1
|
McDonald KS, Kalogeris TJ, Veteto AB, Davis DJ, Hanft LM. Myosin binding protein-C modulates loaded sarcomere shortening in rodent permeabilized cardiac myocytes. J Gen Physiol 2025; 157:e202413678. [PMID: 40126337 PMCID: PMC11932042 DOI: 10.1085/jgp.202413678] [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: 09/27/2024] [Revised: 01/29/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025] Open
Abstract
During the ejection phase of the cardiac cycle, left ventricular (LV) cardiac myocytes undergo loaded shortening and generate power. However, few studies have measured sarcomere shortening during loaded contractions. Here, we simultaneously monitored muscle length (ML) and sarcomere length (SL) during isotonic contractions in rodent permeabilized LV cardiac myocyte preparations. In permeabilized cardiac myocyte preparations from rats, we found that ML and SL traces were closely matched, as SL velocities were within ∼77% of ML velocities during half-maximal Ca2+ activations. We next tested whether cardiac myosin binding protein-C (cMyBP-C) regulates loaded shortening and power output by modulating cross-bridge availability. We characterized force-velocity and power-load relationships in wildtype (WT) and cMyBP-C deficient (Mybpc3-/-) mouse permeabilized cardiac myocyte preparations, at both the ML and SL level, before and after treatment with the small molecule myosin inhibitor, mavacamten. We found that SL traces closely matched ML traces in both WT and Mybpc3-/- cardiac myocytes. However, Mybpc3-/- cardiac myocytes exhibited disproportionately high sarcomere shortening velocities at high loads. Interestingly, in Mybpc3-/- cardiac myocytes, 0.5 µM mavacamten slowed SL-loaded shortening across the force-velocity curve and normalized SL shortening velocity at high loads. Overall, these results suggest that cMyBP-C moderates sarcomere-loaded shortening, especially at high loads, at least in part, by modulating cross-bridge availability.
Collapse
Affiliation(s)
- Kerry S. McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Theodore J. Kalogeris
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Adam B. Veteto
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Daniel J. Davis
- Department of Veterinary Pathology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Laurin M. Hanft
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
2
|
Hayes JB, Willet AH, Caplan LR, Knapik EW, Tyska MJ, Burnette DT. α- and β-myosin II can be non-uniformly distributed within the cardiac sarcomere. iScience 2025; 28:112233. [PMID: 40230528 PMCID: PMC11994913 DOI: 10.1016/j.isci.2025.112233] [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: 10/08/2024] [Revised: 01/29/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
In humans, the forces of the heartbeat are generated by both "fast" α-myosin II and "slow" β-myosin II molecular motors. Here, we report that α-myosin II and β-myosin II can take on an anisotropic arrangement within cardiac sarcomeres, the minimal contractile unit of cardiac muscle. In induced pluripotent stem cell (iPSC)-derived cardiac myocytes, we find that α-myosin II is distributed primarily within the central region of the filaments, being mostly absent from the distal ends, whereas β-myosin II is distributed along the entire filament. We measure that the outer bounds of α-myosin II in filament stacks are shorter than those of β-myosin II in both the zebrafish embryo and adult human myocardium. Our findings suggest that the two motors that drive the human heartbeat can have a specific and non-uniform arrangement within cardiac thick filaments, which may be thus divided into two distinct mechanical regions based on myosin II motor composition. We suggest potential roles for each motor during contraction-relaxation based on these distributions and recent literature.
Collapse
Affiliation(s)
- James B. Hayes
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Alaina H. Willet
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Leah R. Caplan
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Ela W. Knapik
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Dylan T. Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| |
Collapse
|
3
|
Ramadan MM, Alshawi AL, Mostafa YA, Al-Obeid MT, Elmahal M. Omecamtiv Mecarbil in Systolic Heart Failure: Clinical Efficacy and Future Directions of a Novel Myosin-Activating Inotropic Agent. Cureus 2025; 17:e82128. [PMID: 40357100 PMCID: PMC12067359 DOI: 10.7759/cureus.82128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Heart failure with reduced ejection fraction (HFrEF) remains a global health challenge, associated with high morbidity, mortality, and rising healthcare costs. Despite advances in guideline-directed therapy, many patients, especially those with severely reduced ejection fraction, remain symptomatic. Traditional inotropes, including β-agonists and phosphodiesterase inhibitors, are limited in chronic HFrEF due to risks of arrhythmias, calcium overload, and increased myocardial oxygen demand. Omecamtiv mecarbil (OM) is a novel cardiac myosin activator that enhances systolic contraction by increasing the efficiency of actin-myosin interactions. It prolongs systolic ejection time and improves stroke volume without elevating intracellular calcium or energy consumption. Although theoretical concerns exist regarding impaired diastolic filling, clinical trials have not confirmed such adverse effects in most patients. This narrative review discusses OM's pharmacologic profile, clinical trial data, and its potential as an adjunct therapy in advanced HFrEF. While not yet guideline-recommended, OM may benefit patients who remain symptomatic despite optimal treatment.
Collapse
Affiliation(s)
- Mahmoud M Ramadan
- Cardiology, Faculty of Medicine, Mansoura University, Mansoura, EGY
- Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, ARE
| | - Abdullah L Alshawi
- Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, ARE
| | - Yasmeen A Mostafa
- Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, ARE
| | | | - Mohammed Elmahal
- Diabetes and Endocrinology, Khartoum North Teaching Hospital, Khartoum, SDN
| |
Collapse
|
4
|
Landim-Vieira M, Nieto Morales PF, ElSafty S, Kahmini AR, Ranek MJ, Solís C. The role of mechanosignaling in the control of myocardial mass. Am J Physiol Heart Circ Physiol 2025; 328:H622-H638. [PMID: 39739566 DOI: 10.1152/ajpheart.00277.2024] [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: 04/30/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Regulation of myocardial mass is key for maintaining cardiovascular health. This review highlights the complex and regulatory relationship between mechanosignaling and myocardial mass, influenced by many internal and external factors including hemodynamic and microgravity, respectively. The heart is a dynamic organ constantly adapting to changes in workload (preload and afterload) and mechanical stress exerted on the myocardium, influencing both physiological adaptations and pathological remodeling. Mechanosignaling pathways, such as the mitogen-activated protein kinases (MAPKs) and the phosphoinositide 3-kinases and serine/threonine kinase (PI3K/Akt) pathways, mediate downstream effects on gene expression and play key roles in transducing mechanical cues into biochemical signals, thereby modulating cellular processes, including control of myocardial mass. Dysregulation of these processes can lead to pathological cardiac remodeling, such as hypertrophic cardiomyopathy. Furthermore, recent studies have highlighted the importance of protein quality control mechanisms, such as the ubiquitin-proteasome system, in settings of extreme physiological conditions that alter the heart workload such as pregnancy and microgravity. Overall, this review provides a thorough insight into how mechanical signals are converted into chemical signals to regulate myocardial mass in both healthy and diseased conditions.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Paula F Nieto Morales
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Summer ElSafty
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Aida Rahimi Kahmini
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher Solís
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
5
|
McMillan SN, Pitts JRT, Barua B, Winkelmann DA, Scarff CA. Mavacamten inhibits myosin activity by stabilising the myosin interacting-heads motif and stalling motor force generation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637875. [PMID: 39990378 PMCID: PMC11844505 DOI: 10.1101/2025.02.12.637875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Most sudden cardiac deaths in young people arise from hypertrophic cardiomyopathy, a genetic disease of the heart muscle, with many causative mutations found in the molecular motor beta-cardiac myosin that drives contraction. Therapeutic intervention has until recently been limited to symptomatic relief or invasive procedures. However, small molecule modulators of cardiac myosin are promising therapeutic options to target disease progression. Mavacamten is the first example to gain FDA approval but its molecular mode of action remains unclear, limiting our understanding of its functional effects in disease. To better understand this, we solved the cryoEM structures of beta-cardiac heavy meromyosin in three ADP.Pi-bound states, the primed motor domain in the presence and absence of mavacamten, and the sequestered autoinhibited interacting-heads motif (IHM) in complex with mavacamten, to 2.9 Å, 3.4 Å and 3.7 Å global resolution respectively. Together with quantitative crosslinking mass spectrometric analysis, these structures reveal how mavacamten inhibits myosin. Mavacamten stabilises ADP.Pi binding, stalling the motor domain in a primed state, reducing motor dynamics required for actin-binding cleft closure, and slowing progression through the force generation cycle. Within the two-headed myosin molecule, these effects are propagated and lead to stabilisation of the IHM, through increased contacts at the motor-motor interface. Critically, while mavacamten treatment can thus rescue cardiac muscle relaxation in diastole, it can also reduce contractile output in systole in the heart.
Collapse
Affiliation(s)
- Sean N McMillan
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, Faculty of Medicine and Health, University of Leeds (UoL), UK
- Astbury Centre for Structural Molecular Biology, UoL, UK
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, UoL, UK
| | - Jaime R T Pitts
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, Faculty of Medicine and Health, University of Leeds (UoL), UK
- Astbury Centre for Structural Molecular Biology, UoL, UK
| | - Bipasha Barua
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Donald A Winkelmann
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Charlotte A Scarff
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, Faculty of Medicine and Health, University of Leeds (UoL), UK
- Astbury Centre for Structural Molecular Biology, UoL, UK
| |
Collapse
|
6
|
Morotti I, Marcello M, Sautariello G, Pertici I, Bianco P, Piazzesi G, Linari M, Lombardi V, Reconditi M, Caremani M. The Mechanism of Modulation of Cardiac Force by Temperature. Int J Mol Sci 2025; 26:469. [PMID: 39859186 PMCID: PMC11764908 DOI: 10.3390/ijms26020469] [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: 12/17/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
In maximally Ca2+-activated demembranated fibres from the mammalian skeletal muscle, the depression of the force by lowering the temperature below the physiological level (~35 °C) is explained by the reduction of force in the myosin motor. Instead, cooling is reported to not affect the force per motor in Ca2+-activated cardiac trabeculae from the rat ventricle. Here, the mechanism of the cardiac performance depression by cooling is reinvestigated with fast sarcomere-level mechanics. We determine the changes in the half-sarcomere compliance of maximally Ca2+-activated demembranated rat trabeculae in the range of temperatures of 10-30 °C and analyse the data in terms of a simplified mechanical model of the half-sarcomere to extract the contribution of myofilaments and myosin motors. We find that the changes in the ensemble force are due to changes in the force per motor, while the fraction of actin-attached motors remains constant independent of temperature. The results demonstrate that in the cardiac myosin, as in the skeletal muscle myosin, the force-generating transition is endothermic. The underlying large heat absorption indicates the interaction of extended hydrophobic surfaces within the myosin motor, like those suggested by the crystallographic model of the working stroke.
