1
|
Jani V, Kass DA. What can skinned cardiomyocytes teach us about heart disease? Response to letter to the editor: Challenges in assessing myoflament calcium sensitivity and contractile function in HFpEF by Christophe Maack and Vasco Sequeira. J Mol Cell Cardiol 2025; 204:32-34. [PMID: 40381935 DOI: 10.1016/j.yjmcc.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Affiliation(s)
- Vivek Jani
- Department of Medicine, Division of Cardiology and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, United States of America
| | - David A Kass
- Department of Medicine, Division of Cardiology and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, United States of America.
| |
Collapse
|
2
|
Asatryan B, Muller SA, Barth AS. Not All Pathogenic Variants Are Equal: How Founder Variants Lead Precision Medicine Journey in Hypertrophic Cardiomyopathy. JACC. HEART FAILURE 2025:102462. [PMID: 40372295 DOI: 10.1016/j.jchf.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 05/16/2025]
Affiliation(s)
- Babken Asatryan
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Steven A Muller
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Andreas S Barth
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Reil JC, Sequeira V, Reil GH, Scholtz S, Rudolph V, Maack C, Serruys P, Steendijk P. Investigating the Anrep Effect in Hypertrophic Obstructive Cardiomyopathy With Invasive Pressure-Volume Analysis. JACC. ADVANCES 2025; 4:101728. [PMID: 40286379 PMCID: PMC12103075 DOI: 10.1016/j.jacadv.2025.101728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The Anrep effect, an adaptation enhancing left ventricular (LV) contractility in response to raised afterload, is hypothesized to influence hypertrophic cardiomyopathy pathophysiology. OBJECTIVES This study investigated the activation and reversibility of the Anrep effect in obstructive hypertrophic cardiomyopathy (HOCM) patients undergoing percutaneous transluminal septal myocardial ablation (PTSMA) to relieve LV outflow tract obstruction. METHODS Invasive pressure-volume (PV) analysis was performed on 14 HOCM patients before and after PTSMA. The "Anrep Triad," defined by elevated afterload (higher LV end-systolic pressure and effective arterial elastance), augmented contractility (higher end-systolic elastance and maximum LV pressure rise [dP/dtmax]), and prolonged systolic duration (dTes), was assessed via direct hemodynamic comparison preprocedure and postprocedure. Stroke work (SW), potential energy, and total PV area (PVA) quantified mechanical work and efficiency (SW/PVA). RESULTS Postprocedure reversal of the Anrep effect was confirmed (pre- vs post-PTSMA), with reductions in afterload (LV end-systolic pressure: 180 vs 138 mm Hg, P = 0.0001; effective arterial elastance: 2.5 vs 1.9 mm Hg/mL, P = 0.002), contractility (end-systolic elastance: 2.0 vs 1.5 mm Hg/mL, P = 0.0001; dP/dtmax: 1,775 vs 1,560 mm Hg/s, P = 0.017), and systolic duration (dTes: 371 vs 327 ms, P = 0.002). Preprocedure, HOCM patients exhibited higher mechanical workload (SW: 8,161 vs 7,495 mm Hg·mL, P = 0.004; potential energy: 7,837 vs 4,915 mm Hg·mL, P = 0.002; PVA: 16,135 vs 11,742 mm Hg·mL, P = 0.0002) and lower efficiency (SW/PVA: 50% vs 59%, P = 0.03). CONCLUSIONS The Anrep effect is an energy-demanding compensatory mechanism that maintains stroke volume under elevated afterload by increasing contractility and prolonging systole. This study confirms its chronic activation in HOCM and its immediate reversal post-PTSMA.
Collapse
Affiliation(s)
- Jan-Christian Reil
- Klinik für allgemeine und interventionelle Kardiologie, Herz-und Diabetes Zentrum Nordrhein-Westphalen, Bad Oeynhausen, Germany.
| | - Vasco Sequeira
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany.
| | - Gert-Hinrich Reil
- Universitätsklinik für Innere Medizin-Kardiologie, Klinikum Oldenburg, Oldenburg, Germany
| | - Smita Scholtz
- Klinik für allgemeine und interventionelle Kardiologie, Herz-und Diabetes Zentrum Nordrhein-Westphalen, Bad Oeynhausen, Germany
| | - Volker Rudolph
- Klinik für allgemeine und interventionelle Kardiologie, Herz-und Diabetes Zentrum Nordrhein-Westphalen, Bad Oeynhausen, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Patrick Serruys
- CORRIB Research Centre for Advanced Imaging and Core Laboratory, University of Galway, Galway, Ireland
| | - Paul Steendijk
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
4
|
Wang X, Solaro RJ, Dong WJ. Myosin-actin crossbridge independent sarcomere length induced Ca 2+ sensitivity changes in skinned myocardial fibers: Role of myosin heads. J Mol Cell Cardiol 2025; 202:90-101. [PMID: 40073932 DOI: 10.1016/j.yjmcc.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Sarcomere length-dependent activation (LDA) is essential to engaging the Frank-Starling mechanism in the beat-to-beat regulation of cardiac output. Through LDA, the heart increases the Ca2+ sensitivity of myocardial contraction at a longer sarcomere length, leading to an enhanced maximal force at the same level of Ca2+. Despite its importance in both normal and pathological states, the molecular mechanism underlying LDA, especially the origin of the sarcomere length (SL) induced increase in myofilament Ca2+sensitivity, remains elusive. The aim of this study is to interrogate the role of changes in the state of myosin heads during diastole as well as effects of strong force-generating cross-bridges (XB) as determinants of SL-induced Ca2+ sensitivity of troponin in membrane-free (skinned) rat myocardial fibers. Skinned myocardial fibers were reconstituted with troponin complex containing a fluorophore-modified cardiac troponin C, cTnC(13C/51C)AEDANS-DDPM, and recombinant cardiac troponin I (cTnI) mutant, ΔSP-cTnI, in which the switch peptide (Sp) of cTnI was replaced by a non-functional peptide link to partially block the force-generating reaction of myosin with actin. We used the reconstituted myocardial fibers as a platform to investigate how Ca2+ sensitivity of troponin within skinned myocardial fibers responds to sarcomere stretch with variations in the status of myosin-actin XBs. Muscle mechanics and fluorescence measurements clearly showed similar SL-induced increases in troponin Ca2+ sensitivity in either the presence or the absence of strong XBs, suggesting that the SL-induced Ca2+ sensitivity change is independent of reactions of force generating XB with the thin filament. The presence of mavacamten, a selective myosin-motor inhibitor known to promote transition of myosin heads from the weakly actin-bound state (ON or disordered relaxed (DRX) state) to the ordered off state (OFF or super-relaxed (SRX) state), blunted the observed SL-induced increases in Ca2+ sensitivity of troponin regardless of the presence of XBs, suggesting that the presence of the myosin heads in the weakly actin bound state, is essential for Ca2+-troponin to sense the sarcomere stretch. Results from skinned myocardial fibers reconstituted with troponin containing engineered TEV digestible mutant cTnI and cTnT suggest that the observed SL effect on Ca2+ sensitivity may involve potential interactions of weakly bound myosin heads with troponin in the actin/Tm cluster region interacting with cTnT-T1 and residues 182-229 of cTnT-T2. The mechanical stretch effects may then be subsequently transmitted to the N-cTnC via the IT arm of troponin and the N-terminus of cTnI. Our findings strongly indicate that SL-induced potential myosin-troponin interaction in diastole, rather than strong myosin-actin XBs, may be an essential molecular mechanism underlying LDA of myofilament.
