1
|
Lynn ML, Jimenez J, Castillo RL, Vasquez C, Klass MM, Baldo A, Kim A, Gibson C, Murphy AM, Tardiff JC. Arg92Leu-cTnT Alters the cTnC-cTnI Interface Disrupting PKA-Mediated Relaxation. Circ Res 2024; 135:974-989. [PMID: 39328062 PMCID: PMC11502267 DOI: 10.1161/circresaha.124.325223] [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: 07/16/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Impaired left ventricular relaxation, high filling pressures, and dysregulation of Ca2+ homeostasis are common findings contributing to diastolic dysfunction in hypertrophic cardiomyopathy (HCM). Studies have shown that impaired relaxation is an early observation in the sarcomere-gene-positive preclinical HCM cohort, which suggests the potential involvement of myofilament regulators in relaxation. A molecular-level understanding of mechanism(s) at the level of the myofilament is lacking. We hypothesized that mutation-specific, allosterically mediated, changes to the cTnC (cardiac troponin C)-cTnI (cardiac troponin I) interface can account for the development of early-onset diastolic dysfunction via decreased PKA accessibility to cTnI. METHODS HCM mutations R92L-cTnT (cardiac troponin T; Arg92Leu) and Δ160E-cTnT (Glu160 deletion) were studied in vivo, in vitro, and in silico via 2-dimensional echocardiography, Western blotting, ex vivo hemodynamics, stopped-flow kinetics, time-resolved fluorescence resonance energy transfer, and molecular dynamics simulations. RESULTS The HCM-causative mutations R92L-cTnT and Δ160E-cTnT result in different time-of-onset diastolic dysfunction. R92L-cTnT demonstrated early-onset diastolic dysfunction accompanied by a localized decrease in phosphorylation of cTnI. Constitutive phosphorylation of cTnI (cTnI-D23D24) was sufficient to recover diastolic function to non-Tg levels only for R92L-cTnT. Mutation-specific changes in Ca2+ dissociation rates associated with R92L-cTnT reconstituted with cTnI-D23D24 led us to investigate potential involvement of structural changes in the cTnC-cTnI interface as an explanation for these observations. We probed the interface via time-resolved fluorescence resonance energy transfer revealing a repositioning of the N-terminus of cTnI, closer to cTnC, and concomitant decreases in distance distributions at sites flanking the PKA consensus sequence. Implementing time-resolved fluorescence resonance energy transfer distances as constraints into our atomistic model identified additional electrostatic interactions at the consensus sequence. CONCLUSIONS These data show that the early diastolic dysfunction observed in a subset of HCM is attributable to allosterically mediated structural changes at the cTnC-cTnI interface that impair accessibility of PKA, thereby blunting β-adrenergic responsiveness and identifying a potential molecular target for therapeutic intervention.
Collapse
Affiliation(s)
- Melissa L. Lynn
- Department of Biomedical Engineering, University of Arizona, Tucson AZ
| | - Jesus Jimenez
- Department of Medicine, Washington University at St. Louis, St. Louis, MO
| | - Romi L. Castillo
- Department of Biomedical Engineering, University of Arizona, Tucson AZ
| | - Catherine Vasquez
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Matthew M. Klass
- Department of Physiological Sciences, University of Arizona, Tucson, AZ
| | - Anthony Baldo
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ
| | - Andrew Kim
- Department of Physiology, University of Arizona, Tucson, AZ
| | - Cyonna Gibson
- Department of Biomedical Engineering, University of Arizona, Tucson AZ
| | - Anne M. Murphy
- Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jil C. Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson AZ
- Department of Medicine, Washington University at St. Louis, St. Louis, MO
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
- Department of Physiological Sciences, University of Arizona, Tucson, AZ
- Department of Physiology, University of Arizona, Tucson, AZ
| |
Collapse
|
2
|
Cubuk J, Greenberg L, Greenberg AE, Emenecker RJ, Stuchell-Brereton MD, Holehouse AS, Soranno A, Greenberg MJ. Structural dynamics of the intrinsically disordered linker region of cardiac troponin T. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596451. [PMID: 38853835 PMCID: PMC11160775 DOI: 10.1101/2024.05.30.596451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The cardiac troponin complex, composed of troponins I, T, and C, plays a central role in regulating the calcium-dependent interactions between myosin and the thin filament. Mutations in troponin can cause cardiomyopathies; however, it is still a major challenge to connect how changes in sequence affect troponin's function. Recent high-resolution structures of the thin filament revealed critical insights into the structure-function relationship of troponin, but there remain large, unresolved segments of troponin, including the troponin-T linker region that is a hotspot for cardiomyopathy mutations. This linker region is predicted to be intrinsically disordered, with behaviors that are not well described by traditional structural approaches; however, this proposal has not been experimentally verified. Here, we used a combination of single-molecule Förster resonance energy transfer (FRET), molecular dynamics simulations, and functional reconstitution assays to investigate the troponin-T linker region. We show that in the context of both isolated troponin and the fully regulated troponin complex, the linker behaves as a dynamic, intrinsically disordered region. This region undergoes polyampholyte expansion in the presence of high salt and distinct conformational changes during the assembly of the troponin complex. We also examine the ΔE160 hypertrophic cardiomyopathy mutation in the linker and demonstrate that it does not affect the conformational dynamics of the linker, rather it allosterically affects interactions with other troponin complex subunits, leading to increased molecular contractility. Taken together, our data clearly demonstrate the importance of disorder within the troponin-T linker and provide new insights into the molecular mechanisms driving the pathogenesis of cardiomyopathies.
Collapse
Affiliation(s)
- Jasmine Cubuk
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, 660 Euclid Ave, 63110, Saint Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St Louis, 1 Brookings Drive, 63130, Saint Louis, MO, USA
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, 660 Euclid Ave, 63110, Saint Louis, MO, USA
| | - Akiva E. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, 660 Euclid Ave, 63110, Saint Louis, MO, USA
| | - Ryan J. Emenecker
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, 660 Euclid Ave, 63110, Saint Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St Louis, 1 Brookings Drive, 63130, Saint Louis, MO, USA
| | - Melissa D. Stuchell-Brereton
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, 660 Euclid Ave, 63110, Saint Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St Louis, 1 Brookings Drive, 63130, Saint Louis, MO, USA
| | - Alex S. Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, 660 Euclid Ave, 63110, Saint Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St Louis, 1 Brookings Drive, 63130, Saint Louis, MO, USA
| | - Andrea Soranno
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, 660 Euclid Ave, 63110, Saint Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St Louis, 1 Brookings Drive, 63130, Saint Louis, MO, USA
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, 660 Euclid Ave, 63110, Saint Louis, MO, USA
| |
Collapse
|
3
|
Vaniya A, Karlstaedt A, Gulkok D, Thottakara T, Liu Y, Fan S, Eades H, Vakrou S, Fukunaga R, Vernon HJ, Fiehn O, Abraham MR. Allele-specific dysregulation of lipid and energy metabolism in early-stage hypertrophic cardiomyopathy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 8:100073. [PMID: 39430912 PMCID: PMC11485168 DOI: 10.1016/j.jmccpl.2024.100073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 10/22/2024]
Abstract
Introduction Hypertrophic cardiomyopathy (HCM) results from pathogenic variants in sarcomeric protein genes that increase myocyte energy demand and lead to cardiac hypertrophy. However, it is unknown whether a common metabolic trait underlies cardiac phenotype at the early disease stage. To address this question and define cardiac biochemical pathology in early-stage HCM, we studied two HCM mouse models that express pathogenic variants in cardiac troponin T (Tnt2) or myosin heavy chain (Myh6) genes, and have marked differences in cardiac imaging phenotype, mitochondrial function at early disease stage. Methods We used a combination of echocardiography, transcriptomics, mass spectrometry-based untargeted metabolomics (GC-TOF, HILIC, CSH-QTOF), and computational modeling (CardioNet) to examine cardiac structural and metabolic remodeling at early disease stage (5 weeks of age) in R92W-TnT+/- and R403Q-MyHC+/- mutant mice. Data from mutants was compared with respective littermate controls (WT). Results Allele-specific differences in cardiac phenotype, gene expression and metabolites were observed at early disease stage. LV diastolic dysfunction was prominent in TnT mutants. Differentially-expressed genes in TnT mutant hearts were predominantly enriched in the Krebs cycle, respiratory electron transport, and branched-chain amino acid metabolism, whereas MyHC mutants were enriched in mitochondrial biogenesis, calcium homeostasis, and liver-X-receptor signaling. Both mutant hearts demonstrated significant alterations in levels of purine nucleosides, trisaccharides, dicarboxylic acids, acylcarnitines, phosphatidylethanolamines, phosphatidylinositols, ceramides and triglycerides; 40.4 % of lipids and 24.7 % of metabolites were significantly different in TnT mutants, whereas 10.4 % of lipids and 5.8 % of metabolites were significantly different in MyHC mutants. Both mutant hearts had a lower abundance of unsaturated long-chain acyl-carnitines (18:1, 18:2, 20:1), but only TnT mutants showed enrichment of FA18:0 in ceramide and cardiolipin species. CardioNet predicted impaired energy substrate metabolism and greater phospholipid remodeling in TnT mutants than in MyHC mutants. Conclusions Our systems biology approach revealed marked differences in metabolic remodeling in R92W-TnT and R403Q-MyHC mutant hearts, with TnT mutants showing greater derangements than MyHC mutants, at early disease stage. Changes in cardiolipin composition in TnT mutants could contribute to impairment of energy metabolism and diastolic dysfunction observed in this study, and predispose to energetic stress, ventricular arrhythmias under high workloads such as exercise.
Collapse
Affiliation(s)
- Arpana Vaniya
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, United States of America
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Damla Gulkok
- Hypertrophic Cardiomyopathy Center of Excellence, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Tilo Thottakara
- Hypertrophic Cardiomyopathy Center of Excellence, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Yamin Liu
- Hypertrophic Cardiomyopathy Center of Excellence, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Sili Fan
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, United States of America
| | - Hannah Eades
- Hypertrophic Cardiomyopathy Center of Excellence, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Styliani Vakrou
- Hypertrophic Cardiomyopathy Center of Excellence, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins University, Baltimore, MD, United States of America
| | - Hilary J. Vernon
- McKusick Nathans Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States of America
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, United States of America
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, United States of America
| | - M. Roselle Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
4
|
Lynn ML, Jimenez J, Castillo RL, Klass MM, Vasquez C, Baldo A, Gibson C, Murphy AM, Tardiff JC. The HCM - Linked Mutation Arg92Leu in TNNT2 Allosterically Alters the cTnC - cTnI Interface and Disrupts the PKA-mediated Regulation of Myofilament Relaxation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.18.549569. [PMID: 37503299 PMCID: PMC10370115 DOI: 10.1101/2023.07.18.549569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background Impaired left ventricular relaxation, high filling pressures, and dysregulation of Ca 2+ homeostasis are common findings contributing to diastolic dysfunction in hypertrophic cardiomyopathy (HCM). Studies have shown that impaired relaxation is an early observation in the sarcomere-gene-positive preclinical HCM cohort which suggests potential involvement of myofilament regulators of relaxation. Yet, a molecular level understanding of mechanism(s) at the level of the myofilament is lacking. We hypothesized that mutation-specific, allosterically mediated, changes to the cardiac troponin C-cardiac troponin I (cTnC-cTnI) interface can account for the development of early-onset diastolic dysfunction via decreased PKA accessibility to cTnI. Methods HCM mutations R92L-cTnT (Arg92Leu) and Δ160E-cTnT (Glu160 deletion) were studied in vivo , in vitro, and in silico via 2D echocardiography, western blotting, ex vivo hemodynamics, stopped-flow kinetics, time resolved fluorescence resonance energy transfer (TR-FRET), and molecular dynamics simulations. Results The HCM-causative mutations R92L-cTnT and Δ160E-cTnT result in different time-of-onset of diastolic dysfunction. R92L-cTnT demonstrated early-onset diastolic dysfunction accompanied by a localized decrease in phosphorylation of cTnI. Constitutive phosphorylation of cTnI (cTnI-D 23 D 24 ) was sufficient to recover diastolic function to Non-Tg levels only for R92L-cTnT. Mutation-specific changes in Ca 2+ dissociation rates associated with R92L-cTnT reconstituted with cTnI-D 23 D 24 led us to investigate potential involvement of structural changes in the cTnC-cTnI interface as an explanation for these observations. We probed the interface via TR-FRET revealing a repositioning of the N-terminus of cTnI, closer to cTnC, and concomitant decreases in distance distributions at sites flanking the PKA consensus sequence. Implementing TR-FRET distances as constraints into our atomistic model identified additional electrostatic interactions at the consensus sequence. Conclusion These data indicate that the early diastolic dysfunction observed in a subset of HCM is likely attributable to structural changes at the cTnC-cTnI interface that impair accessibility of PKA thereby blunting β-adrenergic responsiveness and identifying a potential molecular target for therapeutic intervention.