Collapse
Affiliation(s)
- Ilaria Morotti
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Matteo Marcello
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Giulia Sautariello
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Irene Pertici
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Pasquale Bianco
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Gabriella Piazzesi
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Marco Linari
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Vincenzo Lombardi
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Massimo Reconditi
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Marco Caremani
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
7
|
Stehle R. Phosphate rebinding induces force reversal via slow backward cycling of cross-bridges. Front Physiol 2025; 15:1476876. [PMID: 39839531 PMCID: PMC11747208 DOI: 10.3389/fphys.2024.1476876] [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: 08/06/2024] [Accepted: 11/28/2024] [Indexed: 01/23/2025] Open
Abstract
Objective Previous studies on muscle fibers, myofibrils, and myosin revealed that the release of inorganic phosphate (Pi) and the force-generating step(s) are reversible, with cross-bridges also cycling backward through these steps by reversing force-generating steps and rebinding Pi. The aim was to explore the significance of force redevelopment kinetics (rate constant k TR) in cardiac myofibrils for the coupling between the Pi binding induced force reversal and the rate-limiting transition f - for backward cycling of cross-bridges from force-generating to non-force-generating states. Methods k TR and force generation of cardiac myofibrils from guinea pigs were investigated at 0.015-20 mM Pi. The observed force-[Pi], force-log [Pi], k TR-[Pi], and k TR-force relations were assessed with various single-pathway models of the cross-bridge cycle that differed in sequence and kinetics of reversible Pi release, reversible force-generating step and reversible rate-limiting transition. Based on the interpretation that k TR reflects the sum of rate-limiting transitions in the cross-bridge cycle, an indicator, the coupling strength, was defined to quantify the contribution of Pi binding induced force reversal to the rate-limiting transition f - from the [Pi]-modulated k TR-force relation. Results Increasing [Pi] decreased force by a bi-linear force-log [Pi] relation, increased k TR in a slightly downward curved dependence with [Pi], and altered k TR almost reciprocally to force reflected by the k TR-force relation. Force-[Pi] and force-log [Pi] relations provided less selectivity for the exclusion of models than the k TR-[Pi] and k TR-force relations. The k TR-force relation observed in experiments with cardiac myofibrils yielded the coupling strength +0.84 ± 0.08 close to 1, the maximum coupling strength expected for the reciprocal k TR-force relationship. Single pathway models consisting of fast reversible force generation before or after rapid reversible Pi release failed to describe the observed k TR-force relation. Single pathway models consistent with the observed k TR-force relation had either slow Pi binding or slow force reversal, i.e., in the consistent single pathway models, f - was assigned to the rate of either Pi binding or force reversal. Conclusion Backward flux of cross-bridges from force-generating to non-force-generating states is limited by the rates of Pi binding or force reversal ruling out other rate-limiting steps uncoupled from Pi binding induced force reversal.
Collapse
Affiliation(s)
- Robert Stehle
- Institute of Vegetative Physiology, University of Cologne, Köln, Germany
| |
Collapse
|
8
|
Olalekan SO, Bakare OO, Okwute PG, Osonuga IO, Adeyanju MM, Edema VB. Hypertrophic cardiomyopathy: insights into pathophysiology and novel therapeutic strategies from clinical studies. Egypt Heart J 2025; 77:5. [PMID: 39776022 PMCID: PMC11706819 DOI: 10.1186/s43044-024-00600-4] [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: 08/20/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a frequently encountered cardiac condition worldwide, often inherited, and characterized by intricate phenotypic and genetic manifestations. The natural progression of HCM is diverse, largely due to mutations in the contractile and relaxation proteins of the heart. These mutations disrupt the normal structure and functioning of the heart muscle, particularly affecting genes that encode proteins involved in the contraction and relaxation of cardiac muscle. MAIN BODY This review focused on understanding the role of contractile and relaxation proteins in the pathogenesis of hypertrophic cardiomyopathy. Mutations in contractile proteins such as myosin, actin, tropomyosin, and troponin are associated with hypercontractility and increased sensitivity of the heart muscle, leading to HCM. Additionally, impaired relaxation of the heart muscle, linked to abnormalities in proteins like phospholamban, sarcolipin, titin, myosin binding protein-C, and calsequestrin, contributes significantly to the disease. The review also explored the impact of targeted therapeutic approaches aimed at modulating these proteins to improve patient outcomes. Recent advances in therapeutic strategies, including novel pharmacological agents like mavacamten and aficamten, were examined for their potential to help patients manage the disease and lead more accommodating lifestyles. CONCLUSIONS The review underscored the significance of early diagnosis and personalized treatment approaches in managing HCM. Future research should prioritize the development of robust biomarkers for early detection and risk stratification, particularly in diverse populations, to enhance clinical outcomes. Furthermore, it is imperative to delve deeper into the genetic mutations and molecular mechanisms associated with HCM, with a focus on exploring the roles of less-studied myocardial relaxation proteins and their interactions with sarcomere constituents.
Collapse
Affiliation(s)
- Samuel Oluwadare Olalekan
- Department of Physiology, Faculty of Basic Medical Sciences, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State, Nigeria.
| | | | | | - Ifabunmi Oduyemi Osonuga
- Department of Physiology, Faculty of Basic Medical Sciences, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State, Nigeria
| | - Muinat Moronke Adeyanju
- Department of Biochemistry, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State, Nigeria
| | - Victoria Biola Edema
- Department of Physiology, Faculty of Basic Medical Sciences, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State, Nigeria
| |
Collapse
|
9
|
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
|
10
|
Irving M. Functional control of myosin motors in the cardiac cycle. Nat Rev Cardiol 2025; 22:9-19. [PMID: 39030271 DOI: 10.1038/s41569-024-01063-5] [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: 07/02/2024] [Indexed: 07/21/2024]
Abstract
Contraction of the heart is driven by cyclical interactions between myosin and actin filaments powered by ATP hydrolysis. The modular structure of heart muscle and the organ-level synchrony of the heartbeat ensure tight reciprocal coupling between this myosin ATPase cycle and the macroscopic cardiac cycle. The myosin motors respond to the cyclical activation of the actin and myosin filaments to drive the pressure changes that control the inflow and outflow valves of the heart chambers. Opening and closing of the valves in turn switches the myosin motors between roughly isometric and roughly isotonic contraction modes. Peak filament stress in the heart is much smaller than in fully activated skeletal muscle, although the myosin filaments in the two muscle types have the same number of myosin motors. Calculations indicate that only ~5% of the myosin motors in the heart are needed to generate peak systolic pressure, although many more motors are needed to drive ejection. Tight regulation of the number of active motors is essential for the efficient functioning of the healthy heart - this control is commonly disrupted by gene variants associated with inherited heart disease, and its restoration might be a useful end point in the development of novel therapies.
Collapse
Affiliation(s)
- Malcolm Irving
- Randall Centre for Cell and Molecular Biophysics and BHF Centre for Research Excellence, King's College London, London, UK.
| |
Collapse
|
11
|
Morotti I, Caremani M, Marcello M, Pertici I, Squarci C, Bianco P, Narayanan T, Piazzesi G, Reconditi M, Lombardi V, Linari M. An integrated picture of the structural pathways controlling the heart performance. Proc Natl Acad Sci U S A 2024; 121:e2410893121. [PMID: 39630866 DOI: 10.1073/pnas.2410893121] [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: 05/31/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
The regulation of heart function is attributed to a dual filament mechanism: i) the Ca2+-dependent structural changes in the regulatory proteins of the thin, actin-containing filament making actin available for myosin motor attachment, and ii) the release of motors from their folded (OFF) state on the surface of the thick filament allowing them to attach and pull the actin filament. Thick filament mechanosensing is thought to control the number of motors switching ON in relation to the systolic performance, but its molecular basis is still controversial. Here, we use high spatial resolution X-ray diffraction data from electrically paced rat trabeculae and papillary muscles to provide a molecular explanation of the modulation of heart performance that calls for a revision of the mechanosensing hypothesis. We find that upon stimulation, titin-mediated structural changes in the thick filament switch motors ON throughout the filament within ~½ the maximum systolic force. These structural changes also drive Myosin Binding Protein-C (MyBP-C) to promote first motor attachments to actin from the central 1/3 of the half-thick filament. Progression of attachments toward the periphery of half-thick filament with increase in systolic force is carried on by near-neighbor cooperative thin filament activation by attached motors. The identification of the roles of MyBP-C, titin, thin and thick filaments in heart regulation enables their targeting for potential therapeutic interventions.
Collapse
Affiliation(s)
- Ilaria Morotti
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Marco Caremani
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Matteo Marcello
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Irene Pertici
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Caterina Squarci
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Pasquale Bianco
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | | | - Gabriella Piazzesi
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Massimo Reconditi
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Vincenzo Lombardi
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Marco Linari
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| |
Collapse
|
12
|
Wang Y, Fusi L, Ovejero JG, Hill C, Juma S, Cullup FP, Ghisleni A, Park-Holohan SJ, Ma W, Irving T, Narayanan T, Irving M, Brunello E. Load-dependence of the activation of myosin filaments in heart muscle. J Physiol 2024; 602:6889-6907. [PMID: 39552044 DOI: 10.1113/jp287434] [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: 08/05/2024] [Accepted: 10/03/2024] [Indexed: 11/19/2024] Open
Abstract
Contraction of heart muscle requires activation of both the actin and myosin filaments. The mechanism of myosin filament activation is unknown, but the leading candidate hypothesis is direct mechano-sensing by the filaments. Here, we tested this hypothesis by activating intact trabeculae from rat heart by electrical stimulation under different loads and measuring myosin filament activation by X-ray diffraction. Unexpectedly, we found that the distinct structural changes in the myosin filament associated with activation had different dependences on the load. In early activation, all the structural changes indicated faster activation at higher load, as expected from the mechano-sensing hypothesis, but, at later times, the helical order of the myosin motors characteristic of the inactivated state was lost even at very low load. We conclude that mechano-sensing does operate in heart muscle, but it is supplemented by a previously undescribed mechanism that links myosin filament activation to actin filament activation. KEY POINTS: Myosin filament activation controls the strength and speed of contraction in heart muscle. Early activation of the myosin filament is determined by the filament load. At later times, myosin filament activation is controlled by a load independent pathway. This load independent pathway provides new targets and assays for drug development.