Collapse
Affiliation(s)
- Xutu Wang
- Voiland School of Chemical and Bioengineering, Washington State University, Pullman, WA 99163-1062, USA
| | - R John Solaro
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Wen-Ji Dong
- Voiland School of Chemical and Bioengineering, Washington State University, Pullman, WA 99163-1062, USA; Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99163-1062, USA.
| |
Collapse
|
5
|
Wang X, Kallish N, Solaro RJ, Dong WJ. Ca 2+ sensitivity changes in skinned myocardial fibers induced by myosin-actin crossbridge-independent sarcomere stretch: Role of N-domain of MyBP-C. J Mol Cell Cardiol 2025; 202:24-34. [PMID: 40068771 DOI: 10.1016/j.yjmcc.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Sarcomere length-dependent activation (LDA) is the key cellular mechanism underlying the Frank-Starling law of the heart, in which sarcomere stretch leads to increased Ca2+ sensitivity of myofilament and force of contraction. Despite its key role in both normal and pathological states, the precise mechanisms underlying LDA remain unclear but are thought to involve multiple interactions among sarcomere proteins, including troponin of the thin filament, myosin, titin and myosin binding protein C (MyBP-C). Our previous study with permeabilized rat cardiac fibers demonstrated that the mechanism underlying the increase in Ca2+ sensitivity of thin filament induced by sarcomere stretch may involve sarcomere length (SL)-induced interactions between troponin and weakly bound, disordered relaxed state (DRX) myosin heads in diastole, rather than strong myosin-actin crossbridge interactions. In this study we investigated the role of the N-domains of MyBP-C in this newly discovered mechanism. To examine the potential role of the N-domain of MyBP-C in SL-induced myosin-troponin interactions, skinned myocardial fibers from a transgenic ΔN-MyBP-C rat with deleted N-terminal C0-C2 domains and a non-transgenic rat were reconstituted with troponin containing wild-type cTnT, cTnC(13C/51C)AEDANS-DDPM and mutant ΔSP-cTnI or wild-type cTnI. Because the switching peptide (SP) of ΔS-cTnI is replaced by a nonfunctional peptide linker, force-generating actin-myosin crossbridge interactions of the reconstituted skinned fibers with mutant ΔSP-cTnI are inhibited regardless of the presence of Ca2+. This approach allowed us to examine the sensitivity of troponin/thin filament to Ca2+ binding in response to sarcomere stretch by monitoring Ca2+-induced changes in fluorescence resonance energy transfer (FRET) between AEDANS and DDPM attached to the N-domain of cTnC in the presence/absence of myosin-actin crossbridge interaction with or without deletion of C0-C2 domains of MyBP-C. Our measurements of SL-induced changes in muscle fiber mechanics and FRET Ca2+ sensitivities provide strong evidence that both the weakly bound myosin heads and the N-terminus of MyBP-C are critical for SL to activate troponin in the diastolic state. A model based on the results is proposed for the mechanism underlying LDA of myofilament.
Collapse
Affiliation(s)
- Xutu Wang
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99163-1062, USA
| | - Nathan Kallish
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99163-1062, USA
| | - R John Solaro
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Wen-Ji Dong
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99163-1062, USA; Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99163-1062, USA.
| |
Collapse
|
6
|
Chen J, Xing Y, Sun J, Liu Y, Lang Z, Zhang L, Yang J. Hypertrophic Cardiomyopathy: Genes and Mechanisms. FRONT BIOSCI-LANDMRK 2025; 30:25714. [PMID: 40018920 DOI: 10.31083/fbl25714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/28/2024] [Accepted: 09/10/2024] [Indexed: 03/01/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a hereditary disease of the myocardium characterized by asymmetric hypertrophy (mainly the left ventricle) not caused by pressure or volume load. Most cases of HCM are caused by genetic mutations, particularly in the gene encoding cardiac myosin, such as MYH7, TNNT2, and MYBPC3. These mutations are usually inherited autosomal dominantly. Approximately 30-60% of HCM patients have a family history of similar cases among their immediate relatives. This underscores the significance of genetic factors in the development of HCM. Therefore, we summarized the gene mutation mechanisms associated with the onset of HCM and potential treatment directions. We aim to improve patient outcomes by increasing doctors' awareness of genetic counseling, early diagnosis, and identification of asymptomatic patients. Additionally, we offer valuable insights for future research directions, as well as for early diagnosis and intervention.