Collapse
|
5
|
Pioner JM, Vitale G, Gentile F, Scellini B, Piroddi N, Cerbai E, Olivotto I, Tardiff J, Coppini R, Tesi C, Poggesi C, Ferrantini C. Genotype-Driven Pathogenesis of Atrial Fibrillation in Hypertrophic Cardiomyopathy: The Case of Different TNNT2 Mutations. Front Physiol 2022; 13:864547. [PMID: 35514357 PMCID: PMC9062294 DOI: 10.3389/fphys.2022.864547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/28/2022] [Indexed: 11/25/2022] Open
Abstract
Atrial dilation and atrial fibrillation (AF) are common in Hypertrophic CardioMyopathy (HCM) patients and associated with a worsening of prognosis. The pathogenesis of atrial myopathy in HCM remains poorly investigated and no specific association with genotype has been identified. By re-analysis of our cohort of thin-filament HCM patients (Coppini et al. 2014) AF was identified in 10% of patients with sporadic mutations in the cardiac Troponin T gene (TNNT2), while AF occurrence was much higher (25-75%) in patients carrying specific "hot-spot" TNNT2 mutations. To determine the molecular basis of arrhythmia occurrence, two HCM mouse models expressing human TNNT2 variants (a "hot-spot" one, R92Q, and a "sporadic" one, E163R) were selected according to the different pathophysiological pathways previously demonstrated in ventricular tissue. Echocardiography studies showed a significant left atrial dilation in both models, but more pronounced in the R92Q. In E163R atrial trabeculae, in line with what previously observed in ventricular preparations, the energy cost of tension generation was markedly increased. However, no changes of twitch amplitude and kinetics were observed, and there was no atrial arrhythmic propensity. R92Q atrial trabeculae, instead, displayed normal ATP consumption but markedly increased myofilament calcium sensitivity, as previously observed in ventricular preparations. This was associated with reduced inotropic reserve and slower kinetics of twitch contractions and, importantly, with an increased occurrence of spontaneous beats and triggered contractions that represent an intrinsic arrhythmogenic mechanism promoting AF. The association of specific TNNT2 mutations with AF occurrence depends on the mutation-driven pathomechanism (i.e., increased atrial myofilament calcium sensitivity rather than increased myofilament tension cost) and may influence the individual response to treatment.
Collapse
Affiliation(s)
| | - Giulia Vitale
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesca Gentile
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Jil Tardiff
- Department of Medicine and Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
6
|
Greenwell AA, Gopal K, Ussher JR. Myocardial Energy Metabolism in Non-ischemic Cardiomyopathy. Front Physiol 2020; 11:570421. [PMID: 33041869 PMCID: PMC7526697 DOI: 10.3389/fphys.2020.570421] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
As the most metabolically demanding organ in the body, the heart must generate massive amounts of energy adenosine triphosphate (ATP) from the oxidation of fatty acids, carbohydrates and other fuels (e.g., amino acids, ketone bodies), in order to sustain constant contractile function. While the healthy mature heart acts omnivorously and is highly flexible in its ability to utilize the numerous fuel sources delivered to it through its coronary circulation, the heart’s ability to produce ATP from these fuel sources becomes perturbed in numerous cardiovascular disorders. This includes ischemic heart disease and myocardial infarction, as well as in various cardiomyopathies that often precede the development of overt heart failure. We herein will provide an overview of myocardial energy metabolism in the healthy heart, while describing the numerous perturbations that take place in various non-ischemic cardiomyopathies such as hypertrophic cardiomyopathy, diabetic cardiomyopathy, arrhythmogenic cardiomyopathy, and the cardiomyopathy associated with the rare genetic disease, Barth Syndrome. Based on preclinical evidence where optimizing myocardial energy metabolism has been shown to attenuate cardiac dysfunction, we will discuss the feasibility of myocardial energetics optimization as an approach to treat the cardiac pathology associated with these various non-ischemic cardiomyopathies.
Collapse
Affiliation(s)
- Amanda A Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
7
|
Lavine KJ, Greenberg MJ. Beyond genomics-technological advances improving the molecular characterization and precision treatment of heart failure. Heart Fail Rev 2020; 26:405-415. [PMID: 32885327 DOI: 10.1007/s10741-020-10021-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 01/04/2023]
Abstract
Dilated cardiomyopathy (DCM) is a major cause of heart failure and cardiovascular mortality. In the past 20 years, there has been an overwhelming focus on developing therapeutics that target common downstream disease pathways thought to be involved in all forms of heart failure independent of the initial etiology. While this strategy is effective at the population level, individual responses vary tremendously and only approximately one third of patients receive benefit from modern heart failure treatments. In this perspective, we propose that DCM should be considered as a collection of diseases with a common phenotype of left ventricular dilation and systolic dysfunction rather than a single disease entity, and that mechanism-based classification of disease subtypes will revolutionize our understanding and clinical approach towards DCM. We discuss how these efforts are central to realizing the potential of precision medicine and how they are empowered by the development of new tools that allow investigators to strategically employ genomic and transcriptomic information. Finally, we outline an investigational strategy to (1) define DCM at the patient level, (2) develop new tools to model and mechanistically dissect subtypes of human heart failure, and (3) harness these insights for the development of precision therapeutics.
Collapse
Affiliation(s)
- Kory J Lavine
- Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8086, St. Louis, MO, 63110, USA.
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8231, St. Louis, MO, 63110, USA.
| |
Collapse
|
8
|
Wijnker PJM, van der Velden J. Mutation-specific pathology and treatment of hypertrophic cardiomyopathy in patients, mouse models and human engineered heart tissue. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165774. [PMID: 32217077 DOI: 10.1016/j.bbadis.2020.165774] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 01/04/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiomyopathy and is characterized by asymmetric left ventricular hypertrophy and diastolic dysfunction, and a frequent cause of sudden cardiac death at young age. Pharmacological treatment to prevent or reverse HCM is lacking. This may be partly explained by the variety of underlying disease causes. Over 1500 mutations have been associated with HCM, of which the majority reside in genes encoding sarcomere proteins, the cardiac contractile building blocks. Several mutation-mediated disease mechanisms have been identified, with proof for gene- and mutation-specific cellular perturbations. In line with mutation-specific changes in cellular pathology, the response to treatment may depend on the underlying sarcomere gene mutation. In this review, we will discuss evidence for mutation-specific pathology and treatment responses in HCM patients, mouse models and engineered heart tissue. The pros and cons of these experimental models for studying mutation-specific HCM pathology and therapies will be outlined.
Collapse
Affiliation(s)
- Paul J M Wijnker
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, Amsterdam, the Netherlands.
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, Amsterdam, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands.
| |
Collapse
|
9
|
Ezekian JE, Clippinger SR, Garcia JM, Yang Q, Denfield S, Jeewa A, Dreyer WJ, Zou W, Fan Y, Allen HD, Kim JJ, Greenberg MJ, Landstrom AP. Variant R94C in TNNT2-Encoded Troponin T Predisposes to Pediatric Restrictive Cardiomyopathy and Sudden Death Through Impaired Thin Filament Relaxation Resulting in Myocardial Diastolic Dysfunction. J Am Heart Assoc 2020; 9:e015111. [PMID: 32098556 PMCID: PMC7335540 DOI: 10.1161/jaha.119.015111] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Pediatric‐onset restrictive cardiomyopathy (RCM) is associated with high mortality, but underlying mechanisms of disease are under investigated. RCM‐associated diastolic dysfunction secondary to variants in TNNT2‐encoded cardiac troponin T (TNNT2) is poorly described. Methods and Results Genetic analysis of a proband and kindred with RCM identified TNNT2‐R94C, which cosegregated in a family with 2 generations of RCM, ventricular arrhythmias, and sudden death. TNNT2‐R94C was absent among large, population‐based cohorts Genome Aggregation Database (gnomAD) and predicted to be pathologic by in silico modeling. Biophysical experiments using recombinant human TNNT2‐R94C demonstrated impaired cardiac regulation at the molecular level attributed to reduced calcium‐dependent blocking of myosin's interaction with the thin filament. Computational modeling predicted a shift in the force‐calcium curve for the R94C mutant toward submaximal calcium activation compared within the wild type, suggesting low levels of muscle activation even at resting calcium concentrations and hypercontractility following activation by calcium. Conclusions The pathogenic TNNT2‐R94C variant activates thin‐filament–mediated sarcomeric contraction at submaximal calcium concentrations, likely resulting in increased muscle tension during diastole and hypercontractility during systole. This describes the proximal biophysical mechanism for development of RCM in this family.
Collapse
Affiliation(s)
- Jordan E Ezekian
- Division of Paediatric Cardiology Department of Pediatrics Duke University School of Medicine Durham NC
| | - Sarah R Clippinger
- Department of Biochemistry and Molecular Biophysics Washington University in St. Louis St. Louis MO
| | - Jaquelin M Garcia
- Department of Biochemistry and Molecular Biophysics Washington University in St. Louis St. Louis MO
| | - Qixin Yang
- Division of Paediatric Cardiology Department of Pediatrics Duke University School of Medicine Durham NC
| | - Susan Denfield
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Aamir Jeewa
- Department of Pediatrics The Hospital for Sick Children Toronto Ontario Canada
| | - William J Dreyer
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Wenxin Zou
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Yuxin Fan
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Hugh D Allen
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Jeffrey J Kim
- Department of Pediatrics The Lillie Frank Abercrombie Section of Pediatric Cardiology Baylor College of Medicine Houston TX
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics Washington University in St. Louis St. Louis MO
| | - Andrew P Landstrom
- Division of Paediatric Cardiology Department of Pediatrics Duke University School of Medicine Durham NC.,Department of Cell Biology Duke University School of Medicine Durham NC
| |
Collapse
|
10
|
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common cardiovascular disease with genetic transmission, characterized by the hypertrophy of any segment of the left ventricle (LV), not totally explained by improper loading conditions, with LV systolic function preserved, increased, or reduced. The histopathological mechanism involved in HCM refers to the primary injury of the myocardium, as follows: disorganized array of myocytes, extracellular matrix modification, microvascular dysfunction, with subsequent appearance of myocardial fibrosis. Multiple sarcomere proteins mutations are responsible for HCM, but two of them are involved in 70% of the cases of HCM: β-myosin heavy chain (MYH7) and myosin-binding protein C (MYBPC3). The development of new genetic techniques involving genome editing is promising to discover a gene therapy for patients with HCM. Clinical presentation may differ from asymptomatic to sudden cardiac death (SCD), the last one targeting younger adults. In this case, the diagnosis and evaluation of SCD risk factors is extremely important. The common method of diagnosis is transthoracic echocardiography, but cardiac magnetic resonance (CMR) imaging represents "gold standard" in the evaluation of HCM patients. Treatment includes pharmacological therapy, surgery, alcohol ablation, and not least SCD prevention.
Collapse
Affiliation(s)
- Ioana Danuta Muresan
- 2nd Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 2-4 Clinicilor, 400006, Cluj-Napoca, Romania
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 2-4 Clinicilor, 400006, Cluj-Napoca, Romania.
| |
Collapse
|
11
|
The relation between sarcomere energetics and the rate of isometric tension relaxation in healthy and diseased cardiac muscle. J Muscle Res Cell Motil 2019; 42:47-57. [PMID: 31745760 PMCID: PMC7932984 DOI: 10.1007/s10974-019-09566-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/13/2019] [Indexed: 12/24/2022]
Abstract
Full muscle relaxation happens when [Ca2+] falls below the threshold for force activation. Several experimental models, from whole muscle organs and intact muscle down to skinned fibers, have been used to explore the cascade of kinetic events leading to mechanical relaxation. The use of single myofibrils together with fast solution switching techniques, has provided new information about the role of cross-bridge (CB) dissociation in the time course of isometric force decay. Myofibril’s relaxation is biphasic starting with a slow seemingly linear phase, with a rate constant, slow kREL, followed by a fast mono-exponential phase. Sarcomeres remain isometric during the slow force decay that reflects CB detachment under isometric conditions while the final fast relaxation phase begins with a sudden give of few sarcomeres and is then dominated by intersarcomere dynamics. Based on a simple two-state model of the CB cycle, myofibril slow kREL represents the apparent forward rate with which CBs leave force generating states (gapp) under isometric conditions and correlates with the energy cost of tension generation (ATPase/tension ratio); in short slow kREL ~ gapp ~ tension cost. The validation of this relationship is obtained by simultaneously measuring maximal isometric force and ATP consumption in skinned myocardial strips that provide an unambiguous determination of the relation between contractile and energetic properties of the sarcomere. Thus, combining kinetic experiments in isolated myofibrils and mechanical and energetic measurements in multicellular cardiac strips, we are able to provide direct evidence for a positive linear correlation between myofibril isometric relaxation kinetics (slow kREL) and the energy cost of force production both measured in preparations from the same cardiac sample. This correlation remains true among different types of muscles with different ATPase activities and also when CB kinetics are altered by cardiomyopathy-related mutations. Sarcomeric mutations associated to hypertrophic cardiomyopathy (HCM), a primary cardiac disorder caused by mutations in genes encoding sarcomeric proteins, have been often found to accelerate CB turnover rate and increase the energy cost of myocardial contraction. Here we review data showing that faster CB detachment results in a proportional increase in the energetic cost of tension generation in heart samples from both HCM patients and mouse models of the disease.