Collapse
Affiliation(s)
- Yanhong Wang
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Luca Fusi
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
- Centre for Human and Applied Physiological Sciences, King's College London, London, UK
| | - Jesus G Ovejero
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Cameron Hill
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Samina Juma
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Flair Paradine Cullup
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Andrea Ghisleni
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - So-Jin Park-Holohan
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Weikang Ma
- BioCAT, Dept of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Thomas Irving
- BioCAT, Dept of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | | | - Malcolm Irving
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Elisabetta Brunello
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| |
Collapse
|
13
|
Sun ZG, Murrell M. Cofilin-Mediated Filament Softening and Crosslinking Counterbalance to Enhance Actin Network Flexibility. PHYSICAL REVIEW LETTERS 2024; 133:218402. [PMID: 39642486 DOI: 10.1103/physrevlett.133.218402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/30/2024] [Indexed: 12/09/2024]
Abstract
Filamentous-actin (F-actin) crosslinking within the cell cytoskeleton mediates the transmission of mechanical forces, enabling changes in cell shape, as occurs during cell division and cell migration. Crosslinking by actin binding proteins (ABPs) generally increases the connectivity of the F-actin network, but also increases network rigidity. As a result, there is a narrow range in the concentration of crosslinker protein at which F-actin networks are both connected and labile. Another ABP, cofilin, severs F-actin filaments at high pH through increasing their bending flexibility and concentrating mechanical stress, inducing fragmentation. By contrast, at lower pH, cofilin increases filament flexibility yet does not sever. Instead, it forms disulfide bonds, which crosslink F-actin into bundles, and bundles into networks. Here, we combine light microscopy and rheology to determine the impact of two potentially opposing effects on the mechanics of F-actin networks-increased flexibility at the filament level, and increased connectivity at the network level. Indeed, by linear rheology, we find that these mechanisms are counterbalanced, such that cofilactin network moduli are only weakly dependent on cofilin concentration over a broad range, in contrast to the dramatic stiffening that occurs with F-actin crosslinking protein. Further, by nonlinear rheology, the network stiffens at a higher stress than crosslinking protein, indicative of a broader range in which the material remains flexible. These results may enable F-actin networks to increase connectivity without heavy penalties to rigidity, and thus provide a new route to modulating active polymer mechanics unseen using traditional F-actin accessory proteins.
Collapse
Affiliation(s)
- Zachary Gao Sun
- Department of Physics, Yale University, 217 Prospect Street, New Haven, Connecticut 06511, USA
- Systems Biology Institute, Yale University, West Haven, Connecticut 06516, USA
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, Connecticut 06520, USA
| | - Michael Murrell
- Department of Physics, Yale University, 217 Prospect Street, New Haven, Connecticut 06511, USA
- Systems Biology Institute, Yale University, West Haven, Connecticut 06516, USA
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, Connecticut 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
14
|
Childers MC, Geeves MA, Regnier M. Interacting myosin head dynamics and their modification by 2'-deoxy-ADP. Biophys J 2024; 123:3997-4008. [PMID: 39444161 PMCID: PMC11617627 DOI: 10.1016/j.bpj.2024.10.013] [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: 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
|
15
|
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
|
16
|
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
|
17
|
Bruns H, Czajka TS, Sztucki M, Brandenburg S, Salditt T. Sarcomere, troponin, and myosin X-ray diffraction signals can be resolved in single cardiomyocytes. Biophys J 2024; 123:3024-3037. [PMID: 38956875 PMCID: PMC11427778 DOI: 10.1016/j.bpj.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/18/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024] Open
Abstract
Cardiac function relies on the autonomous molecular contraction mechanisms in the ventricular wall. Contraction is driven by ordered motor proteins acting in parallel to generate a macroscopic force. The averaged structure can be investigated by diffraction from model tissues such as trabecular and papillary cardiac muscle using collimated synchrotron beams, offering high resolution in reciprocal space. In the ventricular wall, however, the muscle tissue is compartmentalized into smaller branched cardiomyocytes, with a higher degree of disorder. We show that X-ray diffraction is now also capable of resolving the structural organization of actomyosin in single isolated cardiomyocytes of the ventricular wall. In addition to the hexagonal arrangement of thick and thin filaments, the diffraction signal of the hydrated and fixated cardiomyocytes was sufficient to reveal the myosin motor repeat (M3), the troponin complex repeat (Tn), and the sarcomere length. The sarcomere length signal comprised up to 13 diffraction orders, which were used to compute the sarcomere density profile based on Fourier synthesis. The Tn and M3 spacings were found in the same range as previously reported for other muscle types. The approach opens up a pathway to record the structural dynamics of living cells during the contraction cycle, toward a more complete understanding of cardiac muscle function.
Collapse
Affiliation(s)
| | | | - Michael Sztucki
- ESRF - European Synchrotron Radiation Facility, Grenoble, France
| | - Sören Brandenburg
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Tim Salditt
- Institute for X-ray Physics, Göttingen, Germany.
| |
Collapse
|
18
|
Tomalka A, Weidner S, Hahn D, Seiberl W, Siebert T. Force re-development after shortening reveals a role for titin in stretch-shortening performance enhancement in skinned muscle fibres. J Exp Biol 2024; 227:jeb247377. [PMID: 39119673 DOI: 10.1242/jeb.247377] [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: 01/22/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
Stretch-shortening cycles (SSCs) involve muscle lengthening (eccentric contractions) instantly followed by shortening (concentric contractions). This combination enhances force, work and power output compared with pure shortening contractions, which is known as the SSC effect. Recent evidence indicates both cross-bridge (XB)-based and non-XB-based (e.g. titin) structures contribute to this effect. This study analysed force re-development following SSCs and pure shortening contractions to gain further insight into the roles of XB and non-XB structures regarding the SSC effect. Experiments were conducted on rat soleus muscle fibres (n=16) with different SSC velocities (30%, 60% and 85% of maximum shortening velocity) and constant stretch-shortening magnitudes (18% of optimum length). The XB inhibitor blebbistatin was used to distinguish between XB and non-XB contributions to force generation. The results showed SSCs led to significantly greater [mean±s.d. 1.02±0.15 versus 0.68±0.09 (ΔF/Δt); t62=8.61, P<0.001, d=2.79) and faster (75 ms versus 205 ms; t62=-6.37, P<0.001, d=-1.48) force re-development compared with pure shortening contractions in the control treatment. In the blebbistatin treatment, SSCs still resulted in greater [0.11±0.03 versus 0.06±0.01 (ΔF/Δt); t62=8.00, P<0.001, d=2.24) and faster (3010±1631 versus 7916±3230 ms; t62=-8.00, P<0.001, d=-1.92) force re-development compared with pure shortening contractions. These findings deepen our understanding of the SSC effect, underscoring the involvement of non-XB structures such as titin in modulating force production. This modulation is likely to involve complex mechanosensory coupling from stretch to signal transmission during muscle contraction.
Collapse
Affiliation(s)
- André Tomalka
- Motion and Exercise Science, University of Stuttgart, 70569 Stuttgart, Germany
| | - Sven Weidner
- Motion and Exercise Science, University of Stuttgart, 70569 Stuttgart, Germany
| | - Daniel Hahn
- Human Movement Science, Faculty of Sports Science, Ruhr University Bochum, 44801 Bochum, Germany
- School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, QLD 4067, Australia
| | - Wolfgang Seiberl
- Human Movement Science, University of the Bundeswehr Munich, 85579 Neubiberg, Germany
| | - Tobias Siebert
- Motion and Exercise Science, University of Stuttgart, 70569 Stuttgart, Germany
- Stuttgart Center for Simulation Science, University of Stuttgart, 70569 Stuttgart, Germany
| |
Collapse
|
19
|
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
|
20
|
Childers MC, Geeves MA, Regnier M. An atomistic model of myosin interacting heads motif dynamics and their modification by 2'-deoxy-ADP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597809. [PMID: 38895221 PMCID: PMC11185614 DOI: 10.1101/2024.06.06.597809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
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 unable to perform 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 via missense mutations can pathologically disrupt the mechanical performance of muscle tissue. Thus, the IHM state has become a target for therapeutic intervention. An ATP analogue called 2'-deoxy-ATP (dATP) is a potent myosin activator which 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 with IHM containing ADP.Pi in both nucleotide binding pockets revealed residual dynamics in an otherwise 'inactive' and 'sequestered' state of a motor protein. Replacement of ADP.Pi by dADP.Pi triggered a series of structural changes that modify the protein-protein interface that stabilizes the sequestered state, and changes to this interface were accompanied by allosteric changes in remote regions of the protein complex. A comparative analysis of these dynamics predicted new structural sites that may affect IHM stability.
Collapse
|
21
|
Zhao J, Qi L, Yuan S, Irving TC, Ma W. Differences in thick filament activation in fast rodent skeletal muscle and slow porcine cardiac muscle. J Physiol 2024; 602:2751-2762. [PMID: 38695322 PMCID: PMC11178443 DOI: 10.1113/jp286072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/16/2024] [Indexed: 06/15/2024] Open
Abstract
There is a growing appreciation that regulation of muscle contraction requires both thin filament and thick filament activation in order to fully activate the sarcomere. The prevailing mechano-sensing model for thick filament activation was derived from experiments on fast-twitch muscle. We address the question whether, or to what extent, this mechanism can be extrapolated to the slow muscle in the hearts of large mammals, including humans. We investigated the similarities and differences in structural signatures of thick filament activation in porcine myocardium as compared to fast rat extensor digitorum longus (EDL) skeletal muscle under relaxed conditions and sub-maximal contraction using small angle X-ray diffraction. Thick and thin filaments were found to adopt different structural configurations under relaxing conditions, and myosin heads showed different changes in configuration upon sub-maximal activation, when comparing the two muscle types. Titin was found to have an X-ray diffraction signature distinct from those of the overall thick filament backbone, and its spacing change appeared to be positively correlated to the force exerted on the thick filament. Structural changes in fast EDL muscle were found to be consistent with the mechano-sensing model. In porcine myocardium, however, the structural basis of mechano-sensing is blunted suggesting the need for additional activation mechanism(s) in slow cardiac muscle. These differences in thick filament regulation can be related to their different physiological roles where fast muscle is optimized for rapid, burst-like, contractions, and the slow cardiac muscle in large mammalian hearts adopts a more finely tuned, graded response to allow for their substantial functional reserve. KEY POINTS: Both thin filament and thick filament activation are required to fully activate the sarcomere. Thick and thin filaments adopt different structural configurations under relaxing conditions, and myosin heads show different changes in configuration upon sub-maximal activation in fast extensor digitorum longus muscle and slow porcine cardiac muscle. Titin has an X-ray diffraction signature distinct from those of the overall thick filament backbone and this titin reflection spacing change appeared to be directly proportional to the force exerted on the thick filament. Mechano-sensing is blunted in porcine myocardium suggesting the need for additional activation mechanism(s) in slow cardiac muscle. Fast skeletal muscle is optimized for rapid, burst-like contractions, and the slow cardiac muscle in large mammalian hearts adopts a more finely tuned graded response to allow for their substantial functional reserve.
Collapse
Affiliation(s)
- Jing Zhao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Lin Qi
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Shengyao Yuan
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, 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
| | - 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
|
22
|
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] [Grants] [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
|
23
|
Dababneh S, Hamledari H, Maaref Y, Jayousi F, Hosseini DB, Khan A, Jannati S, Jabbari K, Arslanova A, Butt M, Roston TM, Sanatani S, Tibbits GF. Advances in Hypertrophic Cardiomyopathy Disease Modelling Using hiPSC-Derived Cardiomyocytes. Can J Cardiol 2024; 40:766-776. [PMID: 37952715 DOI: 10.1016/j.cjca.2023.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/21/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
The advent of human induced pluripotent stem cells (hiPSCs) and their capacity to be differentiated into beating human cardiomyocytes (CMs) in vitro has revolutionized human disease modelling, genotype-phenotype predictions, and therapeutic testing. Hypertrophic cardiomyopathy (HCM) is a common inherited cardiomyopathy and the leading known cause of sudden cardiac arrest in young adults and athletes. On a molecular level, HCM is often driven by single pathogenic genetic variants, usually in sarcomeric proteins, that can alter the mechanical, electrical, signalling, and transcriptional properties of the cell. A deeper knowledge of these alterations is critical to better understanding HCM manifestation, progression, and treatment. Leveraging hiPSC-CMs to investigate the molecular mechanisms driving HCM presents a unique opportunity to dissect the consequences of genetic variants in a sophisticated and controlled manner. In this review, we summarize the molecular underpinnings of HCM and the role of hiPSC-CM studies in advancing our understanding, and we highlight the advances in hiPSC-CM-based modelling of HCM, including maturation, contractility, multiomics, and genome editing, with the notable exception of electrophysiology, which has been previously covered.