Collapse
Affiliation(s)
- Jinli Chen
- Department of Geriatric Medicine, First Hospital of Lanzhou University, 730000 Lanzhou, Gansu, China
- Gansu Provincial Clinical Medical Research Center for Geriatric Diseases, 730000 Lanzhou, Gansu, China
| | - Yang Xing
- Department of Anesthesia and Surgery, First Hospital of Lanzhou University, 730000 Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Jie Sun
- Department of Geriatric Medicine, First Hospital of Lanzhou University, 730000 Lanzhou, Gansu, China
- Gansu Provincial Clinical Medical Research Center for Geriatric Diseases, 730000 Lanzhou, Gansu, China
| | - Yongming Liu
- Department of Geriatric Medicine, First Hospital of Lanzhou University, 730000 Lanzhou, Gansu, China
- Gansu Provincial Clinical Medical Research Center for Geriatric Diseases, 730000 Lanzhou, Gansu, China
| | - Zekun Lang
- The First Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Lei Zhang
- The First Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Jinggang Yang
- Department of Geriatric Medicine, First Hospital of Lanzhou University, 730000 Lanzhou, Gansu, China
- Gansu Provincial Clinical Medical Research Center for Geriatric Diseases, 730000 Lanzhou, Gansu, China
| |
Collapse
|
7
|
Halder SS, Rynkiewicz MJ, Kim L, Barry ME, Zied AG, Sewanan LR, Kirk JA, Moore JR, Lehman WJ, Campbell SG. Distinct mechanisms drive divergent phenotypes in hypertrophic and dilated cardiomyopathy-associated TPM1 variants. J Clin Invest 2024; 134:e179135. [PMID: 39436707 PMCID: PMC11645150 DOI: 10.1172/jci179135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
Heritable forms of hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) represent starkly diverging clinical phenotypes, yet may be caused by mutations to the same sarcomeric protein. The precise mechanisms by which point mutations within the same gene bring about phenotypic diversity remain unclear. Our objective was to develop a mechanistic explanation of diverging phenotypes in two TPM1 mutations, E62Q (HCM) and E54K (DCM). Drawing on data from the literature and experiments with stem cell-derived cardiomyocytes expressing the TPM1 mutations of interest, we constructed computational simulations that provide plausible explanations of the distinct muscle contractility caused by each variant. In E62Q, increased calcium sensitivity and hypercontractility was explained most accurately by a reduction in effective molecular stiffness of tropomyosin and alterations in its interactions with the actin thin filament that favor the "closed" regulatory state. By contrast, the E54K mutation appeared to act via long-range allosteric interactions to increase the association rate of the C-terminal troponin I mobile domain to tropomyosin/actin. These mutation-linked molecular events produced diverging alterations in gene expression that can be observed in human engineered heart tissues. Modulators of myosin activity confirmed our proposed mechanisms by rescuing normal contractile behavior in accordance with predictions.
Collapse
Affiliation(s)
- Saiti S. Halder
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Michael J. Rynkiewicz
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Lynne Kim
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Meaghan E. Barry
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Ahmed G.A. Zied
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois, USA
| | - Lorenzo R. Sewanan
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Jonathan A. Kirk
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Chicago, Illinois, USA
| | - Jeffrey R. Moore
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - William J. Lehman
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Stuart G. Campbell
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
8
|
Federspiel JM, Pfeifer J, Ramsthaler F, Reil JC, Schmidt PH, Sequeira V. Definitional Challenges in Understanding Hypertrophic Cardiomyopathy. Diagnostics (Basel) 2024; 14:2534. [PMID: 39594199 PMCID: PMC11592529 DOI: 10.3390/diagnostics14222534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common hereditary cardiomyopathy. It is often caused by mutations of genes encoding for sarcomeric or sarcomere-associated proteins. Despite its clinical importance, divergent definitions are published by major cardiology societies. Some regard HCM as a specific genetic disease, whereas others define it as a broad 'spectrum of the thick heart'. The present narrative review aimed to assess both definitions from a pathoanatomical perspective. As a conjoint interdisciplinary and translational approach is needed to further increase knowledge and improve the understanding of HCM, the PubMed database was searched using several advanced search algorithms to explore the perspectives of the (forensic) pathologist, clinician, and basic researcher regarding the difference between the definitions of HCM. This discrepancy between definitions can impact critical data, such as prevalence and mortality rate, and complicate the understanding of the disease. For example, due to the different definitions, research findings regarding molecular changes from studies applying the narrow definition cannot be simply extended to the 'spectrum' of HCM.
Collapse
Affiliation(s)
- Jan M. Federspiel
- Institute for Legal Medicine, Faculty of Medicine, Saarland University, Campus Homburg, Building 49.1, Kirrberger Straße 100, 66421 Homburg/Saar, Germany
| | - Jochen Pfeifer
- Department for Pediatric Cardiology, Saarland University Medical Centre, Building 9, Kirrberger Straße 100, 66421 Homburg/Saar, Germany
| | - Frank Ramsthaler
- Institute for Legal Medicine, Faculty of Medicine, Saarland University, Campus Homburg, Building 49.1, Kirrberger Straße 100, 66421 Homburg/Saar, Germany
| | - Jan-Christian Reil
- Department of General and Interventional Cardiology, Heart and Diabetes Centre North Rhine-Westphalia, Ruhr University Bochum, 32545 Bad Oeynhausen, Germany
| | - Peter H. Schmidt
- Institute for Legal Medicine, Faculty of Medicine, Saarland University, Campus Homburg, Building 49.1, Kirrberger Straße 100, 66421 Homburg/Saar, Germany
| | - Vasco Sequeira
- Department for Translational Research, Congestive Heart Failure Centre, University Clinic Wuerzburg, Building A15, Am Schwarzberg 15, 97078 Wuerzburg, Germany;
| |
Collapse
|
9
|
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
|
10
|
Mertens J, De Lange WJ, Farrell ET, Harbaugh EC, Gauchan A, Fitzsimons DP, Moss RL, Ralphe JC. The W792R HCM missense mutation in the C6 domain of cardiac myosin binding protein-C increases contractility in neonatal mouse myocardium. J Mol Cell Cardiol 2024; 195:14-23. [PMID: 39059462 DOI: 10.1016/j.yjmcc.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Missense mutations in cardiac myosin binding protein C (cMyBP-C) are known to cause hypertrophic cardiomyopathy (HCM). The W792R mutation in the C6 domain of cMyBP-C causes severe, early onset HCM in humans, yet its impact on the function of cMyBP-C and the mechanism through which it causes disease remain unknown. To fully characterize the effect of the W792R mutation on cardiac morphology and function in vivo, we generated a murine knock-in model. We crossed heterozygous W792RWR mice to produce homozygous mutant W792RRR, heterozygous W792RWR, and control W792RWW mice. W792RRR mice present with cardiac hypertrophy, myofibrillar disarray and fibrosis by postnatal day 10 (PND10), and do not survive past PND21. Full-length cMyBP-C is present at similar levels in W792RWW, W792RWR and W792RRR mice and is properly incorporated into the sarcomere. Heterozygous W792RWR mice displayed normal heart morphology and contractility. Permeabilized myocardium from PND10 W792RRR mice showed increased Ca2+ sensitivity, accelerated cross-bridge cycling kinetics, decreased cooperativity in the activation of force, and increased expression of hypertrophy-related genes. In silico modeling suggests that the W792R mutation destabilizes the fold of the C6 domain and increases torsion in the C5-C7 region, possibly impacting regulatory interactions of cMyBP-C with myosin and actin. Based on the data presented here, we propose a model in which mutant W792R cMyBP-C preferentially forms Ca2+ sensitizing interactions with actin, rather than inhibitory interactions with myosin. The W792R-cMyBP-C mouse model provides mechanistic insights into the pathology of this mutation and may provide a mechanism by which other central domain missense mutations in cMyBP-C may alter contractility, leading to HCM.