Collapse
|
12
|
van der Velden J, Tocchetti CG, Varricchi G, Bianco A, Sequeira V, Hilfiker-Kleiner D, Hamdani N, Leite-Moreira AF, Mayr M, Falcão-Pires I, Thum T, Dawson DK, Balligand JL, Heymans S. Metabolic changes in hypertrophic cardiomyopathies: scientific update from the Working Group of Myocardial Function of the European Society of Cardiology. Cardiovasc Res 2019; 114:1273-1280. [PMID: 29912308 PMCID: PMC6054261 DOI: 10.1093/cvr/cvy147] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022] Open
Abstract
Disturbed metabolism as a consequence of obesity and diabetes may cause cardiac diseases (recently highlighted in the cardiovascular research spotlight issue on metabolic cardiomyopathies).1 In turn, the metabolism of the heart may also be disturbed in genetic and acquired forms of hypertrophic cardiac disease. Herein, we provide an overview of recent insights on metabolic changes in genetic hypertrophic cardiomyopathy and discuss several therapies, which may be explored to target disturbed metabolism and prevent onset of cardiac hypertrophy. This article is part of the Mini Review Series from the Varenna 2017 meeting of the Working Group of Myocardial Function of the European Society of Cardiology.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, NA, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Federico II University, Naples, NA, Italy
| | - Anna Bianco
- Department of Translational Medical Sciences, Federico II University, Naples, NA, Italy.,Department of Cardiology, Maastricht University Medical Center & CARIM, Maastricht University, Maastricht, The Netherlands
| | - Vasco Sequeira
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Denise Hilfiker-Kleiner
- Molecular Cardiology, Department of Cardiology and Angiology, Medical School Hannover, Germany
| | - Nazha Hamdani
- Department of Systems Physiology, Ruhr University Bochum, Bochum, Germany
| | - Adelino F Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research Centre, University of Porto, Porto, Portugal
| | - Manuel Mayr
- The James Black Centre & King's British Heart Foundation Centre, King's College, University of London, London, UK
| | - Ines Falcão-Pires
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research Centre, University of Porto, Porto, Portugal
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,National Heart and Lung Institute, Imperial College London, London, UK.,REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany
| | - Dana K Dawson
- School of Medicine & Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics, Institut de Recherche Experimentale et Clinique (IREC), and Clinique Universitaire Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Stephane Heymans
- Netherlands Heart Institute, Utrecht, The Netherlands.,Department of Cardiology, Maastricht University Medical Center & CARIM, Maastricht University, Maastricht, The Netherlands.,Department of Cardiovascular Sciences, Leuven University, Leuven, Belgium
| |
Collapse
|
13
|
Abdullah S, Lynn ML, McConnell MT, Klass MM, Baldo AP, Schwartz SD, Tardiff JC. FRET-based analysis of the cardiac troponin T linker region reveals the structural basis of the hypertrophic cardiomyopathy-causing Δ160E mutation. J Biol Chem 2019; 294:14634-14647. [PMID: 31387947 DOI: 10.1074/jbc.ra118.005098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 07/12/2019] [Indexed: 11/06/2022] Open
Abstract
Mutations in the cardiac thin filament (TF) have highly variable effects on the regulatory function of the cardiac sarcomere. Understanding the molecular-level dysfunction elicited by TF mutations is crucial to elucidate cardiac disease mechanisms. The hypertrophic cardiomyopathy-causing cardiac troponin T (cTnT) mutation Δ160Glu (Δ160E) is located in a putative "hinge" adjacent to an unstructured linker connecting domains TNT1 and TNT2. Currently, no high-resolution structure exists for this region, limiting significantly our ability to understand its role in myofilament activation and the molecular mechanism of mutation-induced dysfunction. Previous regulated in vitro motility data have indicated mutation-induced impairment of weak actomyosin interactions. We hypothesized that cTnT-Δ160E repositions the flexible linker, altering weak actomyosin electrostatic binding and acting as a biophysical trigger for impaired contractility and the observed remodeling. Using time-resolved FRET and an all-atom TF model, here we first defined the WT structure of the cTnT-linker region and then identified Δ160E mutation-induced positional changes. Our results suggest that the WT linker runs alongside the C terminus of tropomyosin. The Δ160E-induced structural changes moved the linker closer to the tropomyosin C terminus, an effect that was more pronounced in the presence of myosin subfragment (S1) heads, supporting previous findings. Our in silico model fully supported this result, indicating a mutation-induced decrease in linker flexibility. Our findings provide a framework for understanding basic pathogenic mechanisms that drive severe clinical hypertrophic cardiomyopathy phenotypes and for identifying structural targets for intervention that can be tested in silico and in vitro.
Collapse
Affiliation(s)
- Salwa Abdullah
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, 85721
| | - Melissa L Lynn
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, 85721
| | - Mark T McConnell
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, 85721
| | - Matthew M Klass
- Department of Physiological Sciences, University of Arizona, Tucson, Arizona, 85721
| | - Anthony P Baldo
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Steven D Schwartz
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85721
| | - Jil C Tardiff
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, 85721 .,Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, 85721.,Department of Physiological Sciences, University of Arizona, Tucson, Arizona, 85721.,Department of Medicine, University of Arizona, Tucson, Arizona 85721
| |
Collapse
|
14
|
Yotti R, Seidman CE, Seidman JG. Advances in the Genetic Basis and Pathogenesis of Sarcomere Cardiomyopathies. Annu Rev Genomics Hum Genet 2019; 20:129-153. [PMID: 30978303 DOI: 10.1146/annurev-genom-083118-015306] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are common heart muscle disorders that are caused by pathogenic variants in sarcomere protein genes. HCM is characterized by unexplained cardiac hypertrophy (increased chamber wall thickness) that is accompanied by enhanced cardiac contractility and impaired relaxation. DCM is defined as increased ventricular chamber volume with contractile impairment. In this review, we discuss recent analyses that provide new insights into the molecular mechanisms that cause these conditions. HCM studies have uncovered the critical importance of conformational changes that occur during relaxation and enable energy conservation, which are frequently disturbed by HCM mutations. DCM studies have demonstrated the considerable prevalence of truncating variants in titin and have discerned that these variants reduce contractile function by impairing sarcomerogenesis. These new pathophysiologic mechanisms open exciting opportunities to identify new pharmacological targets and develop future cardioprotective strategies.
Collapse
Affiliation(s)
- Raquel Yotti
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; .,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA; , .,Cardiovascular Division and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA; ,
| |
Collapse
|
15
|
Lehman SJ, Tal-Grinspan L, Lynn ML, Strom J, Benitez GE, Anderson ME, Tardiff JC. Chronic Calmodulin-Kinase II Activation Drives Disease Progression in Mutation-Specific Hypertrophic Cardiomyopathy. Circulation 2019; 139:1517-1529. [PMID: 30586744 PMCID: PMC6461395 DOI: 10.1161/circulationaha.118.034549] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/01/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Although the genetic causes of hypertrophic cardiomyopathy (HCM) are widely recognized, considerable lag in the development of targeted therapeutics has limited interventions to symptom palliation. This is in part attributable to an incomplete understanding of how point mutations trigger pathogenic remodeling. As a further complication, similar mutations within sarcomeric genes can result in differential disease severity, highlighting the need to understand the mechanism of progression at the molecular level. One pathway commonly linked to HCM progression is calcium homeostasis dysregulation, though how specific mutations disrupt calcium homeostasis remains unclear. METHODS To evaluate the effects of early intervention in calcium homeostasis, we used 2 mouse models of sarcomeric HCM (cardiac troponin T R92L and R92W) with differential myocellular calcium dysregulation and disease presentation. Two modes of intervention were tested: inhibition of the autoactivated calcium-dependent kinase (calmodulin kinase II [CaMKII]) via the AC3I peptide and diltiazem, an L-type calcium channel antagonist. Two-dimensional echocardiography was used to determine cardiac function and left ventricular remodeling, and atrial remodeling was monitored via atrial mass. Sarcoplasmic reticulum Ca2+ATPase activity was measured as an index of myocellular calcium handling and coupled to its regulation via the phosphorylation status of phospholamban. RESULTS We measured an increase in phosphorylation of CaMKII in R92W animals by 6 months of age, indicating increased autonomous activity of the kinase in these animals. Inhibition of CaMKII led to recovery of diastolic function and partially blunted atrial remodeling in R92W mice. This improved function was coupled to increased sarcoplasmic reticulum Ca2+ATPase activity in the R92W animals despite reduction of CaMKII activation, likely indicating improvement in myocellular calcium handling. In contrast, inhibition of CaMKII in R92L animals led to worsened myocellular calcium handling, remodeling, and function. Diltiazem-HCl arrested diastolic dysfunction progression in R92W animals only, with no improvement in cardiac remodeling in either genotype. CONCLUSIONS We propose a highly specific, mutation-dependent role of activated CaMKII in HCM progression and a precise therapeutic target for clinical management of HCM in selected cohorts. Moreover, the mutation-specific response elicited with diltiazem highlights the necessity to understand mutation-dependent progression at a molecular level to precisely intervene in disease progression.
Collapse
Affiliation(s)
- Sarah J. Lehman
- Department of Physiological Sciences, University of Arizona, Tucson, Arizona 85724, USA
| | - Lauren Tal-Grinspan
- Department of Medicine, Columbia University Medical Center, New York, New York 10032, USA
| | - Melissa L. Lynn
- Department of Medicine, University of Arizona, Tucson, Arizona, 85724, USA
| | - Joshua Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona 85724, USA
| | - Grace E. Benitez
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, 85724, USA
| | - Mark E. Anderson
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Jil C. Tardiff
- Department of Medicine, University of Arizona, Tucson, Arizona, 85724, USA
| |
Collapse
|
16
|
van Driel BO, van Rossum AC, Michels M, Huurman R, van der Velden J. Extra energy for hearts with a genetic defect: ENERGY trial. Neth Heart J 2019; 27:200-205. [PMID: 30767167 PMCID: PMC6439046 DOI: 10.1007/s12471-019-1239-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Aims Previous studies have shown that hypertrophic cardiomyopathy mutation carriers have a decreased myocardial energy efficiency, which is thought to play a key role in the pathomechanism of hypertrophic cardiomyopathy (HCM). The ENERGY trial aims to determine whether metabolic drugs correct decreased myocardial energy efficiency in HCM mutation carriers at an early disease stage. Methods 40 genotype-positive, phenotype-negative MYH7 mutation carriers will be treated for two months with trimetazidine or placebo in a double-blind randomised study design. Directly before and after treatment, study subjects will undergo an [11C]-acetate positron emission tomography/computed tomography (PET/CT) and cardiac magnetic resonance (CMR) scan to measure myocardial energy efficiency. Myocardial efficiency will be calculated as the amount of oxygen the heart consumes to perform work. Conclusion The ENERGY trial will be the first proof of concept study to determine whether metabolic drugs are a potential preventive therapy for HCM. Given that trimetazidine is already being used in clinical practice, there is large potential to swiftly implement this drug in HCM therapy.
Collapse
Affiliation(s)
- B O van Driel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands.
