1
|
Elst T, Weidner S, Tomalka A, Hahn D, Paternoster FK, Seiberl W, Siebert T. Consecutive SSCs increase the SSC effect in skinned rat muscle fibres. Pflugers Arch 2025:10.1007/s00424-025-03088-2. [PMID: 40338284 DOI: 10.1007/s00424-025-03088-2] [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: 11/26/2024] [Revised: 04/03/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
Muscle function is essential for generating force and movement, with stretch-shortening cycles (SSCs) playing a fundamental role in the economy of everyday locomotion. Compared with pure shortening contractions, the SSC effect is characterised by increased force, work, and power output during the SSC shortening phase. Few studies have investigated whether SSC effects transfer across consecutive SSCs. Therefore, we investigated SSC effects over three consecutive SSCs in skinned rat muscle fibres by analysing the isometric force immediately before stretch onset (Fonset), the peak force at the end of stretching (Fpeak), and the minimum force at the end of shortening (Fmin), along with mechanical (WorkSSC) and shortening work (WorkSHO) at different activation levels (20%, 60%, and 100%). Each SSC was followed by an isometric hold phase, allowing force to return to a steady state. Results indicated an increase in both Fpeak (20.3%) and WorkSSC (60.9%) from SSC1 to SSC3 across all activation levels tested. At 20% and 60% activation, Fonset, Fmin, and WorkSHO increased (range: 4.5-28.5%) from SSC1 to SSC3. However, at 100% activation, Fonset and WorkSHO remained unchanged, while Fmin decreased (- 8.5%) from SSC1 to SSC3. These results suggest that the increase in SSC effects at submaximal activation may be primarily due to increased cross-bridge forces. The absence of increases in Fonset, Fmin, and WorkSHO at 100% activation suggests that increases in Fpeak and WorkSSC may not be attributed to increased cross-bridge force but could instead be caused by additional effects, possibly involving modulation of non-cross-bridge structures, likely titin, and their stiffness.
Collapse
Affiliation(s)
- Tobias Elst
- Motion and Exercise Science, University of Stuttgart, Stuttgart, Germany.
| | - Sven Weidner
- Motion and Exercise Science, University of Stuttgart, Stuttgart, Germany
| | - André Tomalka
- Motion and Exercise Science, University of Stuttgart, Stuttgart, Germany
| | - Daniel Hahn
- Human Movement Science, Faculty of Sports Science, Ruhr University Bochum, Bochum, Germany
- School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Florian Kurt Paternoster
- Biomechanics in Sports, Department of Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Wolfgang Seiberl
- Human Movement Science, University of the Bundeswehr Munich, Neubiberg, Germany
| | - Tobias Siebert
- Motion and Exercise Science, University of Stuttgart, Stuttgart, Germany
- Stuttgart Center for Simulation Science, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
2
|
Lewis CTA, Moreno-Justicia R, Savoure L, Calvo E, Bak A, Laitila J, Seaborne RAE, Larsen S, Iwamoto H, Cefis M, Morais JA, Gouspillou G, Alegre-Cebollada J, Hawke TJ, Vazquez J, Adrover M, Marcangeli V, Hammad R, Granet J, Gaudreau P, Aubertin-Leheudre M, Bélanger M, Robitaille R, Deshmukh AS, Ochala J. Dysregulated skeletal muscle myosin super-relaxation and energetics in male participants with type 2 diabetes mellitus. Diabetologia 2025:10.1007/s00125-025-06436-0. [PMID: 40295335 DOI: 10.1007/s00125-025-06436-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/04/2025] [Indexed: 04/30/2025]
Abstract
AIMS/HYPOTHESIS Disrupted energy balance is critical for the onset and development of type 2 diabetes mellitus. Understanding of the exact underlying metabolic mechanisms remains incomplete, but skeletal muscle is thought to play an important pathogenic role. As the super-relaxed state of its most abundant protein, myosin, regulates cellular energetics, we aimed to investigate whether it is altered in individuals with type 2 diabetes. METHODS We used vastus lateralis biopsy specimens (obtained from patients with type 2 diabetes and control participants with similar characteristics), and ran a combination of structural and functional assays consisting of loaded 2'- (or 3')-O-(N-methylanthraniloyl)-ATP (Mant-ATP) chase experiments, x-ray diffraction and LC-MS/MS proteomics in isolated muscle fibres. RESULTS Our studies revealed a greater muscle myosin super-relaxation and decreased ATP demand in male participants with type 2 diabetes than in control participants. Subsequent proteomic analyses indicated that these (mal)adaptations probably originated from remodelled sarcomeric proteins and greater myosin glycation levels in patients than in control participants. CONCLUSIONS/INTERPRETATION Overall, our findings indicate a complex molecular dysregulation of myosin super-relaxed state and energy consumption in male participants with type 2 diabetes. Ultimately, pharmacological targeting of myosin could benefit skeletal muscle and whole-body metabolic health through enhancement of ATP consumption. DATA AVAILABILITY The raw MS data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD053022.
Collapse
Affiliation(s)
| | - Roger Moreno-Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lola Savoure
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Enrique Calvo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Agata Bak
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jenni Laitila
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert A E Seaborne
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Centre for Human and Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Steen Larsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Hiroyuki Iwamoto
- SPring-8, Japan Synchrotron Radiation Research Institute, Hyogo, Japan
| | - Marina Cefis
- Département des Sciences de l'Activité Physique, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
- Groupe de Recherche en Activité Physique Adaptée, Montréal, PQ, Canada
| | - Jose A Morais
- Department of Medicine, Research Institute of the McGill University Health Centre, Montréal, PQ, Canada
| | - Gilles Gouspillou
- Département des Sciences de l'Activité Physique, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
- Groupe de Recherche en Activité Physique Adaptée, Montréal, PQ, Canada
| | | | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jesús Vazquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Miquel Adrover
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Institut d'Investigació Sanitària Illes Balears (IdISBa), Departament de Química, Universitat de les Illes Balears, Palma de Mallorca, Spain
| | - Vincent Marcangeli
- Département des Sciences de l'Activité Physique, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
- Groupe de Recherche en Activité Physique Adaptée, Montréal, PQ, Canada
- Département des Sciences Biologiques, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
| | - Rami Hammad
- Département des Sciences de l'Activité Physique, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
- Groupe de Recherche en Activité Physique Adaptée, Montréal, PQ, Canada
- Département des Sciences Biologiques, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, PQ, Canada
- Al-Ahliyya Amman University, Faculty of Educational Sciences, Department of Physical and Health Education, Amman, Jordan
| | - Jordan Granet
- Département des Sciences Biologiques, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, PQ, Canada
| | - Pierrette Gaudreau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Département de médecine, Université de Montréal, Montréal, PQ, Canada
| | - Mylène Aubertin-Leheudre
- Département des Sciences de l'Activité Physique, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
- Groupe de Recherche en Activité Physique Adaptée, Montréal, PQ, Canada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, PQ, Canada
| | - Marc Bélanger
- Département des Sciences de l'Activité Physique, Faculté des Sciences, L'Université du Québec à Montréal (UQAM), Montréal, PQ, Canada
- Groupe de Recherche en Activité Physique Adaptée, Montréal, PQ, Canada
| | - Richard Robitaille
- Groupe de Recherche en Activité Physique Adaptée, Montréal, PQ, Canada
- Département de Neurosciences, Université de Montréal, Montréal, PQ, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l'Apprentissage, Université de Montréal, Montréal, PQ, Canada
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Julien Ochala
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
3
|
Jani VP, Ma W. Thick-Filament-Based Regulation and the Determinants of Force Generation. Biomedicines 2025; 13:703. [PMID: 40149679 PMCID: PMC11939844 DOI: 10.3390/biomedicines13030703] [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: 01/29/2025] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Thick-filament-based regulation in muscle is generally conceived as processes that modulate the number of myosin heads capable of force generation. It has been generally assumed that biochemical and structural assays of myosin active and inactive states provide equivalent measures of myosin recruitment, but recent studies indicate that this may not always be the case. Here, we studied the steady-state and dynamic mechanical changes in skinned porcine myocardium before and after treatment with omecamtiv mecarbil (OM) or piperine to help decipher how the biochemical and structural states of myosin separately affect contractile force. Methods: Force-Ca2+ relationships were obtained from skinned cardiomyocytes isolated from porcine myocardium before and after exposure to 1 μM OM and 7 μM piperine. Crossbridge kinetics were acquired using a step response stretch activation protocol allowing myosin attachment and detachment rates to be calculated. Results: OM augmented calcium-activated force at submaximal calcium levels that can be attributed to increased thick filament recruitment, increases in calcium sensitivity, an increased duty ratio, and from decelerated crossbridge detachment resulting in slowed crossbridge cycling kinetics. Piperine, in contrast, was able to increase activated force at submaximal calcium levels without appreciably affecting crossbridge cycling kinetics. Conclusions: Our study supports the notion that thick filament activation is primarily a process of myosin recruitment that is not necessarily coupled with the chemo-cycling of crossbridges. These new insights into thick filament activation mechanisms will need to be considered in the design of sarcomere-based therapies for treatment of myopathies.
Collapse
Affiliation(s)
- Vivek P. Jani
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Weikang Ma
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60016, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL 60016, USA
| |
Collapse
|
4
|
Kochurova AM, Beldiia EA, Antonets JY, Nefedova VV, Ryabkova NS, Katrukha IA, Bershitsky SY, Matyushenko AM, Kopylova GV, Shchepkin DV. Mavacamten Inhibits the Effect of the N-Terminal Fragment of Cardiac Myosin-Binding Protein C with the L352P Mutation on the Actin-Myosin Interaction at Low Calcium Concentrations. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:389-399. [PMID: 40367081 DOI: 10.1134/s0006297924604131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 05/16/2025]
Abstract
Hypertrophic cardiomyopathy (HCM)-associated mutations in sarcomeric proteins lead to the disruption of the actin-myosin interaction and its calcium regulation and cause myocardial hypercontractility. About half of such mutations are found in the MYBPC3 gene encoding cardiac myosin-binding protein C (cMyBP-C). A new approach to normalize cardiac contractile function in HCM is the use of β-cardiac myosin function inhibitors, one of which is mavacamten. We studied the effect of mavacamten on the calcium regulation of the actin-myosin interaction using isolated cardiac contractile proteins in the in vitro motility assay. The L352P mutation did not affect the maximum sliding velocity of regulated thin filaments on myosin in the in vitro motility assay and the calcium sensitivity of the velocity but led to the underinhibition of the actin-myosin interaction at low calcium concentrations. Mavacamten decreased the maximum sliding velocity of thin filaments in the presence of the WT and L352P C0-C2 fragments, and abolished their movement in the presence of the L352P C0-C2 fragment at low calcium concentrations. Slowing down the kinetics of cross-bridges and inhibition of actin-myosin interaction at low calcium concentrations by mavacamten may reduce the hypercontractility in HCM and the degree of myocardial hypertrophy.