Collapse
Affiliation(s)
- Jasmine Mertens
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America; UW Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America
| | - Willem J De Lange
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America; UW Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America
| | - Emily T Farrell
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America; UW Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America
| | - Ella C Harbaugh
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America; UW Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America
| | - Angeela Gauchan
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America; UW Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America
| | - Daniel P Fitzsimons
- UW Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America
| | - Richard L Moss
- UW Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America
| | - J Carter Ralphe
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America; UW Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, United States of America.
| |
Collapse
|
11
|
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
|
12
|
Federspiel JM, Reil JC, Xu A, Scholtz S, Batzner A, Maack C, Sequeira V. Retrofitting the Heart: Explaining the Enigmatic Septal Thickening in Hypertrophic Cardiomyopathy. Circ Heart Fail 2024; 17:e011435. [PMID: 38695186 DOI: 10.1161/circheartfailure.123.011435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/26/2024] [Indexed: 05/23/2024]
Abstract
Hypertrophic cardiomyopathy is the most common genetic cardiac disease and is characterized by left ventricular hypertrophy. Although this hypertrophy often associates with sarcomeric gene mutations, nongenetic factors also contribute to the disease, leading to diastolic dysfunction. Notably, this dysfunction manifests before hypertrophy and is linked to hypercontractility, as well as nonuniform contraction and relaxation (myofibril asynchrony) of the myocardium. Although the distribution of hypertrophy in hypertrophic cardiomyopathy can vary both between and within individuals, in most cases, it is primarily confined to the interventricular septum. The reasons for septal thickening remain largely unknown. In this article, we propose that alterations in muscle fiber geometry, present from birth, dictate the septal shape. When combined with hypercontractility and exacerbated by left ventricular outflow tract obstruction, these factors predispose the septum to an isometric type of contraction during systole, consequently constraining its mobility. This contraction, or more accurately, this focal increase in biomechanical stress, prompts the septum to adapt and undergo remodeling. Drawing a parallel, this is reminiscent of how earthquake-resistant buildings are retrofitted with vibration dampers to absorb the majority of the shock motion and load. Similarly, the heart adapts by synthesizing viscoelastic elements such as microtubules, titin, desmin, collagen, and intercalated disc components. This pronounced remodeling in the cytoskeletal structure leads to noticeable septal hypertrophy. This structural adaptation acts as a protective measure against damage by attenuating myofibril shortening while reducing cavity tension according to Laplace Law. By examining these events, we provide a coherent explanation for the septum's predisposition toward hypertrophy.
Collapse
Affiliation(s)
- Jan M Federspiel
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
- Saarland University, Faculty of Medicine, Institute for Legal Medicine, Homburg (Saar), Germany (J.M.F.)
| | - Jan-Christian Reil
- Klinik für allgemeine und interventionelle Kardiologie, Herz- und Diabetes-Zentrum Nordrhein-Westphalen, Germany (J.-C.R., S.S.)
| | - Anton Xu
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
| | - Smita Scholtz
- Klinik für allgemeine und interventionelle Kardiologie, Herz- und Diabetes-Zentrum Nordrhein-Westphalen, Germany (J.-C.R., S.S.)
| | - Angelika Batzner
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
- Department of Internal Medicine I, University Hospital Würzburg, Germany (A.B.)
| | - Christoph Maack
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
| | - Vasco Sequeira
- Comprehensive Heart Failure Center, Department of Translational Science University Clinic Würzburg, Germany (J.M.F., A.X., A.B., C.M., V.S.)
| |
Collapse
|
13
|
Wijnker PJM, Dinani R, van der Laan NC, Algül S, Knollmann BC, Verkerk AO, Remme CA, Zuurbier CJ, Kuster DWD, van der Velden J. Hypertrophic cardiomyopathy dysfunction mimicked in human engineered heart tissue and improved by sodium-glucose cotransporter 2 inhibitors. Cardiovasc Res 2024; 120:301-317. [PMID: 38240646 PMCID: PMC10939456 DOI: 10.1093/cvr/cvae004] [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: 12/01/2022] [Revised: 11/15/2023] [Accepted: 11/29/2023] [Indexed: 03/16/2024] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiomyopathy, often caused by pathogenic sarcomere mutations. Early characteristics of HCM are diastolic dysfunction and hypercontractility. Treatment to prevent mutation-induced cardiac dysfunction is lacking. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) are a group of antidiabetic drugs that recently showed beneficial cardiovascular outcomes in patients with acquired forms of heart failure. We here studied if SGLT2i represent a potential therapy to correct cardiomyocyte dysfunction induced by an HCM sarcomere mutation. METHODS AND RESULTS Contractility was measured of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) harbouring an HCM mutation cultured in 2D and in 3D engineered heart tissue (EHT). Mutations in the gene encoding β-myosin heavy chain (MYH7-R403Q) or cardiac troponin T (TNNT2-R92Q) were investigated. In 2D, intracellular [Ca2+], action potential and ion currents were determined. HCM mutations in hiPSC-CMs impaired relaxation or increased force, mimicking early features observed in human HCM. SGLT2i enhance the relaxation of hiPSC-CMs, to a larger extent in HCM compared to control hiPSC-CMs. Moreover, SGLT2i-effects on relaxation in R403Q EHT increased with culture duration, i.e. hiPSC-CMs maturation. Canagliflozin's effects on relaxation were more pronounced than empagliflozin and dapagliflozin. SGLT2i acutely altered Ca2+ handling in HCM hiPSC-CMs. Analyses of SGLT2i-mediated mechanisms that may underlie enhanced relaxation in mutant hiPSC-CMs excluded SGLT2, Na+/H+ exchanger, peak and late Nav1.5 currents, and L-type Ca2+ current, but indicate an important role for the Na+/Ca2+ exchanger. Indeed, electrophysiological measurements in mutant hiPSC-CM indicate that SGLT2i altered Na+/Ca2+ exchange current. CONCLUSION SGLT2i (canagliflozin > dapagliflozin > empagliflozin) acutely enhance relaxation in human EHT, especially in HCM and upon prolonged culture. SGLT2i may represent a potential therapy to correct early cardiac dysfunction in HCM.
