1
|
Wen J, Campbell S, Moore J, Lehman W, Rynkiewicz M. Screening single nucleotide changes to tropomyosin to identify novel cardiomyopathy mutants. J Mol Cell Cardiol 2025; 203:82-90. [PMID: 40268117 DOI: 10.1016/j.yjmcc.2025.04.009] [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: 11/07/2024] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
Inherited cardiomyopathy is a broad class of heart disease that includes pathological cardiac remodeling such as hypertrophic and dilated cardiomyopathy, affecting 1/250-1/500 people worldwide. In many cases, mutations in proteins that make up the sarcomere, the basic subcellular unit of contraction, alter thin filament regulation and are the root cause of hypertrophic and dilated cardiomyopathy. Initially, compensations can maintain cardiac function, so patients may remain asymptomatic for years before a major cardiac episode. Early therapeutic intervention could rescue the deleterious effects of mutations thereby avoiding pathological remodeling, so prediction of potential outcomes and severity of as yet uncharacterized and known mutants of uncertain significance is critical. To accomplish this goal, we begin with the structure of the thin filament containing actin, tropomyosin, and troponin in its regulatory B- and C-states, incorporate all potential single nucleotide mutations to the tropomyosin sequence (over 1700 unique mutations), and then measure the interaction energy between tropomyosin and actin after energy minimization. Analysis of the database thus generated shows the tropomyosin residues resulting in large changes in tropomyosin-actin interaction, and therefore most likely to be deleterious to function. Some of these mutants have been observed in human patients, whereas others are novel. Global analysis further refines hotspots of mutation-sensitive, coiled-coil tropomyosin residues affecting actin interactions. Altogether, the database will allow research to focus in great depth on key candidates for functional analysis, for instance, by assaying in vitro motility and engineered heart tissue mechanics and assessing outcomes in animal models.
Collapse
Affiliation(s)
- Jian Wen
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Stuart Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Jeffrey Moore
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - William Lehman
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Michael Rynkiewicz
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
2
|
Moo EK, Joumaa V, Herzog W. Effect of pre-activation force on active force generation in skeletal muscle. J Biomech 2025; 186:112744. [PMID: 40344920 DOI: 10.1016/j.jbiomech.2025.112744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Cross bridges play a central role in skeletal muscle force generation. The level of force per cross bridge and the number of attached cross bridges are thought to determine muscle performance. Recent studies propose the so-called myosin-activation hypothesis, which suggests that stress exerted on myosin filaments increases the number of attached cross bridges and, hence, active force. This study was aimed at investigating the influence of passive stress magnitude exerted at the onset of activation on active force in a whole muscle preparation. The tibialis anterior (TA) muscle-tendon unit (MTU) of mice (N = 8) was stretched uniaxially in situ to long lengths where substantial viscoelastic passive force relaxation occurs. Muscle stress upon activation was varied by activating the TA either immediately at the end of the passive stretch (high passive force), or following nearly complete passive force relaxation (low passive force). Total forces with and without activation were measured from every MTU. Active forces were calculated by subtracting the passive force relaxation curve from the total force measured over a 1.13-s activation. We found that active force generated by the TA at low passive stress was 5-13 % higher than that at high passive stress. While the results seem contradictory to the myosin-activation hypothesis, we speculate that the results arose either from length adjustments between muscle and tendon during passive force relaxation, from excessive lattice spacing compression, or from unfavourable alterations of myosin conformation by high passive stress. Further research is required to improve our understanding of active force generation under the influence of viscoelasticity of muscle and tendon.
Collapse
Affiliation(s)
- Eng Kuan Moo
- Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, Canada; Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Canada.
| | - Venus Joumaa
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Canada.
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Canada.
| |
Collapse
|
3
|
Talanaite D, Wang H, Qi M, Lan F. Generation of a human embryonic stem cell line (WAe009-A-3B) carrying homozygous TNNT2 gene knockout by CRISPR/Cas9 editing. Stem Cell Res 2025; 86:103729. [PMID: 40344932 DOI: 10.1016/j.scr.2025.103729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/16/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025] Open
Abstract
The TNNT2 gene encodes cardiac troponin T (cTnT), a critical protein in cardiac muscle contraction. Mutations in TNNT2 are associated with various cardiomyopathies, including hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM), which contribute to significant morbidity and mortality. In this study, we established a novel TNNT2 knockout human embryonic stem cell (hESC) line, WAe009-A-3B, utilizing the CRISPR/Cas9 genome editing system. This novel hESC line provides an important tool for investigating the molecular mechanisms underlying TNNT2-related cardiomyopathies and may serve as a promising in vitro model for the development of therapeutic strategies targeting TNNT2 mutations in cardiac diseases.
Collapse
Affiliation(s)
- Didaer Talanaite
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Key Laboratory of Application of Pluripotent Stem Cells in Heart Regeneration, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Hongyue Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Key Laboratory of Application of Pluripotent Stem Cells in Heart Regeneration, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Man Qi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Key Laboratory of Application of Pluripotent Stem Cells in Heart Regeneration, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China; Chinese PLA General Hospital 8th medical center, Beijing 100091, PR China; State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, PR China.
| | - Feng Lan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Key Laboratory of Application of Pluripotent Stem Cells in Heart Regeneration, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China; Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen Key Laboratory of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, PR China; National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, PR China.
| |
Collapse
|
4
|
Wang X, Solaro RJ, Dong WJ. Myosin-actin crossbridge independent sarcomere length induced Ca 2+ sensitivity changes in skinned myocardial fibers: Role of myosin heads. J Mol Cell Cardiol 2025; 202:90-101. [PMID: 40073932 DOI: 10.1016/j.yjmcc.2025.03.003] [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: 08/24/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Sarcomere length-dependent activation (LDA) is essential to engaging the Frank-Starling mechanism in the beat-to-beat regulation of cardiac output. Through LDA, the heart increases the Ca2+ sensitivity of myocardial contraction at a longer sarcomere length, leading to an enhanced maximal force at the same level of Ca2+. Despite its importance in both normal and pathological states, the molecular mechanism underlying LDA, especially the origin of the sarcomere length (SL) induced increase in myofilament Ca2+sensitivity, remains elusive. The aim of this study is to interrogate the role of changes in the state of myosin heads during diastole as well as effects of strong force-generating cross-bridges (XB) as determinants of SL-induced Ca2+ sensitivity of troponin in membrane-free (skinned) rat myocardial fibers. Skinned myocardial fibers were reconstituted with troponin complex containing a fluorophore-modified cardiac troponin C, cTnC(13C/51C)AEDANS-DDPM, and recombinant cardiac troponin I (cTnI) mutant, ΔSP-cTnI, in which the switch peptide (Sp) of cTnI was replaced by a non-functional peptide link to partially block the force-generating reaction of myosin with actin. We used the reconstituted myocardial fibers as a platform to investigate how Ca2+ sensitivity of troponin within skinned myocardial fibers responds to sarcomere stretch with variations in the status of myosin-actin XBs. Muscle mechanics and fluorescence measurements clearly showed similar SL-induced increases in troponin Ca2+ sensitivity in either the presence or the absence of strong XBs, suggesting that the SL-induced Ca2+ sensitivity change is independent of reactions of force generating XB with the thin filament. The presence of mavacamten, a selective myosin-motor inhibitor known to promote transition of myosin heads from the weakly actin-bound state (ON or disordered relaxed (DRX) state) to the ordered off state (OFF or super-relaxed (SRX) state), blunted the observed SL-induced increases in Ca2+ sensitivity of troponin regardless of the presence of XBs, suggesting that the presence of the myosin heads in the weakly actin bound state, is essential for Ca2+-troponin to sense the sarcomere stretch. Results from skinned myocardial fibers reconstituted with troponin containing engineered TEV digestible mutant cTnI and cTnT suggest that the observed SL effect on Ca2+ sensitivity may involve potential interactions of weakly bound myosin heads with troponin in the actin/Tm cluster region interacting with cTnT-T1 and residues 182-229 of cTnT-T2. The mechanical stretch effects may then be subsequently transmitted to the N-cTnC via the IT arm of troponin and the N-terminus of cTnI. Our findings strongly indicate that SL-induced potential myosin-troponin interaction in diastole, rather than strong myosin-actin XBs, may be an essential molecular mechanism underlying LDA of myofilament.
Collapse
Affiliation(s)
- Xutu Wang
- Voiland School of Chemical and Bioengineering, Washington State University, Pullman, WA 99163-1062, USA
| | - R John Solaro
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Wen-Ji Dong
- Voiland School of Chemical and Bioengineering, Washington State University, Pullman, WA 99163-1062, USA; Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99163-1062, USA.
| |
Collapse
|
5
|
Hayes JB, Willet AH, Caplan LR, Knapik EW, Tyska MJ, Burnette DT. α- and β-myosin II can be non-uniformly distributed within the cardiac sarcomere. iScience 2025; 28:112233. [PMID: 40230528 PMCID: PMC11994913 DOI: 10.1016/j.isci.2025.112233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/29/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
In humans, the forces of the heartbeat are generated by both "fast" α-myosin II and "slow" β-myosin II molecular motors. Here, we report that α-myosin II and β-myosin II can take on an anisotropic arrangement within cardiac sarcomeres, the minimal contractile unit of cardiac muscle. In induced pluripotent stem cell (iPSC)-derived cardiac myocytes, we find that α-myosin II is distributed primarily within the central region of the filaments, being mostly absent from the distal ends, whereas β-myosin II is distributed along the entire filament. We measure that the outer bounds of α-myosin II in filament stacks are shorter than those of β-myosin II in both the zebrafish embryo and adult human myocardium. Our findings suggest that the two motors that drive the human heartbeat can have a specific and non-uniform arrangement within cardiac thick filaments, which may be thus divided into two distinct mechanical regions based on myosin II motor composition. We suggest potential roles for each motor during contraction-relaxation based on these distributions and recent literature.
Collapse
Affiliation(s)
- James B. Hayes
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Alaina H. Willet
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Leah R. Caplan
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Ela W. Knapik
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Dylan T. Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| |
Collapse
|
6
|
Severn J, Vacus T, Lauga E. Fluid mechanics of sarcomeres as porous media. SOFT MATTER 2025; 21:2849-2867. [PMID: 40125718 PMCID: PMC11931608 DOI: 10.1039/d4sm01327a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Muscle contraction, both in skeletal and cardiac tissue, is driven by sarcomeres, the microscopic units inside muscle cells where thick myosin and thin actin filaments slide past each other. During contraction and relaxation, the sarcomere's volume changes, causing sarcoplasm (intra-sarcomeric fluid) to flow out during contraction and back in as the sarcomere relaxes. We present a quantitative model of this sarcoplasmic flow, treating the sarcomere as an anisotropic porous medium with regions defined by the presence and absence of thick and thin filaments. Using semi-analytic methods, we solve for axial and lateral fluid flow within the filament lattice, calculating the permeabilities of this porous structure. We then apply these permeabilities within a Darcy model to determine the flow field generated during contraction. The predictions of our continuum model show excellent agreement with finite element simulations, reducing computational time by several orders of magnitude while maintaining accuracy in modelling the biophysical flow dynamics.
Collapse
Affiliation(s)
- John Severn
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge CB3 0WA, UK.
| | - Thomas Vacus
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge CB3 0WA, UK.
- Laboratoire de Physique de l'Ecole Normale Supérieure, Université PSL, 75005 Paris, France
| | - Eric Lauga
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge CB3 0WA, UK.
| |
Collapse
|
7
|
Ramachandran B, Rynkiewicz M, Lehman W. Velcro-binding by cardiac troponin-I traps tropomyosin on actin in a low-energy relaxed state. Biochem Biophys Res Commun 2025; 757:151595. [PMID: 40088678 PMCID: PMC11938286 DOI: 10.1016/j.bbrc.2025.151595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
During muscle relaxation at low sarcoplasmic Ca2+-concentration, the 40-nm long tropomyosin coiled coil is attracted by the C-terminal regulatory domain of troponin subunit-I to a "steric-blocking" B-state position on actin subunits of cardiac and skeletal muscle thin filaments. Tropomyosin located in this B-state position obstructs myosin-binding sites on actin, limiting access of myosin-crossbridge heads on actin. In turn, the steric-hindrance imposed on myosin-binding diminishes actomyosin ATPase, crossbridge movement along actin, and contractility, thus causing relaxation. In contrast, during muscle activation, at high sarcoplasmic Ca2+ levels, the troponin-induced tropomyosin interference is relieved, the tropomyosin coiled coil returns to its default C-state position on actin, and contractility proceeds. In the current study, we examined the energetics associated with tropomyosin's shift in position from its C-state to its B-state on actin and the influence of troponin-I on this relaxed state transition. Control studies showed that in the absence of troponin, the free energy difference between B- and C-state positions of tropomyosin on actin is negligible, i.e. neither B- nor C-state is obviously preferred on troponin-free actin. In contrast, widely separated sites along the C-terminal regulatory domain of troponin-I are responsible for a favorable free energy change of about -0.75 kcal/mol, driving the tropomyosin C-state to B-state shift. Corresponding truncation and point mutations along C-terminal region of TnI lead to a less favorable regulatory transition and are linked to cardiac muscle dysfunction.
