1
|
Rassier DE, Månsson A. Mechanisms of myosin II force generation: insights from novel experimental techniques and approaches. Physiol Rev 2025; 105:1-93. [PMID: 38451233 DOI: 10.1152/physrev.00014.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
Myosin II is a molecular motor that converts chemical energy derived from ATP hydrolysis into mechanical work. Myosin II isoforms are responsible for muscle contraction and a range of cell functions relying on the development of force and motion. When the motor attaches to actin, ATP is hydrolyzed and inorganic phosphate (Pi) and ADP are released from its active site. These reactions are coordinated with changes in the structure of myosin, promoting the so-called "power stroke" that causes the sliding of actin filaments. The general features of the myosin-actin interactions are well accepted, but there are critical issues that remain poorly understood, mostly due to technological limitations. In recent years, there has been a significant advance in structural, biochemical, and mechanical methods that have advanced the field considerably. New modeling approaches have also allowed researchers to understand actomyosin interactions at different levels of analysis. This paper reviews recent studies looking into the interaction between myosin II and actin filaments, which leads to power stroke and force generation. It reviews studies conducted with single myosin molecules, myosins working in filaments, muscle sarcomeres, myofibrils, and fibers. It also reviews the mathematical models that have been used to understand the mechanics of myosin II in approaches focusing on single molecules to ensembles. Finally, it includes brief sections on translational aspects, how changes in the myosin motor by mutations and/or posttranslational modifications may cause detrimental effects in diseases and aging, among other conditions, and how myosin II has become an emerging drug target.
Collapse
Affiliation(s)
- Dilson E Rassier
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Alf Månsson
- Physiology, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
2
|
Mohran S, McMillen TS, Mandrycky C, Tu AY, Kooiker KB, Qian W, Neys S, Osegueda B, Moussavi-Harami F, Irving TC, Regnier M, Ma W. Calcium has a direct effect on thick filament activation in porcine myocardium. J Gen Physiol 2024; 156:e202413545. [PMID: 39302315 PMCID: PMC11415303 DOI: 10.1085/jgp.202413545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/03/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
Sarcomere activation in striated muscle requires both thin filament-based and thick filament-based activation mechanisms. Recent studies have shown that myosin heads on the thick filaments undergo OFF to ON structural transitions in response to calcium (Ca2+) in permeabilized porcine myocardium in the presence of a small molecule inhibitor that eliminated active force. The changes in X-ray diffraction signatures of OFF to ON transitions were interpreted as Ca2+ acting to activate the thick filaments. Alternatively, Ca2+ binding to troponin could initiate a Ca2+-dependent crosstalk from the thin filament to the thick filament via interfilament connections such as the myosin binding protein-C. Here, we exchanged native troponin in permeabilized porcine myocardium for troponin containing the cTnC D65A mutation, which disallows the activation of troponin through Ca2+ binding to determine if Ca2+-dependent thick filament activation persists in the absence of thin filament activation. After the exchange protocol, over 95% of the Ca2+-activated force was eliminated. Equatorial intensity ratio increased significantly in both WT and D65A exchanged myocardium with increasing Ca2+ concentration. The degree of helical ordering of the myosin heads decreased by the same amount in WT and D65A myocardium when Ca2+ concentration increased. These results are consistent with a direct effect of Ca2+ in activating the thick filament rather than an indirect effect due to Ca2+-mediated crosstalk between the thick and thin filaments.
Collapse
Affiliation(s)
- Saffie Mohran
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Timothy S. McMillen
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
| | - Christian Mandrycky
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
| | - An-Yue Tu
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kristina B. Kooiker
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Wenjing Qian
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Stephanie Neys
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Brayan Osegueda
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Farid Moussavi-Harami
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Thomas C. Irving
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Weikang Ma
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
3
|
Childers MC, Geeves MA, Regnier M. Interacting myosin head dynamics and their modification by 2'-deoxy-ADP. Biophys J 2024:S0006-3495(24)00685-4. [PMID: 39444161 DOI: 10.1016/j.bpj.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/22/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024] Open
Abstract
The contraction of striated muscle is driven by cycling myosin motor proteins embedded within the thick filaments of sarcomeres. In addition to cross-bridge cycling with actin, these myosin proteins can enter an inactive, sequestered state in which the globular S1 heads rest along the thick filament surface and are inhibited from performing motor activities. Structurally, this state is called the interacting heads motif (IHM) and is a critical conformational state of myosin that regulates muscle contractility and energy expenditure. Structural perturbation of the sequestered state can pathologically disrupt IHM structure and the mechanical performance of muscle tissue. Thus, the IHM state has become a target for therapeutic intervention. An ATP analog called 2'-deoxy-ATP (dATP) is a potent myosin activator that destabilizes the IHM. Here, we use molecular dynamics simulations to study the molecular mechanisms by which dATP modifies the structure and dynamics of myosin in a sequestered state. Simulations of the IHM state containing ADP.Pi in both nucleotide binding pockets revealed dynamic motions of the blocked head-free head interface, light chain binding domain, and S2 in this "inactive" state of myosin. Replacement of ADP.Pi by dADP.Pi triggered a series of structural changes that increased heterogeneity among residue contact pairs at the blocked head-free head interface and a 14% decrease in the interaction energy at the interface. Dynamic changes to this interface were accompanied by dynamics in the light chain binding region. A comparative analysis of these dynamics predicted new structural sites that may affect IHM stability.
Collapse
Affiliation(s)
- Matthew Carter Childers
- Department of Bioengineering, School of Medicine, University of Washington, Seattle, Washington
| | - Michael A Geeves
- Department of Biosciences, University of Kent, Kent, United Kingdom
| | - Michael Regnier
- Department of Bioengineering, School of Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
4
|
Szczesna-Cordary D. Novel cardiac myosin inhibitor for hypertrophic cardiomyopathy. J Gen Physiol 2024; 156:e202413640. [PMID: 39136654 PMCID: PMC11318667 DOI: 10.1085/jgp.202413640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
New RLC-1 small-molecule inhibits actomyosin interactions, reduces contractile force, and speeds up myosin cross-bridge kinetics.
Collapse
Affiliation(s)
- Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
5
|
Laitila J, Seaborne RAE, Ranu N, Kolb JS, Wallgren-Pettersson C, Witting N, Vissing J, Vilchez JJ, Zanoteli E, Palmio J, Huovinen S, Granzier H, Ochala J. Myosin ATPase inhibition fails to rescue the metabolically dysregulated proteome of nebulin-deficient muscle. J Physiol 2024; 602:5229-5245. [PMID: 39216086 DOI: 10.1113/jp286870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Nemaline myopathy (NM) is a genetic muscle disease, primarily caused by mutations in the NEB gene (NEB-NM) and with muscle myosin dysfunction as a major molecular pathogenic mechanism. Recently, we have observed that the myosin biochemical super-relaxed state was significantly impaired in NEB-NM, inducing an aberrant increase in ATP consumption and remodelling of the energy proteome in diseased muscle fibres. Because the small-molecule Mavacamten is known to promote the myosin super-relaxed state and reduce the ATP demand, we tested its potency in the context of NEB-NM. We first conducted in vitro experiments in isolated single myofibres from patients and found that Mavacamten successfully reversed the myosin ATP overconsumption. Following this, we assessed its short-term in vivo effects using the conditional nebulin knockout (cNeb KO) mouse model and subsequently performing global proteomics profiling in dissected soleus myofibres. After a 4 week treatment period, we observed a remodelling of a large number of proteins in both cNeb KO mice and their wild-type siblings. Nevertheless, these changes were not related to the energy proteome, indicating that short-term Mavacamten treatment is not sufficient to properly counterbalance the metabolically dysregulated proteome of cNeb KO mice. Taken together, our findings emphasize Mavacamten potency in vitro but challenge its short-term efficacy in vivo. KEY POINTS: No cure exists for nemaline myopathy, a type of genetic skeletal muscle disease mainly derived from mutations in genes encoding myofilament proteins. Applying Mavacamten, a small molecule directly targeting the myofilaments, to isolated membrane-permeabilized muscle fibres from human patients restored myosin energetic disturbances. Treating a mouse model of nemaline myopathy in vivo with Mavacamten for 4 weeks, remodelled the skeletal muscle fibre proteome without any noticeable effects on energetic proteins. Short-term Mavacamten treatment may not be sufficient to reverse the muscle phenotype in nemaline myopathy.
Collapse
Affiliation(s)
- Jenni Laitila
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert A E Seaborne
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Natasha Ranu
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Justin S Kolb
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, MO, USA
| | - Carina Wallgren-Pettersson
- The Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland and Department of Medical and Clinical Genetics, Medicum, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Nanna Witting
- Copenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Juan Jesus Vilchez
- Neuromuscular and Ataxias Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Spain, Valencia, Spain
| | - Edmar Zanoteli
- Department of Neurology, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Johanna Palmio
- Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland
| | - Sanna Huovinen
- Department of Pathology, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Henk Granzier
- The Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland and Department of Medical and Clinical Genetics, Medicum, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Julien Ochala
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Spudich JA, Nandwani N, Robert-Paganin J, Houdusse A, Ruppel KM. Reassessing the unifying hypothesis for hypercontractility caused by myosin mutations in hypertrophic cardiomyopathy. EMBO J 2024; 43:4139-4155. [PMID: 39192034 PMCID: PMC11445530 DOI: 10.1038/s44318-024-00199-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024] Open
Affiliation(s)
- James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Neha Nandwani
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France
| | - Kathleen M Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
7
|
Almansouri NE, Nadeem Bukhari SAU, Qureshi MH, Idrees M, Riaz CZ, Asghar AR, Habib A, Ikram J, Ehsan M, Rehman WU, Cheema HA, Ayyan M, Kandel K, Iqbal S, Pasha A, Patel K, Sabouni MA. Efficacy and safety of mavacamten for the treatment of hypertrophic cardiomyopathy: an updated systematic review and meta-analysis of randomized controlled trials. Ann Med Surg (Lond) 2024; 86:6097-6104. [PMID: 39359828 PMCID: PMC11444588 DOI: 10.1097/ms9.0000000000002466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/01/2024] [Indexed: 10/04/2024] Open
Abstract
The efficacy and safety profile of mavacamten, a cardiac myosin inhibitor for the treatment of hypertrophic cardiomyopathy (HCM) is not well-established, prompting the need for an updated meta-analysis. The authors conducted an extensive search across multiple electronic databases, including Embase, MEDLINE (via Pubmed), and CENTRAL, to identify randomized controlled trials (RCTs) assessing the efficacy and safety of mavacamten in HCM. Review Manager 5.4 (Revman) was employed to pool risk ratios (RR) and mean differences (MD). Our literature search yielded 4 RCTs with a total of 503 patients. Mavacamten was found to be associated with higher rates of greater than or equal to 1 New York Heart Association (NYHA) class improvement (RR 2.20, 95% CI: 1.48-3.28; I2=51%) and change from baseline in the Kansas City Cardiomyopathy Questionnaire- Clinical Summary Score (KCCQ-CSS) (MD 7.50, 95% CI: 3.44-11.55; I2 =50%). Mavacamten was also associated with improved resting left ventricular outflow tract (LVOT) gradient (MD -38.33, 95% CI: -49.38 to -27.28; I2 =75%), Valsalva LVOT gradient (MD -48.08, 95% CI: -62.21 to -33.96; I2 =78%), post-exercise LVOT gradient (MD -37.1, 95% CI: -44.37 to -29.84; I2 =0%), LVMI (MD -16.91, 95% CI: -28.29 to -5.54; I2 =88%), and lower rates of septal reduction therapy (SRT) (RR 0.30, 95% CI: 0.22-0.40; I2 =0%). There were no significant differences between mavacamten and placebo regarding the composite functional outcome, greater than or equal to 1 treatment-emergent adverse event, greater than or equal to 1 serious adverse event, and atrial fibrillation. The authors; findings suggest that mavacamten contributes to improvements in NYHA class, KCCQ-CSS scores, and LVOT gradients while reducing the incidence of SRT in patients with HCM.