Collapse
Affiliation(s)
- Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Homa Hamledari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Yasaman Maaref
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Farah Jayousi
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Dina B Hosseini
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Aasim Khan
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Shayan Jannati
- Faculty of Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kosar Jabbari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Alia Arslanova
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Mariam Butt
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Thomas M Roston
- Division of Cardiology and Centre for Cardiovascular Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shubhayan Sanatani
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Glen F Tibbits
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.
| |
Collapse
|
24
|
Arts T, Lyon A, Delhaas T, Kuster DWD, van der Velden J, Lumens J. Translating myosin-binding protein C and titin abnormalities to whole-heart function using a novel calcium-contraction coupling model. J Mol Cell Cardiol 2024; 190:13-23. [PMID: 38462126 DOI: 10.1016/j.yjmcc.2024.03.001] [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: 08/01/2023] [Revised: 01/15/2024] [Accepted: 03/01/2024] [Indexed: 03/12/2024]
Abstract
Mutations in cardiac myosin-binding protein C (cMyBP-C) or titin may respectively lead to hypertrophic (HCM) or dilated (DCM) cardiomyopathies. The mechanisms leading to these phenotypes remain unclear because of the challenge of translating cellular abnormalities to whole-heart and system function. We developed and validated a novel computer model of calcium-contraction coupling incorporating the role of cMyBP-C and titin based on the key assumptions: 1) tension in the thick filament promotes cross-bridge attachment mechanochemically, 2) with increasing titin tension, more myosin heads are unlocked for attachment, and 3) cMyBP-C suppresses cross-bridge attachment. Simulated stationary calcium-tension curves, isotonic and isometric contractions, and quick release agreed with experimental data. The model predicted that a loss of cMyBP-C function decreases the steepness of the calcium-tension curve, and that more compliant titin decreases the level of passive and active tension and its dependency on sarcomere length. Integrating this cellular model in the CircAdapt model of the human heart and circulation showed that a loss of cMyBP-C function resulted in HCM-like hemodynamics with higher left ventricular end-diastolic pressures and smaller volumes. More compliant titin led to higher diastolic pressures and ventricular dilation, suggesting DCM-like hemodynamics. The novel model of calcium-contraction coupling incorporates the role of cMyBP-C and titin. Its coupling to whole-heart mechanics translates changes in cellular calcium-contraction coupling to changes in cardiac pump and circulatory function and identifies potential mechanisms by which cMyBP-C and titin abnormalities may develop into HCM and DCM phenotypes. This modeling platform may help identify distinct mechanisms underlying clinical phenotypes in cardiac diseases.
Collapse
Affiliation(s)
- Theo Arts
- Department of Biomedical Engineering, Cardiovascular Research Center Maastricht (CARIM), Maastricht University, 6200MD Maastricht, the Netherlands.
| | - Aurore Lyon
- Department of Biomedical Engineering, Cardiovascular Research Center Maastricht (CARIM), Maastricht University, 6200MD Maastricht, the Netherlands
| | - Tammo Delhaas
- Department of Biomedical Engineering, Cardiovascular Research Center Maastricht (CARIM), Maastricht University, 6200MD Maastricht, the Netherlands
| | - Diederik W D Kuster
- Department of Physiology, Amsterdam University Medical Center, 1081HZ Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Center, 1081HZ Amsterdam, the Netherlands
| | - Joost Lumens
- Department of Biomedical Engineering, Cardiovascular Research Center Maastricht (CARIM), Maastricht University, 6200MD Maastricht, the Netherlands
| |
Collapse
|
25
|
van Dover G, Javor J, Ewoldt JK, Zhernenkov M, Wąsik P, Freychet G, Lee J, Brown D, Chen CS, Bishop DJ. Structural maturation of myofilaments in engineered 3D cardiac microtissues characterized using small angle x-ray scattering. Phys Biol 2024; 21:036001. [PMID: 38452380 DOI: 10.1088/1478-3975/ad310e] [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: 10/23/2023] [Accepted: 03/07/2024] [Indexed: 03/09/2024]
Abstract
Understanding the structural and functional development of human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) is essential to engineering cardiac tissue that enables pharmaceutical testing, modeling diseases, and designing therapies. Here we use a method not commonly applied to biological materials, small angle x-ray scattering, to characterize the structural development of hiPSC-CMs within three-dimensional engineered tissues during their preliminary stages of maturation. An x-ray scattering experimental method enables the reliable characterization of the cardiomyocyte myofilament spacing with maturation time. The myofilament lattice spacing monotonically decreases as the tissue matures from its initial post-seeding state over the span of 10 days. Visualization of the spacing at a grid of positions in the tissue provides an approach to characterizing the maturation and organization of cardiomyocyte myofilaments and has the potential to help elucidate mechanisms of pathophysiology, and disease progression, thereby stimulating new biological hypotheses in stem cell engineering.
Collapse
Affiliation(s)
| | - Josh Javor
- Boston University, Boston, MA 02215, United States of America
| | | | - Mikhail Zhernenkov
- Brookhaven National Laboratory, Upton, NY 11973, United States of America
| | - Patryk Wąsik
- Brookhaven National Laboratory, Upton, NY 11973, United States of America
| | - Guillaume Freychet
- Brookhaven National Laboratory, Upton, NY 11973, United States of America
| | - Josh Lee
- Boston University, Boston, MA 02215, United States of America
| | - Dana Brown
- Fort Valley State University, Fort Valley, GA 31030, United States of America
| | | | - David J Bishop
- Boston University, Boston, MA 02215, United States of America
| |
Collapse
|
26
|
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: 12] [Impact Index Per Article: 12.0] [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
|
27
|
Abstract
Force generation in striated muscle is primarily controlled by structural changes in the actin-containing thin filaments triggered by an increase in intracellular calcium concentration. However, recent studies have elucidated a new class of regulatory mechanisms, based on the myosin-containing thick filament, that control the strength and speed of contraction by modulating the availability of myosin motors for the interaction with actin. This review summarizes the mechanisms of thin and thick filament activation that regulate the contractility of skeletal and cardiac muscle. A novel dual-filament paradigm of muscle regulation is emerging, in which the dynamics of force generation depends on the coordinated activation of thin and thick filaments. We highlight the interfilament signaling pathways based on titin and myosin-binding protein-C that couple thin and thick filament regulatory mechanisms. This dual-filament regulation mediates the length-dependent activation of cardiac muscle that underlies the control of the cardiac output in each heartbeat.
Collapse
Affiliation(s)
- Elisabetta Brunello
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom; ,
| | - Luca Fusi
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom; ,
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| |
Collapse
|
28
|
Tanner BCW. Design Principles and Benefits of Spatially Explicit Models of Myofilament Function. Methods Mol Biol 2024; 2735:43-62. [PMID: 38038843 DOI: 10.1007/978-1-0716-3527-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Spatially explicit models of muscle contraction include fine-scale details about the spatial, kinetic, and/or mechanical properties of the biological processes being represented within the model network. Over the past 25 years, this has primarily consisted of a set of mathematical and computational algorithms representing myosin cross-bridge activity, Ca2+-activation of contraction, and ensemble force production within a half-sarcomere representation of the myofilament network. Herein we discuss basic design principles associated with creating spatially explicit models of myofilament function, as well as model assumptions underlying model development. A brief overview of computational approaches is introduced. Opportunities for new model directions that could investigate coupled regulatory pathways between the thick-filament and thin-filaments are also presented. Given the modular design and flexibility associated with spatially explicit models, we highlight some advantages of this approach compared to other model formulations.
Collapse
Affiliation(s)
- Bertrand C W Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA.
| |
Collapse
|
29
|
Heeley DH, Belknap B, Atherton JL, Hasan SC, White HD. Effect of the N-terminal extension in myosin essential light chain A1 on the mechanism of actomyosin ATP hydrolysis. J Biol Chem 2024; 300:105521. [PMID: 38042484 PMCID: PMC10777021 DOI: 10.1016/j.jbc.2023.105521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023] Open
Abstract
Myosin essential light chains A1 and A2 are identical isoforms except for an extension of ∼40 amino acids at the N terminus of A1 that binds F-actin. The extension has no bearing on the burst hydrolysis rate (M-ATP → M-ADP-Pi) as determined by chemical quench flow (100 μM isoenzyme). Whereas actomyosin-S1A2 steady state MgATPase (low ionic strength, 20 °C) is hyperbolically dependent on concentration: Vmax 7.6 s-1, Kapp 6.4 μM (F-actin) and Vmax 10.1 s-1, Kapp 5.5 μM (native thin filaments, pCa 4), the relationship for myosin-S1A1 is bimodal; an initial rise at low concentration followed by a decline to one-third the Vmax of S1A2, indicative of more than one rate-limiting step and A1-enforced flux through the slower actomyosin-limited hydrolysis pathway. In double-mixing stopped-flow with an indicator, Ca(II)-mediated activation of Pi dissociation (regulatedAM-ADP-Pi → regulatedAM-ADP + Pi) is attenuated by A1 attachment to thin filaments (pCa 4). The maximum accelerated rates of Pi dissociation are: 81 s-1 (S1A1, Kapp 8.9 μM) versus 129 s-1 (S1A2, Kapp 58 μM). To investigate apomyosin-S1-mediated activation, thin filaments (EGTA) are premixed with a given isomyosin-S1 and double-mixing is repeated with myosin-S1A1 in the first mix. Similar maximum rates of Pi dissociation are observed, 44.5 s-1 (S1A1) and 47.1 s-1 (S1A2), which are lower than for Ca(II) activation. Overall, these results biochemically demonstrate how the longer light chain A1 can contribute to slower contraction and higher force and the shorter version A2 to faster contraction and lower force, consistent with their distribution in different types of striated muscle.