| | - A C van Rossum
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - M Michels
- Department of Cardiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - R Huurman
- Department of Cardiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
17
|
van der Velden J, Stienen GJM. Cardiac Disorders and Pathophysiology of Sarcomeric Proteins. Physiol Rev 2019; 99:381-426. [PMID: 30379622 DOI: 10.1152/physrev.00040.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The sarcomeric proteins represent the structural building blocks of heart muscle, which are essential for contraction and relaxation. During recent years, it has become evident that posttranslational modifications of sarcomeric proteins, in particular phosphorylation, tune cardiac pump function at rest and during exercise. This delicate, orchestrated interaction is also influenced by mutations, predominantly in sarcomeric proteins, which cause hypertrophic or dilated cardiomyopathy. In this review, we follow a bottom-up approach starting from a description of the basic components of cardiac muscle at the molecular level up to the various forms of cardiac disorders at the organ level. An overview is given of sarcomere changes in acquired and inherited forms of cardiac disease and the underlying disease mechanisms with particular reference to human tissue. A distinction will be made between the primary defect and maladaptive/adaptive secondary changes. Techniques used to unravel functional consequences of disease-induced protein changes are described, and an overview of current and future treatments targeted at sarcomeric proteins is given. The current evidence presented suggests that sarcomeres not only form the basis of cardiac muscle function but also represent a therapeutic target to combat cardiac disease.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Ger J M Stienen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| |
Collapse
|
18
|
Piroddi N, Witjas-Paalberends ER, Ferrara C, Ferrantini C, Vitale G, Scellini B, Wijnker PJM, Sequiera V, Dooijes D, Dos Remedios C, Schlossarek S, Leung MC, Messer A, Ward DG, Biggeri A, Tesi C, Carrier L, Redwood CS, Marston SB, van der Velden J, Poggesi C. The homozygous K280N troponin T mutation alters cross-bridge kinetics and energetics in human HCM. J Gen Physiol 2018; 151:18-29. [PMID: 30578328 PMCID: PMC6314385 DOI: 10.1085/jgp.201812160] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/29/2018] [Indexed: 01/24/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomeric proteins, but the pathogenic mechanism is unclear. Piroddi et al. find impairment of cross-bridge kinetics and energetics in human sarcomeres with a TNNT2 mutation, suggesting that HCM involves inefficient ATP utilization. Hypertrophic cardiomyopathy (HCM) is a genetic form of left ventricular hypertrophy, primarily caused by mutations in sarcomere proteins. The cardiac remodeling that occurs as the disease develops can mask the pathogenic impact of the mutation. Here, to discriminate between mutation-induced and disease-related changes in myofilament function, we investigate the pathogenic mechanisms underlying HCM in a patient carrying a homozygous mutation (K280N) in the cardiac troponin T gene (TNNT2), which results in 100% mutant cardiac troponin T. We examine sarcomere mechanics and energetics in K280N-isolated myofibrils and demembranated muscle strips, before and after replacement of the endogenous troponin. We also compare these data to those of control preparations from donor hearts, aortic stenosis patients (LVHao), and HCM patients negative for sarcomeric protein mutations (HCMsmn). The rate constant of tension generation following maximal Ca2+ activation (kACT) and the rate constant of isometric relaxation (slow kREL) are markedly faster in K280N myofibrils than in all control groups. Simultaneous measurements of maximal isometric ATPase activity and Ca2+-activated tension in demembranated muscle strips also demonstrate that the energy cost of tension generation is higher in the K280N than in all controls. Replacement of mutant protein by exchange with wild-type troponin in the K280N preparations reduces kACT, slow kREL, and tension cost close to control values. In donor myofibrils and HCMsmn demembranated strips, replacement of endogenous troponin with troponin containing the K280N mutant increases kACT, slow kREL, and tension cost. The K280N TNNT2 mutation directly alters the apparent cross-bridge kinetics and impairs sarcomere energetics. This result supports the hypothesis that inefficient ATP utilization by myofilaments plays a central role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Nicoletta Piroddi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - E Rosalie Witjas-Paalberends
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Claudia Ferrara
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Cecilia Ferrantini
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy.,LENS, Sesto Fiorentino (Firenze), Florence, Italy
| | - Giulia Vitale
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Beatrice Scellini
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Paul J M Wijnker
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Vasco Sequiera
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Dennis Dooijes
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Clinical Genetics, University Medical Center, Utrecht, Netherlands
| | - Cristobal Dos Remedios
- Department of Anatomy and Histology, Bosch Institute, The University of Sydney, Sydney, Australia
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Man Ching Leung
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Andrew Messer
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Douglas G Ward
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | - Chiara Tesi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | | | - Steven B Marston
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Clinical Genetics, University Medical Center, Utrecht, Netherlands.,ICIN-Netherlands, Heart Institute, Utrecht, Netherlands
| | - Corrado Poggesi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy .,LENS, Sesto Fiorentino (Firenze), Florence, Italy
| |
Collapse
|
19
|
Eschenhagen T, Carrier L. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes-a systematic review. Pflugers Arch 2018; 471:755-768. [PMID: 30324321 PMCID: PMC6475632 DOI: 10.1007/s00424-018-2214-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Human-induced pluripotent stem cells (hiPSC) can be differentiated to cardiomyocytes at high efficiency and are increasingly used to study cardiac disease in a human context. This review evaluated 38 studies on hypertrophic (HCM) and dilated cardiomyopathy (DCM) of different genetic causes asking to which extent published data allow the definition of an in vitro HCM/DCM hiPSC-CM phenotype. The data are put in context with the prevailing hypotheses on HCM/DCM dysfunction and pathophysiology. Relatively consistent findings in HCM not reported in DCM were larger cell size (156 ± 85%, n = 15), more nuclear localization of nuclear factor of activated T cells (NFAT; 175 ± 65%, n = 3), and higher β-myosin heavy chain gene expression levels (500 ± 547%, n = 8) than respective controls. Conversely, DCM lines showed consistently less force development than controls (47 ± 23%, n = 9), while HCM forces scattered without clear trend. Both HCM and DCM lines often showed sarcomere disorganization, higher NPPA/NPPB expression levels, and arrhythmic beating behaviour. The data have to be taken with the caveat that reporting frequencies of the various parameters (e.g. cell size, NFAT expression) differ widely between HCM and DCM lines, in which data scatter is large and that only 9/38 studies used isogenic controls. Taken together, the current data provide interesting suggestions for disease-specific phenotypes in HCM/DCM hiPSC-CM but indicate that the field is still in its early days. Systematic, quantitative comparisons and robust, high content assays are warranted to advance the field.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| |
Collapse
|
20
|
Regnier M. Mechanistic complexity of contractile dysfunction in hypertrophic cardiomyopathy. J Gen Physiol 2018; 150:1051-1053. [PMID: 30037852 PMCID: PMC6080894 DOI: 10.1085/jgp.201812091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Reflections on recent work providing mechanistic insight into the pathological effects of a cardiac troponin T mutation.
Collapse
|
21
|
Reda SM, Chandra M. Cardiomyopathy mutation (F88L) in troponin T abolishes length dependency of myofilament Ca 2+ sensitivity. J Gen Physiol 2018; 150:809-819. [PMID: 29776992 PMCID: PMC5987878 DOI: 10.1085/jgp.201711974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/09/2018] [Accepted: 04/24/2018] [Indexed: 01/14/2023] Open
Abstract
The F88L mutation in cardiac troponin T (TnTF88L) is associated with hypertrophic cardiomyopathy. Reda and Chandra reveal that it abolishes length-mediated increase in myofilament Ca2+ sensitivity and attenuates cooperative mechanisms governing length-dependent activation. Recent clinical studies have revealed a new hypertrophic cardiomyopathy–associated mutation (F87L) in the central region of human cardiac troponin T (TnT). However, despite its implication in several incidences of sudden cardiac death in young and old adults, whether F87L is associated with cardiac contractile dysfunction is unknown. Because the central region of TnT is important for modulating the muscle length–mediated recruitment of new force-bearing cross-bridges (XBs), we hypothesize that the F87L mutation causes molecular changes that are linked to the length-dependent activation of cardiac myofilaments. Length-dependent activation is important because it contributes significantly to the Frank–Starling mechanism, which enables the heart to vary stroke volume as a function of changes in venous return. We measured steady-state and dynamic contractile parameters in detergent-skinned guinea pig cardiac muscle fibers reconstituted with recombinant guinea pig wild-type TnT (TnTWT) or the guinea pig analogue (TnTF88L) of the human mutation at two different sarcomere lengths (SLs): short (1.9 µm) and long (2.3 µm). TnTF88L increases pCa50 (−log [Ca2+]free required for half-maximal activation) to a greater extent at short SL than at long SL; for example, pCa50 increases by 0.25 pCa units at short SL and 0.17 pCa units at long SL. The greater increase in pCa50 at short SL leads to the abolishment of the SL-dependent increase in myofilament Ca2+ sensitivity (ΔpCa50) in TnTF88L fibers, ΔpCa50 being 0.10 units in TnTWT fibers but only 0.02 units in TnTF88L fibers. Furthermore, at short SL, TnTF88L attenuates the negative impact of strained XBs on force-bearing XBs and augments the magnitude of muscle length–mediated recruitment of new force-bearing XBs. Our findings suggest that the TnTF88L-mediated effects on cardiac thin filaments may lead to a negative impact on the Frank–Starling mechanism.
Collapse
Affiliation(s)
- Sherif M Reda
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA
| | - Murali Chandra
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA
| |
Collapse
|
22
|
Abstract
This article focuses on three "bins" that comprise sets of biophysical derangements elicited by cardiomyopathy-associated mutations in the myofilament. Current therapies focus on symptom palliation and do not address the disease at its core. We and others have proposed that a more nuanced classification could lead to direct interventions based on early dysregulation changing the trajectory of disease progression in the preclinical cohort. Continued research is necessary to address the complexity of cardiomyopathic progression and develop efficacious therapeutics.
Collapse
Affiliation(s)
- Melissa L Lynn
- Department of Medicine, University of Arizona, Room 317, 1656 East Mabel Street, Tucson, AZ 85724, USA
| | - Sarah J Lehman
- Department of Physiological Sciences, University of Arizona, Room 317, 1656 East Mabel Street, Tucson, AZ 85724, USA
| | - Jil C Tardiff
- Department of Medicine, University of Arizona, Room 312, 1656 East Mabel Street, Tucson, AZ 85724, USA.
| |
Collapse
|
23
|
Vakrou S, Fukunaga R, Foster DB, Sorensen L, Liu Y, Guan Y, Woldemichael K, Pineda-Reyes R, Liu T, Tardiff JC, Leinwand LA, Tocchetti CG, Abraham TP, O'Rourke B, Aon MA, Abraham MR. Allele-specific differences in transcriptome, miRNome, and mitochondrial function in two hypertrophic cardiomyopathy mouse models. JCI Insight 2018; 3:94493. [PMID: 29563334 DOI: 10.1172/jci.insight.94493] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 02/14/2018] [Indexed: 01/06/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) stems from mutations in sarcomeric proteins that elicit distinct biophysical sequelae, which in turn may yield radically different intracellular signaling and molecular pathologic profiles. These signaling events remain largely unaddressed by clinical trials that have selected patients based on clinical HCM diagnosis, irrespective of genotype. In this study, we determined how two mouse models of HCM differ, with respect to cellular/mitochondrial function and molecular biosignatures, at an early stage of disease. We show that hearts from young R92W-TnT and R403Q-αMyHC mutation-bearing mice differ in their transcriptome, miRNome, intracellular redox environment, mitochondrial antioxidant defense mechanisms, and susceptibility to mitochondrial permeability transition pore opening. Pathway analysis of mRNA-sequencing data and microRNA profiles indicate that R92W-TnT mutants exhibit a biosignature consistent with activation of profibrotic TGF-β signaling. Our results suggest that the oxidative environment and mitochondrial impairment in young R92W-TnT mice promote activation of TGF-β signaling that foreshadows a pernicious phenotype in young individuals. Of the two mutations, R92W-TnT is more likely to benefit from anti-TGF-β signaling effects conferred by angiotensin receptor blockers and may be responsive to mitochondrial antioxidant strategies in the early stage of disease. Molecular and functional profiling may therefore serve as aids to guide precision therapy for HCM.
Collapse
Affiliation(s)
- Styliani Vakrou
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - D Brian Foster
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lars Sorensen
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yamin Liu
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| | - Yufan Guan
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kirubel Woldemichael
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Roberto Pineda-Reyes
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ting Liu
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jill C Tardiff
- Department of Internal Medicine and Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular, and Developmental Biology and the BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Carlo G Tocchetti
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Theodore P Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Miguel A Aon
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - M Roselle Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| |
Collapse
|
24
|
Mickelson AV, Chandra M. Hypertrophic cardiomyopathy mutation in cardiac troponin T (R95H) attenuates length-dependent activation in guinea pig cardiac muscle fibers. Am J Physiol Heart Circ Physiol 2017; 313:H1180-H1189. [PMID: 28842439 DOI: 10.1152/ajpheart.00369.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/07/2017] [Accepted: 08/22/2017] [Indexed: 01/14/2023]
Abstract
The central region of cardiac troponin T (TnT) is important for modulating the dynamics of muscle length-mediated cross-bridge recruitment. Therefore, hypertrophic cardiomyopathy mutations in the central region may affect cross-bridge recruitment dynamics to alter myofilament Ca2+ sensitivity and length-dependent activation of cardiac myofilaments. Given the importance of the central region of TnT for cardiac contractile dynamics, we studied if hypertrophic cardiomyopathy-linked mutation (TnTR94H)-induced effects on contractile function would be differently modulated by sarcomere length (SL). Recombinant wild-type TnT (TnTWT) and the guinea pig analog of the human R94H mutation (TnTR95H) were reconstituted into detergent-skinned cardiac muscle fibers from guinea pigs. Steady-state and dynamic contractile measurements were made at short and long SLs (1.9 and 2.3 µm, respectively). Our results demonstrated that TnTR95H increased pCa50 (-log of free Ca2+ concentration) to a greater extent at short SL; TnTR95H increased pCa50 by 0.11 pCa units at short SL and 0.07 pCa units at long SL. The increase in pCa50 associated with an increase in SL from 1.9 to 2.3 µm (ΔpCa50) was attenuated nearly twofold in TnTR95H fibers; ΔpCa50 was 0.09 pCa units for TnTWT fibers but only 0.05 pCa units for TnTR95H fibers. The SL dependency of rate constants of cross-bridge distortion dynamics and tension redevelopment was also blunted by TnTR95H Collectively, our observations on the SL dependency of pCa50 and rate constants of cross-bridge distortion dynamics and tension redevelopment suggest that mechanisms underlying the length-dependent activation cardiac myofilaments are attenuated by TnTR95HNEW & NOTEWORTHY Mutant cardiac troponin T (TnTR95H) differently affects myofilament Ca2+ sensitivity at short and long sarcomere length, indicating that mechanisms underlying length-dependent activation are altered by TnTR95H TnTR95H enhances myofilament Ca2+ sensitivity to a greater extent at short sarcomere length, thus attenuating the length-dependent increase in myofilament Ca2+ sensitivity.
Collapse
Affiliation(s)
- Alexis V Mickelson
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Murali Chandra
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
25
|
Güçlü A, Knaapen P, Harms HJ, Parbhudayal RY, Michels M, Lammertsma AA, van Rossum AC, Germans T, van der Velden J. Disease Stage-Dependent Changes in Cardiac Contractile Performance and Oxygen Utilization Underlie Reduced Myocardial Efficiency in Human Inherited Hypertrophic Cardiomyopathy. Circ Cardiovasc Imaging 2017; 10:CIRCIMAGING.116.005604. [PMID: 28476777 DOI: 10.1161/circimaging.116.005604] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/03/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Reduced myocardial efficiency represents a target for therapy in hypertrophic cardiomyopathy although therapeutic benefit may depend on disease stage. Here, we determined disease stage-dependent changes in myocardial efficiency and effects of myectomy surgery. METHODS AND RESULTS Myocardial external efficiency (MEE) was determined in 27 asymptomatic mutation carriers (genotype positive/phenotype negative), 10 patients with hypertrophic obstructive cardiomyopathy (HOCM), 10 patients with aortic valve stenosis, and 14 healthy individuals using [11C]-acetate positron emission tomography and cardiovascular magnetic resonance imaging. Follow-up measurements were performed in HOCM and aortic valve stenosis patients 4 months after surgery. External work did not differ in HOCM compared with controls, whereas myocardial oxygen consumption was lower in HOCM. Because of a higher cardiac mass, total cardiac oxygen consumption was significantly higher in HOCM than in controls and genotype positive/phenotype negative. MEE was significantly lower in genotype positive/phenotype negative than in controls (28±6% versus 42±6%) and was further decreased in HOCM (22±5%). In contrast to patients with aortic valve stenosis, MEE was not improved in patients with HOCM after surgery, which was explained by opposite changes in the septum (decrease) and lateral (increase) wall. CONCLUSIONS Different mechanisms underlie reduced MEE at the early and advanced stage of hypertrophic cardiomyopathy. The initial increase and subsequent reduction in myocardial oxygen consumption during disease progression indicates that energy deficiency is a primary mutation-related event, whereas mechanisms secondary to disease remodeling underlie low MEE in HOCM. Our data highlight that the benefit of therapies to improve energetic status of the heart may vary depending on the disease stage and that treatment should be initiated before cardiac remodeling.