Collapse
Affiliation(s)
- Anastasia M Kochurova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Evgenia A Beldiia
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Julia Y Antonets
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Victoria V Nefedova
- Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Natalia S Ryabkova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Hytest Ltd., Turku, 20520, Finland
| | - Ivan A Katrukha
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Hytest Ltd., Turku, 20520, Finland
| | - Sergey Y Bershitsky
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | | | - Galina V Kopylova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Daniil V Shchepkin
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia.
| |
Collapse
|
5
|
Granzier HL, Labeit S. Discovery of Titin and Its Role in Heart Function and Disease. Circ Res 2025; 136:135-157. [PMID: 39745989 DOI: 10.1161/circresaha.124.323051] [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: 09/08/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/04/2025]
Abstract
This review examines the giant elastic protein titin and its critical roles in heart function, both in health and disease, as discovered since its identification nearly 50 years ago. Encoded by the TTN (titin gene), titin has emerged as a major disease locus for cardiac disorders. Functionally, titin acts as a third myofilament type, connecting sarcomeric Z-disks and M-bands, and regulating myocardial passive stiffness and stretch sensing. Its I-band segment, which includes the N2B element and the PEVK (proline, glutamate, valine, and lysine-rich regions), serves as a viscoelastic spring, adjusting sarcomere length and force in response to cardiac stretch. The review details how alternative splicing of titin pre-mRNA produces different isoforms that greatly impact passive tension and cardiac function, under physiological and pathological conditions. Key posttranslational modifications, especially phosphorylation, play crucial roles in adjusting titin's stiffness, allowing for rapid adaptation to changing hemodynamic demands. Abnormal titin modifications and dysregulation of isoforms are linked to cardiac diseases such as heart failure with preserved ejection fraction, where increased stiffness impairs diastolic function. In addition, the review discusses the importance of the A-band region of titin in setting thick filament length and enhancing Ca²+ sensitivity, contributing to the Frank-Starling Mechanism of the heart. TTN truncating variants are frequently associated with dilated cardiomyopathy, and the review outlines potential disease mechanisms, including haploinsufficiency, sarcomere disarray, and altered thick filament regulation. Variants in TTN have also been linked to conditions such as peripartum cardiomyopathy and chemotherapy-induced cardiomyopathy. Therapeutic avenues are explored, including targeting splicing factors such as RBM20 (RNA binding motif protein 20) to adjust isoform ratios or using engineered heart tissues to study disease mechanisms. Advances in genetic engineering, including CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats), offer promise for modifying TTN to treat titin-related cardiomyopathies. This comprehensive review highlights titin's structural, mechanical, and signaling roles in heart function and the impact of TTN mutations on cardiac diseases.
Collapse
Affiliation(s)
- Henk L Granzier
- Department of Cellular and Molecular Medicine, Molecular Cardiovascular Research Program, The University of Arizona, Tucson (H.L.G.)
| | - Siegfried Labeit
- Department of Integrative Pathophysiology, Medical Faculty Mannheim, DZHK Partnersite Mannheim-Heidelberg, University of Heidelberg, Germany (S.L.)
| |
Collapse
|
6
|
De Napoli C, Schmidt L, Montesel M, Cussonneau L, Sanniti S, Marcucci L, Germinario E, Kindberg J, Evans AL, Gauquelin-Koch G, Narici M, Bertile F, Lefai E, Krüger M, Nogara L, Blaauw B. Reduced ATP turnover during hibernation in relaxed skeletal muscle. Nat Commun 2025; 16:80. [PMID: 39747078 PMCID: PMC11696273 DOI: 10.1038/s41467-024-55565-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Hibernating brown bears, due to a drastic reduction in metabolic rate, show only moderate muscle wasting. Here, we evaluate if ATPase activity of resting skeletal muscle myosin can contribute to this energy sparing. By analyzing single muscle fibers taken from the same bears, either during hibernation or in summer, we find that fibers from hibernating bears have a mild decline in force production and a significant reduction in ATPase activity. Single fiber proteomics, western blotting, and immunohistochemical analyses reveal major remodeling of the mitochondrial proteome during hibernation. Furthermore, using bioinformatical approaches and western blotting we find that phosphorylated myosin light chain, a known stimulator of basal myosin ATPase activity, is decreased in hibernating and disused muscles. These results suggest that skeletal muscle limits energy loss by reducing myosin ATPase activity, indicating a possible role for myosin ATPase activity modulation in multiple muscle wasting conditions.
Collapse
Affiliation(s)
- Cosimo De Napoli
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, 35131, University of Padova, Padova, Italy
| | - Luisa Schmidt
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Mauro Montesel
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, 35131, University of Padova, Padova, Italy
| | - Laura Cussonneau
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, 35131, University of Padova, Padova, Italy
| | - Samuele Sanniti
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Lorenzo Marcucci
- Department of Biomedical Sciences, 35131, University of Padova, Padova, Italy
| | - Elena Germinario
- Department of Biomedical Sciences, 35131, University of Padova, Padova, Italy
| | - Jonas Kindberg
- Norwegian Institute for Nature Research, Trondheim, Norway
- Department of Wildlife, Fish and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Alina Lynn Evans
- Department of Forestry and Wildlife Management, Faculty of Applied Ecology and Biotechnology, Inland Norway University of Applied Sciences, Koppang, Norway
| | | | - Marco Narici
- Department of Biomedical Sciences, 35131, University of Padova, Padova, Italy
| | - Fabrice Bertile
- Université de Strasbourg, CNRS, IPHC UMR 7178, 7, Strasbourg, Cedex 2, France
- National Proteomics Infrastructure, ProFi, Strasbourg, France
| | - Etienne Lefai
- Université Clermont Auvergne, INRAE, UNH UMR 1019, CRNH Auvergne, Clermont-Ferrand, France
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| | - Leonardo Nogara
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.
- Department of Biomedical Sciences, 35131, University of Padova, Padova, Italy.
- Department of Pharmaceutical Sciences, 35131, University of Padova, Padova, Italy.
| | - Bert Blaauw
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.
- Department of Biomedical Sciences, 35131, University of Padova, Padova, Italy.
| |
Collapse
|
7
|
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
|
8
|
Goluguri RR, Guhathakurta P, Nandwani N, Dawood A, Yakota S, Roopnarine O, Thomas DD, Spudich JA, Ruppel KM. A FRET assay to quantitate levels of the human β-cardiac myosin interacting heads motif based on its near-atomic resolution structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.626936. [PMID: 39713291 PMCID: PMC11661104 DOI: 10.1101/2024.12.05.626936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
In cardiac muscle, many myosin molecules are in a resting or "OFF" state with their catalytic heads in a folded structure known as the interacting heads motif (IHM). Many mutations in the human β-cardiac myosin gene that cause hypertrophic cardiomyopathy (HCM) are thought to destabilize (decrease the population of) the IHM state. The effects of pathogenic mutations on the IHM structural state are often studied using indirect assays, including a single-ATP turnover assay that detects the super-relaxed (SRX) biochemical state of myosin functionally. Here we develop and use a fluorescence resonance energy transfer (FRET) based sensor for direct quantification of the IHM state in solution. The FRET sensor was able to quantify destabilization of the IHM state in solution, induced by (a) increasing salt concentration, (b) altering proximal S2 tail length, or (c) introducing the HCM mutation P710R, as well as stabilization of the IHM state by introducing a dilated cardiomyopathy-causing mutation (E525K). Our FRET sensor conclusively showed that these perturbations indeed alter the structural IHM state. These results establish that the structural IHM state is one of the structural correlates of the biochemical SRX state in solution.
Collapse
Affiliation(s)
- Rama Reddy Goluguri
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, United States
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
| | - Piyali Guhathakurta
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Neha Nandwani
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, United States
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
| | - Aminah Dawood
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, United States
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
| | - Seiji Yakota
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, United States
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
| | - Osha Roopnarine
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - David D Thomas
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, United States
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
| | - Kathleen M. Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, United States
- Stanford Cardiovascular Institute, Stanford, CA 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, United States
| |
Collapse
|
9
|
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
|
10
|
van der Zee TJ, Wong JD, Kuo AD. On the rate-limiting dynamics of force development in muscle. J Exp Biol 2024; 227:jeb247436. [PMID: 39263848 DOI: 10.1242/jeb.247436] [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: 03/19/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024]
Abstract
Skeletal muscles produce forces relatively slowly compared with the action potentials that excite them. The dynamics of force production are governed by multiple processes, such as calcium activation, cycling of cross-bridges between myofilaments, and contraction against elastic tissues and the body. These processes have been included piecemeal in some muscle models, but not integrated to reveal which are the most rate limiting. We therefore examined their integrative contributions to force development in two conventional types of muscle models: Hill-type and cross-bridge. We found that no combination of these processes can self-consistently reproduce classic data such as twitch and tetanus. Rather, additional dynamics are needed following calcium activation and facilitating cross-bridge cycling, such as for cooperative myofilament interaction and reconfiguration. We provisionally lump such processes into a simple first-order model of 'force facilitation dynamics' that integrate into a cross-bridge-type muscle model. The proposed model self-consistently reproduces force development for a range of excitations including twitch and tetanus and electromyography-to-force curves. The model's step response reveals relatively small timing contributions of calcium activation (3%), cross-bridge cycling (3%) and contraction (27%) to overall force development of human quadriceps, with the remainder (67%) explained by force facilitation. The same set of model parameters predicts the change in force magnitude (gain) and timing (phase delay) as a function of excitatory firing rate, or as a function of cyclic contraction frequency. Although experiments are necessary to reveal the dynamics of muscle, integrative models are useful for identifying the main rate-limiting processes.
