [Indexed: 01/10/2023]
Abstract
Introduction: Heart failure remains one of the largest clinical challenges in the United States. Researchers have continually searched for more effective heart failure treatments that target the cardiac sarcomere but have found few successes despite numerous expensive cardiovascular clinical trials. Among many reasons, the high failure rate of cardiovascular clinical trials may be partly due to incomplete characterization of a drug candidate's complex interaction with cardiac physiology.Areas covered: In this review, the authors address the issue of preclinical cardiovascular studies of sarcomere-targeting heart failure therapies. The authors consider inherent tradeoffs made between mechanistic transparency and physiological fidelity for several relevant preclinical techniques at the atomic, molecular, heart muscle fiber, whole heart, and whole-organism levels. Thus, the authors suggest a comprehensive, bottom-up approach to preclinical cardiovascular studies that fosters scientific rigor and hypothesis-driven drug discovery.Expert opinion: In the authors' opinion, the implementation of hypothesis-driven drug discovery practices, such as the bottom-up approach to preclinical cardiovascular studies, will be imperative for the successful development of novel heart failure treatments. However, additional changes to clinical definitions of heart failure and current drug discovery culture must accompany the bottom-up approach to maximize the effectiveness of hypothesis-driven drug discovery.
Collapse
Affiliation(s)
- Joshua B Holmes
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Chang Yoon Doh
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
42
|
Teerlink JR, Diaz R, Felker GM, McMurray JJV, Metra M, Solomon SD, Legg JC, Büchele G, Varin C, Kurtz CE, Malik FI, Honarpour N. Omecamtiv Mecarbil in Chronic Heart Failure With Reduced Ejection Fraction: Rationale and Design of GALACTIC-HF. JACC-HEART FAILURE 2020; 8:329-340. [PMID: 32035892 DOI: 10.1016/j.jchf.2019.12.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 01/10/2023]
Abstract
A central factor in the pathogenesis of heart failure (HF) with reduced ejection fraction is the initial decrease in systolic function. Prior attempts at increasing cardiac contractility with oral drugs have uniformly resulted in signals of increased mortality at pharmacologically effective doses. Omecamtiv mecarbil is a novel, selective cardiac myosin activator that has been shown to improve cardiac function and to decrease ventricular volumes, heart rate, and N-terminal pro-B-type natriuretic peptide in patients with chronic HF. The GALACTIC-HF (Global Approach to Lowering Adverse Cardiac outcomes Through Improving Contractility in Heart Failure) trial tests the hypotheses that omecamtiv mecarbil can safely improve symptoms, prevent clinical HF events, and delay CV death in patients with chronic HF. The GALACTIC-HF trial is an international, multicenter, randomized, double-blind, placebo-controlled, event-driven cardiovascular outcomes trial. More than 8,000 patients with chronic symptomatic (New York Heart Association functional class II to IV) HF, left ventricular ejection fraction ≤35%, elevated natriuretic peptides, and either current hospitalization for HF or history of hospitalization or emergency department visit for HF within a year of screening will be randomized to either oral placebo or omecamtiv mecarbil employing a pharmacokinetic-guided dose titration strategy using doses of 25, 37.5, or 50 mg twice daily. The primary efficacy outcome is the time to cardiovascular death or first HF event. The study has 90% power to assess a final hazard ratio of approximately 0.80 in cardiovascular death, the first secondary outcome. The GALACTIC-HF trial is the first trial examining whether selectively increasing cardiac contractility in patients with HF with reduced ejection fraction will result in improved clinical outcomes. (Registrational Study With Omecamtiv Mecarbil/AMG 423 to Treat Chronic Heart Failure With Reduced Ejection Fraction [GALACTIC-HF]; NCT02929329).
Collapse
Affiliation(s)
- John R Teerlink
- Section of Cardiology, San Francisco Veterans Affairs Medical Center, School of Medicine, University of California-San Francisco, San Francisco, California.