Collapse
Affiliation(s)
| | | | | | - Muhammad Idrees
- Department of Medicine, Multan Institute of Kidney Diseases, Multan
| | | | | | - Ayesha Habib
- Department of Medicine, Faisalabad Medical University, Faisalabad
| | - Jibran Ikram
- Department of Medicine, Lifecare Hospital & Research Institute, Peshawar, Pakistan
| | - Muhammad Ehsan
- Department of Medicine, King Edward Medical University, Lahore
| | - Wajeeh Ur Rehman
- Department of Medicine, United Health Services, Johnson City, New York
| | | | - Muhammad Ayyan
- Department of Medicine, King Edward Medical University, Lahore
| | - Kamal Kandel
- Department of Medicine, Kathmandu University, Nepal
| | | | - Ahmed Pasha
- Heart and Vascular Institute, United Health Services, Johnson City, NY
| | - Keyoor Patel
- Heart and Vascular Institute, United Health Services, Johnson City, NY
| | | |
Collapse
|
8
|
Lewalle A, Milburn G, Campbell KS, Niederer SA. Cardiac length-dependent activation driven by force-dependent thick-filament dynamics. Biophys J 2024; 123:2996-3009. [PMID: 38807364 PMCID: PMC11428202 DOI: 10.1016/j.bpj.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/17/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The length-dependent activation (LDA) of maximum force and calcium sensitivity are established features of cardiac muscle contraction but the dominant underlying mechanisms remain to be fully clarified. Alongside the well-documented regulation of contraction via the thin filaments, experiments have identified an additional force-dependent thick-filament activation, whereby myosin heads parked in a so-called off state become available to generate force. This process produces a feedback effect that may potentially drive LDA. Using biomechanical modeling of a human left-ventricular myocyte, this study investigates the extent to which the off-state dynamics could, by itself, plausibly account for LDA, depending on the specific mathematical formulation of the feedback. We hypothesized four different models of the off-state regulatory feedback based on (A) total force, (B) active force, (C) sarcomere strain, and (D) passive force. We tested if these models could reproduce the isometric steady-state and dynamic LDA features predicted by an earlier published model of a human left-ventricle myocyte featuring purely phenomenological length dependences. The results suggest that only total-force feedback (A) is capable of reproducing the expected behaviors, but that passive tension could provide a length-dependent signal on which to initiate the feedback. Furthermore, by attributing LDA to off-state dynamics, our proposed model also qualitatively reproduces experimentally observed effects of the off-state-stabilizing drug mavacamten. Taken together, these results support off-state dynamics as a plausible primary mechanism underlying LDA.
Collapse
Affiliation(s)
- Alexandre Lewalle
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom.
| | - Gregory Milburn
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, Kentucky
| | - Steven A Niederer
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
9
|
Greenberg MJ. Super-relaxed or super-stressed: Modeling length-dependent activation in cardiac muscle. Biophys J 2024; 123:2957-2960. [PMID: 39068513 PMCID: PMC11427768 DOI: 10.1016/j.bpj.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024] Open
Affiliation(s)
- Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
10
|
Maurizi N, Antiochos P, Muller O, Wuerzner G, Monney P. Accelerated hypertension following mavacamten introduction in severe obstructive hypertrophic cardiomyopathy with hypertension: a case report. Eur Heart J Case Rep 2024; 8:ytae450. [PMID: 39258018 PMCID: PMC11384883 DOI: 10.1093/ehjcr/ytae450] [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: 03/26/2024] [Revised: 05/16/2024] [Accepted: 08/15/2024] [Indexed: 09/12/2024]
Abstract
Background Mavacamten in Phase 2 and 3 clinical trials was well tolerated, reduced left ventricular outflow tract obstruction (LVOTO), and improved exercise capacity and symptoms. However, due to its recent introduction in the market, there is limited evidence from real-world patients with severe/multiple comorbidities and/or who are exposed to potential treatment interactions. Hypertension is common in patients with hypertrophic cardiomyopathy (HCM), but its impact on the treatment of LVOTO is undefined. Case summary A 55-year-old man with severely obstructive symptomatic HCM and Grade I arterial hypertension underwent treatment with mavacamten 5 mg. He presented an accelerated hypertension from Day 10 of treatment. On admission, he reported improvement of his dyspnoea [New York Heart Association (NYHA) Class II] and NT-pro BNP decreased to 1646 ng/L. Echocardiography showed a left ventricular ejection fraction of 60% with reduced systolic anterior motion and LVOTO (max 21 mmHg). Causes of secondary hypertension were excluded, and blood pressure (BP) was controlled by eplerenone and amlodipine introduction. Accelerated hypertension was concluded as a final diagnosis, and a potential causal link with the introduction of mavacamten was made. Evolution up to Day 135 proved a stabilization of the BP profile and of the LVOT gradient (max 36 mmHg) as well as improvement in functional capacity (NYHA Class I). Discussion We hypothesize that rapid relief of excess afterload may induce alterations potentially leading to high BP in patients with impaired peripheral vascular resistances. Patients with severe obstructive HCM and hypertension should be given special attention during mavacamten titration and should self-monitor the BP during this phase.
Collapse
Affiliation(s)
- Niccolò Maurizi
- Service of Cardiology, Lausanne University Hospital, Rue de Bugnon 46, 1011 Lausanne, Switzerland
| | - Panagiotis Antiochos
- Service of Cardiology, Lausanne University Hospital, Rue de Bugnon 46, 1011 Lausanne, Switzerland
| | - Olivier Muller
- Service of Cardiology, Lausanne University Hospital, Rue de Bugnon 46, 1011 Lausanne, Switzerland
| | - Gregory Wuerzner
- Service of Nephrology, Lausanne University Hospital, Rue de Bugnon 46, 1011 Lausanne, Switzerland
| | - Pierre Monney
- Service of Cardiology, Lausanne University Hospital, Rue de Bugnon 46, 1011 Lausanne, Switzerland
| |
Collapse
|
11
|
Eiswirth C, Gilliland YE. Reflections on community experience with Mavacamten. Prog Cardiovasc Dis 2024; 86:69-72. [PMID: 39153535 DOI: 10.1016/j.pcad.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/19/2024]
Affiliation(s)
- Clement Eiswirth
- Cardiomyopathy and Heart Failure Program & Coronary Critical Care, Department of Cardiovascular Diseases, John Ochsner Heart & Vascular Institute, Ochsner Medical Center, New Orleans, LA 70121, United States of America.
| | - Yvonne E Gilliland
- Department of Cardiovascular Diseases, John Ochsner Heart & Vascular Institute, Ochsner Medical Center, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, LA 70121, United States of America.
| |
Collapse
|
12
|
Alharbi TY, Alnadawi HA, Almutairi GM, Altheyab FY, Aldoweesh OH, Alfehaid OS, Alhaj AA, Alotaibi AM, Al Zweihary AM. Safety and Effectiveness of Mavacamten Use in Hypertrophic Obstructive Cardiomyopathy: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e70550. [PMID: 39479079 PMCID: PMC11524545 DOI: 10.7759/cureus.70550] [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] [Accepted: 09/27/2024] [Indexed: 11/02/2024] Open
Abstract
Hypertrophic obstructive cardiomyopathy (HOCM) is a complex genetic cardiac disease that causes left ventricular hypertrophy and obstruction of the outflow tract. Mavacamten, a novel cardiac myosin inhibitor, has emerged as a potentially beneficial therapeutic option. This meta-analysis aimed to determine whether mavacamten is effective and safe for use in patients with HOCM. A systematic literature search was performed in PubMed and the Cochrane Central Register of Controlled Trials to identify randomized controlled trials (RCTs) that compared mavacamten to placebo in patients with HOCM. The primary objectives were changes in the gradients associated with the Valsalva maneuver and resting left ventricular outflow tract (LVOT). Alterations in the left atrial volume index (LAVI), left ventricular mass index (LVMI), and NT-proBNP level were secondary outcomes. Safety outcomes were also evaluated. Random effects models were used in the meta-analysis. Two RCTs comprising 332 patients were included. Mavacamten significantly reduced the Valsalva LVOT gradient (mean difference (MD) = -54.94 mmHg; 95% CI: -70.32, -39.56; P = 0.13) and resting LVOT gradient (MD = -42.44 mmHg; 95% CI: -67.52, -17.36; P<0.001) compared to placebo. Significant improvements were also observed in LAVI (MD = -7.18 mL/m²; 95% CI: -11.00, -3.37; P = 0.24) and NT-proBNP levels (RR = 0.58; 95% CI: 0.39, 0.84; P<0.001). LVMI showed a trend toward reduction (MD = -19.15 g/m²; 95% CI: -41.98, 3.69; P<0.001). Mavacamten demonstrated a favorable safety profile with few reported adverse events. This meta-analysis aimed to demonstrate the efficacy and short-term safety of mavacamten in patients with HOCM. Considerable improvement was observed in the LVOT gradients, cardiac remodeling measures, and indicators of cardiac stress when mavacamten was administered. Based on this data, mavacamten appears to offer the potential for a paradigm shift in the management of HOCM. However, studies conducted over an extended period are required to validate its long-term effectiveness and safety profile.
Collapse
|
13
|
Merali S, Salinger DH, Palmisano M, Sehnert AJ, Thanneer N, Back H, Seroogy JD, Gretler DD, Roy A, Perera V. Recommendation of mavacamten posology by model-based analyses in adults with obstructive hypertrophic cardiomyopathy. CPT Pharmacometrics Syst Pharmacol 2024; 13:1448-1461. [PMID: 38695527 PMCID: PMC11533099 DOI: 10.1002/psp4.13138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 11/05/2024] Open
Abstract
Mavacamten is the first cardiac myosin inhibitor approved by the US Food and Drug Administration for the treatment of adults with symptomatic obstructive hypertrophic cardiomyopathy (HCM). The phase III EXPLORER-HCM (NCT03470545) study used a dose-titration scheme based on mavacamten exposure and echocardiographic assessment of Valsalva left ventricular outflow tract gradient (VLVOTg) and left ventricular ejection fraction (LVEF). Using population pharmacokinetic/exposure-response modeling and simulations of virtual patients, this in silico study evaluated alternative dose-titration regimens for mavacamten, including regimens that were guided by echocardiographic measures only. Mavacamten exposure-response models for VLVOTg (efficacy) and LVEF (safety) were developed using patient data from five clinical studies and characterized using nonlinear mixed-effects models. Simulations of five echocardiography-guided regimens were performed in virtual cohorts constructed based on either expected or equal population distributions of cytochrome P450 2C19 (CYP2C19) metabolizer phenotypes. Each regimen aimed to maximize the proportions of patients who achieved a VLVOTg below 30 mm Hg while maintaining LVEF above 50% over 40 weeks and 104 weeks, respectively. The exposure-response models successfully characterized mavacamten efficacy and safety parameters. Overall, the simulated regimen with the optimal benefit-risk profile across CYP2C19 phenotypes had steps for down-titration at weeks 4 and 8 (for VLVOTg <20 mm Hg), and up-titration at week 12 (for VLVOTg ≥30 mm Hg and LVEF ≥55%), and every 12 weeks thereafter. This simulation-optimized regimen is recommended in the mavacamten US prescribing information.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Amit Roy
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | | |
Collapse
|
14
|
Liang LW, Lumish HS, Sewanan LR, Shimada YJ, Maurer MS, Weiner SD, Clerkin KJ. Evolving Strategies for the Management of Obstructive Hypertrophic Cardiomyopathy. J Card Fail 2024; 30:1136-1153. [PMID: 38777216 PMCID: PMC11415289 DOI: 10.1016/j.cardfail.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/25/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
For many years, treatment of hypertrophic cardiomyopathy (HCM) has focused on non-disease-specific therapies. Cardiac myosin modulators (ie, mavacamten and aficamten) reduce the pathologic actin-myosin interactions that are characteristic of HCM, leading to improved cardiac energetics and reduction in hypercontractility. Several recently published randomized clinical trials have demonstrated that mavacamten improves exercise capacity, left ventricular outflow tract obstruction and symptoms in patients with obstructive HCM and may delay the need for septal-reduction therapy. Long-term data in real-world populations will be needed to fully assess the safety and efficacy of mavacamten. Importantly, HCM is a complex and heterogeneous disease, and not all patients will respond to mavacamten; therefore, careful patient selection and shared decision making will be necessary in guiding the use of mavacamten in obstructive HCM.