Collapse
Affiliation(s)
- Ahmet Güçlü
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.).
| | - Paul Knaapen
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.)
| | - Hendrik J Harms
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.)
| | - Rahana Y Parbhudayal
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.)
| | - Michelle Michels
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.)
| | - Adriaan A Lammertsma
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.)
| | - Albert C van Rossum
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.)
| | - Tjeerd Germans
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.)
| | - Jolanda van der Velden
- From the Department of Cardiology (A.G., P.K., R.Y.P., A.C.v.R., T.G.), Department of Physiology (A.G., R.Y.P., J.v.d.V.), Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU) (H.J.H., A.A.L.), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiology, Erasmus Medical Center Rotterdam, The Netherlands (M.M.); Department of Cardiology, Medical Center Alkmaar, The Netherlands (T.G.); and ICIN-The Netherlands Heart Institute, Utrecht (A.G., J.v.d.V.)
| |
Collapse
|
26
|
Mickelson AV, Gollapudi SK, Chandra M. Cardiomyopathy-related mutation (A30V) in mouse cardiac troponin T divergently alters the magnitude of stretch activation in α- and β-myosin heavy chain fibers. Am J Physiol Heart Circ Physiol 2017; 312:H141-H149. [PMID: 27769999 PMCID: PMC5283911 DOI: 10.1152/ajpheart.00487.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/29/2016] [Accepted: 10/15/2016] [Indexed: 01/19/2023]
Abstract
The present study investigated the functional consequences of the human hypertrophic cardiomyopathy (HCM) mutation A28V in cardiac troponin T (TnT). The A28V mutation is located within the NH2 terminus of TnT, a region known to be important for full activation of cardiac thin filaments. The functional consequences of the A28V mutation in TnT remain unknown. Given how α- and β-myosin heavy chain (MHC) isoforms differently alter the functional effect of the NH2 terminus of TnT, we hypothesized that the A28V-induced effects would be differently modulated by α- and β-MHC isoforms. Recombinant wild-type mouse TnT (TnTWT) and the mouse equivalent of the human A28V mutation (TnTA30V) were reconstituted into detergent-skinned cardiac muscle fibers extracted from normal (α-MHC) and transgenic (β-MHC) mice. Dynamic and steady-state contractile parameters were measured in reconstituted muscle fibers. Step-like length perturbation experiments demonstrated that TnTA30V decreased the magnitude of the muscle length-mediated recruitment of new force-bearing cross bridges (ER) by 30% in α-MHC fibers. In sharp contrast, TnTA30V increased ER by 55% in β-MHC fibers. Inferences drawn from other dynamic contractile parameters suggest that directional changes in ER in TnTA30V + α-MHC and TnTA30V + β-MHC fibers result from a divergent impact on cross bridge-regulatory unit (troponin-tropomyosin complex) cooperativity. TnTA30V-mediated effects on Ca2+-activated maximal tension and instantaneous muscle fiber stiffness (ED) were also divergently affected by α- and β-MHC. Our study demonstrates that TnTA30V + α-MHC and TnTA30V + β-MHC fibers show contrasting contractile phenotypes; however, only the observations from β-MHC fibers are consistent with the clinical data for A28V in humans. NEW & NOTEWORTHY The differential impact of α- and β-myosin heavy chain (MHC) on contractile dynamics causes a mutant cardiac troponin T (TnTA30V) to differently modulate cardiac contractile function. TnTA30V attenuated Ca2+-activated maximal tension and length-mediated cross-bridge recruitment against α-MHC but augmented these parameters against β-MHC, suggesting divergent contractile phenotypes.
Collapse
Affiliation(s)
- Alexis V Mickelson
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Sampath K Gollapudi
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Murali Chandra
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
27
|
L71F mutation in rat cardiac troponin T augments crossbridge recruitment and detachment dynamics against α-myosin heavy chain, but not against β-myosin heavy chain. J Muscle Res Cell Motil 2016; 37:215-223. [PMID: 27975185 DOI: 10.1007/s10974-016-9460-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/23/2016] [Indexed: 10/20/2022]
Abstract
The N-terminal extension of human cardiac troponin T (TnT), which modulates myofilament Ca2+ sensitivity, contains several hypertrophic cardiomyopathy (HCM)-causing mutations including S69F. However, the functional consequence of S69F mutation is unknown. The human analog of S69F in rat TnT is L71F (TnTL71F). Because the functional consequences due to structural changes in the N-terminal extension are influenced by the type of myosin heavy chain (MHC) isoform, we hypothesized that the TnTL71F-mediated effect would be differently modulated by α- and β-MHC isoforms. TnTL71F and wild-type rat TnT were reconstituted into de-membranated muscle fibers from normal (α-MHC) and propylthiouracil-treated rat hearts (β-MHC) to measure steady-state and dynamic contractile parameters. The magnitude of the TnTL71F-mediated attenuation of Ca2+-activated maximal tension was greater in α- than in β-MHC fibers. For example, TnTL71F attenuated maximal tension by 31% in α-MHC fibers but only by 10% in β-MHC fibers. Furthermore, TnTL71F reduced myofilament Ca2+ sensitivity by 0.11 pCa units in α-MHC fibers but only by 0.05 pCa units in β-MHC fibers. TnTL71F augmented rate constants of crossbridge recruitment and crossbridge detachment dynamics in α-MHC fibers but not in β-MHC fibers. Collectively, our data demonstrate that TnTL71F induces greater contractile deficits against α-MHC than against β-MHC background.
Collapse
|
28
|
Kraker J, Viswanathan SK, Knöll R, Sadayappan S. Recent Advances in the Molecular Genetics of Familial Hypertrophic Cardiomyopathy in South Asian Descendants. Front Physiol 2016; 7:499. [PMID: 27840609 PMCID: PMC5083855 DOI: 10.3389/fphys.2016.00499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/12/2016] [Indexed: 12/14/2022] Open
Abstract
The South Asian population, numbered at 1.8 billion, is estimated to comprise around 20% of the global population and 1% of the American population, and has one of the highest rates of cardiovascular disease. While South Asians show increased classical risk factors for developing heart failure, the role of population-specific genetic risk factors has not yet been examined for this group. Hypertrophic cardiomyopathy (HCM) is one of the major cardiac genetic disorders among South Asians, leading to contractile dysfunction, heart failure, and sudden cardiac death. This disease displays autosomal dominant inheritance, and it is associated with a large number of variants in both sarcomeric and non-sarcomeric proteins. The South Asians, a population with large ethnic diversity, potentially carries region-specific polymorphisms. There is high variability in disease penetrance and phenotypic expression of variants associated with HCM. Thus, extensive studies are required to decipher pathogenicity and the physiological mechanisms of these variants, as well as the contribution of modifier genes and environmental factors to disease phenotypes. Conducting genotype-phenotype correlation studies will lead to improved understanding of HCM and, consequently, improved treatment options for this high-risk population. The objective of this review is to report the history of cardiovascular disease and HCM in South Asians, present previously published pathogenic variants, and introduce current efforts to study HCM using induced pluripotent stem cell-derived cardiomyocytes, next-generation sequencing, and gene editing technologies. The authors ultimately hope that this review will stimulate further research, drive novel discoveries, and contribute to the development of personalized medicine with the aim of expanding therapeutic strategies for HCM.
Collapse
Affiliation(s)
- Jessica Kraker
- Department of Internal Medicine, Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Sciences, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Shiv Kumar Viswanathan
- Department of Internal Medicine, Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Sciences, University of Cincinnati College of Medicine Cincinnati, OH, USA
| | - Ralph Knöll
- AstraZeneca R&D Mölndal, Innovative Medicines and Early Development, Cardiovascular and Metabolic Diseases iMedMölndal, Sweden; Integrated Cardio Metabolic Centre, Karolinska Institutet, Myocardial Genetics, Karolinska University Hospital in HuddingeHuddinge, Sweden
| | - Sakthivel Sadayappan
- Department of Internal Medicine, Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Sciences, University of Cincinnati College of Medicine Cincinnati, OH, USA
| |
Collapse
|
29
|
In Vivo Cannulation Methods for Cardiomyocytes Isolation from Heart Disease Models. PLoS One 2016; 11:e0160605. [PMID: 27500929 PMCID: PMC4976940 DOI: 10.1371/journal.pone.0160605] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/21/2016] [Indexed: 12/13/2022] Open
Abstract
Isolation of high quality cardiomyocytes is critically important for achieving successful experiments in many cellular and molecular cardiology studies. Methods for isolating cardiomyocytes from the murine heart generally are time-sensitive and experience-dependent, and often fail to produce high quality cells. Major technical difficulties can be related to the surgical procedures needed to explant the heart and to cannulate the vessel to mount onto the Langendorff system before in vitro reperfusion can begin. During this period, transient hypoxia and ischemia may damage the heart, resulting in low yield and poor quality of cells, especially for heart disease models that have fragile cells. We have developed novel in vivo cannulation methods to minimize hypoxia and ischemia, and fine-tuned the entire protocol to produce high quality ventricular myocytes. The high cell quality has been confirmed using important structural and functional criteria such as morphology, t-tubule structure, action potential morphology, Ca2+ signaling, responsiveness to beta-adrenergic agonist, and ability to have robust contraction under mechanically loaded condition. Together these assessments show the preservation of the cardiac excitation–contraction machinery in cells isolated using this technique. The in vivo cannulation method enables consistent isolation of high-quality cardiomyocytes, even from heart disease models that were notoriously difficult for cell isolation using traditional methods.
Collapse
|
30
|
Liver Kinase B1 complex acts as a novel modifier of myofilament function and localizes to the Z-disk in cardiac myocytes. Arch Biochem Biophys 2016; 601:32-41. [PMID: 26971467 DOI: 10.1016/j.abb.2016.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/09/2016] [Accepted: 03/08/2016] [Indexed: 01/15/2023]
Abstract
Contractile perturbations downstream of Ca(2+) binding to troponin C, the so-called sarcomere-controlled mechanisms, represent the earliest indicators of energy remodeling in the diseased heart [1]. Central to cellular energy "sensing" is the adenosine monophosphate-activated kinase (AMPK) pathway, which is known to directly target myofilament proteins and alter contractility [2-6]. We previously showed that the upstream AMPK kinase, LKB1/MO25/STRAD, impacts myofilament function independently of AMPK [5]. Therefore, we hypothesized that the LKB1 complex associated with myofilament proteins and that alterations in energy signaling modulated targeting or localization of the LKB1 complex to the myofilament. Using an integrated strategy of myofilament mechanics, immunoblot analysis, co-immunoprecipitation, mass spectroscopy, and immunofluorescence, we showed that 1) LKB1 and MO25 associated with myofibrillar proteins, 2) cellular energy stress re-distributed AMPK/LKB1 complex proteins within the sarcomere, and 3) the LKB1 complex localized to the Z-Disk and interacted with cytoskeletal and energy-regulating proteins, including vinculin and ATP Synthase (Complex V). These data represent a novel role for LKB1 complex proteins in myofilament function and myocellular "energy" sensing in the heart.
Collapse
|
31
|
Cheng Y, Regnier M. Cardiac troponin structure-function and the influence of hypertrophic cardiomyopathy associated mutations on modulation of contractility. Arch Biochem Biophys 2016; 601:11-21. [PMID: 26851561 PMCID: PMC4899195 DOI: 10.1016/j.abb.2016.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 11/29/2022]
Abstract
Cardiac troponin (cTn) acts as a pivotal regulator of muscle contraction and relaxation and is composed of three distinct subunits (cTnC: a highly conserved Ca(2+) binding subunit, cTnI: an actomyosin ATPase inhibitory subunit, and cTnT: a tropomyosin binding subunit). In this mini-review, we briefly summarize the structure-function relationship of cTn and its subunits, its modulation by PKA-mediated phosphorylation of cTnI, and what is known about how these properties are altered by hypertrophic cardiomyopathy (HCM) associated mutations of cTnI. This includes recent work using computational modeling approaches to understand the atomic-based structural level basis of disease-associated mutations. We propose a viewpoint that it is alteration of cTnC-cTnI interaction (rather than the Ca(2+) binding properties of cTn) per se that disrupt the ability of PKA-mediated phosphorylation at cTnI Ser-23/24 to alter contraction and relaxation in at least some HCM-associated mutations. The combination of state of the art biophysical approaches can provide new insight on the structure-function mechanisms of contractile dysfunction resulting cTnI mutations and exciting new avenues for the diagnosis, prevention, and even treatment of heart diseases.