Collapse
Affiliation(s)
- Tim J van der Zee
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada, T2N 1N4
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada, T2N 1N4
| | - Jeremy D Wong
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada, T2N 1N4
| | - Arthur D Kuo
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada, T2N 1N4
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada, T2N 1N4
| |
Collapse
|
11
|
Greenberg MJ. Super-relaxed or super-stressed: Modeling length-dependent activation in cardiac muscle. Biophys J 2024; 123:2957-2960. [PMID: 39068513 PMCID: PMC11427768 DOI: 10.1016/j.bpj.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024] Open
Affiliation(s)
- Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
12
|
Schmidt AA, Grosberg AY, Grosberg A. A novel kinetic model to demonstrate the independent effects of ATP and ADP/Pi concentrations on sarcomere function. PLoS Comput Biol 2024; 20:e1012321. [PMID: 39102392 PMCID: PMC11326600 DOI: 10.1371/journal.pcbi.1012321] [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: 02/26/2024] [Revised: 08/15/2024] [Accepted: 07/12/2024] [Indexed: 08/07/2024] Open
Abstract
Understanding muscle contraction mechanisms is a standing challenge, and one of the approaches has been to create models of the sarcomere-the basic contractile unit of striated muscle. While these models have been successful in elucidating many aspects of muscle contraction, they fall short in explaining the energetics of functional phenomena, such as rigor, and in particular, their dependence on the concentrations of the biomolecules involved in the cross-bridge cycle. Our hypothesis posits that the stochastic time delay between ATP adsorption and ADP/Pi release in the cross-bridge cycle necessitates a modeling approach where the rates of these two reaction steps are controlled by two independent parts of the total free energy change of the hydrolysis reaction. To test this hypothesis, we built a two-filament, stochastic-mechanical half-sarcomere model that separates the energetic roles of ATP and ADP/Pi in the cross-bridge cycle's free energy landscape. Our results clearly demonstrate that there is a nontrivial dependence of the cross-bridge cycle's kinetics on the independent concentrations of ATP, ADP, and Pi. The simplicity of the proposed model allows for analytical solutions of the more basic systems, which provide novel insight into the dominant mechanisms driving some of the experimentally observed contractile phenomena.
Collapse
Affiliation(s)
- Andrew A Schmidt
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, Irvine, California, United States of America
| | - Alexander Y Grosberg
- Department of Physics and Center for Soft Matter Research, New York University, New York, New York, United States of America
| | - Anna Grosberg
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, Irvine, California, United States of America
- Department of Chemical & Biomolecular Engineering, University of California, Irvine, Irvine, California, United States of America
- The NSF-Simons Center for Multiscale Cell Fate Research and Sue and Bill Gross Stem Cell Research Center and Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
| |
Collapse
|
13
|
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
|
14
|
van den Berg M, Shi Z, Claassen WJ, Hooijman P, Lewis CTA, Andersen JL, van der Pijl RJ, Bogaards SJP, Conijn S, Peters EL, Begthel LPL, Uijterwijk B, Lindqvist J, Langlais PR, Girbes ARJ, Stapel S, Granzier H, Campbell KS, Ma W, Irving T, Hwee DT, Hartman JJ, Malik FI, Paul M, Beishuizen A, Ochala J, Heunks L, Ottenheijm CAC. Super-relaxed myosins contribute to respiratory muscle hibernation in mechanically ventilated patients. Sci Transl Med 2024; 16:eadg3894. [PMID: 39083588 PMCID: PMC11586073 DOI: 10.1126/scitranslmed.adg3894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/12/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Patients receiving mechanical ventilation in the intensive care unit (ICU) frequently develop contractile weakness of the diaphragm. Consequently, they may experience difficulty weaning from mechanical ventilation, which increases mortality and poses a high economic burden. Because of a lack of knowledge regarding the molecular changes in the diaphragm, no treatment is currently available to improve diaphragm contractility. We compared diaphragm biopsies from ventilated ICU patients (N = 54) to those of non-ICU patients undergoing thoracic surgery (N = 27). By integrating data from myofiber force measurements, x-ray diffraction experiments, and biochemical assays with clinical data, we found that in myofibers isolated from the diaphragm of ventilated ICU patients, myosin is trapped in an energy-sparing, super-relaxed state, which impairs the binding of myosin to actin during diaphragm contraction. Studies on quadriceps biopsies of ICU patients and on the diaphragm of previously healthy mechanically ventilated rats suggested that the super-relaxed myosins are specific to the diaphragm and not a result of critical illness. Exposing slow- and fast-twitch myofibers isolated from the diaphragm biopsies to small-molecule compounds activating troponin restored contractile force in vitro. These findings support the continued development of drugs that target sarcomere proteins to increase the calcium sensitivity of myofibers for the treatment of ICU-acquired diaphragm weakness.
Collapse
Affiliation(s)
- Marloes van den Berg
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
- Bispebjerg Hospital, Institute of Sports Medicine, Copenhagen 2400, Denmark
| | - Zhonghua Shi
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
- Amsterdam UMC, Location VUmc, Department of Intensive Care Medicine, Amsterdam 1081, HV, Netherlands
- Sanbo Brain Hospital, Capital Medical University, Intensive Care Medicine, Beijing 100093, China
| | - Wout J. Claassen
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Pleuni Hooijman
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Christopher T. A. Lewis
- University of Copenhagen, Department of Biomedical Sciences, Copenhagen 2200, Denmark
- Research and Early Development, Novo Nordisk A/S, Måløv 2760, Denmark
| | - Jesper L. Andersen
- Bispebjerg Hospital, Institute of Sports Medicine, Copenhagen 2400, Denmark
| | | | - Sylvia J. P. Bogaards
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Stefan Conijn
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Eva L. Peters
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| | - Leon P. L. Begthel
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Bas Uijterwijk
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
| | - Johan Lindqvist
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| | - Paul R. Langlais
- Department of Endocrinology, University of Arizona, Tucson, AZ 85721, USA
| | - Armand R. J. Girbes
- Amsterdam UMC, Location VUmc, Department of Intensive Care Medicine, Amsterdam 1081, HV, Netherlands
| | - Sandra Stapel
- Amsterdam UMC, Location VUmc, Department of Intensive Care Medicine, Amsterdam 1081, HV, Netherlands
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| | - Kenneth S. Campbell
- Division of Cardiovascular Medicine, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Weikang Ma
- BioCAT, Illinois Institute of Technology, Lemont, IL 60439, USA
| | - Thomas Irving
- BioCAT, Illinois Institute of Technology, Lemont, IL 60439, USA
| | - Darren T. Hwee
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - James J. Hartman
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Fady I. Malik
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Marinus Paul
- Amsterdam UMC, Location VUmc, Department of Cardiothoracic Surgery, Amsterdam 1081, HV, Netherlands
| | - Albertus Beishuizen
- Medisch Spectrum Twente, Intensive Care Center, Enschede 7511, HN, Netherlands
| | - Julien Ochala
- University of Copenhagen, Department of Biomedical Sciences, Copenhagen 2200, Denmark
| | - Leo Heunks
- Radboud UMC, Department of Intensive Care, Nijmegen 6525, GA, Netherlands
| | - Coen A. C. Ottenheijm
- Amsterdam UMC, Location VUmc, Department of Physiology, Amsterdam 1081, HV, Netherlands
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson AZ 85721, USA
| |
Collapse
|
15
|
Herskind J, Ørtenblad N, Cheng AJ, Pedersen P, Overgaard K. Piperine enhances contractile force in slow- and fast-twitch muscle. J Physiol 2024; 602:2807-2822. [PMID: 38762879 DOI: 10.1113/jp285995] [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: 12/05/2023] [Accepted: 04/30/2024] [Indexed: 05/21/2024] Open
Abstract
Piperine has been shown to bind to myosin and shift the distribution of conformational states of myosin molecules from the super-relaxed state to the disordered relaxed state. However, little is known about the implications for muscle force production and potential underlying mechanisms. Muscle contractility experiments were performed using isolated muscles and single fibres from rats and mice. The dose-response effect of piperine on muscle force was assessed at several stimulation frequencies. The potentiation of muscle force was also tested in muscles fatigued by eccentric contractions. Potential mechanisms of force potentiation were assessed by measuring Ca2+ levels during stimulation in enzymatically dissociated muscle fibres, while myofibrillar Ca2+ sensitivity was assessed in chemically skinned muscle fibres. Piperine caused a dose-dependent increase in low-frequency force with no effect on high-frequency force in both slow- and fast-twitch muscle, with similar relative increases in twitch force, rate of force development and relaxation rate. The potentiating effect of piperine on low-frequency force was reversible, and piperine partially recovered low-frequency force in fatigued muscle. Piperine had no effect on myoplasmic free [Ca2+] levels in mouse muscle fibres, whereas piperine substantially augmented the force response to submaximal levels of [Ca2+] in rat MyHCII fibres and MyHCI fibres along with a minor increase in maximum Ca2+-activated force. Piperine enhances low-frequency force production in both fast- and slow-twitch muscle. The effects are reversible and can counteract muscle fatigue. The primary underlying mechanism appears to be an increase in Ca2+ sensitivity. KEY POINTS: Piperine is a plant alkaloid derived from black pepper. It is known to bind to skeletal muscle myosin and enhance resting ATP turnover but its effects on contractility are not well known. We showed for the first time a piperine-induced force potentiation that was pronounced during low-frequency electrical stimulation of isolated muscles. The effect of piperine was observed in both slow and fast muscle types, was reversible, and could counteract the force decrements observed after fatiguing muscle contractions. Piperine treatment caused an increase in myofibrillar Ca2+ sensitivity in chemically skinned muscle fibres, while we observed no effect on intracellular Ca2+ concentrations during electrical stimulation in enzymatically dissociated muscle fibres.
Collapse
Affiliation(s)
- Jon Herskind
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Niels Ørtenblad
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Arthur J Cheng
- Muscle Health Research Centre, School of Kinesiology and Health Science, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Peter Pedersen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Kristian Overgaard
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
16
|
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
|
17
|
Buvoli M, Wilson GC, Buvoli A, Gugel JF, Hau A, Bönnemann CG, Paradas C, Ryba DM, Woulfe KC, Walker LA, Buvoli T, Ochala J, Leinwand LA. A Laing distal myopathy-associated proline substitution in the β-myosin rod perturbs myosin cross-bridging activity. J Clin Invest 2024; 134:e172599. [PMID: 38690726 PMCID: PMC11060730 DOI: 10.1172/jci172599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/11/2024] [Indexed: 05/03/2024] Open
Abstract
Proline substitutions within the coiled-coil rod region of the β-myosin gene (MYH7) are the predominant mutations causing Laing distal myopathy (MPD1), an autosomal dominant disorder characterized by progressive weakness of distal/proximal muscles. We report that the MDP1 mutation R1500P, studied in what we believe to be the first mouse model for the disease, adversely affected myosin motor activity despite being in the structural rod domain that directs thick filament assembly. Contractility experiments carried out on isolated mutant muscles, myofibrils, and myofibers identified muscle fatigue and weakness phenotypes, an increased rate of actin-myosin detachment, and a conformational shift of the myosin heads toward the more reactive disordered relaxed (DRX) state, causing hypercontractility and greater ATP consumption. Similarly, molecular analysis of muscle biopsies from patients with MPD1 revealed a significant increase in sarcomeric DRX content, as observed in a subset of myosin motor domain mutations causing hypertrophic cardiomyopathy. Finally, oral administration of MYK-581, a small molecule that decreases the population of heads in the DRX configuration, significantly improved the limited running capacity of the R1500P-transgenic mice and corrected the increased DRX state of the myofibrils from patients. These studies provide evidence of the molecular pathogenesis of proline rod mutations and lay the groundwork for the therapeutic advancement of myosin modulators.