Collapse
Affiliation(s)
- Balajee Ramachandran
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, United States
| | - Michael Rynkiewicz
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, United States
| | - William Lehman
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, United States.
| |
Collapse
|
8
|
Qiu X, Wu W, Zhang S, Huang C, Lin D. 3-Hydroxybutyrate Promotes Myoblast Proliferation and Differentiation through Energy Metabolism and GPR109a-Mediated Ca 2+-NFAT Signaling Pathways. J Proteome Res 2025; 24:2063-2080. [PMID: 40099866 DOI: 10.1021/acs.jproteome.4c01150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Skeletal muscle wasting is a critical clinical problem associated with several diseases that significantly impair patient outcomes due to the progressive loss of muscle mass and function. This study explores the potential of 3-hydroxybutyrate (3-HB) as a therapeutic agent to counteract muscle atrophy by promoting the proliferation and differentiation of C2C12 myoblasts. Using nuclear magnetic resonance (NMR)-based metabolomics analysis, we uncover the underlying mechanisms by which 3-HB exerts its effects. Our findings demonstrate that 3-HB exerts its effects through two distinct mechanisms: as a metabolic substrate and as a signaling molecule. As a metabolic substrate, 3-HB enhances myoblast energy efficiency by stimulating the expression of G protein-coupled receptor 109a (GPR109a), which subsequently upregulates the 3-HB transporters MCT1 and CD147, the utilization enzyme OXCT1, and phosphorylated AMPK, thereby increasing ATP production. As a signaling molecule, 3-HB activates GPR109a, promoting calcium influx, improving calcium homeostasis, and increasing the expression of Ca2+-related proteins such as CAMKK2. This signaling cascade activates calcineurin (CaN), facilitating NFAT translocation to the nucleus and gene expression that drives myoblast proliferation and differentiation. By elucidating the dual regulatory roles of 3-HB in energy metabolism and cellular signaling, this study not only advances our understanding of muscle physiology but also highlights the potential of 3-HB as a novel therapeutic approach for the prevention or treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Xu Qiu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Wenfang Wu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuya Zhang
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361024, China
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
9
|
Śmietana P, Śmietana N, Eljasik P, Lisiecki S, Sobczak M, Panicz R. Sexual dimorphism alters seasonal chelae muscle mechanisms in spiny-cheek crayfish ( Faxonius limosus). Front Physiol 2025; 16:1567862. [PMID: 40241719 PMCID: PMC12000753 DOI: 10.3389/fphys.2025.1567862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/05/2025] [Indexed: 04/18/2025] Open
Abstract
Sex-specific behaviours of freshwater crayfish are key elements in sustaining species persistence and successful conquering of new habitats in freshwater ecosystems. However, to date, information on molecular mechanisms that underpin the anatomy and physiology of crayfish sexes in successful mating behaviour was scarcely presented. In this study, Faxonius limosus females and males were sampled in spring and autumn to assess the impact of sexes and seasons on body parameters and activity of arginine kinase (ak), ferritin (fr), crustacean calcium-binding protein 23 (ccbp-23), troponin c (tnnc), and skeletal muscle actin 8 (actinsk8) genes related to the functioning of muscles in chelae. Comparison of body parameters showed significant differences in the weight and size of individuals in two seasons, underlining that large chelae are essential for males in mating behaviours and male-male competitive interactions. The gene expression analysis showed that activities of the five genes in the chelae muscle of F. limosus were influenced by the season- and sex-specific drivers. Multivariate analyses specifically identified the key genes (e.g., tnnc in males from spring) that were directly involved in metabolisms of chelae muscles of males and females collected in spring and autumn. The study, for the first time, described the direct impact of two key seasons and sexes on the anatomical features and molecular mechanisms that shaped the behaviour of F. limosus.
Collapse
Affiliation(s)
- Przemysław Śmietana
- Department of Environmental Ecology, Institute of Marine and Environmental Sciences, University of Szczecin, Szczecin, Poland
| | - Natalia Śmietana
- Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Szczecin, Poland
| | - Piotr Eljasik
- Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Szczecin, Poland
| | - Sławomir Lisiecki
- Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Szczecin, Poland
| | - Małgorzata Sobczak
- Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Szczecin, Poland
| | - Remigiusz Panicz
- Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Szczecin, Poland
| |
Collapse
|
10
|
Foster AD, Straight CR, Woods PC, Lee C, Kent JA, Chipkin SR, Debold EP, Miller MS. Cellular and molecular contractile function in aged human skeletal muscle is altered by phosphate and acidosis and partially reversed with an ATP analog. Am J Physiol Cell Physiol 2025; 328:C1220-C1233. [PMID: 40047118 DOI: 10.1152/ajpcell.00332.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/20/2024] [Accepted: 02/27/2025] [Indexed: 04/01/2025]
Abstract
Skeletal muscle fatigue occurs, in part, from the accumulation of hydrogen (H+) and phosphate (Pi); however, the molecular basis through which these ions inhibit function is not fully understood. Therefore, we examined the effects of these metabolites on myosin-actin cross-bridge kinetics and mechanical properties in skeletal muscle fibers from older (65-75 yr) adults. Slow-contracting myosin heavy chain (MHC) I and fast-contracting MHC IIA fibers were examined under control (5 mM Pi, pH 7.0) and fatigue (30 mM Pi, pH 6.2) conditions at maximal calcium-activation [5 mM adenosine triphosphate (ATP)] and rigor (0 mM ATP). In MHC I and IIA fibers, fatigue decreased force per fiber size (23%-37%), which was accompanied by reduced strongly bound myosin head characteristics (number and/or stiffness; 21%-47%) and slower cross-bridge kinetics [longer myosin attachment times (22%-46%) and reduced rates of force production (20%-33%)] compared with control. MHC I myofilaments became stiffer with fatigue, a potential mechanism to increase force production. In rigor, which causes the myosin that can bind actin to be strongly bound, fatigue decreased force per fiber size (32%-33%) in MHC I and IIA fibers, indicating less force was generated per cross bridge. By replacing ATP with 2-deoxy-ATP, the fatigue-induced slowing of cross-bridge kinetics in MHC I and IIA fibers was reversed, and reduced force production in MHC I fibers was partially improved, revealing potential mechanisms to help mitigate fatigue in older adults. Overall, our results identify novel fiber type-specific changes in cross-bridge kinetics, force per cross bridge, and myofilament stiffness that help explain fatigue in older adults.NEW & NOTEWORTHY Skeletal muscle fatigue is caused, in part, by increased production of phosphate and hydrogen ions, resulting in decreased force generation. We found that reduced force in fibers from older adults was due to altered function of myosin and actin, including slower protein interactions and reduced force per myosin head. Additionally, an ATP analog, dATP, partially reversed contractile dysfunction induced by increased phosphate and hydrogen, improving force production and altering myosin-actin interactions dependent upon fiber type.
Collapse
Affiliation(s)
- Aurora D Foster
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Chad R Straight
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Philip C Woods
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Christopher Lee
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Jane A Kent
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Stuart R Chipkin
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Edward P Debold
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| | - Mark S Miller
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, United States
| |
Collapse
|
11
|
Hill C, Kalakoutis M, Arcidiacono A, Paradine Cullup F, Wang Y, Fukutani A, Narayanan T, Brunello E, Fusi L, Irving M. Dual-filament regulation of relaxation in mammalian fast skeletal muscle. Proc Natl Acad Sci U S A 2025; 122:e2416324122. [PMID: 40073060 PMCID: PMC11929500 DOI: 10.1073/pnas.2416324122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/23/2025] [Indexed: 03/14/2025] Open
Abstract
Muscle contraction is driven by myosin motors from the thick filaments pulling on the actin-containing thin filaments of the sarcomere, and it is regulated by structural changes in both filaments. Thin filaments are activated by an increase in intracellular calcium concentration [Ca2+]i and by myosin binding to actin. Thick filaments are activated by direct sensing of the filament load. However, these mechanisms cannot explain muscle relaxation when [Ca2+]i decreases at high load and myosin motors are attached to actin. There is, therefore, a fundamental gap in our understanding of muscle relaxation, despite its importance for muscle function in vivo, for example, for rapid eye movements or, on slower timescales, for the efficient control of posture. Here, we used time-resolved small-angle X-ray diffraction (SAXD) to determine how muscle thin and thick filaments switch OFF in extensor digitorum longus (EDL) muscles of the mouse in response to decreases in either [Ca2+]i or muscle load and to describe the distribution of muscle sarcomere lengths (SLs) during relaxation. We show that reducing load at high [Ca2+]i is more effective in switching OFF both the thick and thin filaments than reducing [Ca2+]i at high load in normal relaxation. In the latter case, the thick filaments initially remain fully ON, although the number of myosin motors bound to actin decreases and the force per attached motor increases. That initial slow phase of relaxation is abruptly terminated by yielding of one population of sarcomeres, triggering a redistribution of SLs that leads to the rapid completion of mechanical relaxation.
Collapse
Affiliation(s)
- Cameron Hill
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| | - Michaeljohn Kalakoutis
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| | - Alice Arcidiacono
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| | - Flair Paradine Cullup
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| | - Yanhong Wang
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| | - Atsuki Fukutani
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| | | | - Elisabetta Brunello
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| | - Luca Fusi
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
- Centre for Human and Applied Physiological Sciences, Shepherd’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| | - Malcolm Irving
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt’s House, Guy’s Campus, King’s College London, LondonSE1 1UL, United Kingdom
| |
Collapse
|
12
|
Kimmich MJ, Geary MA, Mi-Mi L, Votra SD, Pellenz CD, Sundaramurthy S, Pruyne D. The Sole Essential Low Molecular Weight Tropomyosin Isoform of Caenorhabditis elegans Is Essential for Pharyngeal Muscle Function. Cytoskeleton (Hoboken) 2025. [PMID: 40078096 DOI: 10.1002/cm.22014] [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: 12/17/2024] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Tropomyosin is an actin-binding protein that plays roles ranging from regulating muscle contraction to controlling cytokinesis and cell migration. The simple nematode Caenorhabditis elegans provides a useful model for studying the core functions of tropomyosin in an animal, having a relatively simple anatomy and a single tropomyosin gene, lev-11, that produces seven isoforms. Three higher molecular weight isoforms regulate the contraction of body wall and other muscles, but comparatively less is known of the functions of four lower molecular weight isoforms (LEV-11C, E, T, U). We demonstrate here that C. elegans can survive with a single low molecular weight isoform, LEV-11E. Mutants disrupted for LEV-11E die as young larvae, whereas mutants lacking all other short isoforms are viable, with no overt phenotype. Vertebrate low molecular weight tropomyosins are often considered "nonmuscle" isoforms, but we find LEV-11E localizes to sarcomeric thin filaments in pharyngeal muscle and co-precipitates from worm extracts with the formin FHOD-1, which is also associated with thin filaments in pharyngeal muscle. Pharyngeal sarcomere organization is grossly normal in larvae lacking LEV-11E, indicating that the tropomyosin is not required to stabilize thin filaments, but pharyngeal pumping is absent, suggesting LEV-11E regulates actomyosin activity similar to higher molecular weight sarcomeric tropomyosin isoforms.
Collapse
Affiliation(s)
- Michael J Kimmich
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Meaghan A Geary
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Lei Mi-Mi
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - SarahBeth D Votra
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Christopher D Pellenz
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Sumana Sundaramurthy
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - David Pruyne
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
13
|
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
|
14
|
Childers MC. Modeling cardiac contractile cooperativity across species. J Gen Physiol 2025; 157:e202413722. [PMID: 39887987 PMCID: PMC11784582 DOI: 10.1085/jgp.202413722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025] Open
Abstract
Phan and Fitzsimons (https://doi.org/10.1085/jgp.202413582) develop a new mathematical model of muscle contraction that explores cooperative mechanisms in small (murine) and large (porcine) myocardium.