Collapse
Affiliation(s)
- David H Heeley
- Department of Biochemistry, Memorial University, St John's, Newfoundland, Canada.
| | - Betty Belknap
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Jennifer L Atherton
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Stephanie C Hasan
- Department of Biochemistry, Memorial University, St John's, Newfoundland, Canada
| | - Howard D White
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| |
Collapse
|
30
|
Elkrief D, Matusovsky O, Cheng YS, Rassier DE. From amino-acid to disease: the effects of oxidation on actin-myosin interactions in muscle. J Muscle Res Cell Motil 2023; 44:225-254. [PMID: 37805961 DOI: 10.1007/s10974-023-09658-0] [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: 06/07/2023] [Accepted: 08/15/2023] [Indexed: 10/10/2023]
Abstract
Actin-myosin interactions form the basis of the force-producing contraction cycle within the sarcomere, serving as the primary mechanism for muscle contraction. Post-translational modifications, such as oxidation, have a considerable impact on the mechanics of these interactions. Considering their widespread occurrence, the explicit contributions of these modifications to muscle function remain an active field of research. In this review, we aim to provide a comprehensive overview of the basic mechanics of the actin-myosin complex and elucidate the extent to which oxidation influences the contractile cycle and various mechanical characteristics of this complex at the single-molecule, myofibrillar and whole-muscle levels. We place particular focus on amino acids shown to be vulnerable to oxidation in actin, myosin, and some of their binding partners. Additionally, we highlight the differences between in vitro environments, where oxidation is controlled and limited to actin and myosin and myofibrillar or whole muscle environments, to foster a better understanding of oxidative modification in muscle. Thus, this review seeks to encompass a broad range of studies, aiming to lay out the multi layered effects of oxidation in in vitro and in vivo environments, with brief mention of clinical muscular disorders associated with oxidative stress.
Collapse
Affiliation(s)
- Daren Elkrief
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Oleg Matusovsky
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada
| | - Yu-Shu Cheng
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada
| | - Dilson E Rassier
- Department of Physiology, McGill University, Montreal, QC, Canada.
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada.
- Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
31
|
Parijat P, Attili S, Hoare Z, Shattock M, Kenyon V, Kampourakis T. Discovery of a novel cardiac-specific myosin modulator using artificial intelligence-based virtual screening. Nat Commun 2023; 14:7692. [PMID: 38001148 PMCID: PMC10673995 DOI: 10.1038/s41467-023-43538-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Direct modulation of cardiac myosin function has emerged as a therapeutic target for both heart disease and heart failure. However, the development of myosin-based therapeutics has been hampered by the lack of targeted in vitro screening assays. In this study we use Artificial Intelligence-based virtual high throughput screening (vHTS) to identify novel small molecule effectors of human β-cardiac myosin. We test the top scoring compounds from vHTS in biochemical counter-screens and identify a novel chemical scaffold called 'F10' as a cardiac-specific low-micromolar myosin inhibitor. Biochemical and biophysical characterization in both isolated proteins and muscle fibers show that F10 stabilizes both the biochemical (i.e. super-relaxed state) and structural (i.e. interacting heads motif) OFF state of cardiac myosin, and reduces force and left ventricular pressure development in isolated myofilaments and Langendorff-perfused hearts, respectively. F10 is a tunable scaffold for the further development of a novel class of myosin modulators.
Collapse
Affiliation(s)
- Priyanka Parijat
- Randall Centre for Cell and Molecular Biophysics; and British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, United Kingdom
| | - Seetharamaiah Attili
- Randall Centre for Cell and Molecular Biophysics; and British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, United Kingdom
| | - Zoe Hoare
- School of Cardiovascular and Metabolic Medicine and Sciences; Rayne Institute and British Heart Foundation Centre of Research Excellence, King's College London, London, SE5 9NU, United Kingdom
| | - Michael Shattock
- School of Cardiovascular and Metabolic Medicine and Sciences; Rayne Institute and British Heart Foundation Centre of Research Excellence, King's College London, London, SE5 9NU, United Kingdom
| | | | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics; and British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, United Kingdom.
| |
Collapse
|
32
|
Bodt SML, Ge J, Ma W, Rasicci DV, Desetty R, McCammon JA, Yengo CM. Dilated cardiomyopathy mutation in beta-cardiac myosin enhances actin activation of the power stroke and phosphate release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566646. [PMID: 38014187 PMCID: PMC10680644 DOI: 10.1101/2023.11.10.566646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Inherited mutations in human beta-cardiac myosin (M2β) can lead to severe forms of heart failure. The E525K mutation in M2β is associated with dilated cardiomyopathy (DCM) and was found to stabilize the interacting heads motif (IHM) and autoinhibited super-relaxed (SRX) state in dimeric heavy meromyosin. However, in monomeric M2β subfragment 1 (S1) we found that E525K enhances (3-fold) the maximum steady-state actin-activated ATPase activity (kcat) and decreases (6-fold) the actin concentration at which ATPase is one-half maximal (KATPase). We also found a 3 to 4-fold increase in the actin-activated power stroke and phosphate release rate constants at 30 μM actin, which overall enhanced the duty ratio 3-fold. Loaded motility assays revealed that the enhanced intrinsic motor activity translates to increased ensemble force in M2β S1. Glutamate 525, located near the actin binding region in the so-called activation loop, is highly conserved and predicted to form a salt-bridge with another conserved residue (lysine 484) in the relay helix. Enhanced sampling molecular dynamics simulations predict that the charge reversal mutation disrupts the E525-K484 salt-bridge, inducing conformations with a more flexible relay helix and a wide phosphate release tunnel. Our results highlight a highly conserved allosteric pathway associated with actin activation of the power stroke and phosphate release and suggest an important feature of the autoinhibited IHM is to prevent this region of myosin from interacting with actin. The ability of the E525K mutation to stabilize the IHM likely overrides the enhanced intrinsic motor properties, which may be key to triggering DCM pathogenesis.
Collapse
Affiliation(s)
- Skylar M. L. Bodt
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jinghua Ge
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Wen Ma
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California
| | - David V. Rasicci
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
33
|
Tamborrini D, Wang Z, Wagner T, Tacke S, Stabrin M, Grange M, Kho AL, Rees M, Bennett P, Gautel M, Raunser S. Structure of the native myosin filament in the relaxed cardiac sarcomere. Nature 2023; 623:863-871. [PMID: 37914933 PMCID: PMC10665186 DOI: 10.1038/s41586-023-06690-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/28/2023] [Indexed: 11/03/2023]
Abstract
The thick filament is a key component of sarcomeres, the basic units of striated muscle1. Alterations in thick filament proteins are associated with familial hypertrophic cardiomyopathy and other heart and muscle diseases2. Despite the central importance of the thick filament, its molecular organization remains unclear. Here we present the molecular architecture of native cardiac sarcomeres in the relaxed state, determined by cryo-electron tomography. Our reconstruction of the thick filament reveals the three-dimensional organization of myosin, titin and myosin-binding protein C (MyBP-C). The arrangement of myosin molecules is dependent on their position along the filament, suggesting specialized capacities in terms of strain susceptibility and force generation. Three pairs of titin-α and titin-β chains run axially along the filament, intertwining with myosin tails and probably orchestrating the length-dependent activation of the sarcomere. Notably, whereas the three titin-α chains run along the entire length of the thick filament, titin-β chains do not. The structure also demonstrates that MyBP-C bridges thin and thick filaments, with its carboxy-terminal region binding to the myosin tails and directly stabilizing the OFF state of the myosin heads in an unforeseen manner. These results provide a foundation for future research investigating muscle disorders involving sarcomeric components.
Collapse
Affiliation(s)
- Davide Tamborrini
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Zhexin Wang
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Thorsten Wagner
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Sebastian Tacke
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Markus Stabrin
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Michael Grange
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Structural Biology, The Rosalind Franklin Institute, Didcot, UK
| | - Ay Lin Kho
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Kings College London BHF Centre of Research Excellence, London, UK
| | - Martin Rees
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Kings College London BHF Centre of Research Excellence, London, UK
| | - Pauline Bennett
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Kings College London BHF Centre of Research Excellence, London, UK
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Kings College London BHF Centre of Research Excellence, London, UK
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| |
Collapse
|
34
|
Dutta D, Nguyen V, Campbell KS, Padrón R, Craig R. Cryo-EM structure of the human cardiac myosin filament. Nature 2023; 623:853-862. [PMID: 37914935 PMCID: PMC10846670 DOI: 10.1038/s41586-023-06691-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/28/2023] [Indexed: 11/03/2023]
Abstract
Pumping of the heart is powered by filaments of the motor protein myosin that pull on actin filaments to generate cardiac contraction. In addition to myosin, the filaments contain cardiac myosin-binding protein C (cMyBP-C), which modulates contractility in response to physiological stimuli, and titin, which functions as a scaffold for filament assembly1. Myosin, cMyBP-C and titin are all subject to mutation, which can lead to heart failure. Despite the central importance of cardiac myosin filaments to life, their molecular structure has remained a mystery for 60 years2. Here we solve the structure of the main (cMyBP-C-containing) region of the human cardiac filament using cryo-electron microscopy. The reconstruction reveals the architecture of titin and cMyBP-C and shows how myosin's motor domains (heads) form three different types of motif (providing functional flexibility), which interact with each other and with titin and cMyBP-C to dictate filament architecture and function. The packing of myosin tails in the filament backbone is also resolved. The structure suggests how cMyBP-C helps to generate the cardiac super-relaxed state3; how titin and cMyBP-C may contribute to length-dependent activation4; and how mutations in myosin and cMyBP-C might disturb interactions, causing disease5,6. The reconstruction resolves past uncertainties and integrates previous data on cardiac muscle structure and function. It provides a new paradigm for interpreting structural, physiological and clinical observations, and for the design of potential therapeutic drugs.
Collapse
Affiliation(s)
- Debabrata Dutta
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Vu Nguyen
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kenneth S Campbell
- Department of Physiology and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | - Raúl Padrón
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Roger Craig
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
35
|
Huang X, Torre I, Chiappi M, Yin Z, Vydyanath A, Cao S, Raschdorf O, Beeby M, Quigley B, de Tombe PP, Liu J, Morris EP, Luther PK. Cryo-electron tomography of intact cardiac muscle reveals myosin binding protein-C linking myosin and actin filaments. J Muscle Res Cell Motil 2023; 44:165-178. [PMID: 37115473 PMCID: PMC10542292 DOI: 10.1007/s10974-023-09647-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/29/2023] [Indexed: 04/29/2023]
Abstract
Myosin binding protein C (MyBP-C) is an accessory protein of the thick filament in vertebrate cardiac muscle arranged over 9 stripes of intervals of 430 Å in each half of the A-band in the region called the C-zone. Mutations in cardiac MyBP-C are a leading cause of hypertrophic cardiomyopathy the mechanism of which is unknown. It is a rod-shaped protein composed of 10 or 11 immunoglobulin- or fibronectin-like domains labelled C0 to C10 which binds to the thick filament via its C-terminal region. MyBP-C regulates contraction in a phosphorylation dependent fashion that may be through binding of its N-terminal domains with myosin or actin. Understanding the 3D organisation of MyBP-C in the sarcomere environment may provide new light on its function. We report here the fine structure of MyBP-C in relaxed rat cardiac muscle by cryo-electron tomography and subtomogram averaging of refrozen Tokuyasu cryosections. We find that on average MyBP-C connects via its distal end to actin across a disc perpendicular to the thick filament. The path of MyBP-C suggests that the central domains may interact with myosin heads. Surprisingly MyBP-C at Stripe 4 is different; it has weaker density than the other stripes which could result from a mainly axial or wavy path. Given that the same feature at Stripe 4 can also be found in several mammalian cardiac muscles and in some skeletal muscles, our finding may have broader implication and significance. In the D-zone, we show the first demonstration of myosin crowns arranged on a uniform 143 Å repeat.