Collapse
Affiliation(s)
- Massimo Buvoli
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Genevieve C.K. Wilson
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Ada Buvoli
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Jack F. Gugel
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Abbi Hau
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, and
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, Guy’s Campus, King’s College London, London, United Kingdom
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, Maryland, USA
| | - Carmen Paradas
- Neuromuscular Unit, Department of Neurology, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | | | - Kathleen C. Woulfe
- Division of Cardiology, Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - Lori A. Walker
- Division of Cardiology, Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - Tommaso Buvoli
- Department of Mathematics, Tulane University, New Orleans, Louisiana, USA
| | - Julien Ochala
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, and
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, Guy’s Campus, King’s College London, London, United Kingdom
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leslie A. Leinwand
- Department of Molecular, Cellular and Developmental Biology, and
- BioFrontiers Institute, Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
18
|
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
|
19
|
Li M, Hu Y, Wang Q. Exploring the Super-Relaxed State of Human Cardiac β-Myosin by Molecular Dynamics Simulations. J Phys Chem B 2024; 128:3113-3120. [PMID: 38516963 DOI: 10.1021/acs.jpcb.3c07956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Human β-cardiac myosin plays a critical role in generating the mechanical forces necessary for cardiac muscle contraction. This process relies on a delicate dynamic equilibrium between the disordered relaxed state (DRX) and the super-relaxed state (SRX) of myosin. Disruptions in this equilibrium due to mutations can lead to heart diseases. However, the structural characteristics of SRX and the molecular mechanisms underlying pathogenic mutations have remained elusive. To bridge this gap, we conducted molecular dynamics simulations and free energy calculations to explore the conformational changes in myosin. Our findings indicate that the size of the phosphate-binding pocket can serve as a valuable metric for characterizing the transition from the DRX to SRX state. Importantly, we established a global dynamic coupling network within the myosin motor head at the residue level, elucidating how the pathogenic mutation E483K impacts the equilibrium between SRX and DRX through allosteric effects. Our work illuminates molecular details of SRX and offers valuable insights into disease treatment through the regulation of SRX.
Collapse
Affiliation(s)
- Mingwei Li
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yao Hu
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Qian Wang
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
20
|
Kelly CM, Martin JL, Previs MJ. Myosin folding boosts solubility in cardiac muscle sarcomeres. JCI Insight 2024; 9:e178131. [PMID: 38483507 PMCID: PMC11141871 DOI: 10.1172/jci.insight.178131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/05/2024] [Indexed: 04/23/2024] Open
Abstract
The polymerization of myosin molecules into thick filaments in muscle sarcomeres is essential for cardiac contractility, with the attenuation of interactions between the heads of myosin molecules within the filaments being proposed to result in hypercontractility, as observed in hypertrophic cardiomyopathy (HCM). However, experimental evidence demonstrates that the structure of these giant macromolecular complexes is highly dynamic, with molecules exchanging between the filaments and a pool of soluble molecules on the minute timescale. Therefore, we sought to test the hypothesis that the enhancement of interactions between the heads of myosin molecules within thick filaments limits the mobility of myosin by taking advantage of mavacamten, a small molecule approved for the treatment of HCM. Myosin molecules were labeled in vivo with a green fluorescent protein (GFP) and imaged in intact hearts using multiphoton microscopy. Treatment of the intact hearts with mavacamten resulted in an unexpected > 5-fold enhancement in GFP-myosin mobility within the sarcomere. In vitro biochemical assays suggested that mavacamten enhanced the mobility of GFP-myosin by increasing the solubility of myosin molecules, through the stabilization of a compact/folded conformation of the molecules, once disassociated from the thick filaments. These findings provide alternative insight into the mechanisms by which molecules exchange into and out of thick filaments and have implications for how mavacamten may affect cardiac contractility.
Collapse
Affiliation(s)
- Colleen M Kelly
- Molecular Physiology and Biophysics Department, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Jody L Martin
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Michael J Previs
- Molecular Physiology and Biophysics Department, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
21
|
Garg A, Lavine KJ, Greenberg MJ. Assessing Cardiac Contractility From Single Molecules to Whole Hearts. JACC Basic Transl Sci 2024; 9:414-439. [PMID: 38559627 PMCID: PMC10978360 DOI: 10.1016/j.jacbts.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 04/04/2024]
Abstract
Fundamentally, the heart needs to generate sufficient force and power output to dynamically meet the needs of the body. Cardiomyocytes contain specialized structures referred to as sarcomeres that power and regulate contraction. Disruption of sarcomeric function or regulation impairs contractility and leads to cardiomyopathies and heart failure. Basic, translational, and clinical studies have adapted numerous methods to assess cardiac contraction in a variety of pathophysiological contexts. These tools measure aspects of cardiac contraction at different scales ranging from single molecules to whole organisms. Moreover, these studies have revealed new pathogenic mechanisms of heart disease leading to the development of novel therapies targeting contractility. In this review, the authors explore the breadth of tools available for studying cardiac contractile function across scales, discuss their strengths and limitations, highlight new insights into cardiac physiology and pathophysiology, and describe how these insights can be harnessed for therapeutic candidate development and translational.
Collapse
Affiliation(s)
- Ankit Garg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kory J. Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
22
|
Shen C, Ding X, Ruan J, Ruan F, Hu W, Huang J, He C, Yu Y, Zuo Z. Black phosphorus quantum dots induce myocardial inflammatory responses and metabolic disorders in mice. J Environ Sci (China) 2024; 137:53-64. [PMID: 37980037 DOI: 10.1016/j.jes.2023.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 11/20/2023]
Abstract
As an ultrasmall derivative of black phosphorus (BP) sheets, BP quantum dots (BP-QDs) have been effectively used in many fields. Currently, information on the cardiotoxicity induced by BP-QDs remains limited. We aimed to evaluate BP-QD-induced cardiac toxicity in mice. Histopathological examination of heart tissue sections was performed. Transcriptome sequencing, real-time quantitative PCR (RT‒qPCR), western blotting, and enzyme-linked immunosorbent assay (ELISA) assays were used to detect the mRNA and/or protein expression of proinflammatory cytokines, nuclear factor kappa B (NF-κB), phosphatidylinositol 3 kinase-protein kinase B (PI3K-AKT), peroxisome proliferator-activated receptor gamma (PPARγ), and glucose/lipid metabolism pathway-related genes. We found that heart weight and heart/body weight index (HBI) were significantly reduced in mice after intragastric administration of 0.1 or 1 mg/kg BP-QDs for 28 days. In addition, obvious inflammatory cell infiltration and increased cardiomyocyte diameter were observed in the BP-QD-treated groups. Altered expression of proinflammatory cytokines and genes related to the NF-κB signaling pathway further confirmed that BP-QD exposure induced inflammatory responses. In addition, BP-QD treatment also affected the PI3K-AKT, PPARγ, thermogenesis, oxidative phosphorylation, and cardiac muscle contraction signaling pathways. The expression of genes related to glucose/lipid metabolism signaling pathways was dramatically affected by BP-QD exposure, and the effect was primarily mediated by the PPAR signaling pathway. Our study provides new insights into the toxicity of BP-QDs to human health.
Collapse
Affiliation(s)
- Chao Shen
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xiaoyan Ding
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jinpeng Ruan
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Fengkai Ruan
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Weiping Hu
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jiyi Huang
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Chengyong He
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yi Yu
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| | - Zhenghong Zuo
- Department of Nephrology, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
23
|
Doh CY, Schmidt AV, Chinthalapudi K, Stelzer JE. Bringing into focus the central domains C3-C6 of myosin binding protein C. Front Physiol 2024; 15:1370539. [PMID: 38487262 PMCID: PMC10937550 DOI: 10.3389/fphys.2024.1370539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
Myosin binding protein C (MyBPC) is a multi-domain protein with each region having a distinct functional role in muscle contraction. The central domains of MyBPC have often been overlooked due to their unclear roles. However, recent research shows promise in understanding their potential structural and regulatory functions. Understanding the central region of MyBPC is important because it may have specialized function that can be used as drug targets or for disease-specific therapies. In this review, we provide a brief overview of the evolution of our understanding of the central domains of MyBPC in regard to its domain structures, arrangement and dynamics, interaction partners, hypothesized functions, disease-causing mutations, and post-translational modifications. We highlight key research studies that have helped advance our understanding of the central region. Lastly, we discuss gaps in our current understanding and potential avenues to further research and discovery.
Collapse
Affiliation(s)
- Chang Yoon Doh
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Alexandra V. Schmidt
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
24
|
Ma W, del Rio CL, Qi L, Prodanovic M, Mijailovich S, Zambataro C, Gong H, Shimkunas R, Gollapudi S, Nag S, Irving TC. Myosin in autoinhibited off state(s), stabilized by mavacamten, can be recruited in response to inotropic interventions. Proc Natl Acad Sci U S A 2024; 121:e2314914121. [PMID: 38346202 PMCID: PMC10895252 DOI: 10.1073/pnas.2314914121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Mavacamten is a FDA-approved small-molecule therapeutic designed to regulate cardiac function at the sarcomere level by selectively but reversibly inhibiting the enzymatic activity of myosin. It shifts myosin toward ordered off states close to the thick filament backbone. It remains elusive whether these myosin heads in the off state(s) can be recruited in response to physiological stimuli when required to boost cardiac output. We show that cardiac myosins stabilized in these off state(s) by mavacamten are recruitable by 1) Ca2+, 2) increased chronotropy [heart rate (HR)], 3) stretch, and 4) β-adrenergic (β-AR) stimulation, all known physiological inotropic interventions. At the molecular level, we show that Ca2+ increases myosin ATPase activity by shifting mavacamten-stabilized myosin heads from the inactive super-relaxed state to the active disordered relaxed state. At the myofilament level, both Ca2+ and passive lengthening can shift mavacamten-ordered off myosin heads from positions close to the thick filament backbone to disordered on states closer to the thin filaments. In isolated rat cardiomyocytes, increased stimulation rates enhanced shortening fraction in mavacamten-treated cells. This observation was confirmed in vivo in telemetered rats, where left-ventricular dP/dtmax, an index of inotropy, increased with HR in mavacamten-treated animals. Finally, we show that β-AR stimulation in vivo increases left-ventricular function and stroke volume in the setting of mavacamten. Our data demonstrate that the mavacamten-promoted off states of myosin in the thick filament are at least partially activable, thus preserving cardiac reserve mechanisms.