Collapse
Affiliation(s)
- Paul J M Wijnker
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rafeeh Dinani
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Nico C van der Laan
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Sila Algül
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Bjorn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Arie O Verkerk
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Experimental Cardiology, Amsterdam UMC, Academic Medical Centre, Amsterdam, The Netherlands
| | - Carol Ann Remme
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Experimental Cardiology, Amsterdam UMC, Academic Medical Centre, Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam UMC, Academic Medical Centre, Amsterdam, The Netherlands
| | - Diederik W D Kuster
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
14
|
Abstract
Force generation in striated muscle is primarily controlled by structural changes in the actin-containing thin filaments triggered by an increase in intracellular calcium concentration. However, recent studies have elucidated a new class of regulatory mechanisms, based on the myosin-containing thick filament, that control the strength and speed of contraction by modulating the availability of myosin motors for the interaction with actin. This review summarizes the mechanisms of thin and thick filament activation that regulate the contractility of skeletal and cardiac muscle. A novel dual-filament paradigm of muscle regulation is emerging, in which the dynamics of force generation depends on the coordinated activation of thin and thick filaments. We highlight the interfilament signaling pathways based on titin and myosin-binding protein-C that couple thin and thick filament regulatory mechanisms. This dual-filament regulation mediates the length-dependent activation of cardiac muscle that underlies the control of the cardiac output in each heartbeat.
Collapse
Affiliation(s)
- Elisabetta Brunello
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom; ,
| | - Luca Fusi
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom; ,
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| |
Collapse
|
15
|
Abstract
The Anrep effect is an adaptive response that increases left ventricular contractility following an acute rise in afterload. Although the mechanistic origin remains undefined, recent findings suggest a two-phase activation of resting myosin for contraction, involving strain-sensitive and posttranslational phases. We propose that this mobilization represents a transition among the relaxed states of myosin-specifically, from the super-relaxed (SRX) to the disordered-relaxed (DRX)-with DRX myosin ready to participate in force generation. This hypothesis offers a unified explanation that connects myosin's SRX-DRX equilibrium and the Anrep effect as parts of a singular phenomenon. We underscore the significance of this equilibrium in modulating contractility, primarily studied in the context of hypertrophic cardiomyopathy, the most common inherited cardiomyopathy associated with diastolic dysfunction, hypercontractility, and left ventricular hypertrophy. As we posit that the cellular basis of the Anrep effect relies on a two-phased transition of myosin from the SRX to the contraction-ready DRX configuration, any dysregulation in this equilibrium may result in the pathological manifestation of the Anrep phenomenon. For instance, in hypertrophic cardiomyopathy, hypercontractility is linked to a considerable shift of myosin to the DRX state, implying a persistent activation of the Anrep effect. These valuable insights call for additional research to uncover a clinical Anrep fingerprint in pathological states. Here, we demonstrate through noninvasive echocardiographic pressure-volume measurements that this fingerprint is evident in 12 patients with hypertrophic obstructive cardiomyopathy before septal myocardial ablation. This unique signature is characterized by enhanced contractility, indicated by a leftward shift and steepening of the end-systolic pressure-volume relationship, and a prolonged systolic ejection time adjusted for heart rate, which reverses post-procedure. The clinical application of this concept has potential implications beyond hypertrophic cardiomyopathy, extending to other genetic cardiomyopathies and even noncongenital heart diseases with complex etiologies across a broad spectrum of left ventricular ejection fractions.
Collapse
Affiliation(s)
- Vasco Sequeira
- Department of Translational Science Universitätsklinikum, Deutsche Zentrum für Herzinsuffizienz (DZHI), Würzburg, Germany (V.S., C.M.)
| | - Christoph Maack
- Department of Translational Science Universitätsklinikum, Deutsche Zentrum für Herzinsuffizienz (DZHI), Würzburg, Germany (V.S., C.M.)
| | - Gert-Hinrich Reil
- Klinik für Kardiologie, Klinikum Oldenburg, Innere Medizin I, Germany (G.-H.R.)
| | - Jan-Christian Reil
- Klinik für Allgemeine und Interventionelle Kardiologie, Herz- und Diabetes-Zentrum Nordrhein-Westphalen, Germany (J.-C.R.)
| |
Collapse
|
16
|
Tamargo M, Martínez-Legazpi P, Espinosa MÁ, Lyon A, Méndez I, Gutiérrez-Ibañes E, Fernández AI, Prieto-Arévalo R, González-Mansilla A, Arts T, Delhaas T, Mombiela T, Sanz-Ruiz R, Elízaga J, Yotti R, Tschöpe C, Fernández-Avilés F, Lumens J, Bermejo J. Increased Chamber Resting Tone Is a Key Determinant of Left Ventricular Diastolic Dysfunction. Circ Heart Fail 2023; 16:e010673. [PMID: 38113298 PMCID: PMC10729900 DOI: 10.1161/circheartfailure.123.010673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/22/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Twitch-independent tension has been demonstrated in cardiomyocytes, but its role in heart failure (HF) is unclear. We aimed to address twitch-independent tension as a source of diastolic dysfunction by isolating the effects of chamber resting tone (RT) from impaired relaxation and stiffness. METHODS We invasively monitored pressure-volume data during cardiopulmonary exercise in 20 patients with hypertrophic cardiomyopathy, 17 control subjects, and 35 patients with HF with preserved ejection fraction. To measure RT, we developed a new method to fit continuous pressure-volume measurements, and first validated it in a computational model of loss of cMyBP-C (myosin binding protein-C). RESULTS In hypertrophic cardiomyopathy, RT (estimated marginal mean [95% CI]) was 3.4 (0.4-6.4) mm Hg, increasing to 18.5 (15.5-21.5) mm Hg with exercise (P<0.001). At peak exercise, RT was responsible for 64% (53%-76%) of end-diastolic pressure, whereas incomplete relaxation and stiffness accounted for the rest. RT correlated with the levels of NT-proBNP (N-terminal pro-B-type natriuretic peptide; R=0.57; P=0.02) and with pulmonary wedge pressure but following different slopes at rest and during exercise (R2=0.49; P<0.001). In controls, RT was 0.0 mm Hg and 1.2 (0.3-2.8) mm Hg in HF with preserved ejection fraction patients and was also exacerbated by exercise. In silico, RT increased in parallel to the loss of cMyBP-C function and correlated with twitch-independent myofilament tension (R=0.997). CONCLUSIONS Augmented RT is the major cause of LV diastolic chamber dysfunction in hypertrophic cardiomyopathy and HF with preserved ejection fraction. RT transients determine diastolic pressures, pulmonary pressures, and functional capacity to a greater extent than relaxation and stiffness abnormalities. These findings support antimyosin agents for treating HF.