Collapse
Affiliation(s)
- Xinrui Huang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT, 06516, USA
| | - Iratxe Torre
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Michele Chiappi
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Zhan Yin
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Anupama Vydyanath
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Shuangyi Cao
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | | | - Morgan Beeby
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Bonnie Quigley
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Pieter P de Tombe
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Ave, Chicago, IL, 60612, USA
- Phymedexp, Université de Montpellier, Inserm, CNRS, Montpellier, France
| | - Jun Liu
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT, 06516, USA
| | - Edward P Morris
- Division of Structural Biology, Institute of Cancer Research, London, SW3 6JB, UK
- School of Molecular Biosciences, University of Glasgow, Garscube Campus, Jarrett Building, 351, Bearsden Road, Glasgow, G61 1QH, UK
| | - Pradeep K Luther
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK.
- Cardiac Function Section, National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
36
|
Sebastian SA, Padda I, Lehr EJ, Johal G. Aficamten: A Breakthrough Therapy for Symptomatic Obstructive Hypertrophic Cardiomyopathy. Am J Cardiovasc Drugs 2023; 23:519-532. [PMID: 37526885 DOI: 10.1007/s40256-023-00599-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/02/2023]
Abstract
Aficamten is a novel cardiac myosin inhibitor that has demonstrated its ability to safely lower left ventricular outflow tract (LVOT) gradients and improve heart failure symptoms in patients with obstructive hypertrophic cardiomyopathy (HCM). Based on the REDWOOD-HCM open label extension (OLE) study, participants receiving aficamten had significantly reduced resting and Valsalva LVOT gradient within 2 weeks after initiating treatment, with ongoing improvements over 24 weeks, and recent evidence suggests effects can sustain up to 48 weeks. While beta-blockers, calcium channel blockers, and disopyramide have shown some benefits in managing HCM, they have limited direct impact on the underlying disease process in patients with obstructive HCM. Aficamten achieves its therapeutic effect by reducing hypercontractility and improving diastolic function in obstructive HCM. Mavacamten was the first cardiac myosin inhibitor approved for symptomatic obstructive HCM. However, aficamten has a shorter human half-life (t1/2) and fewer drug-drug interactions, making it a preferable treatment option. This review evaluates the long-term clinical value and safety of aficamten in patients with obstructive HCM based on available data from completed and ongoing clinical trials. Additionally, the molecular basis of sarcomere-targeted therapy in reducing LVOT gradients is explored, and its potential in managing obstructive HCM is discussed.
Collapse
Affiliation(s)
- Sneha Annie Sebastian
- Department of Internal Medicine, Azeezia Medical College, Kollam, Kerala, India.
- , Airdrie, Canada.
| | - Inderbir Padda
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, NY, USA
| | - Eric J Lehr
- Division of Cardiac Surgery, Swedish Heart and Vascular Institute, Seattle, WA, USA
| | - Gurpreet Johal
- Department of Cardiology, University of Washington, Valley Medical Center, Seattle, WA, USA
| |
Collapse
|
37
|
Narayanan T, Chèvremont W, Zinn T. Small-angle X-ray scattering in the era of fourth-generation light sources. J Appl Crystallogr 2023; 56:939-946. [PMID: 37555224 PMCID: PMC10405582 DOI: 10.1107/s1600576723004971] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/05/2023] [Indexed: 08/10/2023] Open
Abstract
Recently, fourth-generation synchrotron sources with several orders of magnitude higher brightness and higher degree of coherence compared with third-generation sources have come into operation. These new X-ray sources offer exciting opportunities for the investigation of soft matter and biological specimens by small-angle X-ray scattering (SAXS) and related scattering methods. The improved beam properties together with the advanced pixel array detectors readily enhance the angular resolution of SAXS and ultra-small-angle X-ray scattering in the pinhole collimation. The high degree of coherence is a major boost for the X-ray photon correlation spectroscopy (XPCS) technique, enabling the equilibrium dynamics to be probed over broader time and length scales. This article presents some representative examples illustrating the performance of SAXS and XPCS with the Extremely Brilliant Source at the European Synchrotron Radiation Facility. The rapid onset of radiation damage is a significant challenge with the vast majority of samples, and appropriate protocols need to be adopted for circumventing this problem.
Collapse
Affiliation(s)
| | | | - Thomas Zinn
- ESRF – The European Synchrotron, 38043 Grenoble, France
- Diamond Light Source, Didcot OX11 0DE, United Kingdom
| |
Collapse
|
38
|
Nag S, Gollapudi SK, Del Rio CL, Spudich JA, McDowell R. Mavacamten, a precision medicine for hypertrophic cardiomyopathy: From a motor protein to patients. SCIENCE ADVANCES 2023; 9:eabo7622. [PMID: 37506209 DOI: 10.1126/sciadv.abo7622] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/23/2023] [Indexed: 07/30/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a primary myocardial disorder characterized by left ventricular hypertrophy, hyperdynamic contraction, and impaired relaxation of the heart. These functional derangements arise directly from altered sarcomeric function due to either mutations in genes encoding sarcomere proteins, or other defects such as abnormal energetics. Current treatment options do not directly address this causal biology but focus on surgical and extra-sarcomeric (sarcolemmal) pharmacological symptomatic relief. Mavacamten (formerly known as MYK-461), is a small molecule designed to regulate cardiac function at the sarcomere level by selectively but reversibly inhibiting the enzymatic activity of myosin, the fundamental motor of the sarcomere. This review summarizes the mechanism and translational progress of mavacamten from proteins to patients, describing how the mechanism of action and pharmacological characteristics, involving both systolic and diastolic effects, can directly target pathophysiological derangements within the cardiac sarcomere to improve cardiac structure and function in HCM. Mavacamten was approved by the Food and Drug Administration in April 2022 for the treatment of obstructive HCM and now goes by the commercial name of Camzyos. Full information about the risks, limitations, and side effects can be found at www.accessdata.fda.gov/drugsatfda_docs/label/2022/214998s000lbl.pdf.
Collapse
Affiliation(s)
- Suman Nag
- MyoKardia Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA 94005, USA
| | - Sampath K Gollapudi
- MyoKardia Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA 94005, USA
| | - Carlos L Del Rio
- MyoKardia Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA 94005, USA
- Cardiac Consulting, 1630 S Delaware St. #56426, San Mateo, CA 94403, USA
| | | | - Robert McDowell
- MyoKardia Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA 94005, USA
| |
Collapse
|
39
|
Lehman SJ, Meller A, Solieva SO, Lotthammer JM, Greenberg L, Langer SJ, Greenberg MJ, Tardiff JC, Bowman GR, Leinwand L. Divergent Molecular Phenotypes in Point Mutations at the Same Residue in Beta-Myosin Heavy Chain Lead to Distinct Cardiomyopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547580. [PMID: 37461648 PMCID: PMC10349964 DOI: 10.1101/2023.07.03.547580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
In genetic cardiomyopathies, a frequently described phenomenon is how similar mutations in one protein can lead to discrete clinical phenotypes. One example is illustrated by two mutations in beta myosin heavy chain (β-MHC) that are linked to hypertrophic cardiomyopathy (HCM) (Ile467Val, I467V) and left ventricular non-compaction (LVNC) (Ile467Thr, I467T). To investigate how these missense mutations lead to independent diseases, we studied the molecular effects of each mutation using recombinant human β-MHC Subfragment 1 (S1) in in vitro assays. Both HCM-I467V and LVNC-I467T S1 mutations exhibited similar mechanochemical function, including unchanged ATPase and enhanced actin velocity but had opposing effects on the super-relaxed (SRX) state of myosin. HCM-I467V S1 showed a small reduction in the SRX state, shifting myosin to a more actin-available state that may lead to the "gain-of-function" phenotype commonly described in HCM. In contrast, LVNC-I467T significantly increased the population of myosin in the ultra-slow SRX state. Interestingly, molecular dynamics simulations reveal that I467T allosterically disrupts interactions between ADP and the nucleotide-binding pocket, which may result in an increased ADP release rate. This predicted change in ADP release rate may define the enhanced actin velocity measured in LVNC-I467T, but also describe the uncoupled mechanochemical function for this mutation where the enhanced ADP release rate may be sufficient to offset the increased SRX population of myosin. These contrasting molecular effects may lead to contractile dysregulation that initiates LVNC-associated signaling pathways that progress the phenotype. Together, analysis of these mutations provides evidence that phenotypic complexity originates at the molecular level and is critical to understanding disease progression and developing therapies.
Collapse
Affiliation(s)
- Sarah J Lehman
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| | - Artur Meller
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
- Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO, USA
| | - Shahlo O Solieva
- University of Pennsylvania, Department of Biochemistry and Biophysics, Philadelphia, PA, USA
| | - Jeffrey M Lotthammer
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Lina Greenberg
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Stephen J Langer
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| | - Michael J Greenberg
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Jil C Tardiff
- University of Arizona, Department of Biomedical Engineering, Tucson, AZ, USA
| | - Gregory R Bowman
- University of Pennsylvania, Department of Biochemistry and Biophysics, Philadelphia, PA, USA
| | - Leslie Leinwand
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| |
Collapse
|
40
|
Fujita H, Kaneshiro J, Takeda M, Sasaki K, Yamamoto R, Umetsu D, Kuranaga E, Higo S, Kondo T, Asano Y, Sakata Y, Miyagawa S, Watanabe TM. Estimation of crossbridge-state during cardiomyocyte beating using second harmonic generation. Life Sci Alliance 2023; 6:e202302070. [PMID: 37236659 PMCID: PMC10215972 DOI: 10.26508/lsa.202302070] [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: 04/02/2023] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Estimation of dynamic change of crossbridge formation in living cardiomyocytes is expected to provide crucial information for elucidating cardiomyopathy mechanisms, efficacy of an intervention, and others. Here, we established an assay system to dynamically measure second harmonic generation (SHG) anisotropy derived from myosin filaments depended on their crossbridge status in pulsating cardiomyocytes. Experiments utilizing an inheritable mutation that induces excessive myosin-actin interactions revealed that the correlation between sarcomere length and SHG anisotropy represents crossbridge formation ratio during pulsation. Furthermore, the present method found that ultraviolet irradiation induced an increased population of attached crossbridges that lost the force-generating ability upon myocardial differentiation. Taking an advantage of infrared two-photon excitation in SHG microscopy, myocardial dysfunction could be intravitally evaluated in a Drosophila disease model. Thus, we successfully demonstrated the applicability and effectiveness of the present method to evaluate the actomyosin activity of a drug or genetic defect on cardiomyocytes. Because genomic inspection alone may not catch the risk of cardiomyopathy in some cases, our study demonstrated herein would be of help in the risk assessment of future heart failure.