Collapse
Affiliation(s)
- Weikang Ma
- Biophysics Collaborative Access Team, Department of Biology, Illinois Institute of Technology, Chicago, IL60616
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL60616
| | - Carlos L. del Rio
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA94005
- Cardiac Consulting, San Mateo, CA94010
| | - Lin Qi
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Momcilo Prodanovic
- Institute for Information Technologies, University of Kragujevac, Kragujevac34000, Serbia
- FilamenTech, Inc., Newtown, MA02458
| | | | | | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Rafael Shimkunas
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA94005
| | - Sampath Gollapudi
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA94005
| | - Suman Nag
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA94005
| | - Thomas C. Irving
- Biophysics Collaborative Access Team, Department of Biology, Illinois Institute of Technology, Chicago, IL60616
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL60616
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| |
Collapse
|
25
|
Jani VP, Song T, Gao C, Gong H, Sadayappan S, Kass DA, Irving TC, Ma W. The structural OFF and ON states of myosin can be decoupled from the biochemical super- and disordered-relaxed states. PNAS NEXUS 2024; 3:pgae039. [PMID: 38328779 PMCID: PMC10849796 DOI: 10.1093/pnasnexus/pgae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
There is a growing awareness that both thick-filament and classical thin-filament regulations play central roles in modulating muscle contraction. Myosin ATPase assays have demonstrated that under relaxed conditions, myosin may reside either in a high-energy-consuming disordered-relaxed (DRX) state available for binding actin to generate force or in an energy-sparing super-relaxed (SRX) state unavailable for actin binding. X-ray diffraction studies have shown that the majority of myosin heads are in a quasi-helically ordered OFF state in a resting muscle and that this helical ordering is lost when myosin heads are turned ON for contraction. It has been assumed that myosin heads in SRX and DRX states are equivalent to the OFF and ON states, respectively, and the terms have been used interchangeably. In this study, we use X-ray diffraction and ATP turnover assays to track the structural and biochemical transitions of myosin heads, respectively, induced with either omecamtiv mecarbil (OM) or piperine in relaxed porcine myocardium. We find that while OM and piperine induce dramatic shifts of myosin heads from the OFF to the ON state, there are no appreciable changes in the population of myosin heads in the SRX and DRX states in both unloaded and loaded preparations. Our results show that biochemically defined SRX and DRX can be decoupled from structurally defined OFF and ON states. In summary, while SRX/DRX and OFF/ON transitions can be correlated in some cases, these two phenomena are measured using different approaches, reflect different properties of the thick filament, and should be investigated and interpreted separately.
Collapse
Affiliation(s)
- Vivek P Jani
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Chengqian Gao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - David A Kass
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thomas C Irving
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Weikang Ma
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
26
|
Sequeira V, Maack C, Reil GH, Reil JC. Exploring the Connection Between Relaxed Myosin States and the Anrep Effect. Circ Res 2024; 134:117-134. [PMID: 38175910 DOI: 10.1161/circresaha.123.323173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
The Anrep effect is an adaptive response that increases left ventricular contractility following an acute rise in afterload. Although the mechanistic origin remains undefined, recent findings suggest a two-phase activation of resting myosin for contraction, involving strain-sensitive and posttranslational phases. We propose that this mobilization represents a transition among the relaxed states of myosin-specifically, from the super-relaxed (SRX) to the disordered-relaxed (DRX)-with DRX myosin ready to participate in force generation. This hypothesis offers a unified explanation that connects myosin's SRX-DRX equilibrium and the Anrep effect as parts of a singular phenomenon. We underscore the significance of this equilibrium in modulating contractility, primarily studied in the context of hypertrophic cardiomyopathy, the most common inherited cardiomyopathy associated with diastolic dysfunction, hypercontractility, and left ventricular hypertrophy. As we posit that the cellular basis of the Anrep effect relies on a two-phased transition of myosin from the SRX to the contraction-ready DRX configuration, any dysregulation in this equilibrium may result in the pathological manifestation of the Anrep phenomenon. For instance, in hypertrophic cardiomyopathy, hypercontractility is linked to a considerable shift of myosin to the DRX state, implying a persistent activation of the Anrep effect. These valuable insights call for additional research to uncover a clinical Anrep fingerprint in pathological states. Here, we demonstrate through noninvasive echocardiographic pressure-volume measurements that this fingerprint is evident in 12 patients with hypertrophic obstructive cardiomyopathy before septal myocardial ablation. This unique signature is characterized by enhanced contractility, indicated by a leftward shift and steepening of the end-systolic pressure-volume relationship, and a prolonged systolic ejection time adjusted for heart rate, which reverses post-procedure. The clinical application of this concept has potential implications beyond hypertrophic cardiomyopathy, extending to other genetic cardiomyopathies and even noncongenital heart diseases with complex etiologies across a broad spectrum of left ventricular ejection fractions.
Collapse
Affiliation(s)
- Vasco Sequeira
- Department of Translational Science Universitätsklinikum, Deutsche Zentrum für Herzinsuffizienz (DZHI), Würzburg, Germany (V.S., C.M.)
| | - Christoph Maack
- Department of Translational Science Universitätsklinikum, Deutsche Zentrum für Herzinsuffizienz (DZHI), Würzburg, Germany (V.S., C.M.)
| | - Gert-Hinrich Reil
- Klinik für Kardiologie, Klinikum Oldenburg, Innere Medizin I, Germany (G.-H.R.)
| | - Jan-Christian Reil
- Klinik für Allgemeine und Interventionelle Kardiologie, Herz- und Diabetes-Zentrum Nordrhein-Westphalen, Germany (J.-C.R.)
| |
Collapse
|
27
|
Kao K, Geeves MA. Protocols for Myosin and Actin-Myosin Assays Using Rapid, Stopped-Flow Kinetics. Methods Mol Biol 2024; 2735:191-211. [PMID: 38038850 DOI: 10.1007/978-1-0716-3527-8_11] [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] [Indexed: 12/02/2023]
Abstract
Fast transient kinetics using stopped-flow fluorimetry is now a powerful method for defining the ATPase cycle of myosin and its subfragments and has found wide use in defining the difference between myosin isoforms, myosins carrying disease linked mutations, and the effect of small molecules on the ATPase cycle. Here the protocols for completing the classical assays of myosin and actin.myosin using the stopped-flow are described. The assays include ATP and ADP binding to myosin and actin.myosin, displacement of ADP from myosin and actin.myosin, and the cleavage of ATP to ADP and phosphate on myosin. Single and multiple turnover assays are also described.
Collapse
Affiliation(s)
- Kerry Kao
- School of Medicine & College of Engineering, University of Washington, Seattle, WA, USA
| | - Michael A Geeves
- School of Biosciences, University of Kent, Canterbury, Kent, UK.
| |
Collapse
|
28
|
Mohran S, Kooiker K, Mahoney-Schaefer M, Mandrycky C, Kao K, Tu AY, Freeman J, Moussavi-Harami F, Geeves M, Regnier M. The biochemically defined super relaxed state of myosin-A paradox. J Biol Chem 2024; 300:105565. [PMID: 38103642 PMCID: PMC10819765 DOI: 10.1016/j.jbc.2023.105565] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/06/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023] Open
Abstract
The biochemical SRX (super-relaxed) state of myosin has been defined as a low ATPase activity state. This state can conserve energy when the myosin is not recruited for muscle contraction. The SRX state has been correlated with a structurally defined ordered (versus disordered) state of muscle thick filaments. The two states may be linked via a common interacting head motif (IHM) where the two heads of heavy meromyosin (HMM), or myosin, fold back onto each other and form additional contacts with S2 and the thick filament. Experimental observations of the SRX, IHM, and the ordered form of thick filaments, however, do not always agree, and result in a series of unresolved paradoxes. To address these paradoxes, we have reexamined the biochemical measurements of the SRX state for porcine cardiac HMM. In our hands, the commonly employed mantATP displacement assay was unable to quantify the population of the SRX state with all data fitting very well by a single exponential. We further show that mavacamten inhibits the basal ATPases of both porcine ventricle HMM and S1 (Ki, 0.32 and 1.76 μM respectively) while dATP activates HMM cooperatively without any evidence of an SRX state. A combination of our experimental observations and theories suggests that the displacement of mantATP in purified proteins is not a reliable assay to quantify the SRX population. This means that while the structurally defined IHM and ordered thick filaments clearly exist, great care must be employed when using the mantATP displacement assay.
Collapse
Affiliation(s)
- Saffie Mohran
- Department of Bioengineering, University of Washington, Seattle, Washington, USA; Center for Translational Muscle Research, University of Washington, Seattle, Washington, USA
| | - Kristina Kooiker
- Center for Translational Muscle Research, University of Washington, Seattle, Washington, USA; Division of Cardiology, University of Washington, Seattle, Washington, USA
| | | | - Christian Mandrycky
- Department of Bioengineering, University of Washington, Seattle, Washington, USA; Center for Translational Muscle Research, University of Washington, Seattle, Washington, USA
| | - Kerry Kao
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - An-Yue Tu
- Department of Bioengineering, University of Washington, Seattle, Washington, USA; Center for Translational Muscle Research, University of Washington, Seattle, Washington, USA
| | - Jeremy Freeman
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Farid Moussavi-Harami
- Department of Bioengineering, University of Washington, Seattle, Washington, USA; Center for Translational Muscle Research, University of Washington, Seattle, Washington, USA; Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Michael Geeves
- School of Biosciences, University of Kent, Canterbury, UK.
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington, USA; Center for Translational Muscle Research, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
29
|
Scott B, Greenberg MJ. Multiscale biophysical models of cardiomyopathies reveal complexities challenging existing dogmas. Biophys J 2023; 122:4632-4634. [PMID: 38006882 PMCID: PMC10754685 DOI: 10.1016/j.bpj.2023.11.014] [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: 11/02/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/27/2023] Open
Abstract
Mutations in sarcomeric proteins, including myosin, cause a variety of cardiomyopathies. A prominent hypothesis has been that myosin mutations causing hypercontractility of the motor lead to hypertrophic cardiomyopathy, while those causing hypocontractility lead to dilated cardiomyopathy; however, recent biophysical studies using multiscale computational and experimental models have revealed complexities not captured by this hypothesis. We summarize recent publications in Biophysical Journal challenging this dogma and highlighting the need for multiscale modeling of these complex diseases.