Collapse
Affiliation(s)
- María Tamargo
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Pablo Martínez-Legazpi
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
- Department of Mathematical Physics and Fluids, Facultad de Ciencias, Universidad Nacional de Educación a Distancia, UNED, Spain (P.M.-L.)
| | - M. Ángeles Espinosa
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Aurore Lyon
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (A.L., T.A., T.D., J.L.)
| | - Irene Méndez
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Enrique Gutiérrez-Ibañes
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Ana I. Fernández
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Raquel Prieto-Arévalo
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Ana González-Mansilla
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Theo Arts
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (A.L., T.A., T.D., J.L.)
| | - Tammo Delhaas
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (A.L., T.A., T.D., J.L.)
| | - Teresa Mombiela
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Ricardo Sanz-Ruiz
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Jaime Elízaga
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Raquel Yotti
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Carsten Tschöpe
- Berlin Institute of Health/Center for Regenerative Therapy (BCRT) at Charite, and Department of Cardiology, Campus Virchow (CVK), Charité Universitätsmedizin, and DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany (C.T.)
| | - Francisco Fernández-Avilés
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| | - Joost Lumens
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (A.L., T.A., T.D., J.L.)
| | - Javier Bermejo
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, and CIBERCV, Spain (M.T., P.M.-L., M.A.E., I.M., E.G.-I., A.I.F., R.P.-A., A.G.-M., T.M., R.S.-R., J.E., R.Y., F.F.-A., J.B.)
| |
Collapse
|
17
|
Hilderink S, Schuldt M, Goebel M, Jansen VJ, Manders E, Moorman S, Dorsch LM, van Steenbeek FG, van der Velden J, Kuster DWD. Characterization of heterozygous and homozygous mouse models with the most common hypertrophic cardiomyopathy mutation MYBPC3 c.2373InsG in the Netherlands. J Mol Cell Cardiol 2023; 185:65-76. [PMID: 37844837 DOI: 10.1016/j.yjmcc.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is frequently caused by mutations in the cardiac myosin binding protein-C (cMyBP-C) encoding gene MYBPC3. In the Netherlands, approximately 25% of patients carry the MYBPC3c.2373InsG founder mutation. Most patients are heterozygous (MYBPC3+/InsG) and have highly variable phenotypic expression, whereas homozygous (MYBPC3InsG/InsG) patients have severe HCM at a young age. To improve understanding of disease progression and genotype-phenotype relationship based on the hallmarks of human HCM, we characterized mice with CRISPR/Cas9-induced heterozygous and homozygous mutations. At 18-28 weeks of age, we assessed the cardiac phenotype of Mybpc3+/InsG and Mybpc3InsG/InsG mice with echocardiography, and performed histological analyses. Cytoskeletal proteins and cardiomyocyte contractility of 3-4 week old and 18-28 week old Mybpc3c.2373InsG mice were compared to wild-type (WT) mice. Expectedly, knock-in of Mybpc3c.2373InsG resulted in the absence of cMyBP-C and our 18-28 week old homozygous Mybpc3c.2373InsG model developed cardiac hypertrophy and severe left ventricular systolic and diastolic dysfunction, whereas HCM was not evident in Mybpc3+/InsG mice. Mybpc3InsG/InsG cardiomyocytes also presented with slowed contraction-relaxation kinetics, to a greater extent in 18-28 week old mice, partially due to increased levels of detyrosinated tubulin and desmin, and reduced cardiac troponin I (cTnI) phosphorylation. Impaired cardiomyocyte contraction-relaxation kinetics were successfully normalized in 18-28 week old Mybpc3InsG/InsG cardiomyocytes by combining detyrosination inhibitor parthenolide and β-adrenergic receptor agonist isoproterenol. Both the 3-4 week old and 18-28 week old Mybpc3InsG/InsG models recapitulate HCM, with a severe phenotype present in the 18-28 week old model.
Collapse
Affiliation(s)
- Sarah Hilderink
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Maike Schuldt
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Max Goebel
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Valentijn J Jansen
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Emmy Manders
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands
| | - Stan Moorman
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Larissa M Dorsch
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Frank G van Steenbeek
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands; Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, the Netherlands
| | - Jolanda van der Velden
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Diederik W D Kuster
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1118, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands.
| |
Collapse
|
18
|
Seo K, Yamamoto Y, Kirillova A, Kawana M, Yadav S, Huang Y, Wang Q, Lane KV, Pruitt BL, Perez MV, Bernstein D, Wu JC, Wheeler MT, Parikh VN, Ashley EA. Improved Cardiac Performance and Decreased Arrhythmia in Hypertrophic Cardiomyopathy With Non-β-Blocking R-Enantiomer Carvedilol. Circulation 2023; 148:1691-1704. [PMID: 37850394 DOI: 10.1161/circulationaha.123.065017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/05/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Hypercontractility and arrhythmia are key pathophysiologic features of hypertrophic cardiomyopathy (HCM), the most common inherited heart disease. β-Adrenergic receptor antagonists (β-blockers) are the first-line therapy for HCM. However, β-blockers commonly selected for this disease are often poorly tolerated in patients, where heart-rate reduction and noncardiac effects can lead to reduced cardiac output and fatigue. Mavacamten, myosin ATPase inhibitor recently approved by the US Food and Drug Administration, has demonstrated the ability to ameliorate hypercontractility without lowering heart rate, but its benefits are so far limited to patients with left ventricular (LV) outflow tract obstruction, and its effect on arrhythmia is unknown. METHODS We screened 21 β-blockers for their impact on myocyte contractility and evaluated the antiarrhythmic properties of the most promising drug in a ventricular myocyte arrhythmia model. We then examined its in vivo effect on LV function by hemodynamic pressure-volume loop analysis. The efficacy of the drug was tested in vitro and in vivo compared with current therapeutic options (metoprolol, verapamil, and mavacamten) for HCM in an established mouse model of HCM (Myh6R403Q/+ and induced pluripotent stem cell (iPSC)-derived cardiomyocytes from patients with HCM (MYH7R403Q/+). RESULTS We identified that carvedilol, a β-blocker not commonly used in HCM, suppresses contractile function and arrhythmia by inhibiting RyR2 (ryanodine receptor type 2). Unlike metoprolol (a β1-blocker), carvedilol markedly reduced LV contractility through RyR2 inhibition, while maintaining stroke volume through α1-adrenergic receptor inhibition in vivo. Clinically available carvedilol is a racemic mixture, and the R-enantiomer, devoid of β-blocking effect, retains the ability to inhibit both α1-receptor and RyR2, thereby suppressing contractile function and arrhythmias without lowering heart rate and cardiac output. In Myh6R403Q/+ mice, R-carvedilol normalized hyperdynamic contraction, suppressed arrhythmia, and increased cardiac output better than metoprolol, verapamil, and mavacamten. The ability of R-carvedilol to suppress contractile function was well retained in MYH7R403Q/+ iPSC-derived cardiomyocytes. CONCLUSIONS R-enantiomer carvedilol attenuates hyperdynamic contraction, suppresses arrhythmia, and at the same time, improves cardiac output without lowering heart rate by dual blockade of α1-adrenergic receptor and RyR2 in mouse and human models of HCM. This combination of therapeutic effects is unique among current therapeutic options for HCM and may particularly benefit patients without LV outflow tract obstruction.