Collapse
Affiliation(s)
- Lusha W Liang
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Heidi S Lumish
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Lorenzo R Sewanan
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Yuichi J Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Shepard D Weiner
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Kevin J Clerkin
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
15
|
Marian AJ. "The unbearable lightness" of the primary end point in clinical trials. Mol Cell Biochem 2024:10.1007/s11010-024-05098-7. [PMID: 39212891 DOI: 10.1007/s11010-024-05098-7] [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: 07/23/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
In this Perspective, I discuss the limitations of a soft primary endpoint that is used in some of the recent randomized phase II/III clinical trials. Unfortunately, many clinicians and investigators do not interpret the data critically to recognize the limitations of such findings. I advise against over-interpreting the effects of an intervention on a soft primary endpoint.
Collapse
Affiliation(s)
- A J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Owens AT, Desai M, Wheeler MT, Rodonski A, Merali S, Sehnert AJ, Saberi S. Mavacamten for Obstructive Hypertrophic Cardiomyopathy: Rationale for Clinically Guided Dose Titration to Optimize Individual Response. J Am Heart Assoc 2024; 13:e033767. [PMID: 39206723 DOI: 10.1161/jaha.124.033767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Mavacamten is the first and only cardiac myosin inhibitor approved in 5 continents for the treatment of adults with symptomatic New York Heart Association class II and III obstructive hypertrophic cardiomyopathy. An evidence-based rationale was used to develop individualized mavacamten dosing, guided by commonly used clinical parameters. Echocardiography is recommended as part of routine clinical assessment of patients with hypertrophic cardiomyopathy, and left ventricular (LV) outflow tract gradient and LV ejection fraction are parameters that can be readily assessed and monitored by echocardiography. Therefore, an echocardiography-based, clinically guided dose-titration strategy was developed to optimize patient benefit from mavacamten for the treatment of symptomatic obstructive hypertrophic cardiomyopathy while minimizing the risk of LV ejection fraction reduction. Results from clinical trials paired with extensive modeling and simulation analyses support a dose-titration and monitoring strategy based on serial echocardiographic measures of Valsalva LV outflow tract gradient and LV ejection fraction. This dosing approach allows for the identification of the lowest individualized mavacamten dose and exposure required to provide improvements in LV outflow tract obstruction, functional capacity, and symptoms. Mavacamten is primarily metabolized by CYP2C19 (cytochrome P450 2C19), and CYP2C19 metabolizer phenotype has an effect on mavacamten exposure. Therefore, this approach has also been demonstrated to provide a favorable safety profile irrespective of patients' CYP2C19 metabolizer status. The dose-titration strategy includes additional considerations for the potential onset of systolic dysfunction in the context of intercurrent illness, and for the potential of drug-drug interactions with inhibitors and substrates of cytochrome P450 enzymes. This posology is reflected in the mavacamten US prescribing information.
Collapse
Affiliation(s)
- Anjali T Owens
- University of Pennsylvania Perelman School of Medicine Philadelphia PA USA
| | - Milind Desai
- Heart, Vascular, and Thoracic Institute Cleveland Clinic Cleveland OH USA
| | - Matthew T Wheeler
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA USA
| | | | | | | | - Sara Saberi
- Division of Cardiovascular Medicine University of Michigan Ann Arbor MI USA
| |
Collapse
|
17
|
Sitbon YH, Kazmierczak K, Liang J, Kloehn AJ, Vinod J, Kanashiro-Takeuchi R, Szczesna-Cordary D. Dual effect of N-terminal deletion of cardiac myosin essential light chain in mitigating cardiomyopathy. iScience 2024; 27:110591. [PMID: 39211545 PMCID: PMC11357882 DOI: 10.1016/j.isci.2024.110591] [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] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/14/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
We investigated the role of the N-terminus (residues 1-43) of the myosin essential light chain (N-ELC) in regulating cardiac function in hypertrophic (HCM-A57G) and restrictive (RCM-E143K) cardiomyopathy mice. Both models were cross-genotyped with N-ELC-truncated Δ43 mice, and the offspring were studied using echocardiography and muscle contractile mechanics. In A57G×Δ43 mice, Δ43 expression improved heart function and reduced hypertrophy and fibrosis. No improvements were seen in E143K×Δ43 compared to RCM-E143K mice. HCM-mutant pathology involved an impaired N-ELC tension sensor, disrupted N-ELC-actin interactions, an altered force-pCa relationship, and a destabilized myosin's super-relaxed state. Removal of the malfunctioning N-ELC sensor led to functional rescue in HCM-truncated mutant hearts. However, the RCM mutation could not be rescued by N-ELC deletion, likely due to its proximity to the myosin motor domain, affecting lever-arm rigidity and myosin function. This study provides insights into the role of N-ELC in the development and potential rescue of ELC-mutant cardiomyopathy.
Collapse
Affiliation(s)
- Yoel H. Sitbon
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew J. Kloehn
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Judith Vinod
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rosemeire Kanashiro-Takeuchi
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
18
|
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
|
19
|
Topriceanu CC, Moon JC, Raja AA, Captur G, Ho CY. Phenotypic Spectrum of Subclinical Sarcomere-Related Hypertrophic Cardiomyopathy and Transition to Overt Disease. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004580. [PMID: 38910555 PMCID: PMC11335455 DOI: 10.1161/circgen.124.004580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Genetic hypertrophic cardiomyopathy (HCM) is classically caused by pathogenic/likely pathogenic variants in sarcomere genes (G+). Currently, HCM is diagnosed if there is unexplained left ventricular (LV) hypertrophy with LV wall thickness ≥15 mm in probands or ≥13 mm in at-risk relatives. Although LV hypertrophy is a key feature, this binary metric does not encompass the full constellation of phenotypic features, particularly in the subclinical stage of the disease. Subtle phenotypic manifestations can be identified in sarcomere variant carriers with normal LV wall thickness, before diagnosis with HCM (G+/LV hypertrophy-; subclinical HCM). We conducted a systematic review to summarize current knowledge about the phenotypic spectrum of subclinical HCM and factors influencing penetrance and expressivity. Although the mechanisms driving the development of LV hypertrophy are yet to be elucidated, activation of profibrotic pathways, impaired relaxation, abnormal Ca2+ signaling, altered myocardial energetics, and microvascular dysfunction have all been identified in subclinical HCM. Progression from subclinical to clinically overt HCM may be more likely if early phenotypic manifestations are present, including ECG abnormalities, longer mitral valve leaflets, lower global E' velocities on Doppler echocardiography, and higher serum N-terminal propeptide of B-type natriuretic peptide. Longitudinal studies of variant carriers are critically needed to improve our understanding of penetrance, characterize the transition to disease, identify risk predictors of phenotypic evolution, and guide the development of novel treatment strategies aimed at influencing disease trajectory.
Collapse
Affiliation(s)
- Constantin-Cristian Topriceanu
- Dept of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- UCL Institute of Cardiovascular Science
- Cardiac MRI Unit, Barts Heart Centre, London, United Kingdom
| | - James C. Moon
- UCL Institute of Cardiovascular Science
- Cardiac MRI Unit, Barts Heart Centre, London, United Kingdom
| | - Anna Axelsson Raja
- Dept of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Gabriella Captur
- UCL Institute of Cardiovascular Science
- UCL MRC Unit for Lifelong Health and Ageing University College London
- The Royal Free Hospital, Centre for Inherited Heart Muscle Conditions, Cardiology Dept, London, United Kingdom
| | - Carolyn Y. Ho
- Dept of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
20
|
Hartman JJ, Hwee DT, Robert-Paganin J, Chuang C, Chin ER, Edell S, Lee KH, Madhvani R, Paliwal P, Pernier J, Sarkar SS, Schaletzky J, Schauer K, Taheri KD, Wang J, Wehri E, Wu Y, Houdusse A, Morgan BP, Malik FI. Aficamten is a small-molecule cardiac myosin inhibitor designed to treat hypertrophic cardiomyopathy. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1003-1016. [PMID: 39196032 PMCID: PMC11358156 DOI: 10.1038/s44161-024-00505-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 06/06/2024] [Indexed: 08/29/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disease of the sarcomere resulting in excessive cardiac contractility. The first-in-class cardiac myosin inhibitor, mavacamten, improves symptoms in obstructive HCM. Here we present aficamten, a selective small-molecule inhibitor of cardiac myosin that diminishes ATPase activity by strongly slowing phosphate release, stabilizing a weak actin-binding state. Binding to an allosteric site on the myosin catalytic domain distinct from mavacamten, aficamten prevents the conformational changes necessary to enter the strongly actin-bound force-generating state. In doing so, aficamten reduces the number of functional myosin heads driving sarcomere shortening. The crystal structure of aficamten bound to cardiac myosin in the pre-powerstroke state provides a basis for understanding its selectivity over smooth and fast skeletal muscle. Furthermore, in cardiac myocytes and in mice bearing the hypertrophic R403Q cardiac myosin mutation, aficamten reduces cardiac contractility. Our findings suggest aficamten holds promise as a therapy for HCM.
Collapse
Affiliation(s)
- James J Hartman
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA.
| | - Darren T Hwee
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, France
| | - Chihyuan Chuang
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Eva R Chin
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Samantha Edell
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Ken H Lee
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Roshni Madhvani
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Preeti Paliwal
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Julien Pernier
- Tumor Cell Dynamics Unit, Inserm U1279 Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | | | - Julia Schaletzky
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Kristine Schauer
- Tumor Cell Dynamics Unit, Inserm U1279 Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Khanha D Taheri
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Jingying Wang
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Eddie Wehri
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Yangsong Wu
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Anne Houdusse
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, France
| | - Bradley P Morgan
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| | - Fady I Malik
- Research and Non-Clinical Development, Cytokinetics, South San Francisco, CA, USA
| |
Collapse
|
21
|
van den Berg M, Shi Z, Claassen WJ, Hooijman P, Lewis CTA, Andersen JL, van der Pijl RJ, Bogaards SJP, Conijn S, Peters EL, Begthel LPL, Uijterwijk B, Lindqvist J, Langlais PR, Girbes ARJ, Stapel S, Granzier H, Campbell KS, Ma W, Irving T, Hwee DT, Hartman JJ, Malik FI, Paul M, Beishuizen A, Ochala J, Heunks L, Ottenheijm CAC. Super-relaxed myosins contribute to respiratory muscle hibernation in mechanically ventilated patients. Sci Transl Med 2024; 16:eadg3894. [PMID: 39083588 DOI: 10.1126/scitranslmed.adg3894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/12/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Patients receiving mechanical ventilation in the intensive care unit (ICU) frequently develop contractile weakness of the diaphragm. Consequently, they may experience difficulty weaning from mechanical ventilation, which increases mortality and poses a high economic burden. Because of a lack of knowledge regarding the molecular changes in the diaphragm, no treatment is currently available to improve diaphragm contractility. We compared diaphragm biopsies from ventilated ICU patients (N = 54) to those of non-ICU patients undergoing thoracic surgery (N = 27). By integrating data from myofiber force measurements, x-ray diffraction experiments, and biochemical assays with clinical data, we found that in myofibers isolated from the diaphragm of ventilated ICU patients, myosin is trapped in an energy-sparing, super-relaxed state, which impairs the binding of myosin to actin during diaphragm contraction. Studies on quadriceps biopsies of ICU patients and on the diaphragm of previously healthy mechanically ventilated rats suggested that the super-relaxed myosins are specific to the diaphragm and not a result of critical illness. Exposing slow- and fast-twitch myofibers isolated from the diaphragm biopsies to small-molecule compounds activating troponin restored contractile force in vitro. These findings support the continued development of drugs that target sarcomere proteins to increase the calcium sensitivity of myofibers for the treatment of ICU-acquired diaphragm weakness.