Collapse
Affiliation(s)
- Brent Scott
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
30
|
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
|
31
|
Ma W, Jani VP, Song T, Gao C, Gong H, Sadayappan S, Kass DA, Irving TC. The structural OFF and ON states of myosin can be decoupled from the biochemical super-relaxed and disordered-relaxed states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562891. [PMID: 37904972 PMCID: PMC10614925 DOI: 10.1101/2023.10.18.562891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
There is a growing awareness that both thick filament and classical thin filament regulation play central roles in modulating muscle contraction. Myosin ATPase assays have demonstrated that under relaxed conditions, myosin may reside in either a high energy-consuming disordered-relaxed (DRX) state available for binding actin to generate force, or in an energy-sparing super-relaxed (SRX) state unavailable for actin binding. X-ray diffraction studies have shown the majority of myosin heads are in a quasi-helically ordered OFF state in a resting muscle and that this helical ordering is lost when myosin heads are turned ON for contraction. It has been assumed that myosin heads in SRX and DRX states are equivalent to the OFF and ON state respectively and the terms have been used interchangeably. Here, we use X-ray diffraction and ATP turnover assays to track the structural and biochemical transitions of myosin heads respectively induced with either omecamtiv mecarbil (OM) or piperine in relaxed porcine myocardium. We find that while OM and piperine induce dramatic shifts of myosin heads from the OFF to ON states, there are no appreciable changes in the population of myosin heads in the SRX and DRX states in both unloaded and loaded preparations. Our results show that biochemically defined SRX and DRX can be decoupled from structurally-defined OFF and ON states. In summary, while SRX/DRX and OFF/ON transitions can be correlated in some cases, these two phenomena are measured using different approaches, do not necessarily reflect the same properties of the thick filament and should be investigated and interpreted separately.
Collapse
Affiliation(s)
- Weikang Ma
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Vivek P. Jani
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, OH, USA
| | - Chengqian Gao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, OH, USA
| | - David A. Kass
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Thomas C. Irving
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
32
|
Ochala J, Lewis CTA, Beck T, Iwamoto H, Hessel AL, Campbell KS, Pyle WG. Predominant myosin superrelaxed state in canine myocardium with naturally occurring dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2023; 325:H585-H591. [PMID: 37505469 PMCID: PMC11932529 DOI: 10.1152/ajpheart.00369.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Dilated cardiomyopathy (DCM) is a naturally occurring heart failure condition in humans and dogs, notably characterized by a reduced contractility and ejection fraction. As the identification of its underlying cellular and molecular mechanisms remain incomplete, the aim of the present study was to assess whether the molecular motor myosin and its known relaxed conformational states are altered in DCM. For that, we dissected and skinned thin cardiac strips from left ventricle obtained from six DCM Doberman Pinschers and six nonfailing (NF) controls. We then used a combination of Mant-ATP chase experiments and X-ray diffraction to assess both energetic and structural changes of myosin. Using the Mant-ATP chase protocol, we observed that in DCM dogs, the amount of myosin molecules in the ATP-conserving conformational state, also known as superrelaxed (SRX), is significantly increased when compared with NF dogs. This alteration can be rescued by applying EMD-57033, a small molecule activating myosin. Conversely, with X-ray diffraction, we found that in DCM dogs, there is a higher proportion of myosin heads in the vicinity of actin when compared with NF dogs (1,0 to 1,1 intensity ratio). Hence, we observed an uncoupling between energetic (Mant-ATP chase) and structural (X-ray diffraction) data. Taken together, these results may indicate that in the heart of Doberman Pinschers with DCM, myosin molecules are potentially stuck in a nonsequestered but ATP-conserving SRX state, that can be counterbalanced by EMD-57033 demonstrating the potential for a myosin-centered pharmacological treatment of DCM.NEW & NOTEWORTHY The key finding of the present study is that, in left ventricles of dogs with a naturally occurring dilated cardiomyopathy, relaxed myosin molecules favor a nonsequestered superrelaxed state potentially impairing sarcomeric contractility. This alteration is rescuable by applying a small molecule activating myosin known as EMD-57033.
Collapse
Affiliation(s)
- Julien Ochala
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Thomas Beck
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hiroyuki Iwamoto
- SPring-8, Japan Synchrotron Radiation Research Institute, Hyogo, Japan
| | - Anthony L Hessel
- Institute of Physiology II, University of Muenster, Muenster, Germany
- Accelerated Muscle Biotechnologies, Boston, Massachusetts, United States
| | - Kenneth S Campbell
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - W Glen Pyle
- IMPART Investigator Team, Dalhousie Medicine, Saint John, New Brunswick, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
33
|
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
|
34
|
Ma W, del Rio CL, Qi L, Prodanovic M, Mijailovich S, Zambataro C, Gong H, Shimkunas R, Gollapudi S, Nag S, Irving TC. Myosin in autoinhibited off state(s), stabilized by mavacamten, can be recruited via inotropic effectors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536292. [PMID: 37090664 PMCID: PMC10120679 DOI: 10.1101/2023.04.10.536292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Mavacamten is a novel, FDA-approved, small molecule therapeutic designed to regulate cardiac function by selectively but reversibly inhibiting the enzymatic activity of myosin. It shifts myosin towards ordered off states close to the thick filament backbone. It remains unresolved whether mavacamten permanently sequesters these myosin heads in the off state(s) or whether these heads can be recruited in response to physiological stimuli when required to boost cardiac output. We show that cardiac myosins stabilized in these off state(s) by mavacamten are recruitable by Ca2+, increased heart rate, stretch, and β-adrenergic (β-AR) stimulation, all known physiological inotropic effectors. At the molecular level, we show that, in presence of mavacamten, Ca2+ increases myosin ATPase activity by shifting myosin heads from the reserve super-relaxed (SRX) state to the active disordered relaxed (DRX) state. At the myofilament level, both Ca2+ and passive lengthening can shift ordered off myosin heads from positions close to the thick filament backbone to disordered on states closer to the thin filaments in the presence of mavacamten. In isolated rat cardiomyocytes, increased stimulation rates enhanced shortening fraction in mavacamten-treated cells. This observation was confirmed in vivo in telemetered rats, where left-ventricular dP/dtmax, an index of inotropy, increased with heart rate in mavacamten treated animals. Finally, we show that β-AR stimulation in vivo increases left-ventricular function and stroke volume in the setting of mavacamten. Our data demonstrate that the mavacamten-promoted off states of myosin in the thick filament are activable, at least partially, thus leading to preservation of cardiac reserve mechanisms.
Collapse
Affiliation(s)
- Weikang Ma
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Carlos L. del Rio
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA 94005
| | - Lin Qi
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Momcilo Prodanovic
- Institute for Information Technologies, University of Kragujevac, Kragujevac, Serbia
- FilamenTech, Inc., Newtown, MA 02458, USA
| | | | | | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Rafael Shimkunas
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA 94005
| | - Sampath Gollapudi
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA 94005
| | - Suman Nag
- Cardiovascular Drug Discovery, Bristol Myers Squibb, Brisbane, CA 94005
| | - Thomas C. Irving
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
35
|
Mi K, Wu S, Lv C, Meng Y, Yin W, Li H, Li J, Yuan H. Comparing the efficacy and safety of medications in adults with hypertrophic cardiomyopathy: a systematic review and network meta-analysis. Front Cardiovasc Med 2023; 10:1190181. [PMID: 37645523 PMCID: PMC10461399 DOI: 10.3389/fcvm.2023.1190181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Background Hypertrophic cardiomyopathy (HCM) is the most common genetic heart disease. The purpose of this study was to evaluate the efficacy and safety of several medications and recommend better drug treatments for adults with HCM. Methods A review of PubMed, Embase, the Cochrane Controlled Register of Trials (CENTRAL), ClinicalTrials.gov and CNKI databases was conducted for studies on the efficacy and safety of drugs for adults with HCM. A frequentist random effects model was used in this network analysis. Results This network meta-analysis included 7 studies assessing seven medications, 6 studies evaluating monotherapy and 1 study evaluating combination therapy. Based on the network meta-analysis results, xiaoxinbi formula plus metoprolol (MD -56.50% [-72.43%, -40.57%]), metoprolol (MD -47.00% [-59.07%, -34.93%]) and mavacamten (MD -34.50% [-44.75%, -24.25%]) significantly reduced the resting left ventricular outflow tract gradient (LVOTG) in comparison with placebo. Resting LVOTG could also be reduced with N-acetylcysteine (NAC). The incidence of adverse drug reactions was not significantly different between the placebo group and the treatment group. Conclusion For adults with HCM, the top 4 treatments included xiaoxinbi formula plus metoprolol, metoprolol, mavacamten and NAC.Systematic Review Registration: [https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=374222], identifier [CRD42022374222].
Collapse
Affiliation(s)
- Keying Mi
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
- JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Sijia Wu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chanyuan Lv
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
- JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Yongkang Meng
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
- JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Wenchao Yin
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
- JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Hongkai Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiangbing Li
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
- JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, China
- JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
Claassen WJ, Baelde RJ, Galli RA, de Winter JM, Ottenheijm CAC. Small molecule drugs to improve sarcomere function in those with acquired and inherited myopathies. Am J Physiol Cell Physiol 2023; 325:C60-C68. [PMID: 37212548 PMCID: PMC10281779 DOI: 10.1152/ajpcell.00047.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Muscle weakness is a hallmark of inherited or acquired myopathies. It is a major cause of functional impairment and can advance to life-threatening respiratory insufficiency. During the past decade, several small-molecule drugs that improve the contractility of skeletal muscle fibers have been developed. In this review, we provide an overview of the available literature and the mechanisms of action of small-molecule drugs that modulate the contractility of sarcomeres, the smallest contractile units in striated muscle, by acting on myosin and troponin. We also discuss their use in the treatment of skeletal myopathies. The first of three classes of drugs discussed here increase contractility by decreasing the dissociation rate of calcium from troponin and thereby sensitizing the muscle to calcium. The second two classes of drugs directly act on myosin and stimulate or inhibit the kinetics of myosin-actin interactions, which may be useful in patients with muscle weakness or stiffness.NEW & NOTEWORTHY During the past decade, several small molecule drugs that improve the contractility of skeletal muscle fibers have been developed. In this review, we provide an overview of the available literature and the mechanisms of action of small molecule drugs that modulate the contractility of sarcomeres, the smallest contractile units in striated muscle, by acting on myosin and troponin.