Collapse
|
15
|
Wei C, Vanhatalo A, Black MI, Rajaram R, Massey G, Jones AM. Dose-response relationship between dietary nitrate intake and nitric oxide congeners in various blood compartments and skeletal muscle: Differential effects on skeletal muscle torque and velocity. Free Radic Biol Med 2025; 229:520-533. [PMID: 39864759 DOI: 10.1016/j.freeradbiomed.2025.01.048] [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: 11/19/2024] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
Plasma nitrate (NO3-) and nitrite (NO2-) increase in a dose-dependent manner following NO3- ingestion. To explore if the same dose-response relationship applies to other nitric oxide (NO) congeners in different blood compartments and skeletal muscle, as well as the subsequent physiological responses, we provided 11 healthy participants with NO3- depleted beetroot juice (placebo), and beetroot juice (BR) containing 6.4, 12.8 and 19.2 mmol NO3- in a randomised, crossover design. Blood and muscle samples were collected, and resting blood pressure (BP) was assessed, before and at 2.5-3 h post-ingestion. Muscle contractile function was assessed using a 5-min all-out maximal voluntary isometric knee extension test at 3.5 h post-ingestion. We found that plasma and skeletal muscle [NO3-], and whole blood S-nitrosothiols ([RSNOs]) increased dose-dependently, while plasma [NO2-] did not increase further with doses above 6.4 mmol NO3-. No significant increases in skeletal muscle [NO2-] were found following ingestion of any of these doses. Resting BP was only reduced after ingestion of 19.2 mmol NO3-. Mean peak torque and mean torque impulse during the first 10 muscle contractions were significantly enhanced following ingestion of both 12.8 mmol and 19.2 mmol NO3- compared to placebo, while the mean absolute rate of torque development (RTD) at 0-50 ms and 0-100 ms was significantly improved following ingestion of 6.4 mmol NO3- compared to placebo and 19.2 mmol NO3-. Significant correlations were found between changes in red blood cell [RSNOs] and changes in absolute RTD at 0-50 ms (rs = -0.70, P = 0.02) and 0-100 ms (rs = -0.84, P < 0.01) following the ingestion of 6.4 mmol NO3-. Our findings suggest that a high dose of 12.8 mmol NO3- is necessary to improve muscle contractile torque, while a lower dose of 6.4 mmol NO3- is sufficient to enhance muscle contractile velocity, at least for the type of exercise employed in the present study.
Collapse
Affiliation(s)
- Chenguang Wei
- University of Exeter, Medical School, Faculty of Health and Life Sciences, St Luke's Campus, Exeter, EX1 2LU, UK
| | - Anni Vanhatalo
- University of Exeter, Medical School, Faculty of Health and Life Sciences, St Luke's Campus, Exeter, EX1 2LU, UK
| | - Matthew I Black
- University of Exeter, Medical School, Faculty of Health and Life Sciences, St Luke's Campus, Exeter, EX1 2LU, UK
| | - Raghini Rajaram
- University of Exeter, Medical School, Faculty of Health and Life Sciences, St Luke's Campus, Exeter, EX1 2LU, UK
| | - Garry Massey
- University of Exeter, Medical School, Faculty of Health and Life Sciences, St Luke's Campus, Exeter, EX1 2LU, UK
| | - Andrew M Jones
- University of Exeter, Medical School, Faculty of Health and Life Sciences, St Luke's Campus, Exeter, EX1 2LU, UK.
| |
Collapse
|
16
|
Doran MH, Rynkiewicz MJ, Despond E, Viswanathan MC, Madan A, Chitre K, Fenwick AJ, Sousa D, Lehman W, Dawson JF, Cammarato A. The hypertrophic cardiomyopathy-associated A331P actin variant enhances basal contractile activity and elicits resting muscle dysfunction. iScience 2025; 28:111816. [PMID: 39981516 PMCID: PMC11841076 DOI: 10.1016/j.isci.2025.111816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/29/2024] [Accepted: 01/13/2025] [Indexed: 02/22/2025] Open
Abstract
Previous studies aimed at defining the mechanistic basis of hypertrophic cardiomyopathy caused by A331P cardiac actin have reported conflicting results. The mutation is located along an actin surface strand, proximal to residues that interact with tropomyosin. These F-actin-tropomyosin associations are vital for proper contractile inhibition. To help resolve disease pathogenesis, we implemented a multidisciplinary approach. Transgenic Drosophila, expressing A331P actin, displayed skeletal muscle hypercontraction and elevated basal myocardial activity. A331P thin filaments, reconstituted using recombinant human cardiac actin, exhibited higher in vitro myosin-based sliding speeds, exclusively at low Ca2+ concentrations. Cryo-EM-based reconstructions revealed no detectable A331P-related structural perturbations in F-actin. In silico, however, the P331-containing actin surface strand was less mobile and established diminished van der Waal's attractive forces with tropomyosin, which correlated with greater variability in inhibitory tropomyosin positioning. Such mutation-induced effects potentially elevate resting contractile activity among our models and may stimulate pathology in patients.
Collapse
Affiliation(s)
- Matthew H. Doran
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord St, Boston, MA 02118, USA
| | - Michael J. Rynkiewicz
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord St, Boston, MA 02118, USA
| | - Evan Despond
- Department Molecular and Cellular Biology, University of Guelph, 50 Stone Road E, Guelph, ON N1G 2W1, Canada
| | - Meera C. Viswanathan
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Aditi Madan
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Kripa Chitre
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Axel J. Fenwick
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Duncan Sousa
- Department of Biophysics, Johns Hopkins University, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - William Lehman
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord St, Boston, MA 02118, USA
| | - John F. Dawson
- Department Molecular and Cellular Biology, University of Guelph, 50 Stone Road E, Guelph, ON N1G 2W1, Canada
| | - Anthony Cammarato
- Department of Medicine, Division of Cardiology, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
17
|
Nguyen A, Leong K, Holt NC. Does the unusual phenomenon of sustained force circumvent the speed-endurance trade-off in the jaw muscle of the southern alligator lizard (Elgaria multicarinata)? J Exp Biol 2025; 228:JEB247979. [PMID: 39690956 PMCID: PMC11832124 DOI: 10.1242/jeb.247979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024]
Abstract
The jaw muscles of the southern alligator lizard, Elgaria multicarinata, are used in prolonged mate-holding behavior, and also to catch fast prey. In both males and females, these muscles exhibit an unusual type of high endurance known as sustained force in which contractile force does not return to baseline between subsequent contractions. This phenomenon is assumed to facilitate the prolonged mate-holding observed in this species. Skeletal muscle is often subject to a speed-endurance trade-off. Here, we determined the isometric twitch, tetanic and isotonic force-velocity properties of the jaw muscles at ∼24°C as metrics of contractile speed and compared these properties with a more typical thigh locomotory muscle to determine whether endurance by sustained force allows for circumvention of the speed-endurance trade-off. The specialized jaw muscle was generally slower than the more typical thigh muscle: time to peak twitch force, twitch 90% relaxation time (P<0.01), and tetanic 90% and 50% relaxation times (P<0.001) were significantly longer, and force-velocity properties were significantly slower (P<0.001) in the jaw than the thigh muscle. However, there seemed to be greater effects on relaxation rates and shortening velocity than on force rise times: there was no effect of muscle on time to peak, or 50% of tetanic force. Hence, the jaw muscle of the southern alligator lizard does not seem to circumvent the speed-endurance trade-off. However, the maintenance of force rise times despite slow relaxation, potentially enabled by the presence of hybrid fibers, may allow this muscle to meet the functional demand of prey capture.
Collapse
Affiliation(s)
- Allyn Nguyen
- Evolution, Ecology, and Organismal Biology Department, University of California, Riverside, Riverside, CA 92521, USA
| | - Kyle Leong
- Evolution, Ecology, and Organismal Biology Department, University of California, Riverside, Riverside, CA 92521, USA
| | - Natalie C. Holt
- Evolution, Ecology, and Organismal Biology Department, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
18
|
Morotti I, Marcello M, Sautariello G, Pertici I, Bianco P, Piazzesi G, Linari M, Lombardi V, Reconditi M, Caremani M. The Mechanism of Modulation of Cardiac Force by Temperature. Int J Mol Sci 2025; 26:469. [PMID: 39859186 PMCID: PMC11764908 DOI: 10.3390/ijms26020469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
In maximally Ca2+-activated demembranated fibres from the mammalian skeletal muscle, the depression of the force by lowering the temperature below the physiological level (~35 °C) is explained by the reduction of force in the myosin motor. Instead, cooling is reported to not affect the force per motor in Ca2+-activated cardiac trabeculae from the rat ventricle. Here, the mechanism of the cardiac performance depression by cooling is reinvestigated with fast sarcomere-level mechanics. We determine the changes in the half-sarcomere compliance of maximally Ca2+-activated demembranated rat trabeculae in the range of temperatures of 10-30 °C and analyse the data in terms of a simplified mechanical model of the half-sarcomere to extract the contribution of myofilaments and myosin motors. We find that the changes in the ensemble force are due to changes in the force per motor, while the fraction of actin-attached motors remains constant independent of temperature. The results demonstrate that in the cardiac myosin, as in the skeletal muscle myosin, the force-generating transition is endothermic. The underlying large heat absorption indicates the interaction of extended hydrophobic surfaces within the myosin motor, like those suggested by the crystallographic model of the working stroke.
Collapse
Affiliation(s)
- Ilaria Morotti
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Matteo Marcello
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Giulia Sautariello
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Irene Pertici
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Pasquale Bianco
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Gabriella Piazzesi
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Marco Linari
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Vincenzo Lombardi
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Massimo Reconditi
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Marco Caremani
- PhysioLab, University of Florence, 50019 Sesto Fiorentino, Italy; (I.M.); (M.M.); (G.S.); (I.P.); (P.B.); (G.P.); (M.L.); (M.R.); (M.C.)
- Department of Biology, University of Florence, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
19
|
Stehle R. Phosphate rebinding induces force reversal via slow backward cycling of cross-bridges. Front Physiol 2025; 15:1476876. [PMID: 39839531 PMCID: PMC11747208 DOI: 10.3389/fphys.2024.1476876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/28/2024] [Indexed: 01/23/2025] Open
Abstract
Objective Previous studies on muscle fibers, myofibrils, and myosin revealed that the release of inorganic phosphate (Pi) and the force-generating step(s) are reversible, with cross-bridges also cycling backward through these steps by reversing force-generating steps and rebinding Pi. The aim was to explore the significance of force redevelopment kinetics (rate constant k TR) in cardiac myofibrils for the coupling between the Pi binding induced force reversal and the rate-limiting transition f - for backward cycling of cross-bridges from force-generating to non-force-generating states. Methods k TR and force generation of cardiac myofibrils from guinea pigs were investigated at 0.015-20 mM Pi. The observed force-[Pi], force-log [Pi], k TR-[Pi], and k TR-force relations were assessed with various single-pathway models of the cross-bridge cycle that differed in sequence and kinetics of reversible Pi release, reversible force-generating step and reversible rate-limiting transition. Based on the interpretation that k TR reflects the sum of rate-limiting transitions in the cross-bridge cycle, an indicator, the coupling strength, was defined to quantify the contribution of Pi binding induced force reversal to the rate-limiting transition f - from the [Pi]-modulated k TR-force relation. Results Increasing [Pi] decreased force by a bi-linear force-log [Pi] relation, increased k TR in a slightly downward curved dependence with [Pi], and altered k TR almost reciprocally to force reflected by the k TR-force relation. Force-[Pi] and force-log [Pi] relations provided less selectivity for the exclusion of models than the k TR-[Pi] and k TR-force relations. The k TR-force relation observed in experiments with cardiac myofibrils yielded the coupling strength +0.84 ± 0.08 close to 1, the maximum coupling strength expected for the reciprocal k TR-force relationship. Single pathway models consisting of fast reversible force generation before or after rapid reversible Pi release failed to describe the observed k TR-force relation. Single pathway models consistent with the observed k TR-force relation had either slow Pi binding or slow force reversal, i.e., in the consistent single pathway models, f - was assigned to the rate of either Pi binding or force reversal. Conclusion Backward flux of cross-bridges from force-generating to non-force-generating states is limited by the rates of Pi binding or force reversal ruling out other rate-limiting steps uncoupled from Pi binding induced force reversal.