Collapse
Affiliation(s)
- Marloes van den Berg
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
- Bispebjerg Hospital, Institute of Sports Medicine, Copenhagen 2400, Denmark
| | - Zhonghua Shi
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
- Amsterdam UMC, Location VUmc, Department of Intensive Care Medicine, Amsterdam 1081, HV, Netherlands
- Sanbo Brain Hospital, Capital Medical University, Intensive Care Medicine, Beijing 100093, China
| | - Wout J Claassen
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Pleuni Hooijman
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Christopher T A Lewis
- University of Copenhagen, Department of Biomedical Sciences, Copenhagen 2200, Denmark
- Research and Early Development, Novo Nordisk A/S, Måløv 2760, Denmark
| | - Jesper L Andersen
- Bispebjerg Hospital, Institute of Sports Medicine, Copenhagen 2400, Denmark
| | - Robbert J van der Pijl
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| | - Sylvia J P Bogaards
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Stefan Conijn
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Eva L Peters
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| | - Leon P L Begthel
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Bas Uijterwijk
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Johan Lindqvist
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| | - Paul R Langlais
- Department of Endocrinology, University of Arizona, Tucson, AZ 85721, USA
| | - Armand R J Girbes
- Amsterdam UMC, Location VUmc, Department of Intensive Care Medicine, Amsterdam 1081, HV, Netherlands
| | - Sandra Stapel
- Amsterdam UMC, Location VUmc, Department of Intensive Care Medicine, Amsterdam 1081, HV, Netherlands
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Weikang Ma
- BioCAT, Illinois Institute of Technology, Lemont, IL 60439, USA
| | - Thomas Irving
- BioCAT, Illinois Institute of Technology, Lemont, IL 60439, USA
| | - Darren T Hwee
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - James J Hartman
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Fady I Malik
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Marinus Paul
- Amsterdam UMC, Location VUmc, Department of Cardiothoracic Surgery, Amsterdam 1081, HV, Netherlands
| | - Albertus Beishuizen
- Medisch Spectrum Twente, Intensive Care Center, Enschede 7511, HN, Netherlands
| | - Julien Ochala
- University of Copenhagen, Department of Biomedical Sciences, Copenhagen 2200, Denmark
| | - Leo Heunks
- Radboud UMC, Department of Intensive Care, Nijmegen 6525, GA, Netherlands
| | - Coen A C Ottenheijm
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| |
Collapse
|
22
|
Vogel A, Arnese R, Gudino Carrillo RM, Sehr D, Deszcz L, Bylicki A, Meinhart A, Clausen T. UNC-45 assisted myosin folding depends on a conserved FX 3HY motif implicated in Freeman Sheldon Syndrome. Nat Commun 2024; 15:6272. [PMID: 39054317 PMCID: PMC11272940 DOI: 10.1038/s41467-024-50442-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Myosin motors are critical for diverse motility functions, ranging from cytokinesis and endocytosis to muscle contraction. The UNC-45 chaperone controls myosin function mediating the folding, assembly, and degradation of the muscle protein. Here, we analyze the molecular mechanism of UNC-45 as a hub in myosin quality control. We show that UNC-45 forms discrete complexes with folded and unfolded myosin, forwarding them to downstream chaperones and E3 ligases. Structural analysis of a minimal chaperone:substrate complex reveals that UNC-45 binds to a conserved FX3HY motif in the myosin motor domain. Disrupting the observed interface by mutagenesis prevents myosin maturation leading to protein aggregation in vivo. We also show that a mutation in the FX3HY motif linked to the Freeman Sheldon Syndrome impairs UNC-45 assisted folding, reducing the level of functional myosin. These findings demonstrate that a faulty myosin quality control is a critical yet unexplored cause of human myopathies.
Collapse
Affiliation(s)
- Antonia Vogel
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Renato Arnese
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Ricardo M Gudino Carrillo
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
- Medical University, Vienna, Austria
| | - Daria Sehr
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Luiza Deszcz
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Andrzej Bylicki
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Anton Meinhart
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Tim Clausen
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria.
- Vienna BioCenter Core Facilities, Vienna, Austria.
| |
Collapse
|
23
|
Merali S, Chiang M, Sychterz C, Chao L, Simmons T, Xu Y, Zhao A, Attanasio M, Murthy B, Perera V. Effect of Activated Charcoal on Mavacamten Pharmacokinetics in Healthy Participants. Am J Cardiovasc Drugs 2024; 24:569-575. [PMID: 38926266 PMCID: PMC11233281 DOI: 10.1007/s40256-024-00659-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE To assess the effect of activated charcoal on the single-dose pharmacokinetics of mavacamten when administered 2 h or 6 h after mavacamten dosing. METHODS In this open-label, randomized, parallel-group study, healthy adults were randomized into three groups to receive mavacamten 15 mg alone or mavacamten 15 mg plus activated charcoal 50 g administered either 2 h or 6 h after mavacamten dosing. Pharmacokinetic parameters were derived from plasma concentration-time data using noncompartmental methods. RESULTS Of the 45 participants randomized, 37 completed the study. When activated charcoal was administered 2 h after mavacamten dosing, mavacamten absorption and exposure were reduced compared with when mavacamten was administered alone: the area under the concentration-time curve from 0 to 72 h (AUC0-72) and area under the concentration-time curve from time 0 extrapolated to infinity (AUCINF) were reduced by 14% and 34%, respectively. The maximum plasma concentration (Cmax) was also slightly lower when activated charcoal was administered 2 h after mavacamten dosing than with mavacamten alone. Pharmacokinetic profiles were similar for mavacamten alone and mavacamten plus activated charcoal administered 6 h after mavacamten dosing. CONCLUSIONS Activated charcoal was successful in reducing mavacamten absorption and exposure when administered as soon as possible after identification of a need for adsorption (2 h after mavacamten dosing). No change in exposure was observed when activated charcoal was administered 6 h after mavacamten dosing. CLINICAL TRIAL REGISTRATION NCT05320094.
Collapse
Affiliation(s)
| | | | | | | | | | - Yiru Xu
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | |
Collapse
|
24
|
Auguin D, Robert-Paganin J, Réty S, Kikuti C, David A, Theumer G, Schmidt AW, Knölker HJ, Houdusse A. Omecamtiv mecarbil and Mavacamten target the same myosin pocket despite opposite effects in heart contraction. Nat Commun 2024; 15:4885. [PMID: 38849353 PMCID: PMC11161628 DOI: 10.1038/s41467-024-47587-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/03/2024] [Indexed: 06/09/2024] Open
Abstract
Inherited cardiomyopathies are common cardiac diseases worldwide, leading in the late stage to heart failure and death. The most promising treatments against these diseases are small molecules directly modulating the force produced by β-cardiac myosin, the molecular motor driving heart contraction. Omecamtiv mecarbil and Mavacamten are two such molecules that completed phase 3 clinical trials, and the inhibitor Mavacamten is now approved by the FDA. In contrast to Mavacamten, Omecamtiv mecarbil acts as an activator of cardiac contractility. Here, we reveal by X-ray crystallography that both drugs target the same pocket and stabilize a pre-stroke structural state, with only few local differences. All-atom molecular dynamics simulations reveal how these molecules produce distinct effects in motor allostery thus impacting force production in opposite way. Altogether, our results provide the framework for rational drug development for the purpose of personalized medicine.
Collapse
Affiliation(s)
- Daniel Auguin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
- Laboratoire de Physiologie, Ecologie et Environnement (P2E), UPRES EA 1207/USC INRAE-1328, UFR Sciences et Techniques, Université d'Orléans, Orléans, France
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | - Stéphane Réty
- Laboratoire de Biologie et Modélisation de la Cellule, ENS de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Lyon, France
| | - Carlos Kikuti
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | - Amandine David
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | | | | | | | - Anne Houdusse
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France.
| |
Collapse
|
25
|
Duno-Miranda S, Nelson SR, Rasicci DV, Bodt SM, Cirilo JA, Vang D, Sivaramakrishnan S, Yengo CM, Warshaw DM. Tail length and E525K dilated cardiomyopathy mutant alter human β-cardiac myosin super-relaxed state. J Gen Physiol 2024; 156:e202313522. [PMID: 38709176 PMCID: PMC11074782 DOI: 10.1085/jgp.202313522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/18/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is a condition characterized by impaired cardiac function, due to myocardial hypo-contractility, and is associated with point mutations in β-cardiac myosin, the molecular motor that powers cardiac contraction. Myocardial function can be modulated through sequestration of myosin motors into an auto-inhibited "super-relaxed" state (SRX), which may be further stabilized by a structural state known as the "interacting heads motif" (IHM). Here, we sought to determine whether hypo-contractility of DCM myocardium results from reduced function of individual myosin molecules or from decreased myosin availability to interact with actin due to increased IHM/SRX stabilization. We used an established DCM myosin mutation, E525K, and characterized the biochemical and mechanical activity of wild-type and mutant human β-cardiac myosin constructs that differed in the length of their coiled-coil tail, which dictates their ability to form the IHM/SRX state. We found that short-tailed myosin constructs exhibited low IHM/SRX content, elevated actin-activated ATPase activity, and fast velocities in unloaded motility assays. Conversely, longer-tailed constructs exhibited higher IHM/SRX content and reduced actomyosin ATPase and velocity. Our modeling suggests that reduced velocities may be attributed to IHM/SRX-dependent sequestration of myosin heads. Interestingly, longer-tailed E525K mutants showed no apparent impact on velocity or actomyosin ATPase at low ionic strength but stabilized IHM/SRX state at higher ionic strength. Therefore, the hypo-contractility observed in DCM may be attributable to reduced myosin head availability caused by enhanced IHM/SRX stability in E525K mutants.
Collapse
Affiliation(s)
- Sebastian Duno-Miranda
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Shane R. Nelson
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - David V. Rasicci
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Skylar M.L. Bodt
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Joseph A. Cirilo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Duha Vang
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Sivaraj Sivaramakrishnan
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - David M. Warshaw
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| |
Collapse
|
26
|
Parodi A, Puscas T, Réant P, Donal E, M'Barek Raboudi D, Billon C, Bacher A, El Hachmi M, Wahbi K, Jeunemaître X, Hagège A. Target population for a selective cardiac myosin inhibitor in hypertrophic obstructive cardiomyopathy: Real-life estimation from the French register of hypertrophic cardiomyopathy (REMY). Arch Cardiovasc Dis 2024; 117:427-432. [PMID: 38762345 DOI: 10.1016/j.acvd.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND The efficacy of current pharmacological therapies in hypertrophic cardiomyopathy is limited. A cardiac myosin inhibitor, mavacamten, has recently been approved as a first-in-class treatment for symptomatic hypertrophic obstructive cardiomyopathy. AIMS To assess the profile and burden of cardiac myosin inhibitor candidates in the hypertrophic cardiomyopathy prospective Register of hypertrophic cardiomyopathy (REMY) held by the French Society of Cardiology. METHODS Data were collected at baseline and during follow-up from patients with hypertrophic cardiomyopathy enrolled in REMY by the three largest participating centres. RESULTS Among 1059 adults with hypertrophic cardiomyopathy, 461 (43.5%) had obstruction; 325 (30.7%) of these were also symptomatic, forming the "cardiac myosin inhibitor candidates" group. Baseline features of this group were: age 58±15years; male sex (n=196; 60.3%); diagnosis-to-inclusion delay 5 (1-12)years; maximum wall thickness 20±6mm; left ventricular ejection fraction 69±6%; family history of hypertrophic cardiomyopathy or sudden cardiac death (n=133; 40.9%); presence of a pathogenic sarcomere gene mutation (n=101; 31.1%); beta-blocker or verapamil treatment (n=304; 93.8%), combined with disopyramide (n=28; 8.7%); and eligibility for septal reduction therapy (n=96; 29%). At the end of a median follow-up of 66 (34-106) months, 319 (98.2%) were treated for obstruction (n=43 [13.2%] received disopyramide), 46 (14.2%) underwent septal reduction therapy and the all-cause mortality rate was 1.9/100 person-years (95% confidence interval 1.4-2.6) (46 deaths). Moreover, 41 (8.9%) patients from the initial hypertrophic obstructive cardiomyopathy group became eligible for a cardiac myosin inhibitor. CONCLUSIONS In this cohort of patients with hypertrophic cardiomyopathy selected from the REMY registry, one third were eligible for a cardiac myosin inhibitor.