| | - Rafael Diaz
- Estudios Clinicos Latinoamérica, Rosario, Argentina
| | - G Michael Felker
- Division of Cardiology, Duke University School of Medicine, Duke Clinical Research Institute, Durham, North Carolina
| | - John J V McMurray
- British Heart Foundation Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Scott D Solomon
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | - Fady I Malik
- Cytokinetics, Inc., South San Francisco, California
| | | |
Collapse
|
43
|
Small Molecule Effectors of Myosin Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1239:61-84. [DOI: 10.1007/978-3-030-38062-5_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Manickam M, Pillaiyar T, Namasivayam V, Boggu PR, Sharma N, Jalani HB, Venkateswararao E, Lee YJ, Jeon ES, Son MJ, Woo SH, Jung SH. Design and synthesis of sulfonamidophenylethylamides as novel cardiac myosin activator. Bioorg Med Chem 2019; 27:4110-4123. [PMID: 31378598 DOI: 10.1016/j.bmc.2019.07.041] [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: 07/01/2019] [Revised: 07/23/2019] [Accepted: 07/25/2019] [Indexed: 01/10/2023]
Abstract
The sulfonamidophenylethylamide analogues were explored for finding novel and potent cardiac myosin activators. Among them, N-(4-(N,N-dimethylsulfamoyl)phenethyl-N-methyl-5-phenylpentanamide (13, CMA at 10 µM = 48.5%; FS = 26.21%; EF = 15.28%) and its isomer, 4-(4-(N,N-dimethylsulfamoyl)phenyl-N-methyl-N-(3-phenylpropyl)butanamide (27, CMA at 10 µM = 55.0%; FS = 24.69%; EF = 14.08%) proved to be efficient cardiac myosin activators both in in vitro and in vivo studies. Compounds 13 (88.2 + 3.1% at 5 µM) and 27 (46.5 + 2.8% at 5 µM) showed positive inotropic effect in isolated rat ventricular myocytes. The potent compounds 13 and 27 were highly selective for cardiac myosin over skeletal and smooth muscle myosin, and therefore these potent and selective amide derivatives could be considered a new class of cardiac myosin activators for the treatment of systolic heart failure.
Collapse
Affiliation(s)
- Manoj Manickam
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Thanigaimalai Pillaiyar
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | | | - Pulla Reddy Boggu
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Niti Sharma
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hitesh B Jalani
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eeda Venkateswararao
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - You-Jung Lee
- Division of Cardiology, Samsung Medical Center, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Eun-Seok Jeon
- Division of Cardiology, Samsung Medical Center, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Republic of Korea
| | - Min-Jeong Son
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sun-Hee Woo
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sang-Hun Jung
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
45
|
Radnai L, Stremel RF, Sellers JR, Rumbaugh G, Miller CA. A Semi-High-Throughput Adaptation of the NADH-Coupled ATPase Assay for Screening Small Molecule Inhibitors. J Vis Exp 2019. [PMID: 31475972 DOI: 10.3791/60017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
ATPase enzymes utilize the free energy stored in adenosine triphosphate to catalyze a wide variety of endergonic biochemical processes in vivo that would not occur spontaneously. These proteins are crucial for essentially all aspects of cellular life, including metabolism, cell division, responses to environmental changes and movement. The protocol presented here describes a nicotinamide adenine dinucleotide (NADH)-coupled ATPase assay that has been adapted to semi-high throughput screening of small molecule ATPase inhibitors. The assay has been applied to cardiac and skeletal muscle myosin II's, two actin-based molecular motor ATPases, as a proof of principle. The hydrolysis of ATP is coupled to the oxidation of NADH by enzymatic reactions in the assay. First, the ADP generated by the ATPase is regenerated to ATP by pyruvate kinase (PK). PK catalyzes the transition of phosphoenolpyruvate (PEP) to pyruvate in parallel. Subsequently, pyruvate is reduced to lactate by lactate dehydrogenase (LDH), which catalyzes the oxidation of NADH in parallel. Thus, the decrease in ATP concentration is directly correlated to the decrease in NADH concentration, which is followed by change to the intrinsic fluorescence of NADH. As long as PEP is available in the reaction system, the ADP concentration remains very low, avoiding inhibition of the ATPase enzyme by its own product. Moreover, the ATP concentration remains nearly constant, yielding linear time courses. The fluorescence is monitored continuously, which allows for easy estimation of the quality of data and helps to filter out potential artifacts (e.g., arising from compound precipitation or thermal changes).