Collapse
Affiliation(s)
- Hideaki Fujita
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Junichi Kaneshiro
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Maki Takeda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kensuke Sasaki
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Rikako Yamamoto
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Daiki Umetsu
- Laboratory for Histogenetic Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Erina Kuranaga
- Laboratory for Histogenetic Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Shuichiro Higo
- Department of Medical Therapeutics for Heart Failure, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takumi Kondo
- Department of Medical Therapeutics for Heart Failure, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomonobu M Watanabe
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
41
|
Kazmierczak K, Liang J, Maura LG, Scott NK, Szczesna-Cordary D. Phosphorylation Mimetic of Myosin Regulatory Light Chain Mitigates Cardiomyopathy-Induced Myofilament Impairment in Mouse Models of RCM and DCM. Life (Basel) 2023; 13:1463. [PMID: 37511838 PMCID: PMC10381296 DOI: 10.3390/life13071463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
This study focuses on mimicking constitutive phosphorylation in the N-terminus of the myosin regulatory light chain (S15D-RLC) as a rescue strategy for mutation-induced cardiac dysfunction in transgenic (Tg) models of restrictive (RCM) and dilated (DCM) cardiomyopathy caused by mutations in essential (ELC, MYL3 gene) or regulatory (RLC, MYL2 gene) light chains of myosin. Phosphomimetic S15D-RLC was reconstituted in left ventricular papillary muscle (LVPM) fibers from two mouse models of cardiomyopathy, RCM-E143K ELC and DCM-D94A RLC, along with their corresponding Tg-ELC and Tg-RLC wild-type (WT) mice. The beneficial effects of S15D-RLC in rescuing cardiac function were manifested by the S15D-RLC-induced destabilization of the super-relaxed (SRX) state that was observed in both models of cardiomyopathy. S15D-RLC promoted a shift from the SRX state to the disordered relaxed (DRX) state, increasing the number of heads readily available to interact with actin and produce force. Additionally, S15D-RLC reconstituted with fibers demonstrated significantly higher maximal isometric force per cross-section of muscle compared with reconstitution with WT-RLC protein. The effects of the phosphomimetic S15D-RLC were compared with those observed for Omecamtiv Mecarbil (OM), a myosin activator shown to bind to the catalytic site of cardiac myosin and increase myocardial contractility. A similar SRX↔DRX equilibrium shift was observed in OM-treated fibers as in S15D-RLC-reconstituted preparations. Additionally, treatment with OM resulted in significantly higher maximal pCa 4 force per cross-section of muscle fibers in both cardiomyopathy models. Our results suggest that both treatments with S15D-RLC and OM may improve the function of myosin motors and cardiac muscle contraction in RCM-ELC and DCM-RLC mice.
Collapse
Affiliation(s)
- Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, School of Medicine, University of Miami Miller, Miami, FL 33136, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, School of Medicine, University of Miami Miller, Miami, FL 33136, USA
| | - Luis G Maura
- Department of Molecular and Cellular Pharmacology, School of Medicine, University of Miami Miller, Miami, FL 33136, USA
| | - Natissa K Scott
- Department of Molecular and Cellular Pharmacology, School of Medicine, University of Miami Miller, Miami, FL 33136, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, School of Medicine, University of Miami Miller, Miami, FL 33136, USA
| |
Collapse
|
42
|
Grinzato A, Auguin D, Kikuti C, Nandwani N, Moussaoui D, Pathak D, Kandiah E, Ruppel KM, Spudich JA, Houdusse A, Robert-Paganin J. Cryo-EM structure of the folded-back state of human β-cardiac myosin. Nat Commun 2023; 14:3166. [PMID: 37258552 PMCID: PMC10232470 DOI: 10.1038/s41467-023-38698-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/11/2023] [Indexed: 06/02/2023] Open
Abstract
To save energy and precisely regulate cardiac contractility, cardiac muscle myosin heads are sequestered in an 'off' state that can be converted to an 'on' state when exertion is increased. The 'off' state is equated with a folded-back structure known as the interacting-heads motif (IHM), which is a regulatory feature of all class-2 muscle and non-muscle myosins. We report here the human β-cardiac myosin IHM structure determined by cryo-electron microscopy to 3.6 Å resolution, providing details of all the interfaces stabilizing the 'off' state. The structure shows that these interfaces are hot spots of hypertrophic cardiomyopathy mutations that are thought to cause hypercontractility by destabilizing the 'off' state. Importantly, the cardiac and smooth muscle myosin IHM structures dramatically differ, providing structural evidence for the divergent physiological regulation of these muscle types. The cardiac IHM structure will facilitate development of clinically useful new molecules that modulate IHM stability.
Collapse
Affiliation(s)
- Alessandro Grinzato
- CM01 beamline. European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Daniel Auguin
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France
- Laboratoire de Biologie des Ligneux et des Grandes Cultures, Université d'Orléans, UPRES EA 1207, INRA-USC1328, F-45067, Orléans, France
| | - Carlos Kikuti
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France
| | - Neha Nandwani
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dihia Moussaoui
- BM29 BIOSAXS beamline, European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Divya Pathak
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Eaazhisai Kandiah
- CM01 beamline. European Synchrotron Radiation Facility (ESRF), Grenoble, 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.
| | - James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France.
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France.
| |
Collapse
|
43
|
Prodanovic M, Wang Y, Mijailovich SM, Irving T. Using Multiscale Simulations as a Tool to Interpret Equatorial X-ray Fiber Diffraction Patterns from Skeletal Muscle. Int J Mol Sci 2023; 24:8474. [PMID: 37239821 PMCID: PMC10218096 DOI: 10.3390/ijms24108474] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Synchrotron small-angle X-ray diffraction is the method of choice for nm-scale structural studies of striated muscle under physiological conditions and on millisecond time scales. The lack of generally applicable computational tools for modeling X-ray diffraction patterns from intact muscles has been a significant barrier to exploiting the full potential of this technique. Here, we report a novel "forward problem" approach using the spatially explicit computational simulation platform MUSICO to predict equatorial small-angle X-ray diffraction patterns and the force output simultaneously from resting and isometrically contracting rat skeletal muscle that can be compared to experimental data. The simulation generates families of thick-thin filament repeating units, each with their individually predicted occupancies of different populations of active and inactive myosin heads that can be used to generate 2D-projected electron density models based on known Protein Data Bank structures. We show how, by adjusting only a few selected parameters, we can achieve a good correspondence between experimental and predicted X-ray intensities. The developments presented here demonstrate the feasibility of combining X-ray diffraction and spatially explicit modeling to form a powerful hypothesis-generating tool that can be used to motivate experiments that can reveal emergent properties of muscle.
Collapse
Affiliation(s)
- Momcilo Prodanovic
- Institute for Information Technologies, University of Kragujevac, 34000 Kragujevac, Serbia;
- FilamenTech, Inc., Newton, MA 02458, USA;
| | - Yiwei Wang
- Department of Applied Mathematics, Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
- Department of Mathematics, University of California, Riverside, CA 92521, USA
| | | | - Thomas Irving
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
44
|
Grinzato A, Auguin D, Kikuti C, Nandwani N, Moussaoui D, Pathak D, Kandiah E, Ruppel KM, Spudich JA, Houdusse A, Robert-Paganin J. Cryo-EM structure of the folded-back state of human β-cardiac myosin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.15.536999. [PMID: 37131793 PMCID: PMC10153137 DOI: 10.1101/2023.04.15.536999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
During normal levels of exertion, many cardiac muscle myosin heads are sequestered in an off-state even during systolic contraction to save energy and for precise regulation. They can be converted to an on-state when exertion is increased. Hypercontractility caused by hypertrophic cardiomyopathy (HCM) myosin mutations is often the result of shifting the equilibrium toward more heads in the on-state. The off-state is equated with a folded-back structure known as the interacting head motif (IHM), which is a regulatory feature of all muscle myosins and class-2 non-muscle myosins. We report here the human β-cardiac myosin IHM structure to 3.6 Å resolution. The structure shows that the interfaces are hot spots of HCM mutations and reveals details of the significant interactions. Importantly, the structures of cardiac and smooth muscle myosin IHMs are dramatically different. This challenges the concept that the IHM structure is conserved in all muscle types and opens new perspectives in the understanding of muscle physiology. The cardiac IHM structure has been the missing puzzle piece to fully understand the development of inherited cardiomyopathies. This work will pave the way for the development of new molecules able to stabilize or destabilize the IHM in a personalized medicine approach. *This manuscript was submitted to Nature Communications in August 2022 and dealt efficiently by the editors. All reviewers received this version of the manuscript before 9 208 August 2022. They also received coordinates and maps of our high resolution structure on the 18 208 August 2022. Due to slowness of at least one reviewer, this contribution was delayed for acceptance by Nature Communications and we are now depositing in bioRxiv the originally submitted version written in July 2022 for everyone to see. Indeed, two bioRxiv contributions at lower resolution but adding similar concepts on thick filament regulation were deposited this week in bioRxiv, one of the contributions having had access to our coordinates. We hope that our data at high resolution will be helpful for all readers that appreciate that high resolution information is required to build accurate atomic models and discuss implications for sarcomere regulation and the effects of cardiomyopathy mutations on heart muscle function.
Collapse
Affiliation(s)
- Alessandro Grinzato
- CM01 beamline. European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Daniel Auguin
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005 Paris, France
- Laboratoire de Biologie des Ligneux et des Grandes Cultures, Université d’Orléans, UPRES EA 1207, INRA-USC1328, F-45067 Orléans, France
| | - Carlos Kikuti
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005 Paris, France
| | - Neha Nandwani
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Dihia Moussaoui
- BM29 BIOSAXS beamline, European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Divya Pathak
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Eaazhisai Kandiah
- CM01 beamline. European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Kathleen M. Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, United States
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005 Paris, France
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005 Paris, France
| |
Collapse
|
45
|
Dutta D, Nguyen V, Campbell KS, Padrón R, Craig R. Cryo-EM structure of the human cardiac myosin filament. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536274. [PMID: 37090534 PMCID: PMC10120621 DOI: 10.1101/2023.04.11.536274] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Pumping of the heart is powered by filaments of the motor protein myosin, which pull on actin filaments to generate cardiac contraction. In addition to myosin, the filaments contain cardiac myosin-binding protein C (cMyBP-C), which modulates contractility in response to physiological stimuli, and titin, which functions as a scaffold for filament assembly 1 . Myosin, cMyBP-C and titin are all subject to mutation, which can lead to heart failure. Despite the central importance of cardiac myosin filaments to life, their molecular structure has remained a mystery for 60 years 2 . Here, we have solved the structure of the main (cMyBP-C-containing) region of the human cardiac filament to 6 Å resolution by cryo-EM. The reconstruction reveals the architecture of titin and cMyBP-C for the first time, and shows how myosin's motor domains (heads) form 3 different types of motif (providing functional flexibility), which interact with each other and with specific domains of titin and cMyBP-C to dictate filament architecture and regulate function. A novel packing of myosin tails in the filament backbone is also resolved. The structure suggests how cMyBP-C helps generate the cardiac super-relaxed state 3 , how titin and cMyBP-C may contribute to length-dependent activation 4 , and how mutations in myosin and cMyBP-C might disrupt interactions, causing disease 5, 6 . A similar structure is likely in vertebrate skeletal myosin filaments. The reconstruction resolves past uncertainties, and integrates previous data on cardiac muscle structure and function. It provides a new paradigm for interpreting structural, physiological and clinical observations, and for the design of potential therapeutic drugs.