Collapse
Affiliation(s)
- Alessandro Parodi
- Département de cardiologie, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France; Università del Piemonte Orientale Amedeo Avogadro, 13100 Vercelli, Italy
| | - Tania Puscas
- Département de cardiologie, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France
| | - Patricia Réant
- Département de cardiologie, hôpital Haut-Lévêque, CHU de Bordeaux, université de Bordeaux, Inserm 1045, IHU Lyric, CIC 1401, 33600 Pessac, France
| | - Erwan Donal
- Service de cardiologie, hôpital Pontchaillou, CHU de Rennes, université de Rennes, Inserm, LTSI-UMR 1099, 35000 Rennes, France
| | - Dorra M'Barek Raboudi
- Département de cardiologie, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France
| | - Clarisse Billon
- Département de cardiologie, hôpital Haut-Lévêque, CHU de Bordeaux, université de Bordeaux, Inserm 1045, IHU Lyric, CIC 1401, 33600 Pessac, France
| | - Anne Bacher
- Département de cardiologie, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France
| | - Mohamed El Hachmi
- Département de génétique, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France; Molecular Medicine, La Sapienza University, 00185 Rome, Italy
| | - Karim Wahbi
- Département de génétique, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France; Inserm U970, Paris Cardiovascular Research Centre, Université Paris Cité, 75015 Paris, France
| | - Xavier Jeunemaître
- Département de génétique, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France; Inserm U970, Paris Cardiovascular Research Centre, Université Paris Cité, 75015 Paris, France
| | - Albert Hagège
- Département de cardiologie, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France; Inserm U970, Paris Cardiovascular Research Centre, Université Paris Cité, 75015 Paris, France.
| |
Collapse
|
27
|
Steczina S, Mohran S, Bailey LRJ, McMillen TS, Kooiker KB, Wood NB, Davis J, Previs MJ, Olivotto I, Pioner JM, Geeves MA, Poggesi C, Regnier M. MYBPC3-c.772G>A mutation results in haploinsufficiency and altered myosin cycling kinetics in a patient induced stem cell derived cardiomyocyte model of hypertrophic cardiomyopathy. J Mol Cell Cardiol 2024; 191:27-39. [PMID: 38648963 PMCID: PMC11116032 DOI: 10.1016/j.yjmcc.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/13/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Approximately 40% of hypertrophic cardiomyopathy (HCM) mutations are linked to the sarcomere protein cardiac myosin binding protein-C (cMyBP-C). These mutations are either classified as missense mutations or truncation mutations. One mutation whose nature has been inconsistently reported in the literature is the MYBPC3-c.772G > A mutation. Using patient-derived human induced pluripotent stem cells differentiated to cardiomyocytes (hiPSC-CMs), we have performed a mechanistic study of the structure-function relationship for this MYBPC3-c.772G > A mutation versus a mutation corrected, isogenic cell line. Our results confirm that this mutation leads to exon skipping and mRNA truncation that ultimately suggests ∼20% less cMyBP-C protein (i.e., haploinsufficiency). This, in turn, results in increased myosin recruitment and accelerated myofibril cycling kinetics. Our mechanistic studies suggest that faster ADP release from myosin is a primary cause of accelerated myofibril cross-bridge cycling due to this mutation. Additionally, the reduction in force generating heads expected from faster ADP release during isometric contractions is outweighed by a cMyBP-C phosphorylation mediated increase in myosin recruitment that leads to a net increase of myofibril force, primarily at submaximal calcium activations. These results match well with our previous report on contractile properties from myectomy samples of the patients from whom the hiPSC-CMs were generated, demonstrating that these cell lines are a good model to study this pathological mutation and extends our understanding of the mechanisms of altered contractile properties of this HCM MYBPC3-c.772G > A mutation.
Collapse
Affiliation(s)
- Sonette Steczina
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Saffie Mohran
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Logan R J Bailey
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Molecular and Cellular Biology, University of Washington, Seattle, WA 98109, USA; Department of Lab Medicine and Pathology, University of Washington, Seattle, WA 98109, USA
| | - Timothy S McMillen
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA
| | - Kristina B Kooiker
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Neil B Wood
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05404, USA
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Lab Medicine and Pathology, University of Washington, Seattle, WA 98109, USA
| | - Michael J Previs
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05404, USA
| | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, Division of Physiology, University of Florence, Italy
| | | | | | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, Division of Physiology, University of Florence, Italy
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
28
|
Spudich JA. One must reconstitute the functions of interest from purified proteins. Front Physiol 2024; 15:1390186. [PMID: 38827995 PMCID: PMC11140241 DOI: 10.3389/fphys.2024.1390186] [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: 02/22/2024] [Accepted: 04/01/2024] [Indexed: 06/05/2024] Open
Abstract
I am often asked by students and younger colleagues and now by the editors of this issue to tell the history of the development of the in vitro motility assay and the dual-beam single-molecule laser trap assay for myosin-driven actin filament movement, used widely as key assays for understanding how both muscle and nonmuscle myosin molecular motors work. As for all discoveries, the history of the development of the myosin assays involves many people who are not authors of the final publications, but without whom the assays would not have been developed as they are. Also, early experiences shape how one develops ideas and experiments, and influence future discoveries in major ways. I am pleased here to trace my own path and acknowledge the many individuals involved and my early science experiences that led to the work I and my students, postdoctoral fellows, and sabbatical visitors did to develop these assays. Mentors are too often overlooked in historical descriptions of discoveries, and my story starts with those who mentored me.
Collapse
Affiliation(s)
- James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
29
|
Chakraborti A, Tardiff JC, Schwartz SD. Myosin-Catalyzed ATP Hydrolysis in the Presence of Disease-Causing Mutations: Mavacamten as a Way to Repair Mechanism. J Phys Chem B 2024; 128:4716-4727. [PMID: 38708944 PMCID: PMC11103257 DOI: 10.1021/acs.jpcb.4c01601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Hypertrophic cardiomyopathy is one of the most common forms of genetic cardiomyopathy. Mavacamten is a first-in-class myosin modulator that was identified via activity screening on the wild type, and it is FDA-approved for the treatment of obstructive hypertrophic cardiomyopathy (HCM). The drug selectively binds to the cardiac β-myosin, inhibiting myosin function to decrease cardiac contractility. Though the drug is thought to affect multiple steps of the myosin cross-bridge cycle, its detailed mechanism of action is still under investigation. Individual steps in the overall cross-bridge cycle must be queried to elucidate the full mechanism of action. In this study, we utilize the rare-event method of transition path sampling to generate reactive trajectories to gain insights into the action of the drug on the dynamics and rate of the ATP hydrolysis step for human cardiac β-myosin. We study three known HCM causative myosin mutations: R453C, P710R, and R712L to observe the effect of the drug on the alterations caused by these mutations in the chemical step. Since the crystal structure of the drug-bound myosin was not available at the time of this work, we created a model of the drug-bound system utilizing a molecular docking approach. We find a significant effect of the drug in one case, where the actual mechanism of the reaction is altered from the wild type by mutation. The drug restores both the rate of hydrolysis to the wildtype level and the mechanism of the reaction. This is a way to check the effect of the drug on untested mutations.
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
|
30
|
Solaro RJ, Goldspink PH, Wolska BM. Emerging Concepts of Mechanisms Controlling Cardiac Tension: Focus on Familial Dilated Cardiomyopathy (DCM) and Sarcomere-Directed Therapies. Biomedicines 2024; 12:999. [PMID: 38790961 PMCID: PMC11117855 DOI: 10.3390/biomedicines12050999] [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: 03/13/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Novel therapies for the treatment of familial dilated cardiomyopathy (DCM) are lacking. Shaping research directions to clinical needs is critical. Triggers for the progression of the disorder commonly occur due to specific gene variants that affect the production of sarcomeric/cytoskeletal proteins. Generally, these variants cause a decrease in tension by the myofilaments, resulting in signaling abnormalities within the micro-environment, which over time result in structural and functional maladaptations, leading to heart failure (HF). Current concepts support the hypothesis that the mutant sarcomere proteins induce a causal depression in the tension-time integral (TTI) of linear preparations of cardiac muscle. However, molecular mechanisms underlying tension generation particularly concerning mutant proteins and their impact on sarcomere molecular signaling are currently controversial. Thus, there is a need for clarification as to how mutant proteins affect sarcomere molecular signaling in the etiology and progression of DCM. A main topic in this controversy is the control of the number of tension-generating myosin heads reacting with the thin filament. One line of investigation proposes that this number is determined by changes in the ratio of myosin heads in a sequestered super-relaxed state (SRX) or in a disordered relaxed state (DRX) poised for force generation upon the Ca2+ activation of the thin filament. Contrasting evidence from nanometer-micrometer-scale X-ray diffraction in intact trabeculae indicates that the SRX/DRX states may have a lesser role. Instead, the proposal is that myosin heads are in a basal OFF state in relaxation then transfer to an ON state through a mechano-sensing mechanism induced during early thin filament activation and increasing thick filament strain. Recent evidence about the modulation of these mechanisms by protein phosphorylation has also introduced a need for reconsidering the control of tension. We discuss these mechanisms that lead to different ideas related to how tension is disturbed by levels of mutant sarcomere proteins linked to the expression of gene variants in the complex landscape of DCM. Resolving the various mechanisms and incorporating them into a unified concept is crucial for gaining a comprehensive understanding of DCM. This deeper understanding is not only important for diagnosis and treatment strategies with small molecules, but also for understanding the reciprocal signaling processes that occur between cardiac myocytes and their micro-environment. By unraveling these complexities, we can pave the way for improved therapeutic interventions for managing DCM.
Collapse
Affiliation(s)
- R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
| | - Paul H. Goldspink
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
| | - Beata M. Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
31
|
Buvoli M, Wilson GC, Buvoli A, Gugel JF, Hau A, Bönnemann CG, Paradas C, Ryba DM, Woulfe KC, Walker LA, Buvoli T, Ochala J, Leinwand LA. A Laing distal myopathy-associated proline substitution in the β-myosin rod perturbs myosin cross-bridging activity. J Clin Invest 2024; 134:e172599. [PMID: 38690726 PMCID: PMC11060730 DOI: 10.1172/jci172599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/11/2024] [Indexed: 05/03/2024] Open
Abstract
Proline substitutions within the coiled-coil rod region of the β-myosin gene (MYH7) are the predominant mutations causing Laing distal myopathy (MPD1), an autosomal dominant disorder characterized by progressive weakness of distal/proximal muscles. We report that the MDP1 mutation R1500P, studied in what we believe to be the first mouse model for the disease, adversely affected myosin motor activity despite being in the structural rod domain that directs thick filament assembly. Contractility experiments carried out on isolated mutant muscles, myofibrils, and myofibers identified muscle fatigue and weakness phenotypes, an increased rate of actin-myosin detachment, and a conformational shift of the myosin heads toward the more reactive disordered relaxed (DRX) state, causing hypercontractility and greater ATP consumption. Similarly, molecular analysis of muscle biopsies from patients with MPD1 revealed a significant increase in sarcomeric DRX content, as observed in a subset of myosin motor domain mutations causing hypertrophic cardiomyopathy. Finally, oral administration of MYK-581, a small molecule that decreases the population of heads in the DRX configuration, significantly improved the limited running capacity of the R1500P-transgenic mice and corrected the increased DRX state of the myofibrils from patients. These studies provide evidence of the molecular pathogenesis of proline rod mutations and lay the groundwork for the therapeutic advancement of myosin modulators.
Collapse
Affiliation(s)
- Massimo Buvoli
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Genevieve C.K. Wilson
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Ada Buvoli
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Jack F. Gugel
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Abbi Hau
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, and
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, Guy’s Campus, King’s College London, London, United Kingdom
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, Maryland, USA
| | - Carmen Paradas
- Neuromuscular Unit, Department of Neurology, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | | | - Kathleen C. Woulfe
- Division of Cardiology, Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - Lori A. Walker
- Division of Cardiology, Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - Tommaso Buvoli
- Department of Mathematics, Tulane University, New Orleans, Louisiana, USA
| | - Julien Ochala
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, and
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, Guy’s Campus, King’s College London, London, United Kingdom
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leslie A. Leinwand
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
32
|
Li J, Fang J, Liu Y, Wei X. Apical hypertrophic cardiomyopathy: pathophysiology, diagnosis and management. Clin Res Cardiol 2024; 113:680-693. [PMID: 37982860 PMCID: PMC11026226 DOI: 10.1007/s00392-023-02328-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/18/2023] [Indexed: 11/21/2023]
Abstract
Since the first description of apical hypertrophic cardiomyopathy (ApHCM) in 1976, contrasting information from all over the world has emerged regarding the natural history of the disease. However, the recommended guidelines on hypertrophic cardiomyopathy (HCM) pay a cursory reference to ApHCM, without ApHCM-specific recommendations to guide the diagnosis and management. In addition, cardiologists may not be aware of certain aspects that are specific to this disease subtype, and a robust understanding of specific disease features can facilitate recognition and timely diagnosis. Therefore, the review covers the incidence, pathogenesis, and characteristics of ApHCM and imaging methods. Echocardiography and cardiovascular magnetic resonance imaging (CMR) are the most commonly used imaging methods. Moreover, this review presents the management strategies of this heterogeneous clinical entity. In this review, we introduce a novel transapical beating-heart septal myectomy procedure for ApHCM patients with a promising short-time result.