Collapse
Affiliation(s)
- Kinya Seo
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Yuta Yamamoto
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Anna Kirillova
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Masataka Kawana
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Sunil Yadav
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Yong Huang
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Qianru Wang
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Kerry V Lane
- Departments of Mechanical Engineering (K.V.L., B.L.P.), University of California, Santa Barbara, CA
| | - Beth L Pruitt
- Departments of Mechanical Engineering (K.V.L., B.L.P.), University of California, Santa Barbara, CA
- BioMolecular Science and Engineering (B.L.P.), University of California, Santa Barbara, CA
| | - Marco V Perez
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | | | - Joseph C Wu
- Cardiovascular Research Institute (J.C.W.), Stanford University School of Medicine, CA
| | - Matthew T Wheeler
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Victoria N Parikh
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
| | - Euan A Ashley
- From the Departments of Medicine (K.S., Y.Y., A.K., M.K., S.Y., Y.H., Q.W., M.V.P., M.T.W., V.N.P., E.A.A.), Stanford University School of Medicine, CA
- Genetics (E.A.A.), Stanford University School of Medicine, CA
| |
Collapse
|
19
|
Vaniya A, Karlstaedt A, Gulkok DA, Thottakara T, Liu Y, Fan S, Eades H, Fukunaga R, Vernon HJ, Fiehn O, Roselle Abraham M. Lipid metabolism drives allele-specific early-stage hypertrophic cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.564562. [PMID: 38014251 PMCID: PMC10680657 DOI: 10.1101/2023.11.10.564562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) results from pathogenic variants in sarcomeric protein genes, that increase myocyte energy demand and lead to cardiac hypertrophy. But it is unknown whether a common metabolic trait underlies the cardiac phenotype at early disease stage. This study characterized two HCM mouse models (R92W-TnT, R403Q-MyHC) that demonstrate differences in mitochondrial function at early disease stage. Using a combination of cardiac phenotyping, transcriptomics, mass spectrometry-based metabolomics and computational modeling, we discovered allele-specific differences in cardiac structure/function and metabolic changes. TnT-mutant hearts had impaired energy substrate metabolism and increased phospholipid remodeling compared to MyHC-mutants. TnT-mutants showed increased incorporation of saturated fatty acid residues into ceramides, cardiolipin, and increased lipid peroxidation, that could underlie allele-specific differences in mitochondrial function and cardiomyopathy.
Collapse
|
20
|
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
|
21
|
Landim-Vieira M, Ma W, Song T, Rastegarpouyani H, Gong H, Coscarella IL, Bogaards SJP, Conijn SP, Ottenheijm CAC, Hwang HS, Papadaki M, Knollmann BC, Sadayappan S, Irving TC, Galkin VE, Chase PB, Pinto JR. Cardiac troponin T N-domain variant destabilizes the actin interface resulting in disturbed myofilament function. Proc Natl Acad Sci U S A 2023; 120:e2221244120. [PMID: 37252999 PMCID: PMC10265946 DOI: 10.1073/pnas.2221244120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Missense variant Ile79Asn in human cardiac troponin T (cTnT-I79N) has been associated with hypertrophic cardiomyopathy and sudden cardiac arrest in juveniles. cTnT-I79N is located in the cTnT N-terminal (TnT1) loop region and is known for its pathological and prognostic relevance. A recent structural study revealed that I79 is part of a hydrophobic interface between the TnT1 loop and actin, which stabilizes the relaxed (OFF) state of the cardiac thin filament. Given the importance of understanding the role of TnT1 loop region in Ca2+ regulation of the cardiac thin filament along with the underlying mechanisms of cTnT-I79N-linked pathogenesis, we investigated the effects of cTnT-I79N on cardiac myofilament function. Transgenic I79N (Tg-I79N) muscle bundles displayed increased myofilament Ca2+ sensitivity, smaller myofilament lattice spacing, and slower crossbridge kinetics. These findings can be attributed to destabilization of the cardiac thin filament's relaxed state resulting in an increased number of crossbridges during Ca2+ activation. Additionally, in the low Ca2+-relaxed state (pCa8), we showed that more myosin heads are in the disordered-relaxed state (DRX) that are more likely to interact with actin in cTnT-I79N muscle bundles. Dysregulation of the myosin super-relaxed state (SRX) and the SRX/DRX equilibrium in cTnT-I79N muscle bundles likely result in increased mobility of myosin heads at pCa8, enhanced actomyosin interactions as evidenced by increased active force at low Ca2+, and increased sinusoidal stiffness. These findings point to a mechanism whereby cTnT-I79N weakens the interaction of the TnT1 loop with the actin filament, which in turn destabilizes the relaxed state of the cardiac thin filament.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL32306
| | - Weikang Ma
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH45267
| | - Hosna Rastegarpouyani
- Department of Biological Science, Florida State University, Tallahassee, FL32306
- Institude of Molecular Biophysics, Florida State University, Tallahassee, FL32306
| | - Henry Gong
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Isabella Leite Coscarella
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL32306
| | - Sylvia J. P. Bogaards
- Department of Physiology, Amsterdam University Medical Center, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Stefan P. Conijn
- Department of Physiology, Amsterdam University Medical Center, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Coen A. C. Ottenheijm
- Department of Physiology, Amsterdam University Medical Center, Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Hyun S. Hwang
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL32306
| | - Maria Papadaki
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Chicago, IL60153
| | - Bjorn C. Knollmann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN37232
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH45267
| | - Thomas C. Irving
- Department of Biology, Illinois Institute of Technology, Chicago, IL60616
| | - Vitold E. Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA23507
| | - P. Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL32306
| |
Collapse
|
22
|