Collapse
Affiliation(s)
- Wout J Claassen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Rianne J Baelde
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Ricardo A Galli
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Josine M de Winter
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| | - Coen A C Ottenheijm
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan, Amsterdam, Netherlands
| |
Collapse
|
39
|
Landim-Vieira M, Ma W, Song T, Rastegarpouyani H, Gong H, Coscarella IL, Bogaards SJP, Conijn SP, Ottenheijm CAC, Hwang HS, Papadaki M, Knollmann BC, Sadayappan S, Irving TC, Galkin VE, Chase PB, Pinto JR. Cardiac troponin T N-domain variant destabilizes the actin interface resulting in disturbed myofilament function. Proc Natl Acad Sci U S A 2023; 120:e2221244120. [PMID: 37252999 PMCID: PMC10265946 DOI: 10.1073/pnas.2221244120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Missense variant Ile79Asn in human cardiac troponin T (cTnT-I79N) has been associated with hypertrophic cardiomyopathy and sudden cardiac arrest in juveniles. cTnT-I79N is located in the cTnT N-terminal (TnT1) loop region and is known for its pathological and prognostic relevance. A recent structural study revealed that I79 is part of a hydrophobic interface between the TnT1 loop and actin, which stabilizes the relaxed (OFF) state of the cardiac thin filament. Given the importance of understanding the role of TnT1 loop region in Ca2+ regulation of the cardiac thin filament along with the underlying mechanisms of cTnT-I79N-linked pathogenesis, we investigated the effects of cTnT-I79N on cardiac myofilament function. Transgenic I79N (Tg-I79N) muscle bundles displayed increased myofilament Ca2+ sensitivity, smaller myofilament lattice spacing, and slower crossbridge kinetics. These findings can be attributed to destabilization of the cardiac thin filament's relaxed state resulting in an increased number of crossbridges during Ca2+ activation. Additionally, in the low Ca2+-relaxed state (pCa8), we showed that more myosin heads are in the disordered-relaxed state (DRX) that are more likely to interact with actin in cTnT-I79N muscle bundles. Dysregulation of the myosin super-relaxed state (SRX) and the SRX/DRX equilibrium in cTnT-I79N muscle bundles likely result in increased mobility of myosin heads at pCa8, enhanced actomyosin interactions as evidenced by increased active force at low Ca2+, and increased sinusoidal stiffness. These findings point to a mechanism whereby cTnT-I79N weakens the interaction of the TnT1 loop with the actin filament, which in turn destabilizes the relaxed state of the cardiac thin filament.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL32306
| | - Weikang Ma
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH45267
| | - Hosna Rastegarpouyani
- Department of Biological Science, Florida State University, Tallahassee, FL32306
- Institude of Molecular Biophysics, Florida State University, Tallahassee, FL32306
| | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Isabella Leite Coscarella
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL32306
| | - Sylvia J. P. Bogaards
- Department of Physiology, Amsterdam University Medical Center, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Stefan P. Conijn
- Department of Physiology, Amsterdam University Medical Center, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Coen A. C. Ottenheijm
- Department of Physiology, Amsterdam University Medical Center, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Hyun S. Hwang
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL32306
| | - Maria Papadaki
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, IL60153
| | - Bjorn C. Knollmann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN37232
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH45267
| | - Thomas C. Irving
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Vitold E. Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA23507
| | - P. Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL32306
| |
Collapse
|
40
|
Asencio A, Malingen S, Kooiker KB, Powers JD, Davis J, Daniel T, Moussavi-Harami F. Machine learning meets Monte Carlo methods for models of muscle's molecular machinery to classify mutations. J Gen Physiol 2023; 155:e202213291. [PMID: 37000171 PMCID: PMC10067704 DOI: 10.1085/jgp.202213291] [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: 10/31/2022] [Revised: 02/14/2023] [Accepted: 03/14/2023] [Indexed: 04/01/2023] Open
Abstract
The timing and magnitude of force generation by a muscle depend on complex interactions in a compliant, contractile filament lattice. Perturbations in these interactions can result in cardiac muscle diseases. In this study, we address the fundamental challenge of connecting the temporal features of cardiac twitches to underlying rate constants and their perturbations associated with genetic cardiomyopathies. Current state-of-the-art metrics for characterizing the mechanical consequence of cardiac muscle disease do not utilize information embedded in the complete time course of twitch force. We pair dimension reduction techniques and machine learning methods to classify underlying perturbations that shape the timing of twitch force. To do this, we created a large twitch dataset using a spatially explicit Monte Carlo model of muscle contraction. Uniquely, we modified the rate constants of this model in line with mouse models of cardiac muscle disease and varied mutation penetrance. Ultimately, the results of this study show that machine learning models combined with biologically informed dimension reduction techniques can yield excellent classification accuracy of underlying muscle perturbations.
Collapse
Affiliation(s)
- Anthony Asencio
- Department of Biology, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Division of Cardiology, University of Washington, Seattle, WA, USA
- Center for Transnational Muscle Research, University of Washington, Seattle, WA, USA
| | - Sage Malingen
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center for Transnational Muscle Research, University of Washington, Seattle, WA, USA
| | - Kristina B. Kooiker
- Division of Cardiology, University of Washington, Seattle, WA, USA
- Center for Transnational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Joseph D. Powers
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center for Transnational Muscle Research, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine Pathology, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Thomas Daniel
- Department of Biology, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center for Transnational Muscle Research, University of Washington, Seattle, WA, USA
| | - Farid Moussavi-Harami
- Division of Cardiology, University of Washington, Seattle, WA, USA
- Center for Transnational Muscle Research, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine Pathology, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| |
Collapse
|
41
|
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: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [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
|
42
|
Månsson A. The potential of myosin and actin in nanobiotechnology. J Cell Sci 2023; 136:292584. [PMID: 36861886 DOI: 10.1242/jcs.261025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Since the late 1990s, efforts have been made to utilize cytoskeletal filaments, propelled by molecular motors, for nanobiotechnological applications, for example, in biosensing and parallel computation. This work has led to in-depth insights into the advantages and challenges of such motor-based systems, and has yielded small-scale, proof-of-principle applications but, to date, no commercially viable devices. Additionally, these studies have also elucidated fundamental motor and filament properties, as well as providing other insights obtained from biophysical assays in which molecular motors and other proteins are immobilized on artificial surfaces. In this Perspective, I discuss the progress towards practically viable applications achieved so far using the myosin II-actin motor-filament system. I also highlight several fundamental pieces of insights derived from the studies. Finally, I consider what may be required to achieve real devices in the future or at least to allow future studies with a satisfactory cost-benefit ratio.
Collapse
Affiliation(s)
- Alf Månsson
- Department of Chemistry and Biomedical Science, Linnaeus University, SE-391 82 Kalmar, Sweden
| |
Collapse
|
43
|
Mhatre KN, Murray JD, Flint G, McMillen TS, Weber G, Shakeri M, Tu AY, Steczina S, Weiss R, Marcinek DJ, Murry CE, Raftery D, Tian R, Moussavi-Harami F, Regnier M. dATP elevation induces myocardial metabolic remodeling to support improved cardiac function. J Mol Cell Cardiol 2023; 175:1-12. [PMID: 36470336 PMCID: PMC9974746 DOI: 10.1016/j.yjmcc.2022.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Hallmark features of systolic heart failure are reduced contractility and impaired metabolic flexibility of the myocardium. Cardiomyocytes (CMs) with elevated deoxy ATP (dATP) via overexpression of ribonucleotide reductase (RNR) enzyme robustly improve contractility. However, the effect of dATP elevation on cardiac metabolism is unknown. Here, we developed proteolysis-resistant versions of RNR and demonstrate that elevation of dATP/ATP to ∼1% in CMs in a transgenic mouse (TgRRB) resulted in robust improvement of cardiac function. Pharmacological approaches showed that CMs with elevated dATP have greater basal respiratory rates by shifting myosin states to more active forms, independent of its isoform, in relaxed CMs. Targeted metabolomic profiling revealed a significant reprogramming towards oxidative phosphorylation in TgRRB-CMs. Higher cristae density and activity in the mitochondria of TgRRB-CMs improved respiratory capacity. Our results revealed a critical property of dATP to modulate myosin states to enhance contractility and induce metabolic flexibility to support improved function in CMs.
Collapse
Affiliation(s)
- Ketaki N Mhatre
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jason D Murray
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Department of Physiology and Biophysics, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Galina Flint
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Timothy S McMillen
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA
| | - Gerhard Weber
- Division of Cardiology, University of Washington, Seattle, WA 98109, USA
| | - Majid Shakeri
- Division of Cardiology, University of Washington, Seattle, WA 98109, USA
| | - An-Yue Tu
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Sonette Steczina
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Robert Weiss
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Charles E Murry
- Division of Cardiology, University of Washington, Seattle, WA 98109, USA; Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA; The Mitochondria and Metabolism Center (MMC), University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA
| | - Rong Tian
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA; The Mitochondria and Metabolism Center (MMC), University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA
| | - Farid Moussavi-Harami
- Division of Cardiology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA.
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Department of Physiology and Biophysics, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
44
|
Rasicci DV, Ge J, Milburn GN, Wood NB, Pruznak AM, Lang CH, Previs MJ, Campbell KS, Yengo CM. Cardiac myosin motor deficits are associated with left ventricular dysfunction in human ischemic heart failure. Am J Physiol Heart Circ Physiol 2023; 324:H198-H209. [PMID: 36525480 PMCID: PMC9829461 DOI: 10.1152/ajpheart.00272.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
During ischemic heart failure (IHF), cardiac muscle contraction is typically impaired, though the molecular changes within the myocardium are not fully understood. Thus, we aimed to characterize the biophysical properties of cardiac myosin in IHF. Cardiac tissue was harvested from 10 age-matched males, either with a history of IHF or nonfailing (NF) controls that had no history of structural or functional cardiac abnormalities. Clinical measures before cardiac biopsy demonstrated significant differences in measures of ejection fraction and left ventricular dimensions. Myofibrils and myosin were extracted from left ventricular free wall cardiac samples. There were no changes in myofibrillar ATPase activity or calcium sensitivity between groups. Using isolated myosin, we found a 15% reduction in the IHF group in actin sliding velocity in the in vitro motility assay, which was observed in the absence of a myosin isoform shift. Oxidative damage (carbonylation) of isolated myosin was compared, in which there were no significant differences between groups. Synthetic thick filaments were formed from purified myosin and the ATPase activity was similar in both basal and actin-activated conditions (20 µM actin). Correlation analysis and Deming linear regression were performed between all studied parameters, in which we found statistically significant correlations between clinical measures of contractility with molecular measures of sliding velocity and ELC carbonylation. Our data indicate that subtle deficits in myosin mechanochemical properties are associated with reduced contractile function and pathological remodeling of the heart, suggesting that the myosin motor may be an effective pharmacological intervention in ischemia.NEW & NOTEWORTHY Ischemic heart failure is associated with impairments in contractile performance of the heart. This study revealed that cardiac myosin isolated from patients with ischemic heart failure had reduced mechanical activity, which correlated with the impaired clinical phenotype of the patients. The results suggest that restoring myosin function with pharmacological intervention may be a viable method for therapeutic intervention.