Collapse
Affiliation(s)
- Yuanhua Cheng
- University of Washington, Department of Bioengineering, Seattle, WA, USA
| | - Michael Regnier
- University of Washington, Department of Bioengineering, Seattle, WA, USA.
| |
Collapse
|
32
|
Messer AE, Bayliss CR, El-Mezgueldi M, Redwood CS, Ward DG, Leung MC, Papadaki M, Dos Remedios C, Marston SB. Mutations in troponin T associated with Hypertrophic Cardiomyopathy increase Ca(2+)-sensitivity and suppress the modulation of Ca(2+)-sensitivity by troponin I phosphorylation. Arch Biochem Biophys 2016; 601:113-20. [PMID: 27036851 PMCID: PMC4909753 DOI: 10.1016/j.abb.2016.03.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/16/2016] [Accepted: 03/26/2016] [Indexed: 11/24/2022]
Abstract
We investigated the effect of 7 Hypertrophic Cardiomyopathy (HCM)-causing mutations in troponin T (TnT) on troponin function in thin filaments reconstituted with actin and human cardiac tropomyosin. We used the quantitative in vitro motility assay to study Ca2+-regulation of unloaded movement and its modulation by troponin I phosphorylation. Troponin from a patient with the K280N TnT mutation showed no difference in Ca2+-sensitivity when compared with donor heart troponin and the Ca2+-sensitivity was also independent of the troponin I phosphorylation level (uncoupled). The recombinant K280N TnT mutation increased Ca2+-sensitivity 1.7-fold and was also uncoupled. The R92Q TnT mutation in troponin from transgenic mouse increased Ca2+-sensitivity and was also completely uncoupled. Five TnT mutations (Δ14, Δ28 + 7, ΔE160, S179F and K273E) studied in recombinant troponin increased Ca2+-sensitivity and were all fully uncoupled. Thus, for HCM-causing mutations in TnT, Ca2+-sensitisation together with uncoupling in vitro is the usual response and both factors may contribute to the HCM phenotype. We also found that Epigallocatechin-3-gallate (EGCG) can restore coupling to all uncoupled HCM-causing TnT mutations. In fact the combination of Ca2+-desensitisation and re-coupling due to EGCG completely reverses both the abnormalities found in troponin with a TnT HCM mutation suggesting it may have therapeutic potential. 7 HCM-causing mutations in cardiac TnT were studied using in vitro motility assay. All the mutations increased myofilament Ca2+-sensitivity (range 1.5–2.7 fold). All mutations suppressed the modulation of Ca2+-sensitivity by TnI phosphorylation. Epigallocatechin-3-gallate (EGCG) restored this modulation to all mutations. This suggests a therapeutic potential for EGCG in the treatment of HCM.
Collapse
|
33
|
Abstract
Calcium binding and dissociation within the cardiac thin filament (CTF) is a fundamental regulator of normal contraction and relaxation. Although the disruption of this complex, allosterically mediated process has long been implicated in human disease, the precise atomic-level mechanisms remain opaque, greatly hampering the development of novel targeted therapies. To address this question, we used a fully atomistic CTF model to test both Ca(2+) binding strength and the energy required to remove Ca(2+) from the N-lobe binding site in WT and mutant troponin complexes that have been linked to genetic cardiomyopathies. This computational approach is combined with measurements of in vitro Ca(2+) dissociation rates in fully reconstituted WT and cardiac troponin T R92L and R92W thin filaments. These human disease mutations represent known substitutions at the same residue, reside at a significant distance from the calcium binding site in cardiac troponin C, and do not affect either the binding pocket affinity or EF-hand structure of the binding domain. Both have been shown to have significantly different effects on cardiac function in vivo. We now show that these mutations independently alter the interaction between the Ca(2+) ion and cardiac troponin I subunit. This interaction is a previously unidentified mechanism, in which mutations in one protein of a complex indirectly affect a third via structural and dynamic changes in a second to yield a pathogenic change in thin filament function that results in mutation-specific disease states. We can now provide atom-level insight that is potentially highly actionable in drug design.
Collapse
|
34
|
Crocini C, Ferrantini C, Scardigli M, Coppini R, Mazzoni L, Lazzeri E, Pioner JM, Scellini B, Guo A, Song LS, Yan P, Loew LM, Tardiff J, Tesi C, Vanzi F, Cerbai E, Pavone FS, Sacconi L, Poggesi C. Novel insights on the relationship between T-tubular defects and contractile dysfunction in a mouse model of hypertrophic cardiomyopathy. J Mol Cell Cardiol 2015; 91:42-51. [PMID: 26714042 PMCID: PMC4767219 DOI: 10.1016/j.yjmcc.2015.12.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 11/16/2015] [Accepted: 12/16/2015] [Indexed: 02/06/2023]
Abstract
Abnormalities of cardiomyocyte Ca2 + homeostasis and excitation–contraction (E–C) coupling are early events in the pathogenesis of hypertrophic cardiomyopathy (HCM) and concomitant determinants of the diastolic dysfunction and arrhythmias typical of the disease. T-tubule remodelling has been reported to occur in HCM but little is known about its role in the E–C coupling alterations of HCM. Here, the role of T-tubule remodelling in the electro-mechanical dysfunction associated to HCM is investigated in the Δ160E cTnT mouse model that expresses a clinically-relevant HCM mutation. Contractile function of intact ventricular trabeculae is assessed in Δ160E mice and wild-type siblings. As compared with wild-type, Δ160E trabeculae show prolonged kinetics of force development and relaxation, blunted force-frequency response with reduced active tension at high stimulation frequency, and increased occurrence of spontaneous contractions. Consistently, prolonged Ca2 + transient in terms of rise and duration are also observed in Δ160E trabeculae and isolated cardiomyocytes. Confocal imaging in cells isolated from Δ160E mice reveals significant, though modest, remodelling of T-tubular architecture. A two-photon random access microscope is employed to dissect the spatio-temporal relationship between T-tubular electrical activity and local Ca2 + release in isolated cardiomyocytes. In Δ160E cardiomyocytes, a significant number of T-tubules (> 20%) fails to propagate action potentials, with consequent delay of local Ca2 + release. At variance with wild-type, we also observe significantly increased variability of local Ca2 + transient rise as well as higher Ca2 +-spark frequency. Although T-tubule structural remodelling in Δ160E myocytes is modest, T-tubule functional defects determine non-homogeneous Ca2 + release and delayed myofilament activation that significantly contribute to mechanical dysfunction. Contraction and Ca2 + transient kinetics are impaired in myocardial preparations from mice carrying the cardiac troponin T ∆ 160E mutation. T-tubules architecture is mildly altered in ∆160E cardiomyocytes. 20% of T-tubules fail to propagate action potential and produce delay of local Ca2 + rise. Higher spatio-temporal variability of local Ca2 + rise and increased Ca2 + sparks frequency are found in ∆160E cardiomyocytes.
Collapse
Affiliation(s)
- C Crocini
- European Laboratory for Non-Linear Spectroscopy, 50019 Florence, Italy
| | - C Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - M Scardigli
- European Laboratory for Non-Linear Spectroscopy, 50019 Florence, Italy
| | - R Coppini
- Division of Pharmacology, Department "NeuroFarBa", University of Florence, 50139 Florence, Italy
| | - L Mazzoni
- Division of Pharmacology, Department "NeuroFarBa", University of Florence, 50139 Florence, Italy
| | - E Lazzeri
- European Laboratory for Non-Linear Spectroscopy, 50019 Florence, Italy
| | - J M Pioner
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - B Scellini
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - A Guo
- Division of Cardiovascular Medicine, Department of Internal Medicine and Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - L S Song
- Division of Cardiovascular Medicine, Department of Internal Medicine and Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - P Yan
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - L M Loew
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - J Tardiff
- Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - C Tesi
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - F Vanzi
- European Laboratory for Non-Linear Spectroscopy, 50019 Florence, Italy
| | - E Cerbai
- Division of Pharmacology, Department "NeuroFarBa", University of Florence, 50139 Florence, Italy
| | - F S Pavone
- European Laboratory for Non-Linear Spectroscopy, 50019 Florence, Italy; Department of Physics and Astronomy, University of Florence, 50019 Sesto Fiorentino, Italy; National Institute of Optics, National Research Council, 50125 Florence, Italy
| | - L Sacconi
- European Laboratory for Non-Linear Spectroscopy, 50019 Florence, Italy; National Institute of Optics, National Research Council, 50125 Florence, Italy.
| | - C Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| |
Collapse
|
35
|
Chandra V, Gollapudi SK, Chandra M. Rat cardiac troponin T mutation (F72L)-mediated impact on thin filament cooperativity is divergently modulated by α- and β-myosin heavy chain isoforms. Am J Physiol Heart Circ Physiol 2015; 309:H1260-70. [PMID: 26342069 DOI: 10.1152/ajpheart.00519.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/31/2015] [Indexed: 11/22/2022]
Abstract
The primary causal link between disparate effects of human hypertrophic cardiomyopathy (HCM)-related mutations in troponin T (TnT) and α- and β-myosin heavy chain (MHC) isoforms on cardiac contractile phenotype remains poorly understood. Given the divergent impact of α- and β-MHC on the NH2-terminal extension (44-73 residues) of TnT, we tested if the effects of the HCM-linked mutation (TnTF70L) were differentially altered by α- and β-MHC. We hypothesized that the emergence of divergent thin filament cooperativity would lead to contrasting effects of TnTF70L on contractile function in the presence of α- and β-MHC. The rat TnT analog of the human F70L mutation (TnTF72L) or the wild-type rat TnT (TnTWT) was reconstituted into demembranated muscle fibers from normal (α-MHC) and propylthiouracil-treated (β-MHC) rat hearts to measure steady-state and dynamic contractile function. TnTF72L-mediated effects on tension, myofilament Ca(2+) sensitivity, myofilament cooperativity, rate constants of cross-bridge (XB) recruitment dynamics, and force redevelopment were divergently modulated by α- and β-MHC. TnTF72L increased the rate of XB distortion dynamics by 49% in α-MHC fibers but had no effect in β-MHC fibers; these observations suggest that TnTF72L augmented XB detachment kinetics in α-MHC, but not β-MHC, fibers. TnTF72L increased the negative impact of strained XBs on the force-bearing XBs by 39% in α-MHC fibers but had no effect in β-MHC fibers. Therefore, TnTF72L leads to contractile changes that are linked to dilated cardiomyopathy in the presence of α-MHC. On the other hand, TnTF72L leads to contractile changes that are linked to HCM in the presence of β-MHC.
Collapse
Affiliation(s)
- Vikram Chandra
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Sampath K Gollapudi
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Murali Chandra
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
36
|
Gollapudi SK, Tardiff JC, Chandra M. The functional effect of dilated cardiomyopathy mutation (R144W) in mouse cardiac troponin T is differently affected by α- and β-myosin heavy chain isoforms. Am J Physiol Heart Circ Physiol 2015; 308:H884-93. [PMID: 25681424 DOI: 10.1152/ajpheart.00528.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 02/04/2015] [Indexed: 11/22/2022]
Abstract
Given the differential impact of α- and β-myosin heavy chain (MHC) isoforms on how troponin T (TnT) modulates contractile dynamics, we hypothesized that the effects of dilated cardiomyopathy (DCM) mutations in TnT would be altered differently by α- and β-MHC. We characterized dynamic contractile features of normal (α-MHC) and transgenic (β-MHC) mouse cardiac muscle fibers reconstituted with a mouse TnT analog (TnTR144W) of the human DCM R141W mutation. TnTR144W did not alter maximal tension but attenuated myofilament Ca(2+) sensitivity (pCa50) to a similar extent in α- and β-MHC fibers. TnTR144W attenuated the speed of cross-bridge (XB) distortion dynamics (c) by 24% and the speed of XB recruitment dynamics (b) by 17% in α-MHC fibers; however, both b and c remained unaltered in β-MHC fibers. Likewise, TnTR144W attenuated the rates of XB detachment (g) and tension redevelopment (ktr) only in α-MHC fibers. TnTR144W also decreased the impact of strained XBs on the recruitment of new XBs (γ) by 30% only in α-MHC fibers. Because c, b, g, ktr, and γ are strongly influenced by thin filament-based cooperative mechanisms, we conclude that the TnTR144W- and β-MHC-mediated changes in the thin filament interact to produce a less severe functional phenotype, compared with that brought about by TnTR144W and α-MHC. These observations provide a basis for lower mortality rates of humans (β-MHC) harboring the TnTR141W mutant compared with transgenic mouse studies. Our findings strongly suggest that some caution is necessary when extrapolating data from transgenic mouse studies to human hearts.
Collapse
Affiliation(s)
- Sampath K Gollapudi
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington; and
| | - Jil C Tardiff
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Murali Chandra
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington; and
| |
Collapse
|
37
|
Effects of pseudo-phosphorylated rat cardiac troponin T are differently modulated by α- and β-myosin heavy chain isoforms. Basic Res Cardiol 2014; 109:442. [PMID: 25301196 DOI: 10.1007/s00395-014-0442-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/31/2014] [Accepted: 09/18/2014] [Indexed: 10/24/2022]
Abstract
Interplay between the protein kinase C (PKC)-mediated phosphorylation of troponin T (TnT)- and myosin heavy chain (MHC)-mediated effects on thin filaments takes on a new significance because: (1) there is significant interaction between the TnT- and MHC-mediated effects on cardiac thin filaments; (2) although the phosphorylation of TnT by PKC isoforms is common to both human and rodent hearts, human hearts predominantly express β-MHC while rodent hearts predominantly express α-MHC. Therefore, we tested how α- and β-MHC isoforms differently affected the functional effects of phosphorylated TnT. Contractile measurements were made on cardiac muscle fibers from normal rats (α-MHC) and propylthiouracil-treated rats (β-MHC), reconstituted with the recombinant phosphomimetic-TnT (T204E; threonine 204 replaced by glutamate). Ca2+ -activated maximal tension decreased differently in α-MHC + T204E (~68%) and β-MHC + T204E (~35%). However, myofilament Ca2+ sensitivity decreased similarly in α-MHC + T204E and β-MHC + T204E, demonstrating that a decrease in Ca2+ sensitivity alone cannot explain the greater attenuation of tension in α-MHC + T204E. Interestingly, dynamic contractile parameters (rates of tension redevelopment, crossbridge (XB) recruitment dynamics, XB distortion dynamics, and XB detachment kinetics) decreased only in α-MHC + T204E. Thus, the transition of thin filaments from the blocked- to closed-state was attenuated in α-MHC + T204E and β-MHC + T204E, but the closed- to open-state transition was attenuated only in α-MHC + T204E. Our study demonstrates that the effects of phosphorylated TnT and MHC isoforms interact to bring about different functional states of cardiac thin filaments.