Collapse
Affiliation(s)
- Jiangtao Li
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Jing Fang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Yani Liu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
| | - Xiang Wei
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.
- NHC Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China.
| |
Collapse
|
33
|
Haraf R, Habib H, Masri A. The Revolution of Cardiac Myosin Inhibitors in Patients With Hypertrophic Cardiomyopathy. Can J Cardiol 2024; 40:800-819. [PMID: 38280487 DOI: 10.1016/j.cjca.2024.01.022] [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: 11/02/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/29/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiomyopathy worldwide and causes significant morbidity and mortality. For decades, medical treatment options have been limited and untargeted, with frequent need for invasive interventions not readily accessible to many HCM patients. More recently, our understanding of the genetic basis and pathophysiologic mechanism of HCM has grown significantly, leading to the discovery of a new class of medications, cardiac myosin inhibitors (CMIs), that shift myosin into the super-relaxed state to counteract the hypercontractility in HCM. Subsequent clinical trials have proven the mechanism and efficacy of CMIs in humans with obstructive HCM, and additional trials are under way in patients with nonobstructive HCM. With favourable results in the completed clinical trials and ongoing research on the horizon, CMIs represent a bright new era in the targeted management of HCM. This review is focused on the discovery of CMIs, provides a summary of the results of clinical trials to date, provides clinicians with a roadmap for implementing CMIs into practice, and identifies gaps in our current understanding as well as areas of ongoing investigation.
Collapse
Affiliation(s)
- Rebecca Haraf
- The Hypertrophic Cardiomyopathy Center, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Hany Habib
- The Hypertrophic Cardiomyopathy Center, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Ahmad Masri
- The Hypertrophic Cardiomyopathy Center, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
34
|
Vyas R, Panchal V, Jain S, Sondhi M, Singh M, Jaisingh K, Thotamgari SR, Thakre A, Modi K. Evaluating the efficacy and safety of mavacamten in hypertrophic cardiomyopathy: A systematic review and meta-analysis focusing on qualitative assessment, biomarkers, and cardiac imaging. PLoS One 2024; 19:e0301704. [PMID: 38635724 PMCID: PMC11025865 DOI: 10.1371/journal.pone.0301704] [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: 12/16/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Hypertrophic Cardiomyopathy (HCM) is a complex cardiac condition characterized by hypercontractility of cardiac muscle leading to a dynamic obstruction of left ventricular outlet tract (LVOT). Mavacamten, a first-in-class cardiac myosin inhibitor, is increasingly being studied in randomized controlled trials. In this meta-analysis, we aimed to analyse the efficacy and safety profile of Mavacamten compared to placebo in patients of HCM. METHOD We carried out a comprehensive search in PubMed, Cochrane, and clinicaltrials.gov to analyze the efficacy and safety of mavacamten compared to placebo from 2010 to 2023. To calculate pooled odds ratio (OR) or risk ratio (RR) at 95% confidence interval (CI), the Mantel-Haenszel formula with random effect was used and Generic Inverse Variance method assessed pooled mean difference value at a 95% CI. RevMan was used for analysis. P<0.05 was considered significant. RESULTS We analyzed five phase 3 RCTs including 609 patients to compare mavacamten with a placebo. New York Heart Association (NYHA) grade improvement and KCCQ score showed the odds ratio as 4.94 and 7.93 with p<0.00001 at random effect, respectively. Cardiac imaging which included LAVI, LVOT at rest, LVOT post valsalva, LVOT post-exercise, and reduction in LVEF showed the pooled mean differences for change as -5.29, -49.72, -57.45, -36.11, and -3.00 respectively. Changes in LVEDV and LVMI were not statistically significant. The pooled mean difference for change in NT-proBNP and Cardiac troponin-I showed 0.20 and 0.57 with p<0.00001. The efficacy was evaluated in 1) A composite score, which was defined as either 1·5 mL/kg per min or greater increase in peak oxygen consumption (pVO2) and at least one NYHA class reduction, or a 3·0 mL/kg per min or greater pVO2 increase without NYHA class worsening and 2) changes in pVO2, which was not statistically significant. Similarly, any treatment-associated emergent adverse effects (TEAE), treatment-associated serious adverse effects (TSAE), and cardiac-related adverse effects were not statistically significant. CONCLUSION Mavacamten influences diverse facets of HCM comprehensively. Notably, our study delved into the drug's impact on the heart's structural and functional aspects, providing insights that complement prior findings. Further large-scale trials are needed to evaluate the safety profile of Mavacamten.
Collapse
Affiliation(s)
- Rahul Vyas
- Department of Internal Medicine, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Viraj Panchal
- Department of Medicine, Smt. NHL Municipal Medical College and SVPISMR, Ahmedabad, Gujarat, India
| | - Shubhika Jain
- Department of Medicine, Kasturba Medical College, Manipal, Karnataka, India
| | - Manush Sondhi
- Department of Internal Medicine, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Mansunderbir Singh
- Department of Internal Medicine, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Keerthish Jaisingh
- Department of Cardiology, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Sahith Reddy Thotamgari
- Department of Cardiology, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Anuj Thakre
- Department of Internal Medicine, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Kalgi Modi
- Department of Cardiology, Louisiana State University, Shreveport, Louisiana, United States of America
| |
Collapse
|
35
|
Masri A, Lester SJ, Stendahl JC, Hegde SM, Sehnert AJ, Balaratnam G, Shah A, Fox S, Wang A. Long-Term Safety and Efficacy of Mavacamten in Symptomatic Obstructive Hypertrophic Cardiomyopathy: Interim Results of the PIONEER-OLE Study. J Am Heart Assoc 2024; 13:e030607. [PMID: 38591260 PMCID: PMC11262496 DOI: 10.1161/jaha.123.030607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/16/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND The phase 2 PIONEER-HCM (Phase 2 Open-label Pilot Study Evaluating Mavacamten in Subjects With Symptomatic Hypertrophic Cardiomyopathy and Left Ventricular Outflow Tract Obstruction) study showed that mavacamten improved left ventricular outflow tract gradients, exercise capacity, and symptoms in patients with obstructive hypertrophic cardiomyopathy (HCM), but the results of longer-term treatment are less well described. We report interim results from the PIONEER-OLE (PIONEER Open-Label Extension) study, the longest-term study of mavacamten in patients with symptomatic obstructive HCM. METHODS AND RESULTS Patients who previously completed PIONEER-HCM (n=20) were eligible to enroll in PIONEER-OLE. Patients received oral mavacamten, 5 mg once daily (starting dose), with individualized dose titration at week 6. Evaluations included serial monitoring of safety, echocardiography, Kansas City Cardiomyopathy Questionnaire-Overall Summary Score, and serum NT-proBNP (N-terminal pro-B-type natriuretic peptide) levels. Thirteen patients enrolled and received mavacamten (median study duration at data cutoff, 201 weeks). Most patients (92.3%) received β-blockers concomitantly. Treatment-emergent adverse events were predominantly mild/moderate. One patient had an isolated reduction in left ventricular ejection fraction to 47%, which recovered and remained normal with continued treatment at a reduced dose. At week 180, mavacamten was associated with New York Heart Association class improvements from baseline (class II to I, n=9; class III to II, n=1; and unchanged, n=2), sustained reductions in left ventricular outflow tract gradients (mean [SD] change from baseline: resting, -50 [55] mm Hg; Valsalva, -70 [41] mm Hg), and serum NT-proBNP levels (median [interquartile range] change from baseline: -498 [-2184 to -76] ng/L), and improved Kansas City Cardiomyopathy Questionnaire-Overall Summary Score (mean [SD] change from baseline: +17 [16]). CONCLUSIONS This long-term analysis supports the continued safety and effectiveness of mavacamten for >3 years in obstructive HCM. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03496168.
Collapse
Affiliation(s)
- Ahmad Masri
- Division of Cardiology, Hypertrophic Cardiomyopathy Center, School of MedicineOregon Health & Science UniversityPortlandOR
| | - Steven J. Lester
- Department of Cardiovascular DiseasesMayo Clinic ArizonaPhoenixAZ
| | - John C. Stendahl
- Section of Cardiovascular Medicine, Department of Internal MedicineYale School of MedicineNew HavenCT
| | - Sheila M. Hegde
- Division of Cardiovascular MedicineBrigham and Women’s HospitalBostonMA
| | | | | | | | | | - Andrew Wang
- Duke CardiologyDuke University HospitalDurhamNC
| |
Collapse
|
36
|
Li M, Hu Y, Wang Q. Exploring the Super-Relaxed State of Human Cardiac β-Myosin by Molecular Dynamics Simulations. J Phys Chem B 2024; 128:3113-3120. [PMID: 38516963 DOI: 10.1021/acs.jpcb.3c07956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Human β-cardiac myosin plays a critical role in generating the mechanical forces necessary for cardiac muscle contraction. This process relies on a delicate dynamic equilibrium between the disordered relaxed state (DRX) and the super-relaxed state (SRX) of myosin. Disruptions in this equilibrium due to mutations can lead to heart diseases. However, the structural characteristics of SRX and the molecular mechanisms underlying pathogenic mutations have remained elusive. To bridge this gap, we conducted molecular dynamics simulations and free energy calculations to explore the conformational changes in myosin. Our findings indicate that the size of the phosphate-binding pocket can serve as a valuable metric for characterizing the transition from the DRX to SRX state. Importantly, we established a global dynamic coupling network within the myosin motor head at the residue level, elucidating how the pathogenic mutation E483K impacts the equilibrium between SRX and DRX through allosteric effects. Our work illuminates molecular details of SRX and offers valuable insights into disease treatment through the regulation of SRX.
Collapse
Affiliation(s)
- Mingwei Li
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yao Hu
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Qian Wang
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
37
|
Donkervoort S, van de Locht M, Ronchi D, Reunert J, McLean CA, Zaki M, Orbach R, de Winter JM, Conijn S, Hoomoedt D, Neto OLA, Magri F, Viaene AN, Foley AR, Gorokhova S, Bolduc V, Hu Y, Acquaye N, Napoli L, Park JH, Immadisetty K, Miles LB, Essawi M, McModie S, Ferreira LF, Zanotti S, Neuhaus SB, Medne L, ElBagoury N, Johnson KR, Zhang Y, Laing NG, Davis MR, Bryson-Richardson RJ, Hwee DT, Hartman JJ, Malik FI, Kekenes-Huskey PM, Comi GP, Sharaf-Eldin W, Marquardt T, Ravenscroft G, Bönnemann CG, Ottenheijm CAC. Pathogenic TNNI1 variants disrupt sarcomere contractility resulting in hypo- and hypercontractile muscle disease. Sci Transl Med 2024; 16:eadg2841. [PMID: 38569017 DOI: 10.1126/scitranslmed.adg2841] [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: 05/23/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
Troponin I (TnI) regulates thin filament activation and muscle contraction. Two isoforms, TnI-fast (TNNI2) and TnI-slow (TNNI1), are predominantly expressed in fast- and slow-twitch myofibers, respectively. TNNI2 variants are a rare cause of arthrogryposis, whereas TNNI1 variants have not been conclusively established to cause skeletal myopathy. We identified recessive loss-of-function TNNI1 variants as well as dominant gain-of-function TNNI1 variants as a cause of muscle disease, each with distinct physiological consequences and disease mechanisms. We identified three families with biallelic TNNI1 variants (F1: p.R14H/c.190-9G>A, F2 and F3: homozygous p.R14C), resulting in loss of function, manifesting with early-onset progressive muscle weakness and rod formation on histology. We also identified two families with a dominantly acting heterozygous TNNI1 variant (F4: p.R174Q and F5: p.K176del), resulting in gain of function, manifesting with muscle cramping, myalgias, and rod formation in F5. In zebrafish, TnI proteins with either of the missense variants (p.R14H; p.R174Q) incorporated into thin filaments. Molecular dynamics simulations suggested that the loss-of-function p.R14H variant decouples TnI from TnC, which was supported by functional studies showing a reduced force response of sarcomeres to submaximal [Ca2+] in patient myofibers. This contractile deficit could be reversed by a slow skeletal muscle troponin activator. In contrast, patient myofibers with the gain-of-function p.R174Q variant showed an increased force to submaximal [Ca2+], which was reversed by the small-molecule drug mavacamten. Our findings demonstrated that TNNI1 variants can cause muscle disease with variant-specific pathomechanisms, manifesting as either a hypo- or a hypercontractile phenotype, suggesting rational therapeutic strategies for each mechanism.