Collapse
Affiliation(s)
- Robert Stehle
- Institute of Vegetative Physiology, University of Cologne, Köln, Germany
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Patsalis C, Kyriakou S, Georgiadou M, Ioannou L, Constantinou L, Soteriou V, Jossif A, Evangelidou P, Sismani C, Kypri E, Ioannides M, Koumbaris G. Investigating TNNC1 gene inheritance and clinical outcomes through a comprehensive familial study. Am J Med Genet A 2025; 197:e63838. [PMID: 39248034 DOI: 10.1002/ajmg.a.63838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 09/10/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) and restrictive cardiomyopathy (RCM) have significant phenotypic overlap and a similar genetic background, both caused mainly by variants in sarcomeric genes. HCM is the most common cardiomyopathy, while RCM is a rare and often underdiagnosed heart condition, with a poor prognosis. This study focuses on a large family with four infants diagnosed with fatal RCM associated with biventricular hypertrophy. Affected infants were found to be homozygous for NM_003280.3(TNNC1):c.23C>T(p.Ala8Val) variant. Interestingly, this variant resulted in a low penetrance and mild form of hypertrophic cardiomyopathy (HCM) in relatives carrying a single copy of the variant. Overall, this study underscores the complex nature of genetic inheritance in cardiomyopathies and the wide range of clinical presentations they can exhibit. This emphasizes the vital role of genetic testing in providing essential insights crucial for diagnosis, prognosis, early intervention, and the development of potential treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Antonis Jossif
- Paedi Center for Specialized Pediatrics, Nicosia, Cyprus
| | - Paola Evangelidou
- Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Carolina Sismani
- Department of Cytogenetics and Genomics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | | | | | | |
Collapse
|
23
|
Kimmich MJ, Geary MA, Mi-Mi L, Votra SD, Pellenz CD, Sundaramurthy S, Pruyne D. The sole essential low molecular weight tropomyosin isoform of Caenorhabditis elegans is essential for pharyngeal muscle function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628433. [PMID: 39764053 PMCID: PMC11702560 DOI: 10.1101/2024.12.13.628433] [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] [Indexed: 01/19/2025]
Abstract
Tropomyosin is an actin-binding protein that plays roles ranging from regulating muscle contraction to controlling cytokinesis and cell migration. The simple nematode Caenorhabditis elegans provides a useful model for studying the core functions of tropomyosin in an animal, having a relatively simple anatomy, and a single tropomyosin gene, lev-11, that produces seven isoforms. Three higher molecular weight isoforms (LEV-11A, D, O) regulate contraction of body wall and other muscles, but comparatively less is known of the functions of four lower molecular weight isoforms (LEV-11C, E, T, U). We demonstrate here C. elegans can survive with a single low molecular weight isoform, LEV-11E. Mutants disrupted for LEV-11E die as young larvae, whereas mutants disrupted for all other short isoforms are viable with no overt phenotype. Vertebrate low molecular weight tropomyosins are often considered "nonmuscle" isoforms, but we find LEV-11E localizes to sarcomeric thin filaments in pharyngeal muscle, and co-precipitates from worm extracts with the formin FHOD-1, which is also associated with thin filaments in pharyngeal muscle. Pharyngeal sarcomere organization is grossly normal in larvae lacking LEV-11E, indicating the tropomyosin is not required to stabilize thin filaments, but pharyngeal pumping is absent, suggesting LEV-11E regulates actomyosin activity similar to higher molecular weight sarcomeric tropomyosin isoforms.
Collapse
Affiliation(s)
- Michael J Kimmich
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - Meaghan A Geary
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - Lei Mi-Mi
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - SarahBeth D Votra
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - Christopher D Pellenz
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - Sumana Sundaramurthy
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| | - David Pruyne
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 766 Irving Avenue, Syracuse, NY 13210
| |
Collapse
|
24
|
Dominic KL, Schmidt AV, Granzier H, Campbell KS, Stelzer JE. Mechanism-based myofilament manipulation to treat diastolic dysfunction in HFpEF. Front Physiol 2024; 15:1512550. [PMID: 39726859 PMCID: PMC11669688 DOI: 10.3389/fphys.2024.1512550] [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: 10/16/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major public health challenge, affecting millions worldwide and placing a significant burden on healthcare systems due to high hospitalization rates and limited treatment options. HFpEF is characterized by impaired cardiac relaxation, or diastolic dysfunction. However, there are no therapies that directly treat the primary feature of the disease. This is due in part to the complexity of normal diastolic function, and the challenge of isolating the mechanisms responsible for dysfunction in HFpEF. Without a clear understanding of the mechanisms driving diastolic dysfunction, progress in treatment development has been slow. In this review, we highlight three key areas of molecular dysregulation directly underlying impaired cardiac relaxation in HFpEF: altered calcium sensitivity in the troponin complex, impaired phosphorylation of myosin-binding protein C (cMyBP-C), and reduced titin compliance. We explore how targeting these pathways can restore normal relaxation, improve diastolic function, and potentially provide new therapeutic strategies for HFpEF treatment. Developing effective HFpEF therapies requires precision targeting to balance systolic and diastolic function, avoiding both upstream non-specificity and downstream rigidity. This review highlights three rational molecular targets with a strong mechanistic basis and potential for therapeutic success.
Collapse
Affiliation(s)
- Katherine L. Dominic
- 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
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Kenneth S. Campbell
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, United States
- Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
25
|
Morotti I, Caremani M, Marcello M, Pertici I, Squarci C, Bianco P, Narayanan T, Piazzesi G, Reconditi M, Lombardi V, Linari M. An integrated picture of the structural pathways controlling the heart performance. Proc Natl Acad Sci U S A 2024; 121:e2410893121. [PMID: 39630866 DOI: 10.1073/pnas.2410893121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
The regulation of heart function is attributed to a dual filament mechanism: i) the Ca2+-dependent structural changes in the regulatory proteins of the thin, actin-containing filament making actin available for myosin motor attachment, and ii) the release of motors from their folded (OFF) state on the surface of the thick filament allowing them to attach and pull the actin filament. Thick filament mechanosensing is thought to control the number of motors switching ON in relation to the systolic performance, but its molecular basis is still controversial. Here, we use high spatial resolution X-ray diffraction data from electrically paced rat trabeculae and papillary muscles to provide a molecular explanation of the modulation of heart performance that calls for a revision of the mechanosensing hypothesis. We find that upon stimulation, titin-mediated structural changes in the thick filament switch motors ON throughout the filament within ~½ the maximum systolic force. These structural changes also drive Myosin Binding Protein-C (MyBP-C) to promote first motor attachments to actin from the central 1/3 of the half-thick filament. Progression of attachments toward the periphery of half-thick filament with increase in systolic force is carried on by near-neighbor cooperative thin filament activation by attached motors. The identification of the roles of MyBP-C, titin, thin and thick filaments in heart regulation enables their targeting for potential therapeutic interventions.
Collapse
Affiliation(s)
- Ilaria Morotti
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Marco Caremani
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Matteo Marcello
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Irene Pertici
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Caterina Squarci
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Pasquale Bianco
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | | | - Gabriella Piazzesi
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Massimo Reconditi
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Vincenzo Lombardi
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| | - Marco Linari
- PhysioLab, University of Florence, Sesto Fiorentino 50019, Italy
- Department of Biology, University of Florence, Sesto Fiorentino 50019, Italy
| |
Collapse
|
26
|
Steinz MM, Beard N, Shorter E, Lanner JT. Stable oxidative posttranslational modifications alter the gating properties of RyR1. J Gen Physiol 2024; 156:e202313515. [PMID: 39499505 PMCID: PMC11540854 DOI: 10.1085/jgp.202313515] [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: 11/29/2023] [Revised: 07/03/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024] Open
Abstract
The ryanodine receptor type 1 (RyR1) is a Ca2+ release channel that regulates skeletal muscle contraction by controlling Ca2+ release from the sarcoplasmic reticulum (SR). Posttranslational modifications (PTMs) of RyR1, such as phosphorylation, S-nitrosylation, and carbonylation are known to increase RyR1 open probability (Po), contributing to SR Ca2+ leak and skeletal muscle dysfunction. PTMs on RyR1 have been linked to muscle dysfunction in diseases like breast cancer, rheumatoid arthritis, Duchenne muscle dystrophy, and aging. While reactive oxygen species (ROS) and oxidative stress induce PTMs, the impact of stable oxidative modifications like 3-nitrotyrosine (3-NT) and malondialdehyde adducts (MDA) on RyR1 gating remains unclear. Mass spectrometry and single-channel recordings were used to study how 3-NT and MDA modify RyR1 and affect Po. Both modifications increased Po in a dose-dependent manner, with mass spectrometry identifying 30 modified residues out of 5035 amino acids per RyR1 monomer. Key modifications were found in domains critical for protein interaction and channel activation, including Y808/3NT in SPRY1, Y1081/3NT and H1254/MDA in SPRY2&3, and Q2107/MDA and Y2128/3NT in JSol, near the binding site of FKBP12. Though these modifications did not directly overlap with FKBP12 binding residues, they promoted FKBP12 dissociation from RyR1. These findings provide detailed insights into how stable oxidative PTMs on RyR1 residues alter channel gating, advancing our understanding of RyR1-mediated Ca2+ release in conditions associated with oxidative stress and muscle weakness.
Collapse
Affiliation(s)
- Maarten M. Steinz
- Department of Physiology and Pharmacology, Molecular Muscle Physiology and Pathophysiology lab, Karolinska Institutet, Stockholm, Sweden
| | - Nicole Beard
- Faculty or Science and Technology, University of Canberra, Canberra, Australia
| | - Emily Shorter
- Department of Physiology and Pharmacology, Molecular Muscle Physiology and Pathophysiology lab, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T. Lanner
- Department of Physiology and Pharmacology, Molecular Muscle Physiology and Pathophysiology lab, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Ghanta KP, Tardiff JC, Schwartz SD. The transmission of mutation effects in a multiprotein machine: A comprehensive metadynamics study of the cardiac thin filament. Protein Sci 2024; 33:e5215. [PMID: 39548812 PMCID: PMC11568392 DOI: 10.1002/pro.5215] [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: 08/29/2024] [Revised: 10/03/2024] [Accepted: 10/24/2024] [Indexed: 11/18/2024]
Abstract
The binding of Ca2+ ions within the troponin core of the cardiac thin filament (CTF) regulates normal contraction and relaxation. Mutations within the troponin complexes are known to alter normal functions and result in the eventual development of cardiomyopathy. However, despite the importance of the problem, detailed microscopic knowledge of the mechanism of pathogenic effect of point mutations and their effects on the conformational free energy surface of CTF remains elusive. Mutations are known to transmit their effects hundreds of angstroms along this protein complex and between different component proteins. To explore the impact of point mutations on the conformational free energy barrier between the closed and blocked state of CTF, and to understand the transmission of mutation, we have carried out metadynamics simulations for the wild-type (WT) and two mutants (cardiac troponin T Arg92Trp (R92W) and Arg92Leu (R92L)). Specifically, we have investigated the conformational modification of the tropomyosin (Tm) and the troponin (Tn) complex during the closed-to-blocked state transition for both the WT and two hypertrophic cardiomyopathy causing mutations. Our calculations demonstrated that mutations within the cardiac troponin T (cTnT) protein alter conformational properties of the Tm and the other proteins of the Tn complex as well as the Ca2+ binding affinity of the cTnC protein through the indirect mediation of cardiac troponin I (cTnI). Importantly, the data revealed a significant influence of the mutations on the conformational transition free energy barriers for both the Tm and cTnC proteins. Furthermore, we found both mutations independently alter the free energy barrier of transitions of cTnT. Such alteration in the free energy upon mutation of one protein in a complex, allosterically affects the others through structural and dynamical changes, leading to a pathogenic effect on the function of the thin filament.