Collapse
Affiliation(s)
- Laszlo Radnai
- Department of Molecular Medicine, The Scripps Research Institute; Department of Neuroscience, The Scripps Research Institute
| | - Rebecca F Stremel
- Department of Molecular Medicine, The Scripps Research Institute; Department of Neuroscience, The Scripps Research Institute
| | - James R Sellers
- Laboratory of Molecular Physiology, NHLBI, National Institutes of Health
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute
| | - Courtney A Miller
- Department of Molecular Medicine, The Scripps Research Institute; Department of Neuroscience, The Scripps Research Institute;
| |
Collapse
|
46
|
Affiliation(s)
- Sarah Chuzi
- Division of Cardiology, Department of Medicine, Northwestern University, Chicago, Illinois
| | - Larry A. Allen
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora
| | - Shannon M. Dunlay
- Division of Cardiology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Haider J. Warraich
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
47
|
Caille S, Allgeier AM, Bernard C, Correll TL, Cosbie A, Crockett RD, Cui S, Faul MM, Hansen KB, Huggins S, Langille N, Mennen SM, Morgan BP, Morrison H, Muci A, Nagapudi K, Quasdorf K, Ranganathan K, Roosen P, Shi X, Thiel OR, Wang F, Tvetan JT, Woo JCS, Wu S, Walker SD. Development of a Factory Process for Omecamtiv Mecarbil, a Novel Cardiac Myosin Activator. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.9b00200] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Seb Caille
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Alan M. Allgeier
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Charles Bernard
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Tiffany L. Correll
- Attribute Sciences, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Andrew Cosbie
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Richard D. Crockett
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Sheng Cui
- Drug Substance Technologies, Process Development, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Margaret M. Faul
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Karl B. Hansen
- Drug Substance Technologies, Process Development, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Seth Huggins
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Neil Langille
- Drug Substance Technologies, Process Development, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Steven M. Mennen
- Drug Substance Technologies, Process Development, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Bradley P. Morgan
- Research & Nonclinical Development, Cytokinetics, 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Henry Morrison
- Drug Product Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Alexander Muci
- Research & Nonclinical Development, Cytokinetics, 280 East Grand Avenue, South San Francisco, California 94080, United States
| | - Karthik Nagapudi
- Drug Product Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Kyle Quasdorf
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Krishnakumar Ranganathan
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Philipp Roosen
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Xianqing Shi
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Oliver R. Thiel
- Drug Substance Technologies, Process Development, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Fang Wang
- Attribute Sciences, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Justin T. Tvetan
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Jacqueline C. S. Woo
- Drug Substance Technologies, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Steven Wu
- Attribute Sciences, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Shawn D. Walker
- Drug Substance Technologies, Process Development, Amgen Inc., 360 Binney Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
48
|
Abstract
Advances in the treatment of heart failure with reduced ejection fraction due to systolic dysfunction are engaging an ever-expanding compendium of molecular signaling targets. Well established approaches modifying hemodynamics and cell biology by neurohumoral receptor blockade are evolving, exploring the role and impact of modulating intracellular signaling pathways with more direct myocardial effects. Even well-tread avenues are being reconsidered with new insights into the signaling engaged and thus opportunity to treat underlying myocardial disease. This review explores therapies that have proven successful, those that have not, those that are moving into the clinic but whose utility remains to be confirmed, and those that remain in the experimental realm. The emphasis is on signaling pathways that are tractable for therapeutic manipulation. Of the approaches yet to be tested in humans, we chose those with a well-established experimental history, where clinical translation may be around the corner. The breadth of opportunities bodes well for the next generation of heart failure therapeutics.
Collapse
Affiliation(s)
| | | | - David A. Kass
- Division of Cardiology, Department of Medicine
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore Maryland, 21205
| |
Collapse
|
49
|
Yotti R, Seidman CE, Seidman JG. Advances in the Genetic Basis and Pathogenesis of Sarcomere Cardiomyopathies. Annu Rev Genomics Hum Genet 2019; 20:129-153. [PMID: 30978303 DOI: 10.1146/annurev-genom-083118-015306] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are common heart muscle disorders that are caused by pathogenic variants in sarcomere protein genes. HCM is characterized by unexplained cardiac hypertrophy (increased chamber wall thickness) that is accompanied by enhanced cardiac contractility and impaired relaxation. DCM is defined as increased ventricular chamber volume with contractile impairment. In this review, we discuss recent analyses that provide new insights into the molecular mechanisms that cause these conditions. HCM studies have uncovered the critical importance of conformational changes that occur during relaxation and enable energy conservation, which are frequently disturbed by HCM mutations. DCM studies have demonstrated the considerable prevalence of truncating variants in titin and have discerned that these variants reduce contractile function by impairing sarcomerogenesis. These new pathophysiologic mechanisms open exciting opportunities to identify new pharmacological targets and develop future cardioprotective strategies.
Collapse
Affiliation(s)
- Raquel Yotti
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; .,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA; , .,Cardiovascular Division and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA; ,
| |
Collapse
|
50
|
van der Velden J, Stienen GJM. Cardiac Disorders and Pathophysiology of Sarcomeric Proteins. Physiol Rev 2019; 99:381-426. [PMID: 30379622 DOI: 10.1152/physrev.00040.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The sarcomeric proteins represent the structural building blocks of heart muscle, which are essential for contraction and relaxation. During recent years, it has become evident that posttranslational modifications of sarcomeric proteins, in particular phosphorylation, tune cardiac pump function at rest and during exercise. This delicate, orchestrated interaction is also influenced by mutations, predominantly in sarcomeric proteins, which cause hypertrophic or dilated cardiomyopathy. In this review, we follow a bottom-up approach starting from a description of the basic components of cardiac muscle at the molecular level up to the various forms of cardiac disorders at the organ level. An overview is given of sarcomere changes in acquired and inherited forms of cardiac disease and the underlying disease mechanisms with particular reference to human tissue. A distinction will be made between the primary defect and maladaptive/adaptive secondary changes. Techniques used to unravel functional consequences of disease-induced protein changes are described, and an overview of current and future treatments targeted at sarcomeric proteins is given. The current evidence presented suggests that sarcomeres not only form the basis of cardiac muscle function but also represent a therapeutic target to combat cardiac disease.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Ger J M Stienen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| |
Collapse
|