Collapse
Affiliation(s)
- Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Martijn van de Locht
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Dario Ronchi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, 20135, Italy
| | - Janine Reunert
- Department of General Pediatrics, University of Münster, Münster, 48149, Germany
| | - Catriona A McLean
- Department of Anatomical Pathology, Alfred Hospital, Melbourne, Victoria, 3004, Australia
- Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, 3168, Australia
| | - Maha Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Rotem Orbach
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Josine M de Winter
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Stefan Conijn
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Daan Hoomoedt
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Osorio Lopes Abath Neto
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Francesca Magri
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, 20122, Italy
| | - Angela N Viaene
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104 PA, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Svetlana Gorokhova
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Medical Genetics, Timone Children's Hospital, APHM, Marseille, 13005, France
- INSERM, U1251-MMG, Aix-Marseille Université, Marseille, 13009, France
| | - Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Acquaye
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura Napoli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Julien H Park
- Department of General Pediatrics, University Hospital Münster, Münster, 48149 Germany
| | - Kalyan Immadisetty
- Department of Cell and Molecular Physiology, Loyola University, Chicago, IL 60153, USA
| | - Lee B Miles
- School of Biological Sciences, Monash University, Melbourne, Victoria, 3800, Australia
| | - Mona Essawi
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Salar McModie
- Department of Neurology, Alfred Health, Melbourne, Victoria, 3004, Australia
| | - Leonardo F Ferreira
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Simona Zanotti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Sarah B Neuhaus
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Livija Medne
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nagham ElBagoury
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Kory R Johnson
- Bioinformatics Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong Zhang
- Bioinformatics Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nigel G Laing
- Neurogenetics Unit, Department of Diagnostic Genomics, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
- Centre for Medical Research University of Western Australia, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Mark R Davis
- Neurogenetics Unit, Department of Diagnostic Genomics, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | | | - Darren T Hwee
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - James J Hartman
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Fady I Malik
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | | | - Giacomo Pietro Comi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, 20135, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Wessam Sharaf-Eldin
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Thorsten Marquardt
- Department of General Pediatrics, University of Münster, Münster, 48149, Germany
| | - Gianina Ravenscroft
- Centre for Medical Research University of Western Australia, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| |
Collapse
|
38
|
Garrido-Casado M, Asensio-Juárez G, Talayero VC, Vicente-Manzanares M. Engines of change: Nonmuscle myosin II in mechanobiology. Curr Opin Cell Biol 2024; 87:102344. [PMID: 38442667 DOI: 10.1016/j.ceb.2024.102344] [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: 10/07/2023] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 03/07/2024]
Abstract
The emergence of mechanobiology has unveiled complex mechanisms by which cells adjust intracellular force production to their needs. Most communicable intracellular forces are generated by myosin II, an actin-associated molecular motor that transforms adenosine triphosphate (ATP) hydrolysis into contraction in nonmuscle and muscle cells. Myosin II-dependent force generation is tightly regulated, and deregulation is associated with specific pathologies. Here, we focus on the role of myosin II (nonmuscle myosin II, NMII) in force generation and mechanobiology. We outline the regulation and molecular mechanism of force generation by NMII, focusing on the actual outcome of contraction, that is, force application to trigger mechanosensitive events or the building of dissipative structures. We describe how myosin II-generated forces drive two major types of events: modification of the cellular morphology and/or triggering of genetic programs, which enhance the ability of cells to adapt to, or modify, their microenvironment. Finally, we address whether targeting myosin II to impair or potentiate its activity at the motor level is a viable therapeutic strategy, as illustrated by recent examples aimed at modulating cardiac myosin II function in heart disease.
Collapse
Affiliation(s)
- Marina Garrido-Casado
- Molecular Mechanisms Program, Centro de Investigación del Cáncer/ Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, 37007 Salamanca, Spain
| | - Gloria Asensio-Juárez
- Molecular Mechanisms Program, Centro de Investigación del Cáncer/ Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, 37007 Salamanca, Spain
| | - Vanessa C Talayero
- Molecular Mechanisms Program, Centro de Investigación del Cáncer/ Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, 37007 Salamanca, Spain
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer/ Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
39
|
Negri F, Sanna GD, Di Giovanna G, Cittar M, Grilli G, De Luca A, Dal Ferro M, Baracchini N, Burelli M, Paldino A, Del Franco A, Pradella S, Todiere G, Olivotto I, Imazio M, Sinagra G, Merlo M. Cardiac Magnetic Resonance Feature-Tracking Identifies Preclinical Abnormalities in Hypertrophic Cardiomyopathy Sarcomere Gene Mutation Carriers. Circ Cardiovasc Imaging 2024; 17:e016042. [PMID: 38563190 DOI: 10.1161/circimaging.123.016042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/05/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Assessing myocardial strain by cardiac magnetic resonance feature tracking (FT) has been found to be useful in patients with overt hypertrophic cardiomyopathy (HCM). Little is known, however, of its role in sarcomere gene mutation carriers without overt left ventricular hypertrophy (subclinical HCM). METHODS Thirty-eight subclinical HCM subjects and 42 healthy volunteers were enrolled in this multicenter case-control study. They underwent a comprehensive cardiac magnetic resonance study. Two-dimensional global radial, circumferential, and longitudinal strain of the left ventricle (LV) were evaluated by FT analysis. RESULTS The subclinical HCM sample was 41 (22-51) years old and 32% were men. FT analysis revealed a reduction in global radial strain (29±7.2 versus 47.9±7.4; P<0.0001), global circumferential strain (-17.3±2.6 -versus -20.8±7.4; P<0.0001) and global longitudinal strain (-16.9±2.4 versus -20.5±2.6; P<0.0001) in subclinical HCM compared with control subjects. The significant differences persisted when considering the 23 individuals free of all the structural and functional ECG and cardiac magnetic resonance abnormalities previously described. Receiver operating characteristic curve analyses showed that the differential diagnostic performances of FT in discriminating subclinical HCM from normal subjects were good to excellent (global radial strain with optimal cut-off value of 40.43%: AUC, 0.946 [95% CI, 0.93-1.00]; sensitivity 90.48%, specificity 94.44%; global circumferential strain with cut-off, -18.54%: AUC, 0.849 [95% CI, 0.76-0.94]; sensitivity, 88.10%; specificity, 72.22%; global longitudinal strain with cut-off, -19.06%: AUC, 0.843 [95% CI, 0.76-0.93]; sensitivity, 78.57%; specificity, 78.95%). Similar values were found for discriminating those subclinical HCM subjects without other phenotypic abnormalities from healthy volunteers (global radial strain with optimal cut-off 40.43%: AUC, 0.966 [95% CI, 0.92-1.00]; sensitivity, 90.48%; specificity, 95.45%; global circumferential strain with cut-off, -18.44%: AUC, 0.866 [95% CI, 0.76-0.96]; sensitivity, 92.86%; specificity, 77.27%; global longitudinal strain with cut-off, -17.32%: AUC, 0.838 [95% CI, 0.73-0.94]; sensitivity, 90.48%; specificity, 65.22%). CONCLUSIONS Cardiac magnetic resonance FT-derived parameters are consistently lower in subclinical patients with HCM, and they could emerge as a good tool for discovering the disease during a preclinical phase.
Collapse
Affiliation(s)
- Francesco Negri
- Cardiology Department, University Hospital "Santa Maria della Misericordia," Azienda Sanitaria Universitaria Integrata Friuli Centrale, Udine, Italy (F.N., M.I.)
| | | | - Giulia Di Giovanna
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Marco Cittar
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Giulia Grilli
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Antonio De Luca
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Matteo Dal Ferro
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Nikita Baracchini
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Massimo Burelli
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Alessia Paldino
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Annamaria Del Franco
- Cardiomyopathy Unit, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy (A.D.F., I.O.)
| | - Silvia Pradella
- Department of Emergency Radiology, University Hospital Careggi, Florence (Italy) (S.P.)
| | | | - Iacopo Olivotto
- Cardiomyopathy Unit, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy (A.D.F., I.O.)
- Department of Experimental and Clinical Medicine, Meyer Children's Hospital, University of Florence, Italy (I.O.)
| | - Massimo Imazio
- Cardiology Department, University Hospital "Santa Maria della Misericordia," Azienda Sanitaria Universitaria Integrata Friuli Centrale, Udine, Italy (F.N., M.I.)
- Department of Medicine, University of Udine, Italy (M.I.)
| | - Gianfranco Sinagra
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| | - Marco Merlo
- Cardiovascular Department, Centre for Diagnosis and Management of Cardiomyopathies, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Italy (G.d.G., M.C., G.G., A.D.L., M.d.F., N.B., M.B., A.P., G.S., M.M.)
| |
Collapse
|
40
|
Kelly CM, Martin JL, Previs MJ. Myosin folding boosts solubility in cardiac muscle sarcomeres. JCI Insight 2024; 9:e178131. [PMID: 38483507 PMCID: PMC11141871 DOI: 10.1172/jci.insight.178131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/05/2024] [Indexed: 04/23/2024] Open
Abstract
The polymerization of myosin molecules into thick filaments in muscle sarcomeres is essential for cardiac contractility, with the attenuation of interactions between the heads of myosin molecules within the filaments being proposed to result in hypercontractility, as observed in hypertrophic cardiomyopathy (HCM). However, experimental evidence demonstrates that the structure of these giant macromolecular complexes is highly dynamic, with molecules exchanging between the filaments and a pool of soluble molecules on the minute timescale. Therefore, we sought to test the hypothesis that the enhancement of interactions between the heads of myosin molecules within thick filaments limits the mobility of myosin by taking advantage of mavacamten, a small molecule approved for the treatment of HCM. Myosin molecules were labeled in vivo with a green fluorescent protein (GFP) and imaged in intact hearts using multiphoton microscopy. Treatment of the intact hearts with mavacamten resulted in an unexpected > 5-fold enhancement in GFP-myosin mobility within the sarcomere. In vitro biochemical assays suggested that mavacamten enhanced the mobility of GFP-myosin by increasing the solubility of myosin molecules, through the stabilization of a compact/folded conformation of the molecules, once disassociated from the thick filaments. These findings provide alternative insight into the mechanisms by which molecules exchange into and out of thick filaments and have implications for how mavacamten may affect cardiac contractility.
Collapse
Affiliation(s)
- Colleen M Kelly
- Molecular Physiology and Biophysics Department, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Jody L Martin
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Michael J Previs
- Molecular Physiology and Biophysics Department, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
41
|
Squarci C, Campbell KS. Myosins may know when to hold and when to fold. Biophys J 2024; 123:525-526. [PMID: 38297835 PMCID: PMC10938075 DOI: 10.1016/j.bpj.2024.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/02/2024] Open
Affiliation(s)
- Caterina Squarci
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, Kentucky
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
42
|
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
|
43
|
Doh CY, Schmidt AV, Chinthalapudi K, Stelzer JE. Bringing into focus the central domains C3-C6 of myosin binding protein C. Front Physiol 2024; 15:1370539. [PMID: 38487262 PMCID: PMC10937550 DOI: 10.3389/fphys.2024.1370539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
Myosin binding protein C (MyBPC) is a multi-domain protein with each region having a distinct functional role in muscle contraction. The central domains of MyBPC have often been overlooked due to their unclear roles. However, recent research shows promise in understanding their potential structural and regulatory functions. Understanding the central region of MyBPC is important because it may have specialized function that can be used as drug targets or for disease-specific therapies. In this review, we provide a brief overview of the evolution of our understanding of the central domains of MyBPC in regard to its domain structures, arrangement and dynamics, interaction partners, hypothesized functions, disease-causing mutations, and post-translational modifications. We highlight key research studies that have helped advance our understanding of the central region. Lastly, we discuss gaps in our current understanding and potential avenues to further research and discovery.