Collapse
Affiliation(s)
| | - Jil C. Tardiff
- Department of Biomedical EngineeringThe University of ArizonaTucsonArizonaUSA
| | - Steven D. Schwartz
- Department of Chemistry and BiochemistryThe University of ArizonaTucsonArizonaUSA
| |
Collapse
|
28
|
Huang HL, Suchenko A, Grandinetti G, Balasubramanian MK, Chinthalapudi K, Heissler SM. Cryo-EM structures of cardiac muscle α-actin mutants M305L and A331P give insights into the structural mechanisms of hypertrophic cardiomyopathy. Eur J Cell Biol 2024; 103:151460. [PMID: 39393252 PMCID: PMC11611453 DOI: 10.1016/j.ejcb.2024.151460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/13/2024] Open
Abstract
Cardiac muscle α-actin is a key protein of the thin filament in the muscle sarcomere that, together with myosin thick filaments, produce force and contraction important for normal heart function. Missense mutations in cardiac muscle α-actin can cause hypertrophic cardiomyopathy, a complex disorder of the heart characterized by hypercontractility at the molecular scale that leads to diverse clinical phenotypes. While the clinical aspects of hypertrophic cardiomyopathy have been extensively studied, the molecular mechanisms of missense mutations in cardiac muscle α-actin that cause the disease remain largely elusive. Here we used cryo-electron microscopy to reveal the structures of hypertrophic cardiomyopathy-associated actin mutations M305L and A331P in the filamentous state. We show that the mutations have subtle impacts on the overall architecture of the actin filament with mutation-specific changes in the nucleotide binding cleft active site, interprotomer interfaces, and local changes around the mutation site. This suggests that structural changes induced by M305L and A331P have implications for the positioning of the thin filament protein tropomyosin and the interaction with myosin motors. Overall, this study supports a structural model whereby altered interactions between thick and thin filament proteins contribute to disease mechanisms in hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Hsiang-Ling Huang
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Andrejus Suchenko
- Centre for Mechanochemical Cell Biology and Warwick Medical School, Division of Biomedical Sciences, Coventry, United Kingdom
| | - Giovanna Grandinetti
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA; Center for Electron Microscopy and Analysis, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Mohan K Balasubramanian
- Centre for Mechanochemical Cell Biology and Warwick Medical School, Division of Biomedical Sciences, Coventry, United Kingdom
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
29
|
Sevrieva IR, Kampourakis T, Irving M. Structural changes in troponin during activation of skeletal and heart muscle determined in situ by polarised fluorescence. Biophys Rev 2024; 16:753-772. [PMID: 39830118 PMCID: PMC11735716 DOI: 10.1007/s12551-024-01245-y] [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: 08/04/2024] [Accepted: 09/30/2024] [Indexed: 01/22/2025] Open
Abstract
Calcium binding to troponin triggers the contraction of skeletal and heart muscle through structural changes in the thin filaments that allow myosin motors from the thick filaments to bind to actin and drive filament sliding. Here, we review studies in which those changes were determined in demembranated fibres of skeletal and heart muscle using fluorescence for in situ structure (FISS), which determines domain orientations using polarised fluorescence from bifunctional rhodamine attached to cysteine pairs in the target domain. We describe the changes in the orientations of the N-terminal lobe of troponin C (TnCN) and the troponin IT arm in skeletal and cardiac muscle cells associated with contraction and compare the orientations with those determined in isolated cardiac thin filaments by cryo-electron microscopy. We show that the orientations of the IT arm determined by the two approaches are essentially the same and that this region acts as an almost rigid scaffold for regulatory changes in the more mobile regions of troponin. However, the TnCN orientations determined by the two methods are clearly distinct in both low- and high-calcium conditions. We discuss the implications of these results for the role of TnCN in mediating the multiple signalling pathways acting through troponin in heart muscle cells and the general advantages and limitations of FISS and cryo-EM for determining protein domain orientations in cells and multiprotein complexes.
Collapse
Affiliation(s)
- Ivanka R Sevrieva
- Randall Centre for Cell & Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, UK
| | - Thomas Kampourakis
- Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Malcolm Irving
- Randall Centre for Cell & Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, UK
| |
Collapse
|
30
|
Morales PN, Coons AN, Koopman AJ, Patel S, Chase PB, Parvatiyar MS, Pinto JR. Post-translational modifications of vertebrate striated muscle myosin heavy chains. Cytoskeleton (Hoboken) 2024; 81:832-842. [PMID: 38587113 PMCID: PMC11458826 DOI: 10.1002/cm.21857] [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: 12/11/2023] [Revised: 03/06/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
Post-translational modifications (PTMs) play a crucial role in regulating the function of many sarcomeric proteins, including myosin. Myosins comprise a family of motor proteins that play fundamental roles in cell motility in general and muscle contraction in particular. A myosin molecule consists of two myosin heavy chains (MyHCs) and two pairs of myosin light chains (MLCs); two MLCs are associated with the neck region of each MyHC's N-terminal head domain, while the two MyHC C-terminal tails form a coiled-coil that polymerizes with other MyHCs to form the thick filament backbone. Myosin undergoes extensive PTMs, and dysregulation of these PTMs may lead to abnormal muscle function and contribute to the development of myopathies and cardiovascular disorders. Recent studies have uncovered the significance of PTMs in regulating MyHC function and showed how these PTMs may provide additional modulation of contractile processes. Here, we discuss MyHC PTMs that have been biochemically and/or functionally studied in mammals' and rodents' striated muscle. We have identified hotspots or specific regions in three isoforms of myosin (MYH2, MYH6, and MYH7) where the prevalence of PTMs is more frequent and could potentially play a significant role in fine-tuning the activity of these proteins.
Collapse
Affiliation(s)
- Paula Nieto Morales
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306 USA
| | - Arianna N. Coons
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Amelia J. Koopman
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Sonu Patel
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - P. Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Michelle S. Parvatiyar
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - Jose R. Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306 USA
| |
Collapse
|
31
|
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
|
32
|
Ragusa R, Caselli C. Focus on cardiac troponin complex: From gene expression to cardiomyopathy. Genes Dis 2024; 11:101263. [PMID: 39211905 PMCID: PMC11357864 DOI: 10.1016/j.gendis.2024.101263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 09/04/2024] Open
Abstract
The cardiac troponin complex (cTn) is a regulatory component of sarcomere. cTn consists of three subunits: cardiac troponin C (cTnC), which confers Ca2+ sensitivity to muscle; cTnI, which inhibits the interaction of cross-bridge of myosin with thin filament during diastole; and cTnT, which has multiple roles in sarcomere, such as promoting the link between the cTnI-cTnC complex and tropomyosin within the thin filament and influencing Ca2+ sensitivity of cTn and force development during contraction. Conditions that interfere with interactions within cTn and/or other thin filament proteins can be key factors in the regulation of cardiac contraction. These conditions include alterations in myofilament Ca2+ sensitivity, direct changes in cTn function, and triggering downstream events that lead to adverse cardiac remodeling and impairment of heart function. This review describes gene expression and post-translational modifications of cTn as well as the conditions that can adversely affect the delicate balance among the components of cTn, thereby promoting contractile dysfunction.
Collapse
Affiliation(s)
- Rosetta Ragusa
- Institute of Clinical Physiology, CNR, via Moruzzi 1, Pisa 56124, Italy
| | - Chiara Caselli
- Institute of Clinical Physiology, CNR, via Moruzzi 1, Pisa 56124, Italy
- Fondazione Toscana Gabriele Monasterio, via Moruzzi 1, Pisa 56124, Italy
| |
Collapse
|
33
|
Kochurova AM, Beldiia EA, Nefedova VV, Yampolskaya DS, Koubassova NA, Kleymenov SY, Antonets JY, Ryabkova NS, Katrukha IA, Bershitsky SY, Matyushenko AM, Kopylova GV, Shchepkin DV. The D75N and P161S Mutations in the C0-C2 Fragment of cMyBP-C Associated with Hypertrophic Cardiomyopathy Disturb the Thin Filament Activation, Nucleotide Exchange in Myosin, and Actin-Myosin Interaction. Int J Mol Sci 2024; 25:11195. [PMID: 39456977 PMCID: PMC11508426 DOI: 10.3390/ijms252011195] [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: 08/28/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
About half of the mutations that lead to hypertrophic cardiomyopathy (HCM) occur in the MYBPC3 gene. However, the molecular mechanisms of pathogenicity of point mutations in cardiac myosin-binding protein C (cMyBP-C) remain poorly understood. In this study, we examined the effects of the D75N and P161S substitutions in the C0 and C1 domains of cMyBP-C on the structural and functional properties of the C0-C1-m-C2 fragment (C0-C2). Differential scanning calorimetry revealed that these mutations disorder the tertiary structure of the C0-C2 molecule. Functionally, the D75N mutation reduced the maximum sliding velocity of regulated thin filaments in an in vitro motility assay, while the P161S mutation increased it. Both mutations significantly reduced the calcium sensitivity of the actin-myosin interaction and impaired thin filament activation by cross-bridges. D75N and P161S C0-C2 fragments substantially decreased the sliding velocity of the F-actin-tropomyosin filament. ADP dose-dependently reduced filament sliding velocity in the presence of WT and P161S fragments, but the velocity remained unchanged with the D75N fragment. We suppose that the D75N mutation alters nucleotide exchange kinetics by decreasing ADP affinity to the ATPase pocket and slowing the myosin cycle. Our molecular dynamics simulations mean that the D75N mutation affects myosin S1 function. Both mutations impair cardiac contractility by disrupting thin filament activation. The results offer new insights into the HCM pathogenesis caused by missense mutations in N-terminal domains of cMyBP-C, highlighting the distinct effects of D75N and P161S mutations on cardiac contractile function.
Collapse
Affiliation(s)
- Anastasia M. Kochurova
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | - Evgenia A. Beldiia
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | - Victoria V. Nefedova
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Daria S. Yampolskaya
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | | | - Sergey Y. Kleymenov
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Julia Y. Antonets
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | - Natalia S. Ryabkova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- HyTest Ltd., 20520 Turku, Finland
| | - Ivan A. Katrukha
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- HyTest Ltd., 20520 Turku, Finland
| | - Sergey Y. Bershitsky
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | | | - Galina V. Kopylova
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | - Daniil V. Shchepkin
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| |
Collapse
|
34
|
Bruns H, Czajka TS, Sztucki M, Brandenburg S, Salditt T. Sarcomere, troponin, and myosin X-ray diffraction signals can be resolved in single cardiomyocytes. Biophys J 2024; 123:3024-3037. [PMID: 38956875 PMCID: PMC11427778 DOI: 10.1016/j.bpj.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/18/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024] Open
Abstract
Cardiac function relies on the autonomous molecular contraction mechanisms in the ventricular wall. Contraction is driven by ordered motor proteins acting in parallel to generate a macroscopic force. The averaged structure can be investigated by diffraction from model tissues such as trabecular and papillary cardiac muscle using collimated synchrotron beams, offering high resolution in reciprocal space. In the ventricular wall, however, the muscle tissue is compartmentalized into smaller branched cardiomyocytes, with a higher degree of disorder. We show that X-ray diffraction is now also capable of resolving the structural organization of actomyosin in single isolated cardiomyocytes of the ventricular wall. In addition to the hexagonal arrangement of thick and thin filaments, the diffraction signal of the hydrated and fixated cardiomyocytes was sufficient to reveal the myosin motor repeat (M3), the troponin complex repeat (Tn), and the sarcomere length. The sarcomere length signal comprised up to 13 diffraction orders, which were used to compute the sarcomere density profile based on Fourier synthesis. The Tn and M3 spacings were found in the same range as previously reported for other muscle types. The approach opens up a pathway to record the structural dynamics of living cells during the contraction cycle, toward a more complete understanding of cardiac muscle function.
Collapse
Affiliation(s)
| | | | - Michael Sztucki
- ESRF - European Synchrotron Radiation Facility, Grenoble, France
| | - Sören Brandenburg
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Tim Salditt
- Institute for X-ray Physics, Göttingen, Germany.
| |
Collapse
|
35
|
Bardova K, Janovska P, Vavrova A, Kopecky J, Zouhar P. Adaptive Induction of Nonshivering Thermogenesis in Muscle Rather Than Brown Fat Could Counteract Obesity. Physiol Res 2024; 73:S279-S294. [PMID: 38752772 PMCID: PMC11412341 DOI: 10.33549/physiolres.935361] [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: 09/04/2024] Open
Abstract
Warm-blooded animals such as birds and mammals are able to protect stable body temperature due to various thermogenic mechanisms. These processes can be facultative (occurring only under specific conditions, such as acute cold) and adaptive (adjusting their capacity according to long-term needs). They can represent a substantial part of overall energy expenditure and, therefore, affect energy balance. Classical mechanisms of facultative thermogenesis include shivering of skeletal muscles and (in mammals) non-shivering thermogenesis (NST) in brown adipose tissue (BAT), which depends on uncoupling protein 1 (UCP1). Existence of several alternative thermogenic mechanisms has been suggested. However, their relative contribution to overall heat production and the extent to which they are adaptive and facultative still needs to be better defined. Here we focus on comparison of NST in BAT with thermogenesis in skeletal muscles, including shivering and NST. We present indications that muscle NST may be adaptive but not facultative, unlike UCP1-dependent NST. Due to its slow regulation and low energy efficiency, reflecting in part the anatomical location, induction of muscle NST may counteract development of obesity more effectively than UCP1-dependent thermogenesis in BAT.