Collapse
Affiliation(s)
- D. V. Rasicci
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University School of Medicine, Morgantown, West Virginia
| | - J. Ge
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - G. N. Milburn
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - N. B. Wood
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont
| | - A. M. Pruznak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - C. H. Lang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - M. J. Previs
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont
| | - K. S. Campbell
- Department of Physiology, University of Kentucky, Lexington, Kentucky
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, Kentucky
| | - C. M. Yengo
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
45
|
Structural OFF/ON transitions of myosin in relaxed porcine myocardium predict calcium-activated force. Proc Natl Acad Sci U S A 2023; 120:e2207615120. [PMID: 36696446 PMCID: PMC9945958 DOI: 10.1073/pnas.2207615120] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Contraction in striated muscle is initiated by calcium binding to troponin complexes, but it is now understood that dynamic transition of myosin between resting, ordered OFF states on thick filaments and active, disordered ON states that can bind to thin filaments is critical in regulating muscle contractility. These structural OFF to ON transitions of myosin are widely assumed to correspond to transitions from the biochemically defined, energy-sparing, super-relaxed (SRX) state to the higher ATPase disordered-relaxed (DRX) state. Here we examined the effect of 2'-deoxy-ATP (dATP), a naturally occurring energy substrate for myosin, on the structural OFF to ON transitions of myosin motors in porcine cardiac muscle thick filaments. Small-angle X-ray diffraction revealed that titrating dATP in relaxation solutions progressively moves the myosin heads from ordered OFF states on the thick filament backbone to disordered ON states closer to thin filaments. Importantly, we found that the structural OFF to ON transitions are not equivalent to the biochemically defined SRX to DRX transitions and that the dATP-induced structural OFF to ON transitions of myosin motors in relaxed muscle are strongly correlated with submaximal force augmentation by dATP. These results indicate that structural OFF to ON transitions of myosin in relaxed muscle can predict the level of force attained in calcium-activated cardiac muscle. Computational modeling and stiffness measurements suggest a final step in the OFF to ON transition may involve a subset of DRX myosins that form weakly bound cross-bridges prior to becoming active force-producing cross-bridges.
Collapse
|
46
|
Lewis CTA, Ochala J. Myosin Heavy Chain as a Novel Key Modulator of Striated Muscle Resting State. Physiology (Bethesda) 2023; 38:0. [PMID: 36067133 DOI: 10.1152/physiol.00018.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
After years of intense research using structural, biological, and biochemical experimental procedures, it is clear that myosin molecules are essential for striated muscle contraction. However, this is just the tip of the iceberg of their function. Interestingly, it has been shown recently that these molecules (especially myosin heavy chains) are also crucial for cardiac and skeletal muscle resting state. In the present review, we first overview myosin heavy chain biochemical states and how they influence the consumption of ATP. We then detail how neighboring partner proteins including myosin light chains and myosin binding protein C intervene in such processes, modulating the ATP demand in health and disease. Finally, we present current experimental drugs targeting myosin ATP consumption and how they can treat muscle diseases.
Collapse
Affiliation(s)
| | - Julien Ochala
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Abstract
A historical perspective of the super-relaxed (SRX) state, interacting heads motif (IHM), and impact of calcium on muscle contractility.
Collapse
Affiliation(s)
- Michael J. Previs
- Molecular Physiology and Biophysics Department, Larner College of Medicine, University of Vermont, Burlington, VT, USA,Correspondence to Michael J. Previs:
| |
Collapse
|
48
|
Ma W, Nag S, Gong H, Qi L, Irving TC. Cardiac myosin filaments are directly regulated by calcium. J Gen Physiol 2022; 154:e202213213. [PMID: 36327149 PMCID: PMC9629851 DOI: 10.1085/jgp.202213213] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/25/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
Classically, striated muscle contraction is initiated by calcium (Ca2+)-dependent structural changes in regulatory proteins on actin-containing thin filaments, which allow the binding of myosin motors to generate force. Additionally, dynamic switching between resting off and active on myosin states has been shown to regulate muscle contractility, a recently validated mechanism by novel myosin-targeted therapeutics. The molecular nature of this switching, however, is not understood. Here, using a combination of small-angle x-ray fiber diffraction and biochemical assays with reconstituted systems, we show that cardiac thick filaments are directly Ca2+-regulated. We find that Ca2+ induces a structural transition of myosin heads from ordered off states close to the thick filament to disordered on states closer to the thin filaments. Biochemical assays show a Ca2+-induced transition from an inactive super-relaxed (SRX) state(s) to an active disordered-relaxed (DRX) state(s) in synthetic thick filaments. We show that these transitions are an intrinsic property of cardiac myosin only when assembled into thick filaments and provide a fresh perspective on nature's two orthogonal mechanisms to regulate muscle contraction through the thin and the thick filaments.
Collapse
Affiliation(s)
- Weikang Ma
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL
| | - Suman Nag
- Department of Biochemistry, Bristol Myers Squibb, Brisbane, CA
| | - Henry Gong
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL
| | - Lin Qi
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL
| | - Thomas C. Irving
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL
| |
Collapse
|
49
|
Awinda PO, Ma W, Turner KL, Zhao J, Gong H, Thompson MS, Campbell KS, Irving TC, Tanner BCW. MgADP Promotes Myosin Head Movement toward Actin at Low [Ca 2+] to Increase Force Production and Ca 2+-Sensitivity of Contraction in Permeabilized Porcine Myocardial Strips. Int J Mol Sci 2022; 23:ijms232315084. [PMID: 36499408 PMCID: PMC9737397 DOI: 10.3390/ijms232315084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 12/04/2022] Open
Abstract
Myosin cross-bridges dissociate from actin following Mg2+-adenosine triphosphate (MgATP) binding. Myosin hydrolyses MgATP into inorganic phosphate (Pi) and Mg2+-adenosine diphosphate (ADP), and release of these hydrolysis products drives chemo-mechanical energy transitions within the cross-bridge cycle to power muscle contraction. Some forms of heart disease are associated with metabolic or enzymatic dysregulation of the MgATP-MgADP nucleotide pool, resulting in elevated cytosolic [MgADP] and impaired muscle relaxation. We investigated the mechanical and structural effects of increasing [MgADP] in permeabilized myocardial strips from porcine left ventricle samples. Sarcomere length was set to 2.0 µm at 28 °C, and all solutions contained 3% dextran T-500 to compress myofilament lattice spacing to near-physiological values. Under relaxing low [Ca2+] conditions (pCa 8.0, where pCa = -log10[Ca2+]), tension increased as [MgADP] increased from 0-5 mM. Complementary small-angle X-ray diffraction measurements show that the equatorial intensity ratio, I1,1/I1,0, also increased as [MgADP] increased from 0 to 5 mM, indicating myosin head movement away from the thick-filament backbone towards the thin-filament. Ca2+-activated force-pCa measurements show that Ca2+-sensitivity of contraction increased with 5 mM MgADP, compared to 0 mM MgADP. These data show that MgADP augments tension at low [Ca2+] and Ca2+-sensitivity of contraction, suggesting that MgADP destabilizes the quasi-helically ordered myosin OFF state, thereby shifting the cross-bridge population towards the disordered myosin ON state. Together, these results indicate that MgADP enhances the probability of cross-bridge binding to actin due to enhancement of both thick and thin filament-based activation mechanisms.
Collapse
Affiliation(s)
- Peter O. Awinda
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Weikang Ma
- The Biophysics Collaborative Access Team (BioCAT), Illinois Institute of Technology, Chicago, IL 60616, USA
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Kyrah L. Turner
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | - Jing Zhao
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Mindy S. Thompson
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Kenneth S. Campbell
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Thomas C. Irving
- The Biophysics Collaborative Access Team (BioCAT), Illinois Institute of Technology, Chicago, IL 60616, USA
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Bertrand C. W. Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
- Correspondence: ; Tel.: +1-(509)-335-7785; Fax: +1-(509)-335-4650
| |
Collapse
|
50
|
Rasicci DV, Tiwari P, Bodt SML, Desetty R, Sadler FR, Sivaramakrishnan S, Craig R, Yengo CM. Dilated cardiomyopathy mutation E525K in human beta-cardiac myosin stabilizes the interacting-heads motif and super-relaxed state of myosin. eLife 2022; 11:e77415. [PMID: 36422472 PMCID: PMC9691020 DOI: 10.7554/elife.77415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
The auto-inhibited, super-relaxed (SRX) state of cardiac myosin is thought to be crucial for regulating contraction, relaxation, and energy conservation in the heart. We used single ATP turnover experiments to demonstrate that a dilated cardiomyopathy (DCM) mutation (E525K) in human beta-cardiac myosin increases the fraction of myosin heads in the SRX state (with slow ATP turnover), especially in physiological ionic strength conditions. We also utilized FRET between a C-terminal GFP tag on the myosin tail and Cy3ATP bound to the active site of the motor domain to estimate the fraction of heads in the closed, interacting-heads motif (IHM); we found a strong correlation between the IHM and SRX state. Negative stain electron microscopy and 2D class averaging of the construct demonstrated that the E525K mutation increased the fraction of molecules adopting the IHM. Overall, our results demonstrate that the E525K DCM mutation may reduce muscle force and power by stabilizing the auto-inhibited SRX state. Our studies also provide direct evidence for a correlation between the SRX biochemical state and the IHM structural state in cardiac muscle myosin. Furthermore, the E525 residue may be implicated in crucial electrostatic interactions that modulate this conserved, auto-inhibited conformation of myosin.
Collapse
Affiliation(s)
- David V Rasicci
- Department of Cellular and Molecular Physiology, Penn State College of MedicineHersheyUnited States
| | - Prince Tiwari
- Department of Radiology, Division of Cell Biology and Imaging, UMass Chan Medical SchoolWorcesterUnited States
| | - Skylar ML Bodt
- Department of Cellular and Molecular Physiology, Penn State College of MedicineHersheyUnited States
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, Penn State College of MedicineHersheyUnited States
| | - Fredrik R Sadler
- Department of Genetics, Cell Biology, and Development, University of Minnesota Twin CitiesMinneapolisUnited States
| | - Sivaraj Sivaramakrishnan
- Department of Genetics, Cell Biology, and Development, University of Minnesota Twin CitiesMinneapolisUnited States
| | - Roger Craig
- Department of Radiology, Division of Cell Biology and Imaging, UMass Chan Medical SchoolWorcesterUnited States
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Penn State College of MedicineHersheyUnited States
| |
Collapse
|