Collapse
|
46
|
Nelson S, Beck-Previs S, Sadayappan S, Tong C, Warshaw DM. Myosin-binding protein C stabilizes, but is not the sole determinant of SRX myosin in cardiac muscle. J Gen Physiol 2023; 155:e202213276. [PMID: 36688870 PMCID: PMC9884578 DOI: 10.1085/jgp.202213276] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/08/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023] Open
Abstract
The myosin super-relaxed (SRX) state is central to striated muscle metabolic and functional regulation. In skeletal muscle, SRX myosin are predominantly colocalized with myosin-binding protein C (MyBP-C) in the sarcomere C-zone. To define how cardiac MyBP-C (cMyBP-C) and its specific domains contribute to stabilizing the SRX state in cardiac muscle, we took advantage of transgenic cMyBP-C null mice and those expressing cMyBP-C with a 271-residue N-terminal truncation. Utilizing super-resolution microscopy, we determined the lifetime and subsarcomeric location of individual fluorescent-ATP turnover events within isolated cardiac myofibrils. The proportion of SRX myosin demonstrated a gradient along the half-thick filament, highest in the P- and C-zones (72 ± 9% and 71 ± 6%, respectively) and lower in the D-zone (45 ± 10%), which lies farther from the sarcomere center and lacks cMyBP-C, suggesting a possible role for cMyBP-C in stabilizing the SRX. However, myofibrils from cMyBP-C null mice demonstrated an ∼40% SRX reduction, not only within the now cMyBP-C-free C-zone (49 ± 9% SRX), but also within the D-zone (22 ± 5% SRX). These data suggest that the influence of cMyBP-C on the SRX state is not limited to the C-zone but extends along the thick filament. Interestingly, myofibrils with N-terminal truncated cMyBP-C had an SRX content and spatial gradient similar to the cMyBP-C null, indicating that the N terminus of cMyBP-C is necessary for cMyBP-C's role in enhancing the SRX gradient along the entire thick filament. Given that SRX myosin exist as a gradient along the thick filament that is highest in the C-zone, even in the absence of cMyBP-C or its N-terminus, an inherent bias must exist in the structure of the thick filament to stabilize the SRX state.
Collapse
Affiliation(s)
- Shane Nelson
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Samantha Beck-Previs
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Carl Tong
- Department of Medical Physiology, Texas A&M University, Bryan, TX, USA
| | - David M. Warshaw
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT, USA
| |
Collapse
|
47
|
Tomalka A. Eccentric muscle contractions: from single muscle fibre to whole muscle mechanics. Pflugers Arch 2023; 475:421-435. [PMID: 36790515 PMCID: PMC10011336 DOI: 10.1007/s00424-023-02794-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/24/2023] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
Eccentric muscle loading encompasses several unique features compared to other types of contractions. These features include increased force, work, and performance at decreased oxygen consumption, reduced metabolic cost, improved energy efficiency, as well as decreased muscle activity. This review summarises explanatory approaches to long-standing questions in terms of muscular contraction dynamics and molecular and cellular mechanisms underlying eccentric muscle loading. Moreover, this article intends to underscore the functional link between sarcomeric components, emphasising the fundamental role of titin in skeletal muscle. The giant filament titin reveals versatile functions ranging from sarcomere organisation and maintenance, providing passive tension and elasticity, and operates as a mechanosensory and signalling platform. Structurally, titin consists of a viscoelastic spring segment that allows activation-dependent coupling to actin. This titin-actin interaction can explain linear force increases in active lengthening experiments in biological systems. A three-filament model of skeletal muscle force production (mediated by titin) is supposed to overcome significant deviations between experimental observations and predictions by the classic sliding-filament and cross-bridge theories. Taken together, this review intends to contribute to a more detailed understanding of overall muscle behaviour and force generation-from a microscopic sarcomere level to a macroscopic multi-joint muscle level-impacting muscle modelling, the understanding of muscle function, and disease.
Collapse
Affiliation(s)
- André Tomalka
- Motion and Exercise Science, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
48
|
Marcucci L. Muscle Mechanics and Thick Filament Activation: An Emerging Two-Way Interaction for the Vertebrate Striated Muscle Fine Regulation. Int J Mol Sci 2023; 24:ijms24076265. [PMID: 37047237 PMCID: PMC10094676 DOI: 10.3390/ijms24076265] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Contraction in striated muscle is classically described as regulated by calcium-mediated structural changes in the actin-containing thin filaments, which release the binding sites for the interaction with myosin motors to produce force. In this view, myosin motors, arranged in the thick filaments, are basically always ready to interact with the thin filaments, which ultimately regulate the contraction. However, a new “dual-filament” activation paradigm is emerging, where both filaments must be activated to generate force. Growing evidence from the literature shows that the thick filament activation has a role on the striated muscle fine regulation, and its impairment is associated with severe pathologies. This review is focused on the proposed mechanical feedback that activates the inactive motors depending on the level of tension generated by the active ones, the so-called mechanosensing mechanism. Since the main muscle function is to generate mechanical work, the implications on muscle mechanics will be highlighted, showing: (i) how non-mechanical modulation of the thick filament activation influences the contraction, (ii) how the contraction influences the activation of the thick filament and (iii) how muscle, through the mechanical modulation of the thick filament activation, can regulate its own mechanics. This description highlights the crucial role of the emerging bi-directional feedback on muscle mechanical performance.
Collapse
Affiliation(s)
- Lorenzo Marcucci
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
- Center for Biosystems Dynamics Research, RIKEN, Suita 565-0874, Japan
| |
Collapse
|
49
|
Fan X, Zheng X, An T, Li X, Leung N, Zhu B, Sui T, Shi N, Fan T, Zhao Q. Light diffraction by sarcomeres produces iridescence in transmission in the transparent ghost catfish. Proc Natl Acad Sci U S A 2023; 120:e2219300120. [PMID: 36913569 PMCID: PMC10041080 DOI: 10.1073/pnas.2219300120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/07/2023] [Indexed: 03/15/2023] Open
Abstract
Despite the elaborate varieties of iridescent colors in biological species, most of them are reflective. Here we show the rainbow-like structural colors found in the ghost catfish (Kryptopterus vitreolus), which exist only in transmission. The fish shows flickering iridescence throughout the transparent body. The iridescence originates from the collective diffraction of light after passing through the periodic band structures of the sarcomeres inside the tightly stacked myofibril sheets, and the muscle fibers thus work as transmission gratings. The length of the sarcomeres varies from ~1 μm from the body neutral plane near the skeleton to ~2 μm next to the skin, and the iridescence of a live fish mainly results from the longer sarcomeres. The length of the sarcomere changes by ~80 nm as it relaxes and contracts, and the fish shows a quickly blinking dynamic diffraction pattern as it swims. While similar diffraction colors are also observed in thin slices of muscles from non-transparent species such as the white crucian carps, a transparent skin is required indeed to have such iridescence in live species. The ghost catfish skin is of a plywood structure of collagen fibrils, which allows more than 90% of the incident light to pass directly into the muscles and the diffracted light to exit the body. Our findings could also potentially explain the iridescence in other transparent aquatic species, including the eel larvae (Leptocephalus) and the icefishes (Salangidae).
Collapse
Affiliation(s)
- Xiujun Fan
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| | - Xuezhi Zheng
- Department of Electrical Engineering, KU Leuven, LeuvenB3001, Belgium
| | - Tong An
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| | - Xiuhong Li
- Shanghai Synchrotron Radiation Facility, Shanghai201204, China
| | - Nathanael Leung
- School of Mechanical Engineering Sciences, University of Surrey, SurreyGU2 7XH, UK
| | - Bin Zhu
- School of Mechanical Engineering Sciences, University of Surrey, SurreyGU2 7XH, UK
| | - Tan Sui
- School of Mechanical Engineering Sciences, University of Surrey, SurreyGU2 7XH, UK
| | - Nan Shi
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| | - Tongxiang Fan
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| | - Qibin Zhao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| |
Collapse
|
50
|
Tanner BCW, Awinda PO, Agonias KB, Attili S, Blair CA, Thompson MS, Walker LA, Kampourakis T, Campbell KS. Sarcomere length affects Ca2+ sensitivity of contraction in ischemic but not non-ischemic myocardium. J Gen Physiol 2023; 155:213800. [PMID: 36633584 PMCID: PMC9859763 DOI: 10.1085/jgp.202213200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/18/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
In healthy hearts, myofilaments become more sensitive to Ca2+ as the myocardium is stretched. This effect is known as length-dependent activation and is an important cellular-level component of the Frank-Starling mechanism. Few studies have measured length-dependent activation in the myocardium from failing human hearts. We investigated whether ischemic and non-ischemic heart failure results in different length-dependent activation responses at physiological temperature (37°C). Myocardial strips from the left ventricular free wall were chemically permeabilized and Ca2+-activated at sarcomere lengths (SLs) of 1.9 and 2.3 µm. Data were acquired from 12 hearts that were explanted from patients receiving cardiac transplants; 6 had ischemic heart failure and 6 had non-ischemic heart failure. Another 6 hearts were obtained from organ donors. Maximal Ca2+-activated force increased at longer SL for all groups. Ca2+ sensitivity increased with SL in samples from donors (P < 0.001) and patients with ischemic heart failure (P = 0.003) but did not change with SL in samples from patients with non-ischemic heart failure. Compared with donors, troponin I phosphorylation decreased in ischemic samples and even more so in non-ischemic samples; cardiac myosin binding protein-C (cMyBP-C) phosphorylation also decreased with heart failure. These findings support the idea that troponin I and cMyBP-C phosphorylation promote length-dependent activation and show that length-dependent activation of contraction is blunted, yet extant, in the myocardium from patients with ischemic heart failure and further reduced in the myocardium from patients with non-ischemic heart failure. Patients who have a non-ischemic disease may exhibit a diminished contractile response to increased ventricular filling.
Collapse
Affiliation(s)
- Bertrand C W Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Peter O Awinda
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Keinan B Agonias
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Seetharamaiah Attili
- Randall Centre for Cell and Molecular Biophysics, King's College London , London, UK
| | - Cheavar A Blair
- Department of Physiology, University of Kentucky , Lexington, KY, USA
| | - Mindy S Thompson
- Department of Physiology, University of Kentucky , Lexington, KY, USA
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus , Aurora, CO, USA
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics, King's College London , London, UK
| | - Kenneth S Campbell
- Department of Physiology, University of Kentucky , Lexington, KY, USA.,Division of Cardiovascular Medicine, University of Kentucky , Lexington, KY, USA
| |
Collapse
|