Collapse
Affiliation(s)
- K Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague 4, Czech Republic. or
| | | | | | | | | |
Collapse
|
36
|
Creso JG, Gokhan I, Rynkiewicz MJ, Lehman W, Moore JR, Campbell SG. In silico and in vitro models reveal the molecular mechanisms of hypocontractility caused by TPM1 M8R. Front Physiol 2024; 15:1452509. [PMID: 39282088 PMCID: PMC11392859 DOI: 10.3389/fphys.2024.1452509] [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: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is an inherited disorder often leading to severe heart failure. Linkage studies in affected families have revealed hundreds of different mutations that can cause DCM, with most occurring in genes associated with the cardiac sarcomere. We have developed an investigational pipeline for discovering mechanistic genotype-phenotype relationships in DCM and here apply it to the DCM-linked tropomyosin mutation TPM1 M8R. Atomistic simulations predict that M8R increases flexibility of the tropomyosin chain and enhances affinity for the blocked or inactive state of tropomyosin on actin. Applying these molecular effects to a Markov model of the cardiac thin filament reproduced the shifts in Ca2+sensitivity, maximum force, and a qualitative drop in cooperativity that were observed in an in vitro system containing TPM1 M8R. The model was then used to simulate the impact of M8R expression on twitch contractions of intact cardiac muscle, predicting that M8R would reduce peak force and duration of contraction in a dose-dependent manner. To evaluate this prediction, TPM1 M8R was expressed via adenovirus in human engineered heart tissues and isometric twitch force was observed. The mutant tissues manifested depressed contractility and twitch duration that agreed in detail with model predictions. Additional exploratory simulations suggest that M8R-mediated alterations in tropomyosin-actin interactions contribute more potently than tropomyosin chain stiffness to cardiac twitch dysfunction, and presumably to the ultimate manifestation of DCM. This study is an example of the growing potential for successful in silico prediction of mutation pathogenicity for inherited cardiac muscle disorders.
Collapse
Affiliation(s)
- Jenette G. Creso
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Ilhan Gokhan
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Michael J. Rynkiewicz
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - William Lehman
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jeffrey R. Moore
- Department of Biological Sciences, University of Massachusetts–Lowell, Lowell, MA, United States
| | - Stuart G. Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
37
|
Silva-Del-Rio N, Solórzano LC, Lago A, Lobo RR, Goff JP, Weiss WP, Tempelman RJ. Relative availability of five inorganic magnesium sources in non-pregnant non-lactating Holstein cows. J Dairy Sci 2024:S0022-0302(24)01052-X. [PMID: 39098497 DOI: 10.3168/jds.2024-25106] [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: 04/29/2024] [Accepted: 07/13/2024] [Indexed: 08/06/2024]
Abstract
Inorganic sources of Mg are commonly used in dairy cow diets, but their availability varies significantly. This study assessed the relative availability of 4 commonly used inorganic Mg sources and a novel alkalinizing proprietary mineral blend [PMB; Multesium (GLC Minerals, LLC, Green Bay, WI, USA)]. The study was a duplicated 6 × 6 Latin square, with 12 nonlactating, non-pregnant Holstein dairy cows assigned to a square based on BW and parity. Cows were fed 90% of their voluntary DMI (diet contained 0.21% Mg). Each experimental period lasted 7 d. On d 2 of each period, urinary catheters were fitted. Total urine collection started on d 3 for 48 h with samples collected and measured every 12 h. On d 4, 30 g of Mg were administered as boluses with gelatin capsules: negative control (one empty capsule), magnesium oxide (MgO), magnesium sulfate (MgSO4), calcium magnesium hydroxide [CaMg(OH)4], calcium magnesium carbonate [CaMg(CO3)2], and PMB [a blend of Ca and Mg sources that includes CaMg(CO3)2, CaMg(OH)4, and MgO]. Blood samples were collected at 0, 1, 2, 3, 12, and 24 h after treatment administration on d 4 of each treatment period. Urine and blood samples were analyzed for Mg and Ca concentration. Statistical analyses were conducted with PROC GLIMMIX including treatment, time, period, square, treatment × time, treatment × period, and time × period as fixed effects, and cow nested within square as a random effect in the model. Urinary Mg excretion for 4 of the Mg sources studied [PMB, MgO, CaMg(OH)4, and MgSO4] increased significantly, representing an increase of at least 40.8% relative to control. The supplementation of CaMg(CO3)2 did not significantly increase relative to control. There were no significant changes in blood Mg concentration with treatment; but, a significant treatment × time effect was observed. Calcium-rich sources [PMB, CaMg(OH)4, CaMg(CO3)2] had lower blood Mg concentrations at 12 or 24 h after treatment than control and CaMg(CO3)2. Based on urinary Mg excretion 24 h after treatment, 4 of the Mg sources evaluated (including PMB) showed a similar availability, however, the availability of the commercial CaMg(CO3)2 source included in our study was similar to the negative control (no-supplemented cows).
Collapse
Affiliation(s)
- N Silva-Del-Rio
- Veterinary Medicine Teaching and Research Center, 18830 Road 112, Tulare, CA 93274; Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616.
| | | | - A Lago
- DairyExperts Inc., Tulare, CA 93274
| | - R R Lobo
- Veterinary Medicine Teaching and Research Center, 18830 Road 112, Tulare, CA 93274
| | - J P Goff
- College of Veterinary Medicine, Iowa State University, Ames, IA 50011
| | - W P Weiss
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691
| | - R J Tempelman
- Department of Animal Science, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
38
|
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
|
39
|
Lorenzetti WR, Ibelli AMG, Peixoto JDO, Savoldi IR, Mores MAZ, de Souza Romano G, do Carmo KB, Ledur MC. The downregulation of genes encoding muscle proteins have a potential role in the development of scrotal hernia in pigs. Mol Biol Rep 2024; 51:822. [PMID: 39023774 DOI: 10.1007/s11033-024-09766-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Testicular descent is a physiological process regulated by many factors. Eventually, disturbances in the embryological/fetal development path facilitate the occurrence of scrotal hernia, a congenital malformation characterized by the presence of intestinal portions within the scrotal sac due to the abnormal expansion of the inguinal ring. In pigs, some genes have been related to this anomaly, but the genetic mechanisms involved remain unclear. This study aimed to investigate the expression profile of a set of genes potentially involved with the manifestation of scrotal hernia in the inguinal ring tissue. METHODS AND RESULTS Tissue samples from the inguinal ring/canal of normal and scrotal hernia-affected male pigs with approximately 30 days of age were used. Relative expression analysis was performed using qPCR to confirm the expression profile of 17 candidate genes previously identified in an RNA-Seq study. Among them, the Myosin heavy chain 1 (MYH1), Desmin (DES), and Troponin 1 (TNNI1) genes were differentially expressed between groups and had reduced levels of expression in the affected animals. These genes encode proteins involved in the formation of muscle tissue, which seems to be important for increasing the resistance of the inguinal ring to the abdominal pressure, which is essential to avoid the occurrence of scrotal hernia. CONCLUSIONS The downregulation of muscular candidate genes in the inguinal tissue clarifies the genetic mechanisms involved with this anomaly in its primary site, providing useful information for developing strategies to control this malformation in pigs and other mammals.
Collapse
Affiliation(s)
- William Raphael Lorenzetti
- Programa de Pós-graduação em Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, Rua Beloni Trombeta Zanin 680E, Chapecó, Santa Catarina, 89815-630, Brazil
| | - Adriana Mércia Guaratini Ibelli
- Embrapa Suínos e Aves, Rodovia BR153, km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina, 89715-899, Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838, Guarapuava, Paraná, 85040-167, Brazil
- Embrapa Pecuária Sudeste, Rodovia Washington Luiz, Km 234, São Carlos, São Paulo, 13560-970, Brazil
| | - Jane de Oliveira Peixoto
- Embrapa Suínos e Aves, Rodovia BR153, km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina, 89715-899, Brazil
- Programa de Pós-Graduação em Ciências Veterinárias, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia, 838, Guarapuava, Paraná, 85040-167, Brazil
| | - Igor Ricardo Savoldi
- Programa de Pós-graduação em Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, Rua Beloni Trombeta Zanin 680E, Chapecó, Santa Catarina, 89815-630, Brazil
- Laudo laboratório Avícola, Rodovia BR-365, Morumbi, Uberlândia, Minas Gerais, 38407180, Brazil
| | - Marcos Antônio Zanella Mores
- Embrapa Suínos e Aves, Rodovia BR153, km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina, 89715-899, Brazil
| | | | - Kamilla Bleil do Carmo
- Universidade do Contestado, Concórdia, Santa Catarina, Brazil
- Instituto Federal Catarinense, Rodovia SC 283, km 17, Concórdia, Santa Catarina, 89703-720, Brazil
| | - Mônica Corrêa Ledur
- Programa de Pós-graduação em Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, Rua Beloni Trombeta Zanin 680E, Chapecó, Santa Catarina, 89815-630, Brazil.
- Embrapa Suínos e Aves, Rodovia BR153, km 110, Distrito de Tamanduá, Caixa Postal: 321, Concórdia, Santa Catarina, 89715-899, Brazil.
| |
Collapse
|
40
|
Gerzen OP, Potoskueva IK, Tzybina AE, Myachina TA, Nikitina LV. Cardiac Myosin and Thin Filament as Targets for Lead and Cadmium Divalent Cations. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1273-1282. [PMID: 39218024 DOI: 10.1134/s0006297924070095] [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: 05/18/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 09/04/2024]
Abstract
Lead and cadmium are heavy metals widely distributed in the environment and contribute significantly to cardiovascular morbidity and mortality. Using Leadmium Green dye, we have shown that lead and cadmium enter cardiomyocytes, distributing throughout the cell. Using an in vitro motility assay, we have shown that sliding velocity of actin and native thin filaments over myosin decreases with increasing concentrations of Pb2+ and Cd2+. Significantly lower concentrations of Pb2+ and Cd2+ (0.6 mM) were required to stop sliding of thin filaments over myosin compared to the stopping actin sliding over the same myosin (1.1-1.6 mM). Lower concentration of Cd2+ (1.1 mM) needed to stop actin sliding over myosin compared to the Pb2++Cd2+ combination (1.3 mM) and lead alone (1.6 mM). There were no differences found in the effects of lead and cadmium cations on relative force developed by myosin heads or number of actin filaments bound to myosin. Sliding velocity of actin over myosin in the left atrium, right and left ventricles changed equally when exposed to the same dose of the same metal. Thus, we have demonstrated for the first time that Pb2+ and Cd2+ can directly affect myosin and thin filament function, with Cd2+ exerting a more toxic influence on myosin function compared to Pb2+.
Collapse
Affiliation(s)
- Oksana P Gerzen
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, 620078, Russia.
| | - Iulia K Potoskueva
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, 620078, Russia
| | - Alena E Tzybina
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, 620078, Russia
| | - Tatiana A Myachina
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, 620078, Russia
| | - Larisa V Nikitina
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, 620078, Russia
| |
Collapse
|
41
|
Fu W, Liao Q, Shi Y, Liu W, Ren H, Xu C, Zeng C. Transient induction of actin cytoskeletal remodeling associated with dedifferentiation, proliferation, and redifferentiation stimulates cardiac regeneration. Acta Pharm Sin B 2024; 14:2537-2553. [PMID: 38828141 PMCID: PMC11143747 DOI: 10.1016/j.apsb.2024.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 06/05/2024] Open
Abstract
The formation of new and functional cardiomyocytes requires a 3-step process: dedifferentiation, proliferation, and redifferentiation, but the critical genes required for efficient dedifferentiation, proliferation, and redifferentiation remain unknown. In our study, a circular trajectory using single-nucleus RNA sequencing of the pericentriolar material 1 positive (PCM1+) cardiomyocyte nuclei from hearts 1 and 3 days after surgery-induced myocardial infarction (MI) on postnatal Day 1 was reconstructed and demonstrated that actin remodeling contributed to the dedifferentiation, proliferation, and redifferentiation of cardiomyocytes after injury. We identified four top actin-remodeling regulators, namely Tmsb4x, Tmsb10, Dmd, and Ctnna3, which we collectively referred to as 2D2P. Transiently expressed changes of 2D2P, using a polycistronic non-integrating lentivirus driven by Tnnt2 (cardiac-specific troponin T) promoters (Tnnt2-2D2P-NIL), efficiently induced transiently proliferative activation and actin remodeling in postnatal Day 7 cardiomyocytes and adult hearts. Furthermore, the intramyocardial delivery of Tnnt2-2D2P-NIL resulted in a sustained improvement in cardiac function without ventricular dilatation, thickened septum, or fatal arrhythmia for at least 4 months. In conclusion, this study highlights the importance of actin remodeling in cardiac regeneration and provides a foundation for new gene-cocktail-therapy approaches to improve cardiac repair and treat heart failure using a novel transient and cardiomyocyte-specific viral construct.