Collapse
|
38
|
Ablation of plasma membrane Ca(2+)-ATPase isoform 4 prevents development of hypertrophy in a model of hypertrophic cardiomyopathy. J Mol Cell Cardiol 2014; 77:53-63. [PMID: 25280781 DOI: 10.1016/j.yjmcc.2014.09.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 09/23/2014] [Accepted: 09/24/2014] [Indexed: 11/20/2022]
Abstract
The mechanisms linking the expression of sarcomeric mutant proteins to the development of pathological hypertrophy in hypertrophic cardiomyopathy (HCM) remain poorly understood. We investigated the role of the plasma membrane Ca(2+)-ATPase PMCA4 in the HCM phenotype using a transgenic model that expresses mutant (Glu180Gly) α-tropomyosin (Tm180) in heart. Immunoblot analysis revealed that cardiac PMCA4 expression was upregulated early in Tm180 disease pathogenesis. This was accompanied by an increase in levels of the L-type Ca(2+)-channel, which is implicated in pathological hypertrophy. When Tm180 mice were crossed with a PMCA4-null line, loss of PMCA4 caused the abrogation of hypertrophy in Tm180/PMCA4-null double mutant mice. RT-PCR analysis of Tm180/PMCA4-null hearts revealed blunting of the fetal program and reversion of pro-fibrotic Col1a1 and Col3a1 gene expression to wild-type levels. This was accompanied by evidence of reduced L-type Ca(2+)-channel expression, and diminished calcineurin activity. Expression of the metabolic substrate transporters glucose transporter 4 and carnitine palmitoyltransferase 1b was preserved and Tm180-related changes in mRNA levels of various contractile stress-related proteins including the cardiac ankyrin protein CARP and the N2B isoform of titin were reversed in Tm180/PMCA4-null hearts. cGMP levels were increased and phosphorylation of vasodilator-stimulated phosphoprotein was elevated in Tm180/PMCA4-null hearts. These changes were associated with a sharp reduction in left ventricular end-diastolic pressure in Tm180/PMCA4-null hearts, which occurred despite persistence of Tm180-related impairment of relaxation dynamics. These results reveal a novel and specific role for PMCA4 in the Tm180 hypertrophic phenotype, with the "protective" effects of PMCA4 deficiency encompassing multiple determinants of HCM-related hypertrophy.
Collapse
|
39
|
Witjas-Paalberends ER, Güçlü A, Germans T, Knaapen P, Harms HJ, Vermeer AMC, Christiaans I, Wilde AAM, Dos Remedios C, Lammertsma AA, van Rossum AC, Stienen GJM, van Slegtenhorst M, Schinkel AF, Michels M, Ho CY, Poggesi C, van der Velden J. Gene-specific increase in the energetic cost of contraction in hypertrophic cardiomyopathy caused by thick filament mutations. Cardiovasc Res 2014; 103:248-57. [PMID: 24835277 DOI: 10.1093/cvr/cvu127] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Disease mechanisms regarding hypertrophic cardiomyopathy (HCM) are largely unknown and disease onset varies. Sarcomere mutations might induce energy depletion for which until now there is no direct evidence at sarcomere level in human HCM. This study investigated if mutations in genes encoding myosin-binding protein C (MYBPC3) and myosin heavy chain (MYH7) underlie changes in the energetic cost of contraction in the development of human HCM disease. METHODS AND RESULTS Energetic cost of contraction was studied in vitro by measurements of force development and ATPase activity in cardiac muscle strips from 26 manifest HCM patients (11 MYBPC3mut, 9 MYH7mut, and 6 sarcomere mutation-negative, HCMsmn). In addition, in vivo, the ratio between external work (EW) and myocardial oxygen consumption (MVO2) to obtain myocardial external efficiency (MEE) was determined in 28 pre-hypertrophic mutation carriers (14 MYBPC3mut and 14 MYH7mut) and 14 healthy controls using [(11)C]-acetate positron emission tomography and cardiovascular magnetic resonance imaging. Tension cost (TC), i.e. ATPase activity during force development, was higher in MYBPC3mut and MYH7mut compared with HCMsmn at saturating [Ca(2+)]. TC was also significantly higher in MYH7mut at submaximal, more physiological [Ca(2+)]. EW was significantly lower in both mutation carrier groups, while MVO2 did not differ. MEE was significantly lower in both mutation carrier groups compared with controls, showing the lowest efficiency in MYH7 mutation carriers. CONCLUSION We provide direct evidence that sarcomere mutations perturb the energetic cost of cardiac contraction. Gene-specific severity of cardiac abnormalities may underlie differences in disease onset and suggests that early initiation of metabolic treatment may be beneficial, in particular, in MYH7 mutation carriers.
Collapse
Affiliation(s)
- E Rosalie Witjas-Paalberends
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Ahmet Güçlü
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands ICIN Netherlands Heart Institute, Utrecht, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Paul Knaapen
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Hendrik J Harms
- Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Alexa M C Vermeer
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Imke Christiaans
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Arthur A M Wilde
- Department of Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Cris Dos Remedios
- Institute for Biomedical Research, Muscle Research Unit, University of Sydney, Sydney, Australia
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Albert C van Rossum
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Ger J M Stienen
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands Department of Physics and Astronomy, VU University, Amsterdam, The Netherlands
| | | | - Arend F Schinkel
- Thorax Center, Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Michelle Michels
- Thorax Center, Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carolyn Y Ho
- Brigham and Women's Hospital, Cardiology, Boston, MA, USA
| | - Corrado Poggesi
- Department of Physiology, University of Florence, Florence, Italy
| | - Jolanda van der Velden
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands ICIN Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
40
|
Poggesi C, Ho CY. Muscle dysfunction in hypertrophic cardiomyopathy: what is needed to move to translation? J Muscle Res Cell Motil 2014; 35:37-45. [PMID: 24493262 DOI: 10.1007/s10974-014-9374-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 01/02/2014] [Indexed: 02/04/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomere genes. As such, HCM provides remarkable opportunities to study how changes to the heart's molecular motor apparatus may influence cardiac structure and function. Although the genetic basis of HCM is well-described, there is much more limited understanding of the precise consequences of sarcomere mutations--how they remodel the heart, and how these changes lead to the dramatic clinical consequences associated with HCM. More precise characterization of the mechanisms leading from sarcomere mutation to altered cardiac muscle function is critical to gain insight into fundamental disease biology and phenotypic evolution. Such knowledge will help foster development of novel treatment strategies aimed at correcting and preventing disease development in HCM.
Collapse
Affiliation(s)
- Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 63, 50134, Florence, Italy,
| | | |
Collapse
|
41
|
Moore RK, Abdullah S, Tardiff JC. Allosteric effects of cardiac troponin TNT1 mutations on actomyosin binding: a novel pathogenic mechanism for hypertrophic cardiomyopathy. Arch Biochem Biophys 2014; 552-553:21-8. [PMID: 24480310 DOI: 10.1016/j.abb.2014.01.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/16/2013] [Accepted: 01/16/2014] [Indexed: 12/13/2022]
Abstract
The majority of hypertrophic cardiomyopathy mutations in (cTnT) occur within the alpha-helical tropomyosin binding TNT1 domain. A highly charged region at the C-terminal end of TNT1 unwinds to create a flexible "hinge". While this region has not been structurally resolved, it likely acts as an extended linker between the two cTnT functional domains. Mutations in this region cause phenotypically diverse and often severe forms of HCM. Mechanistic insight, however, has been limited by the lack of structural information. To overcome this limitation, we evaluated the effects of cTnT 160-163 mutations using regulated in vitro motility (R-IVM) assays and transgenic mouse models. R-IVM revealed that cTnT mutations Δ160E, E163R and E163K disrupted weak electrostatic actomyosin binding. Reducing the ionic strength or decreasing Brownian motion rescued function. This is the first observation of HCM-linked mutations in cTnT disrupting weak interactions between the thin filament and myosin. To evaluate the in vivo effects of altering weak actomyosin binding we generated transgenic mice expressing Δ160E and E163R mutant cTnT and observed severe cardiac remodeling and profound myofilament disarray. The functional changes observed in vitro may contribute to the structural impairment seen in vivo by destabilizing myofilament structure and acting as a constant pathophysiologic stress.
Collapse
Affiliation(s)
- Rachel K Moore
- Department of Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Salwa Abdullah
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Jil C Tardiff
- Department of Medicine, University of Arizona, Tucson, AZ 85724, United States; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States.
| |
Collapse
|
42
|
Viswanathan MC, Kaushik G, Engler AJ, Lehman W, Cammarato A. A Drosophila melanogaster model of diastolic dysfunction and cardiomyopathy based on impaired troponin-T function. Circ Res 2013; 114:e6-17. [PMID: 24221941 DOI: 10.1161/circresaha.114.302028] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
RATIONALE Regulation of striated muscle contraction is achieved by Ca2+ -dependent steric modulation of myosin cross-bridge cycling on actin by the thin filament troponin-tropomyosin complex. Alterations in the complex can induce contractile dysregulation and disease. For example, mutations between or near residues 112 to 136 of cardiac troponin-T, the crucial TnT1 (N-terminal domain of troponin-T)-tropomyosin-binding region, cause cardiomyopathy. The Drosophila upheld(101) Glu/Lys amino acid substitution lies C-terminally adjacent to this phylogenetically conserved sequence. OBJECTIVE Using a highly integrative approach, we sought to determine the molecular trigger of upheld(101) myofibrillar degeneration, to evaluate contractile performance in the mutant cardiomyocytes, and to examine the effects of the mutation on the entire Drosophila heart to elucidate regulatory roles for conserved TnT1 regions and provide possible mechanistic insight into cardiac dysfunction. METHODS AND RESULTS Live video imaging of Drosophila cardiac tubes revealed that the troponin-T mutation prolongs systole and restricts diastolic dimensions of the heart, because of increased numbers of actively cycling myosin cross-bridges. Elevated resting myocardial stiffness, consistent with upheld(101) diastolic dysfunction, was confirmed by an atomic force microscopy-based nanoindentation approach. Direct visualization of mutant thin filaments via electron microscopy and 3-dimensional reconstruction resolved destabilized tropomyosin positioning and aberrantly exposed myosin-binding sites under low Ca2+ conditions. CONCLUSIONS As a result of troponin-tropomyosin dysinhibition, upheld(101) hearts exhibited cardiac dysfunction and remodeling comparable to that observed during human restrictive cardiomyopathy. Thus, reversal of charged residues about the conserved tropomyosin-binding region of TnT1 may perturb critical intermolecular associations required for proper steric regulation, which likely elicits myopathy in our Drosophila model.
Collapse
Affiliation(s)
- Meera Cozhimuttam Viswanathan
- From the Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (M.C.V., A.C.); Department of Bioengineering, University of California, San Diego, La Jolla, CA (G.K., A.J.E.); and Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA (W.L.)
| | | | | | | | | |
Collapse
|
43
|
Moore RK, Grinspan LT, Jimenez J, Guinto PJ, Ertz-Berger B, Tardiff JC. HCM-linked ∆160E cardiac troponin T mutation causes unique progressive structural and molecular ventricular remodeling in transgenic mice. J Mol Cell Cardiol 2013; 58:188-98. [PMID: 23434821 PMCID: PMC3819192 DOI: 10.1016/j.yjmcc.2013.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 01/11/2013] [Accepted: 02/02/2013] [Indexed: 01/27/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a primary disease of the cardiac muscle, and one of the most common causes of sudden cardiac death (SCD) in young people. Many mutations in cardiac troponin T (cTnT) lead to a complex form of HCM with varying degrees of ventricular hypertrophy and ~65% of all cTnT mutations occur within or flanking the elongated N-terminal TNT1 domain. Biophysical studies have predicted that distal TNT1 mutations, including Δ160E, cause disease by a novel, yet unknown mechanism as compared to N-terminal mutations. To begin to address the specific effects of this commonly observed cTnT mutation we generated two independent transgenic mouse lines carrying variant doses of the mutant transgene. Hearts from the 30% and 70% cTnT Δ160E lines demonstrated a highly unique, dose-dependent disruption in cellular and sarcomeric architecture and a highly progressive pattern of ventricular remodeling. While adult ventricular myocytes isolated from Δ160E transgenic mice exhibited dosage-independent mechanical impairments, decreased sarcoplasmic reticulum calcium load and SERCA2a calcium uptake activity, the observed decreases in calcium transients were dosage-dependent. The latter findings were concordant with measures of calcium regulatory protein abundance and phosphorylation state. Finally, studies of whole heart physiology in the isovolumic mode demonstrated dose-dependent differences in the degree of cardiac dysfunction. We conclude that the observed clinical severity of the cTnT Δ160E mutation is caused by a combination of direct sarcomeric disruption coupled to a profound dysregulation of Ca(2+) homeostasis at the cellular level that results in a unique and highly progressive pattern of ventricular remodeling.