Collapse
Affiliation(s)
- Chang Yoon Doh
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Alexandra V. Schmidt
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
44
|
Liu C, Karabina A, Meller A, Bhattacharjee A, Agostino CJ, Bowman GR, Ruppel KM, Spudich JA, Leinwand LA. Homologous mutations in human β, embryonic, and perinatal muscle myosins have divergent effects on molecular power generation. Proc Natl Acad Sci U S A 2024; 121:e2315472121. [PMID: 38377203 PMCID: PMC10907259 DOI: 10.1073/pnas.2315472121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 01/12/2024] [Indexed: 02/22/2024] Open
Abstract
Mutations at a highly conserved homologous residue in three closely related muscle myosins cause three distinct diseases involving muscle defects: R671C in β-cardiac myosin causes hypertrophic cardiomyopathy, R672C and R672H in embryonic skeletal myosin cause Freeman-Sheldon syndrome, and R674Q in perinatal skeletal myosin causes trismus-pseudocamptodactyly syndrome. It is not known whether their effects at the molecular level are similar to one another or correlate with disease phenotype and severity. To this end, we investigated the effects of the homologous mutations on key factors of molecular power production using recombinantly expressed human β, embryonic, and perinatal myosin subfragment-1. We found large effects in the developmental myosins but minimal effects in β myosin, and magnitude of changes correlated partially with clinical severity. The mutations in the developmental myosins dramatically decreased the step size and load-sensitive actin-detachment rate of single molecules measured by optical tweezers, in addition to decreasing overall enzymatic (ATPase) cycle rate. In contrast, the only measured effect of R671C in β myosin was a larger step size. Our measurements of step size and bound times predicted velocities consistent with those measured in an in vitro motility assay. Finally, molecular dynamics simulations predicted that the arginine to cysteine mutation in embryonic, but not β, myosin may reduce pre-powerstroke lever arm priming and ADP pocket opening, providing a possible structural mechanism consistent with the experimental observations. This paper presents direct comparisons of homologous mutations in several different myosin isoforms, whose divergent functional effects are a testament to myosin's highly allosteric nature.
Collapse
Affiliation(s)
- Chao Liu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA94550
| | - Anastasia Karabina
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO80309
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO80309
- Kainomyx, Inc., Palo Alto, CA94304
| | - Artur Meller
- Department of Biochemistry and Biophysics, Washington University in St. Louis, St. Louis, MO63110
- Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO63110
| | - Ayan Bhattacharjee
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Colby J. Agostino
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Greg R. Bowman
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Kathleen M. Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Kainomyx, Inc., Palo Alto, CA94304
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Kainomyx, Inc., Palo Alto, CA94304
| | - Leslie A. Leinwand
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO80309
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO80309
| |
Collapse
|
45
|
Chen L, Liu J, Rastegarpouyani H, Janssen PML, Pinto JR, Taylor KA. Structure of mavacamten-free human cardiac thick filaments within the sarcomere by cryoelectron tomography. Proc Natl Acad Sci U S A 2024; 121:e2311883121. [PMID: 38386705 PMCID: PMC10907299 DOI: 10.1073/pnas.2311883121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Heart muscle has the unique property that it can never rest; all cardiomyocytes contract with each heartbeat which requires a complex control mechanism to regulate cardiac output to physiological requirements. Changes in calcium concentration regulate the thin filament activation. A separate but linked mechanism regulates the thick filament activation, which frees sufficient myosin heads to bind the thin filament, thereby producing the required force. Thick filaments contain additional nonmyosin proteins, myosin-binding protein C and titin, the latter being the protein that transmits applied tension to the thick filament. How these three proteins interact to control thick filament activation is poorly understood. Here, we show using 3-D image reconstruction of frozen-hydrated human cardiac muscle myofibrils lacking exogenous drugs that the thick filament is structured to provide three levels of myosin activation corresponding to the three crowns of myosin heads in each 429Å repeat. In one crown, the myosin heads are almost completely activated and disordered. In another crown, many myosin heads are inactive, ordered into a structure called the interacting heads motif. At the third crown, the myosin heads are ordered into the interacting heads motif, but the stability of that motif is affected by myosin-binding protein C. We think that this hierarchy of control explains many of the effects of length-dependent activation as well as stretch activation in cardiac muscle control.
Collapse
Affiliation(s)
- Liang Chen
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL32306
| | - Jun Liu
- Microbial Sciences Institute, Yale University, West Haven, CT06516
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT06536
| | - Hosna Rastegarpouyani
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL32306
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Paul M. L. Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Jose R. Pinto
- Department of Biomedical Sciences, Florida State College of Medicine, Florida State University, Tallahassee, FL32306
| | - Kenneth A. Taylor
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL32306
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| |
Collapse
|
46
|
Ma W, del Rio CL, Qi L, Prodanovic M, Mijailovich S, Zambataro C, Gong H, Shimkunas R, Gollapudi S, Nag S, Irving TC. Myosin in autoinhibited off state(s), stabilized by mavacamten, can be recruited in response to inotropic interventions. Proc Natl Acad Sci U S A 2024; 121:e2314914121. [PMID: 38346202 PMCID: PMC10895252 DOI: 10.1073/pnas.2314914121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Mavacamten is a FDA-approved small-molecule therapeutic designed to regulate cardiac function at the sarcomere level by selectively but reversibly inhibiting the enzymatic activity of myosin. It shifts myosin toward ordered off states close to the thick filament backbone. It remains elusive whether these myosin heads in the off state(s) can be recruited in response to physiological stimuli when required to boost cardiac output. We show that cardiac myosins stabilized in these off state(s) by mavacamten are recruitable by 1) Ca2+, 2) increased chronotropy [heart rate (HR)], 3) stretch, and 4) β-adrenergic (β-AR) stimulation, all known physiological inotropic interventions. At the molecular level, we show that Ca2+ increases myosin ATPase activity by shifting mavacamten-stabilized myosin heads from the inactive super-relaxed state to the active disordered relaxed state. At the myofilament level, both Ca2+ and passive lengthening can shift mavacamten-ordered off myosin heads from positions close to the thick filament backbone to disordered on states closer to the thin filaments. In isolated rat cardiomyocytes, increased stimulation rates enhanced shortening fraction in mavacamten-treated cells. This observation was confirmed in vivo in telemetered rats, where left-ventricular dP/dtmax, an index of inotropy, increased with HR in mavacamten-treated animals. Finally, we show that β-AR stimulation in vivo increases left-ventricular function and stroke volume in the setting of mavacamten. Our data demonstrate that the mavacamten-promoted off states of myosin in the thick filament are at least partially activable, thus preserving cardiac reserve mechanisms.
Collapse
Affiliation(s)
- Weikang Ma
- Biophysics Collaborative Access Team, Department of Biology, Illinois Institute of Technology, Chicago, IL60616
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL60616
| | - Carlos L. del Rio
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA94005
- Cardiac Consulting, San Mateo, CA94010
| | - Lin Qi
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Momcilo Prodanovic
- Institute for Information Technologies, University of Kragujevac, Kragujevac34000, Serbia
- FilamenTech, Inc., Newtown, MA02458
| | | | | | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Rafael Shimkunas
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA94005
| | - Sampath Gollapudi
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA94005
| | - Suman Nag
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA94005
| | - Thomas C. Irving
- Biophysics Collaborative Access Team, Department of Biology, Illinois Institute of Technology, Chicago, IL60616
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL60616
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| |
Collapse
|
47
|
Braunwald E. Hypertrophic Cardiomyopathy: A Brief Overview. Am J Cardiol 2024; 212S:S1-S3. [PMID: 38368032 DOI: 10.1016/j.amjcard.2023.10.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 02/19/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is a complex, heterogeneous disorder that affects approximately 1 in every 500 persons worldwide and about 750,000 Americans. It is characterized by left ventricular hypertrophy that is usually asymmetric, with enlarged myocytes in disarray, unexplained by loading conditions. Obstruction to left ventricular outflow occurs in approximately 60% of patients. The natural history and cardiac morphology of HCM are quite heterogeneous. Although most patients with HCM are asymptomatic or mildly symptomatic, a minority are disabled by dyspnea, angina, or syncope, develop advanced heart failure, or die suddenly.
Collapse
Affiliation(s)
- Eugene Braunwald
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital; Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
48
|
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
|
49
|
Chen L, Luo Y, Zhang C, Liu X, Fang N, Wang X, Zhao X, Jiang J. Trifloxystrobin induced developmental toxicity by disturbing the ABC transporters, carbohydrate and lipid metabolism in adult zebrafish. CHEMOSPHERE 2024; 349:140747. [PMID: 38000556 DOI: 10.1016/j.chemosphere.2023.140747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
The environmental risks of trifloxystrobin (TR) have drawn attention because of its multiplex toxicity on aquatic organisms, but few studies have paid close attention to its chronic toxicity at environmental concentrations. In present study, histopathology, metabolomics and transcriptomics were comprehensively performed to investigate the toxic effects and biological responses on adult zebrafish after exposure to 0.1, 1 and 10 μg/L TR for 21 d. Results demonstrated long-term exposure of TR affected zebrafish liver, ovary and heart development. Metabolomics revealed 0.1, 1 and 10 μg/L TR simultaneously decreased the carbohydrates enriched in glucose metabolism and ABC transporters pathways, such as glycogen, lactose, lactulose, maltose, maltotriose, d-trehalose, while 1 μg/L and 10 μg/L TR significantly increased many metabolites related to glycerophospholipid and sphingolipid metabolism in zebrafish liver. Transcriptomics showed TR activated the transcription of the Abcb4, Abcb5 and Abcb11 involved in ABC transporters, Pck1, Pfk, Hk, Gyg1a and Pygma related to glucose metabolism, as well as the Lpcat1, Lpcat4, Gpat2, Cers and Sgms in glycerophospholipid and sphingolipid metabolism. Results further demonstrated high concentration of TR strongly affected the DNA repair system, while low dose of TR caused pronounced effects on cardiomyocytes and oocyte regulation pathways at transcriptional levels. The results indicated the abnormal liver, gonad and heart development caused by TR might be ascribed to the disturbance of carbohydrates and lipid metabolism mediating by the Abcb4, Abcb5 and Abcb11 ABC transporters, and long-term exposure of environmental concentration of TR was sufficient to affect zebrafish normal metabolism and development.
Collapse
Affiliation(s)
- Liping Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Yuqin Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Changpeng Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Xingang Liu
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Nan Fang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Xiangyun Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Xueping Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China
| | - Jinhua Jiang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory for Pesticide Residue Detection of Ministry of Agriculture, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, Zhejiang, China.
| |
Collapse
|
50
|
Liu X, Shu S. Suggesting Dictyostelium as a Model for Disease-Related Protein Studies through Myosin II Polymerization Pathway. Cells 2024; 13:263. [PMID: 38334655 PMCID: PMC10854627 DOI: 10.3390/cells13030263] [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: 11/23/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/10/2024] Open
Abstract
Dictyostelium myosin II displays remarkable dynamism within the cell, continually undergoing polymerization and depolymerization processes. Under low-ion conditions, it assumes a folded structure like muscle myosins and forms thick filaments through polymerization. In our study, we presented intermediate structures observed during the early stages of polymerization of purified myosin via negative staining electron microscopy, immediately crosslinked with glutaraldehyde at the onset of polymerization. We identified folded monomers, dimers, and tetramers in the process. Our findings suggest that Dictyostelium myosin II follows a polymerization pathway in vitro akin to muscle myosin, with folded monomers forming folded parallel and antiparallel dimers that subsequently associate to create folded tetramers. These folded tetramers eventually unfold and associate with other tetramers to produce long filaments. Furthermore, our research revealed that ATP influences filament size, reducing it regardless of the status of RLC phosphorylation while significantly increasing the critical polymerization concentrations from 0.2 to 9 nM. In addition, we demonstrate the morphology of fully matured Dictyostelium myosin II filaments.
Collapse
|