Collapse
Affiliation(s)
- Wenbin Fu
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Qiao Liao
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Yu Shi
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Wujian Liu
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Chunmei Xu
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, the Third Military Medical University (Army Medical University), Chongqing 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing 400042, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing 400042, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, the Third Military Medical University, Chongqing 400042, China
- Cardiovascular Research Center, Chongqing College, University of Chinese Academy of Sciences, Chongqing 400042, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing 400042, China
| |
Collapse
|
42
|
Lewis CTA, Melhedegaard EG, Ognjanovic MM, Olsen MS, Laitila J, Seaborne RAE, Gronset M, Zhang C, Iwamoto H, Hessel AL, Kuehn MN, Merino C, Amigo N, Frobert O, Giroud S, Staples JF, Goropashnaya AV, Fedorov VB, Barnes B, Toien O, Drew K, Sprenger RJ, Ochala J. Remodeling of skeletal muscle myosin metabolic states in hibernating mammals. eLife 2024; 13:RP94616. [PMID: 38752835 PMCID: PMC11098559 DOI: 10.7554/elife.94616] [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: 05/18/2024] Open
Abstract
Hibernation is a period of metabolic suppression utilized by many small and large mammal species to survive during winter periods. As the underlying cellular and molecular mechanisms remain incompletely understood, our study aimed to determine whether skeletal muscle myosin and its metabolic efficiency undergo alterations during hibernation to optimize energy utilization. We isolated muscle fibers from small hibernators, Ictidomys tridecemlineatus and Eliomys quercinus and larger hibernators, Ursus arctos and Ursus americanus. We then conducted loaded Mant-ATP chase experiments alongside X-ray diffraction to measure resting myosin dynamics and its ATP demand. In parallel, we performed multiple proteomics analyses. Our results showed a preservation of myosin structure in U. arctos and U. americanus during hibernation, whilst in I. tridecemlineatus and E. quercinus, changes in myosin metabolic states during torpor unexpectedly led to higher levels in energy expenditure of type II, fast-twitch muscle fibers at ambient lab temperatures (20 °C). Upon repeating loaded Mant-ATP chase experiments at 8 °C (near the body temperature of torpid animals), we found that myosin ATP consumption in type II muscle fibers was reduced by 77-107% during torpor compared to active periods. Additionally, we observed Myh2 hyper-phosphorylation during torpor in I. tridecemilineatus, which was predicted to stabilize the myosin molecule. This may act as a potential molecular mechanism mitigating myosin-associated increases in skeletal muscle energy expenditure during periods of torpor in response to cold exposure. Altogether, we demonstrate that resting myosin is altered in hibernating mammals, contributing to significant changes to the ATP consumption of skeletal muscle. Additionally, we observe that it is further altered in response to cold exposure and highlight myosin as a potentially contributor to skeletal muscle non-shivering thermogenesis.
Collapse
Affiliation(s)
| | | | - Marija M Ognjanovic
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| | - Mathilde S Olsen
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| | - Jenni Laitila
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| | - Robert AE Seaborne
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
- Centre for Human and Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King’s College LondonLondonUnited Kingdom
| | - Magnus Gronset
- Department of Cellular and Molecular Medicine, University of CopenhagenCopenhagenDenmark
| | - Changxin Zhang
- Department of Computational Medicine and Bioinformatics, University of MichiganAnn ArborUnited States
| | - Hiroyuki Iwamoto
- Spring-8, Japan Synchrotron Radiation Research InstituteHyogoJapan
| | - Anthony L Hessel
- Institute of Physiology II, University of MuensterMuensterGermany
- Accelerated Muscle Biotechnologies ConsultantsBostonUnited States
| | - Michel N Kuehn
- Institute of Physiology II, University of MuensterMuensterGermany
- Accelerated Muscle Biotechnologies ConsultantsBostonUnited States
| | | | | | - Ole Frobert
- Department of Clinical Medicine, Faculty of Health, Aarhus UniversityAarhusDenmark
- Faculty of Health, Department of Cardiology, Örebro UniversityÖrebroSweden
| | - Sylvain Giroud
- Energetics Lab, Department of Biology, Northern Michigan UniversityMarquetteUnited States
- Research Institute of Wildlife Ecology, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine ViennaViennaAustria
| | - James F Staples
- Department of Biology, University of Western OntarioLondonCanada
| | - Anna V Goropashnaya
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Vadim B Fedorov
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Brian Barnes
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Oivind Toien
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska FairbanksFairbanksUnited States
| | - Ryan J Sprenger
- Department of Zoology, University of British ColumbiaVancouverCanada
| | - Julien Ochala
- Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
43
|
Rynkiewicz MJ, Childers MC, Karpicheva OE, Regnier M, Geeves MA, Lehman W. Myosin's powerstroke transitions define atomic scale movement of cardiac thin filament tropomyosin. J Gen Physiol 2024; 156:e202413538. [PMID: 38607351 PMCID: PMC11010328 DOI: 10.1085/jgp.202413538] [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: 01/02/2024] [Revised: 02/27/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
Dynamic interactions between the myosin motor head on thick filaments and the actin molecular track on thin filaments drive the myosin-crossbridge cycle that powers muscle contraction. The process is initiated by Ca2+ and the opening of troponin-tropomyosin-blocked myosin-binding sites on actin. The ensuing recruitment of myosin heads and their transformation from pre-powerstroke to post-powerstroke conformation on actin produce the force required for contraction. Cryo-EM-based atomic models confirm that during this process, tropomyosin occupies three different average positions on actin. Tropomyosin pivoting on actin away from a TnI-imposed myosin-blocking position accounts for part of the Ca2+ activation observed. However, the structure of tropomyosin on thin filaments that follows pre-powerstroke myosin binding and its translocation during myosin's pre-powerstroke to post-powerstroke transition remains unresolved. Here, we approach this transition computationally in silico. We used the myosin helix-loop-helix motif as an anchor to dock models of pre-powerstroke cardiac myosin to the cleft between neighboring actin subunits along cardiac thin filaments. We then performed targeted molecular dynamics simulations of the transition between pre- and post-powerstroke conformations on actin in the presence of cardiac troponin-tropomyosin. These simulations show Arg 369 and Glu 370 on the tip of myosin Loop-4 encountering identically charged residues on tropomyosin. The charge repulsion between residues causes tropomyosin translocation across actin, thus accounting for the final regulatory step in the activation of the thin filament, and, in turn, facilitating myosin movement along the filament. We suggest that during muscle activity, myosin-induced tropomyosin movement is likely to result in unencumbered myosin head interactions on actin at low-energy cost.
Collapse
Affiliation(s)
- Michael J. Rynkiewicz
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | | | - Olga E. Karpicheva
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - William Lehman
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
44
|
Donkervoort S, van de Locht M, Ronchi D, Reunert J, McLean CA, Zaki M, Orbach R, de Winter JM, Conijn S, Hoomoedt D, Neto OLA, Magri F, Viaene AN, Foley AR, Gorokhova S, Bolduc V, Hu Y, Acquaye N, Napoli L, Park JH, Immadisetty K, Miles LB, Essawi M, McModie S, Ferreira LF, Zanotti S, Neuhaus SB, Medne L, ElBagoury N, Johnson KR, Zhang Y, Laing NG, Davis MR, Bryson-Richardson RJ, Hwee DT, Hartman JJ, Malik FI, Kekenes-Huskey PM, Comi GP, Sharaf-Eldin W, Marquardt T, Ravenscroft G, Bönnemann CG, Ottenheijm CAC. Pathogenic TNNI1 variants disrupt sarcomere contractility resulting in hypo- and hypercontractile muscle disease. Sci Transl Med 2024; 16:eadg2841. [PMID: 38569017 DOI: 10.1126/scitranslmed.adg2841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
Troponin I (TnI) regulates thin filament activation and muscle contraction. Two isoforms, TnI-fast (TNNI2) and TnI-slow (TNNI1), are predominantly expressed in fast- and slow-twitch myofibers, respectively. TNNI2 variants are a rare cause of arthrogryposis, whereas TNNI1 variants have not been conclusively established to cause skeletal myopathy. We identified recessive loss-of-function TNNI1 variants as well as dominant gain-of-function TNNI1 variants as a cause of muscle disease, each with distinct physiological consequences and disease mechanisms. We identified three families with biallelic TNNI1 variants (F1: p.R14H/c.190-9G>A, F2 and F3: homozygous p.R14C), resulting in loss of function, manifesting with early-onset progressive muscle weakness and rod formation on histology. We also identified two families with a dominantly acting heterozygous TNNI1 variant (F4: p.R174Q and F5: p.K176del), resulting in gain of function, manifesting with muscle cramping, myalgias, and rod formation in F5. In zebrafish, TnI proteins with either of the missense variants (p.R14H; p.R174Q) incorporated into thin filaments. Molecular dynamics simulations suggested that the loss-of-function p.R14H variant decouples TnI from TnC, which was supported by functional studies showing a reduced force response of sarcomeres to submaximal [Ca2+] in patient myofibers. This contractile deficit could be reversed by a slow skeletal muscle troponin activator. In contrast, patient myofibers with the gain-of-function p.R174Q variant showed an increased force to submaximal [Ca2+], which was reversed by the small-molecule drug mavacamten. Our findings demonstrated that TNNI1 variants can cause muscle disease with variant-specific pathomechanisms, manifesting as either a hypo- or a hypercontractile phenotype, suggesting rational therapeutic strategies for each mechanism.
Collapse
Affiliation(s)
- Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Martijn van de Locht
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Dario Ronchi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, 20135, Italy
| | - Janine Reunert
- Department of General Pediatrics, University of Münster, Münster, 48149, Germany
| | - Catriona A McLean
- Department of Anatomical Pathology, Alfred Hospital, Melbourne, Victoria, 3004, Australia
- Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, 3168, Australia
| | - Maha Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Rotem Orbach
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Josine M de Winter
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Stefan Conijn
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Daan Hoomoedt
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Osorio Lopes Abath Neto
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Francesca Magri
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, 20122, Italy
| | - Angela N Viaene
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104 PA, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Svetlana Gorokhova
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Medical Genetics, Timone Children's Hospital, APHM, Marseille, 13005, France
- INSERM, U1251-MMG, Aix-Marseille Université, Marseille, 13009, France
| | - Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Acquaye
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura Napoli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Julien H Park
- Department of General Pediatrics, University Hospital Münster, Münster, 48149 Germany
| | - Kalyan Immadisetty
- Department of Cell and Molecular Physiology, Loyola University, Chicago, IL 60153, USA
| | - Lee B Miles
- School of Biological Sciences, Monash University, Melbourne, Victoria, 3800, Australia
| | - Mona Essawi
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Salar McModie
- Department of Neurology, Alfred Health, Melbourne, Victoria, 3004, Australia
| | - Leonardo F Ferreira
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Simona Zanotti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Sarah B Neuhaus
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Livija Medne
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nagham ElBagoury
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Kory R Johnson
- Bioinformatics Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong Zhang
- Bioinformatics Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nigel G Laing
- Neurogenetics Unit, Department of Diagnostic Genomics, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
- Centre for Medical Research University of Western Australia, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Mark R Davis
- Neurogenetics Unit, Department of Diagnostic Genomics, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | | | - Darren T Hwee
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - James J Hartman
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Fady I Malik
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | | | - Giacomo Pietro Comi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, 20135, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Wessam Sharaf-Eldin
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Thorsten Marquardt
- Department of General Pediatrics, University of Münster, Münster, 48149, Germany
| | - Gianina Ravenscroft
- Centre for Medical Research University of Western Australia, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| |
Collapse
|
45
|
Ronca F, Raggi A. Role of the interaction between troponin T and AMP deaminase by zinc bridge in modulating muscle contraction and ammonia production. Mol Cell Biochem 2024; 479:793-809. [PMID: 37184757 PMCID: PMC11016001 DOI: 10.1007/s11010-023-04763-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/06/2023] [Indexed: 05/16/2023]
Abstract
The N-terminal region of troponin T (TnT) does not bind any protein of the contractile machinery and the role of its hypervariability remains uncertain. In this review we report the evidence of the interaction between TnT and AMP deaminase (AMPD), a regulated zinc enzyme localized on the myofibril. In periods of intense muscular activity, a decrease in the ATP/ADP ratio, together with a decrease in the tissue pH, is the stimulus for the activation of the enzyme that deaminating AMP to IMP and NH3 displaces the myokinase reaction towards the formation of ATP. In skeletal muscle subjected to strong tetanic contractions, a calpain-like proteolytic activity produces the removal in vivo of a 97-residue N-terminal fragment from the enzyme that becomes desensitized towards the inhibition by ATP, leading to an unrestrained production of NH3. When a 95-residue N-terminal fragment is removed from AMPD by trypsin, simulating in vitro the calpain action, rabbit fast TnT or its phosphorylated 50-residue N-terminal peptide binds AMPD restoring the inhibition by ATP. Taking in consideration that the N-terminus of TnT expressed in human as well as rabbit white muscle contains a zinc-binding motif, we suggest that TnT might mimic the regulatory action of the inhibitory N-terminal domain of AMPD due to the presence of a zinc ion connecting the N-terminal and C-terminal regions of the enzyme, indicating that the two proteins might physiologically associate to modulate muscle contraction and ammonia production in fast-twitching muscle under strenuous conditions.