Collapse
Affiliation(s)
- Rachel K Moore
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Yeshiva University, 1300 Morris Park Avenue, Ullmann, Room 316, Bronx, NY 10461, USA
| | | | | | | | | | | |
Collapse
|
44
|
The tropomyosin binding region of cardiac troponin T modulates crossbridge recruitment dynamics in rat cardiac muscle fibers. J Mol Biol 2013; 425:1565-81. [PMID: 23357173 DOI: 10.1016/j.jmb.2013.01.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 01/15/2013] [Accepted: 01/17/2013] [Indexed: 11/22/2022]
Abstract
The cardiac muscle comprises dynamically interacting components that use allosteric/cooperative mechanisms to yield unique heart-specific properties. An essential protein in this allosteric/cooperative mechanism is cardiac muscle troponin T (cTnT), the central region (CR) and the T2 region of which differ significantly from those of fast skeletal muscle troponin T (fsTnT). To understand the biological significance of such sequence heterogeneity, we replaced the T1 or T2 domain of rat cTnT (RcT1 or RcT2) with its counterpart from rat fsTnT (RfsT1or RfsT2) to generate RfsT1-RcT2 and RcT1-RfsT2 recombinant proteins. In addition to contractile function measurements, dynamic features of RfsT1-RcT2- and RcT1-RfsT2-reconstituted rat cardiac muscle fibers were captured by fitting the recruitment-distortion model to the force response of small-amplitude (0.5%) muscle length changes. RfsT1-RcT2 fibers showed a 40% decrease in tension and a 44% decrease in ATPase activity, but RcT1-RfsT2 fibers were unaffected. The magnitude of length-mediated increase in crossbridge (XB) recruitment (E0) decreased by ~33% and the speed of XB recruitment (b) increased by ~100% in RfsT1-RcT2 fibers. Our data suggest the following: (1) the CR of cTnT modulates XB recruitment dynamics; (2) the N-terminal end region of cTnT has a synergistic effect on the ability of the CR to modulate XB recruitment dynamics; (3) the T2 region is important for tuning the Ca(2+) regulation of cardiac thin filaments. The combined effects of CR-tropomyosin interactions and the modulating effect of the N-terminal end of cTnT on CR-tropomyosin interactions may lead to the emergence of a unique property that tunes contractile dynamics to heart rates.
Collapse
|
45
|
Cardiomyopathy-Related Mutations in Cardiac Troponin C, L29Q and G159D, Have Divergent Effects on Rat Cardiac Myofiber Contractile Dynamics. Biochem Res Int 2012; 2012:824068. [PMID: 23008774 PMCID: PMC3447348 DOI: 10.1155/2012/824068] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/06/2012] [Accepted: 08/08/2012] [Indexed: 11/17/2022] Open
Abstract
Previous studies of cardiomyopathy-related mutations in cardiac troponin C (cTnC)-L29Q and G159D-have shown diverse findings. The link between such mutant effects and their divergent impact on cardiac phenotypes has remained elusive due to lack of studies on contractile dynamics. We hypothesized that a cTnC mutant-induced change in the thin filament will affect global myofilament mechanodynamics because of the interactions of thin filament kinetics with both Ca(2+) binding and crossbridge (XB) cycling kinetics. We measured pCa-tension relationship and contractile dynamics in detergent-skinned rat cardiac papillary muscle fibers reconstituted with the recombinant wild-type rat cTnC (cTnC(WT)), cTnC(L29Q), and cTnC(G159D) mutants. cTnC(L29Q) fibers demonstrated a significant decrease in Ca(2+) sensitivity, but cTnC(G159D) fibers did not. Both mutants had no effect on Ca(2+)-activated maximal tension. The rate of XB recruitment dynamics increased in cTnC(L29Q) (26%) and cTnC(G159D) (25%) fibers. The rate of XB distortion dynamics increased in cTnC(G159D) fibers (15%). Thus, the cTnC(L29Q) mutant modulates the equilibrium between the non-cycling and cycling pool of XB by affecting the on/off kinetics of the regulatory units (Tropomyosin-Troponin); whereas, the cTnC(G159D) mutant increases XB cycling rate. Different effects on contractile dynamics may offer clue regarding how cTnC(L29Q) and cTnC(G159D) cause divergent effects on cardiac phenotypes.
Collapse
|
46
|
Gollapudi S, Mamidi R, Mallampalli S, Chandra M. The N-terminal extension of cardiac troponin T stabilizes the blocked state of cardiac thin filament. Biophys J 2012; 103:940-8. [PMID: 23009843 PMCID: PMC3433604 DOI: 10.1016/j.bpj.2012.07.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/25/2012] [Accepted: 07/09/2012] [Indexed: 11/21/2022] Open
Abstract
Cardiac troponin T (cTnT) is a key component of contractile regulatory proteins. cTnT is characterized by a ∼32 amino acid N-terminal extension (NTE), the function of which remains unknown. To understand its function, we generated a transgenic (TG) mouse line that expressed a recombinant chimeric cTnT in which the NTE of mouse cTnT was removed by replacing its 1-73 residues with the corresponding 1-41 residues of mouse fast skeletal TnT. Detergent-skinned papillary muscle fibers from non-TG (NTG) and TG mouse hearts were used to measure tension, ATPase activity, Ca(2+) sensitivity (pCa(50)) of tension, rate of tension redevelopment, dynamic muscle fiber stiffness, and maximal fiber shortening velocity at sarcomere lengths (SLs) of 1.9 and 2.3 μm. Ca(2+) sensitivity increased significantly in TG fibers at both short SL (pCa(50) of 5.96 vs. 5.62 in NTG fibers) and long SL (pCa(50) of 6.10 vs. 5.76 in NTG fibers). Maximal cross-bridge turnover and detachment kinetics were unaltered in TG fibers. Our data suggest that the NTE constrains cardiac thin filament activation such that the transition of the thin filament from the blocked to the closed state becomes less responsive to Ca(2+). Our finding has implications regarding the effect of tissue- and disease-related changes in cTnT isoforms on cardiac muscle function.
Collapse
Affiliation(s)
| | | | | | - Murali Chandra
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, Pullman, Washington
| |
Collapse
|
47
|
Ford SJ, Mamidi R, Jimenez J, Tardiff JC, Chandra M. Effects of R92 mutations in mouse cardiac troponin T are influenced by changes in myosin heavy chain isoform. J Mol Cell Cardiol 2012; 53:542-51. [PMID: 22884844 DOI: 10.1016/j.yjmcc.2012.07.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 07/26/2012] [Accepted: 07/29/2012] [Indexed: 11/18/2022]
Abstract
One limitation in understanding how different familial hypertrophic cardiomyopathy (FHC)-related mutations lead to divergent cardiac phenotypes is that such mutations are often studied in transgenic (TG) mouse hearts which contain a fast cycling myosin heavy chain isoform (α-MHC). However, the human heart contains a slow cycling MHC isoform (β-MHC). Given the physiological significance of MHC-troponin interplay effects on cardiac contractile function, we hypothesized that cardiac troponin T (cTnT) mutation-mediated effects on contractile function depend on the type of MHC isoform present in the sarcomere. We tested our hypothesis using two variants of cTnT containing mutations at FHC hotspot R92 (R92L or R92Q), expressed against either an α-MHC or β-MHC background in TG mouse hearts. One finding from our study was that R92L attenuated the length-dependent increase in tension and abolished the length-dependent increase in myofilament Ca(2+) sensitivity only when β-MHC was present. In addition, α- and β-MHC isoforms differentially affected how R92 mutations altered crossbridge (XB) recruitment dynamics. For example, the rate of XB recruitment was faster in R92L or R92Q fibers when β-MHC was present, but was unaffected when α-MHC was present. The R92Q mutation sped XB detachment in the presence of β-MHC, but not in the presence of α-MHC. R92Q affected the XB strain-dependent influence on XB recruitment dynamics, an effect not observed for R92L. Our findings have major implications for understanding not only the divergent effects of R92 mutations on cardiac phenotype, but also the distinct effects of MHC isoforms in determining the outcome of mutations in cTnT.
Collapse
Affiliation(s)
- Steven J Ford
- Department of Veterinary and Comparative Anatomy, Washington State University, Pullman, WA 99164, USA
| | | | | | | | | |
Collapse
|
48
|
Mamidi R, Gollapudi SK, Mallampalli SL, Chandra M. Alanine or aspartic acid substitutions at serine23/24 of cardiac troponin I decrease thin filament activation, with no effect on crossbridge detachment kinetics. Arch Biochem Biophys 2012; 525:1-8. [PMID: 22684024 DOI: 10.1016/j.abb.2012.05.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 05/24/2012] [Accepted: 05/29/2012] [Indexed: 11/16/2022]
Abstract
Ala/Asp substitutions at Ser23/24 have been employed to investigate the functional impact of cardiac troponin I (cTnI) phosphorylation by protein kinase A (PKA). Some limitations of previous studies include the use of heterologous proteins and confounding effects arising from phosphorylation of cardiac myosin binding protein-C. Our goal was to probe the effects of cTnI phosphorylation using a homologous assay, so that altered function could be solely attributed to changes in cTnI. We reconstituted detergent-skinned rat cardiac papillary fibers with homologous rat cardiac troponin subunits to study the impact of Ala and Asp substitutions at Ser23/24 of rat cTnI (RcTnI S23A/24A and RcTnI S23D/24D). Both RcTnI S23A/24A and RcTnI S23D/24D showed a ~36% decrease in Ca(2+)-activated maximal tension. Both RcTnI S23A/24A and RcTnI S23D/24D showed a ~18% decrease in ATPase activity. Muscle fiber stiffness measurements suggested that the decrease in thin filament activation observed in RcTnI S23A/24A and RcTnI S23D/24D was due to a decrease in the number of strongly-bound crossbridges. Another major finding was that Ala and Asp substitutions in cTnI did not affect crossbridge detachment kinetics.
Collapse
Affiliation(s)
- Ranganath Mamidi
- Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology (VCAPP), Washington State University, Pullman, WA 99164-6520, USA
| | | | | | | |
Collapse
|
49
|
Manning EP, Guinto PJ, Tardiff JC. Correlation of molecular and functional effects of mutations in cardiac troponin T linked to familial hypertrophic cardiomyopathy: an integrative in silico/in vitro approach. J Biol Chem 2012; 287:14515-23. [PMID: 22334656 DOI: 10.1074/jbc.m111.257436] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Nearly 70% of all of the known cTnT mutations that cause familial hypertrophic cardiomyopathy fall within the TNT1 region that is critical to cTn-Tm binding. The high resolution structure of this domain has not been determined, and this lack of information has hindered structure-function analysis. In the current study, a coupled computational experimental approach was employed to correlate changes in cTnT dynamics to basic function using the regulated in vitro motility assay (R-IVM). An in silico approach to calculate forces in terms of a bending coordinate was used to precisely identify decreases in bending forces at residues 105 and 106 within the proposed cTnT "hinge" region. Significant functional changes were observed in multiple functional properties, including a decrease in the cooperativity of calcium activation, the calcium sensitivity of sliding speed, and maximum sliding speed. Correlation of the computational and experimental findings revealed an association between TNT1 flexibility and the cooperativity of thin filament calcium activation where an increase in flexibility led to a decrease in cooperativity. Further analysis of the primary sequence of the TNT1 region revealed a unique pattern of conserved charged TNT1 residues altered by the R92W and R92L mutations and may represent the underlying "structure" modulating this central functional domain. These data provide a framework for further integrated in silico/in vitro approaches that may be extended into a high-throughput predictive screen to overcome the current structural limitations in linking molecular phenotype to genotype in thin filament cardiomyopathies.
Collapse
Affiliation(s)
- Edward P Manning
- Department of Physiology and Biophysics, Albert Einstein College of Medicine Bronx, Bronx, New York 10461, USA
| | | | | |
Collapse
|
50
|
Wu B, Wang L, Liu Q, Luo Q. Myocardial contractile and metabolic properties of familial hypertrophic cardiomyopathy caused by cardiac troponin I gene mutations: a simulation study. Exp Physiol 2011; 97:155-69. [PMID: 21967901 DOI: 10.1113/expphysiol.2011.059956] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Familial hypertrophic cardiomyopathy (FHC) is an inherited disease that is caused by sarcomeric protein gene mutations. The mechanism by which these mutant proteins cause disease is uncertain. Experimentally, cardiac troponin I (CTnI) gene mutations mainly alter myocardial performance via increases in the Ca(2+) sensitivity of cardiac contractility. In this study, we used an integrated simulation that links electrophysiology, contractile activity and energy metabolism of the myocardium to investigate alterations in myocardial contractile function and energy metabolism regulation as a result of increased Ca(2+) sensitivity in CTnI mutations. Simulation results reproduced the following typical features of FHC: (1) slower relaxation (diastolic dysfunction) caused by prolonged [Ca(2+)](i) and force transients; (2) higher energy consumption with the increase in Ca(2+) sensitivity; and (3) reduced fatty acid oxidation and enhanced glucose utilization in hypertrophied heart metabolism. Furthermore, the simulation indicated that in conditions of high energy consumption (that is, more than an 18.3% increase in total energy consumption), the myocardial energetic metabolic network switched from a net consumer to a net producer of lactate, resulting in a low coupling of glucose oxidation to glycolysis, which is a common feature of hypertrophied hearts. This study provides a novel systematic myocardial contractile and metabolic analysis to help elucidate the pathogenesis of FHC and suggests that the alterations in resting heart energy supply and demand could contribute to disease progression.
Collapse
Affiliation(s)
- Bo Wu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China
| | | | | | | |
Collapse
|