Collapse
Affiliation(s)
- Francesca Ronca
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Antonio Raggi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| |
Collapse
|
46
|
Schuftan D, Kooh YKG, Guo J, Sun Y, Aryan L, Stottlemire B, Berkland C, Genin GM, Huebsch N. Dynamic control of contractile resistance to iPSC-derived micro-heart muscle arrays. J Biomed Mater Res A 2024; 112:534-548. [PMID: 37952251 PMCID: PMC10922390 DOI: 10.1002/jbm.a.37642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023]
Abstract
Many types of cardiovascular disease are linked to the mechanical forces placed on the heart. However, our understanding of how mechanical forces exactly affect the cellular biology of the heart remains incomplete. In vitro models based on cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CM) enable researchers to develop medium to high-throughput systems to study cardiac mechanobiology at the cellular level. Previous models have been developed to enable the study of mechanical forces, such as cardiac afterload. However, most of these models require exogenous extracellular matrix (ECM) to form cardiac tissues. Recently, a system was developed to simulate changes in afterload by grafting ECM-free micro-heart muscle arrays to elastomeric substrates of discrete stiffnesses. In the present study, we extended this system by combining the elastomer-grafted tissue arrays with a magnetorheological elastomeric substrate. This system allows iPSC-CM based micro-heart muscle arrays to experience dynamic changes in contractile resistance to mimic dynamically altered afterload. Acute changes in substrate stiffness led to acute changes in the calcium dynamics and contractile forces, illustrating the system's ability to dynamically elicit changes in tissue mechanics by dynamically changing contractile resistance.
Collapse
Affiliation(s)
- David Schuftan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yasaman Kargar Gaz Kooh
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jingxuan Guo
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yuwen Sun
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bryce Stottlemire
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
| | - Cory Berkland
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Guy M. Genin
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| |
Collapse
|
47
|
Zhong R, Rua MT, Wei-LaPierre L. Targeting mitochondrial Ca 2+ uptake for the treatment of amyotrophic lateral sclerosis. J Physiol 2024; 602:1519-1549. [PMID: 38010626 PMCID: PMC11032238 DOI: 10.1113/jp284143] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare adult-onset neurodegenerative disease characterized by progressive motor neuron (MN) loss, muscle denervation and paralysis. Over the past several decades, researchers have made tremendous efforts to understand the pathogenic mechanisms underpinning ALS, with much yet to be resolved. ALS is described as a non-cell autonomous condition with pathology detected in both MNs and non-neuronal cells, such as glial cells and skeletal muscle. Studies in ALS patient and animal models reveal ubiquitous abnormalities in mitochondrial structure and function, and disturbance of intracellular calcium homeostasis in various tissue types, suggesting a pivotal role of aberrant mitochondrial calcium uptake and dysfunctional calcium signalling cascades in ALS pathogenesis. Calcium signalling and mitochondrial dysfunction are intricately related to the manifestation of cell death contributing to MN loss and skeletal muscle dysfunction. In this review, we discuss the potential contribution of intracellular calcium signalling, particularly mitochondrial calcium uptake, in ALS pathogenesis. Functional consequences of excessive mitochondrial calcium uptake and possible therapeutic strategies targeting mitochondrial calcium uptake or the mitochondrial calcium uniporter, the main channel mediating mitochondrial calcium influx, are also discussed.
Collapse
Affiliation(s)
- Renjia Zhong
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611
- Department of Emergency Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China, 110001
| | - Michael T. Rua
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611
| | - Lan Wei-LaPierre
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, 32611
- Myology Institute, University of Florida, Gainesville, FL 32611
| |
Collapse
|
48
|
Kondacs L, Parijat P, Cobb AJA, Kampourakis T. Synthesis and Biophysical Characterization of Fingolimod Derivatives as Cardiac Troponin Antagonists. ACS Med Chem Lett 2024; 15:413-417. [PMID: 38505838 PMCID: PMC10945792 DOI: 10.1021/acsmedchemlett.3c00511] [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: 11/13/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/21/2024] Open
Abstract
Calcium binding to cardiac troponin C (cTnC) in the thin filaments acts as a trigger for cardiac muscle contraction. The N-lobe of cTnC (NcTnC) undergoes a conformational change in the presence of calcium that allows for interaction with the switch region of cardiac troponin I (cTnISP), releasing its inhibitory effect on the thin filament structure. The small molecule fingolimod inhibits cTnC-cTnISP interactions via electrostatic repulsion between its positively charged tail and positively charged residues in cTnISP and acts as a calcium desensitizer of the contractile myofilaments. Here we investigate the structure-activity relationship of the fingolimod hydrophobic headgroup and show that increasing the alkyl chain length increases both its affinity for NcTnC and its inhibitory effect on the NcTnC-cTnISP interaction and that decreasing flexibility completely abolishes these effects. Strikingly, the longer derivatives have no effect on the calcium affinity of cTnC, suggesting that they act as better inhibitors.
Collapse
Affiliation(s)
- Laszlo Kondacs
- Department
of Chemistry, King’s College London, Britannia House, London SE1 1DB, United Kingdom
| | - Priyanka Parijat
- Randall
Centre for Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s
College London, London SE1 1UL, United Kingdom
| | - Alexander J. A. Cobb
- Department
of Chemistry, King’s College London, Britannia House, London SE1 1DB, United Kingdom
| | - Thomas Kampourakis
- Randall
Centre for Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s
College London, London SE1 1UL, United Kingdom
| |
Collapse
|
49
|
Barry ME, Rynkiewicz MJ, Pavadai E, Viana A, Lehman W, Moore JR. Glutamate 139 of tropomyosin is critical for cardiac thin filament blocked-state stabilization. J Mol Cell Cardiol 2024; 188:30-37. [PMID: 38266978 PMCID: PMC11654406 DOI: 10.1016/j.yjmcc.2024.01.004] [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: 06/16/2023] [Revised: 12/14/2023] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
The cardiac thin filament proteins troponin and tropomyosin control actomyosin formation and thus cardiac contractility. Calcium binding to troponin changes tropomyosin position along the thin filament, allowing myosin head binding to actin required for heart muscle contraction. The thin filament regulatory proteins are hot spots for genetic mutations causing heart muscle dysfunction. While much of the thin filament structure has been characterized, critical regions of troponin and tropomyosin involved in triggering conformational changes remain unresolved. A poorly resolved region, helix-4 (H4) of troponin I, is thought to stabilize tropomyosin in a position on actin that blocks actomyosin interactions at low calcium concentrations during muscle relaxation. We have proposed that contact between glutamate 139 on tropomyosin and positively charged residues on H4 leads to blocking-state stabilization. In this study, we attempted to disrupt these interactions by replacing E139 with lysine (E139K) to define the importance of this residue in thin filament regulation. Comparison of mutant and wild-type tropomyosin was carried out using in-vitro motility assays, actin co-sedimentation, and molecular dynamics simulations to determine perturbations in troponin-tropomyosin function caused by the tropomyosin mutation. Motility assays revealed that mutant thin filaments moved at higher velocity at low calcium with increased calcium sensitivity demonstrating that tropomyosin residue 139 is vital for proper tropomyosin-mediated inhibition during relaxation. Similarly, molecular dynamic simulations revealed a mutation-induced decrease in interaction energy between tropomyosin-E139K and troponin I (R170 and K174). These results suggest that salt-bridge stabilization of tropomyosin position by troponin IH4 is essential to prevent actomyosin interactions during cardiac muscle relaxation.
Collapse
Affiliation(s)
- Meaghan E Barry
- Department of Biological Sciences, University of Massachusetts Lowell, One University Ave, Lowell, MA 01854, United States of America
| | - Michael J Rynkiewicz
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisan School of Medicine, 700 Albany Street, W-408E, Boston, MA 02118, United States of America
| | - Elumalai Pavadai
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisan School of Medicine, 700 Albany Street, W-408E, Boston, MA 02118, United States of America
| | - Alex Viana
- Department of Biological Sciences, University of Massachusetts Lowell, One University Ave, Lowell, MA 01854, United States of America
| | - William Lehman
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisan School of Medicine, 700 Albany Street, W-408E, Boston, MA 02118, United States of America
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts Lowell, One University Ave, Lowell, MA 01854, United States of America.
| |
Collapse
|
50
|
Lewis CTA, Melhedegaard EG, Ognjanovic MM, Olsen MS, Laitila J, Seaborne RAE, Gronset MN, Zhang C, Iwamoto H, Hessel AL, Kuehn MN, Merino C, Amigo N, Frobert O, Giroud S, Staples JF, Goropashnaya AV, Fedorov VB, Barnes BM, Toien O, Drew KL, Sprenger RJ, Ochala J. Remodelling of Skeletal Muscle Myosin Metabolic States in Hibernating Mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.14.566992. [PMID: 38014200 PMCID: PMC10680686 DOI: 10.1101/2023.11.14.566992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Hibernation is a period of metabolic suppression utilized by many small and large mammal species to survive during winter periods. As the underlying cellular and molecular mechanisms remain incompletely understood, our study aimed to determine whether skeletal muscle myosin and its metabolic efficiency undergo alterations during hibernation to optimize energy utilization. We isolated muscle fibers from small hibernators, Ictidomys tridecemlineatus and Eliomys quercinus and larger hibernators, Ursus arctos and Ursus americanus. We then conducted loaded Mant-ATP chase experiments alongside X-ray diffraction to measure resting myosin dynamics and its ATP demand. In parallel, we performed multiple proteomics analyses. Our results showed a preservation of myosin structure in U. arctos and U. americanus during hibernation, whilst in I. tridecemlineatus and E. quercinus, changes in myosin metabolic states during torpor unexpectedly led to higher levels in energy expenditure of type II, fast-twitch muscle fibers at ambient lab temperatures (20°C). Upon repeating loaded Mant-ATP chase experiments at 8°C (near the body temperature of torpid animals), we found that myosin ATP consumption in type II muscle fibers was reduced by 77-107% during torpor compared to active periods. Additionally, we observed Myh2 hyper-phosphorylation during torpor in I. tridecemilineatus, which was predicted to stabilize the myosin molecule. This may act as a potential molecular mechanism mitigating myosin-associated increases in skeletal muscle energy expenditure during periods of torpor in response to cold exposure. Altogether, we demonstrate that resting myosin is altered in hibernating mammals, contributing to significant changes to the ATP consumption of skeletal muscle. Additionally, we observe that it is further altered in response to cold exposure and highlight myosin as a potentially contributor to skeletal muscle non-shivering thermogenesis.
Collapse
|