Sequeira V, Waddingham MT, Tsuchimochi H, Maack C, Pearson JT. Mechano-energetic uncoupling in hypertrophic cardiomyopathy: Pathophysiological mechanisms and therapeutic opportunities. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 4:100036. [PMID: 39801694 PMCID: PMC11708264 DOI: 10.1016/j.jmccpl.2023.100036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 01/16/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a frequent inherited form of heart failure. The underlying cause of HCM is generally attributed to mutations in genes that encode for sarcomeric proteins, but the pathogenesis of the disease is also influenced by non-genetic factors, which can contribute to diastolic dysfunction and hypertrophic remodeling. Central to the pathogenesis of HCM is hypercontractility, a state that is an antecedent to several key derangements, including increased mitochondrial workload and oxidative stress. As a result, energy depletion and mechano-energetic uncoupling drive cardiac growth through signaling pathways such as ERK and/or potentially AMPK downregulation. Metabolic remodeling also occurs in HCM, characterized by decreased fatty acid oxidation and increased glucose uptake. In some instances, ketones may also feed the heart with energy and act as signaling molecules to reduce oxidative stress and hypertrophic signaling. In addition, arrhythmias are frequently triggered in HCM, resulting from the high Ca2+-buffering of the myofilaments and changes in the ATP/ADP ratio. Understanding the mechanisms driving the progression of HCM is critical to the development of effective therapeutic strategies. This paper presents evidence from both experimental and clinical studies that support the role of hypercontractility and cellular energy alterations in the progression of HCM towards heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Vasco Sequeira
- DZHI, Department of Translational Science Universitätsklinikum, Würzburg, Germany
| | - Mark T. Waddingham
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Christoph Maack
- DZHI, Department of Translational Science Universitätsklinikum, Würzburg, Germany
| | - James T. Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
- Department of Physiology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
23
|
Valtonen J, Prajapati C, Cherian RM, Vanninen S, Ojala M, Leivo K, Heliö T, Koskenvuo J, Aalto-Setälä K. The Junctophilin-2 Mutation p.(Thr161Lys) Is Associated with Hypertrophic Cardiomyopathy Using Patient-Specific iPS Cardiomyocytes and Demonstrates Prolonged Action Potential and Increased Arrhythmogenicity. Biomedicines 2023; 11:1558. [PMID: 37371654 DOI: 10.3390/biomedicines11061558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is one of the most common genetic cardiac diseases; it is primarily caused by mutations in sarcomeric genes. However, HCM is also associated with mutations in non-sarcomeric proteins and a Finnish founder mutation for HCM in non-sarcomeric protein junctophilin-2 (JPH2) has been identified. This study aimed at assessing the issue of modelling the rare Finnish founder mutation in cardiomyocytes (CMs) differentiated from iPSCs; therefore, presenting the same cardiac abnormalities observed in the patients. To explore the abnormal functions in JPH2-HCM, skin fibroblasts from a Finnish patient with JPH2 p.(Thr161Lys) were reprogrammed into iPSCs and further differentiated into CMs. As a control line, an isogenic counterpart was generated using the CRISPR/Cas9 genome editing method. Finally, iPSC-CMs were evaluated for the morphological and functional characteristics associated with JPH2 mutation. JPH2-hiPSC-CMs displayed key HCM hallmarks (cellular hypertrophy, multi-nucleation, sarcomeric disarray). Moreover, JPH2-hiPSC-CMs exhibit a higher degree of arrhythmia and longer action potential duration associated with slower inactivation of calcium channels. Functional evaluation supported clinical observations, with differences in beating characteristics when compared with isogenic-hiPSC-CMs. Thus, the iPSC-derived, disease-specific cardiomyocytes could serve as a translationally relevant platform to study genetic cardiac diseases.
Collapse
Affiliation(s)
- Joona Valtonen
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Chandra Prajapati
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Reeja Maria Cherian
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Sari Vanninen
- Tampere University Heart Hospital, 33520 Tampere, Finland
| | - Marisa Ojala
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Krista Leivo
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, 00290 Helsinki, Finland
| | - Tiina Heliö
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, 00290 Helsinki, Finland
| | | | - Katriina Aalto-Setälä
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
- Tampere University Heart Hospital, 33520 Tampere, Finland
| |
Collapse
|
24
|
Hanft LM, Robinett JC, Kalogeris TJ, Campbell KS, Biesiadecki BJ, McDonald KS. Thin filament regulation of cardiac muscle power output: Implications for targets to improve human failing hearts. J Gen Physiol 2023; 155:e202213290. [PMID: 37000170 PMCID: PMC10067705 DOI: 10.1085/jgp.202213290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/14/2023] [Accepted: 03/13/2023] [Indexed: 04/01/2023] Open
Abstract
The heart's pumping capacity is determined by myofilament power generation. Power is work done per unit time and measured as the product of force and velocity. At a sarcomere level, these contractile properties are linked to the number of attached cross-bridges and their cycling rate, and many signaling pathways modulate one or both factors. We previously showed that power is increased in rodent permeabilized cardiac myocytes following PKA-mediated phosphorylation of myofibrillar proteins. The current study found that that PKA increased power by ∼30% in permeabilized cardiac myocyte preparations (n = 8) from human failing hearts. To address myofilament molecular specificity of PKA effects, mechanical properties were measured in rat permeabilized slow-twitch skeletal muscle fibers before and after exchange of endogenous slow skeletal troponin with recombinant human Tn complex that contains cardiac (c)TnT, cTnC and either wildtype (WT) cTnI or pseudo-phosphorylated cTnI at sites Ser23/24Asp, Tyr26Glu, or the combinatorial Ser23/24Asp and Tyr26Glu. We found that cTnI Ser23/24Asp, Tyr26Glu, and combinatorial Ser23/24Asp and Tyr26Glu were sufficient to increase power by ∼20%. Next, we determined whether pseudo-phosphorylated cTnI at Ser23/24 was sufficient to increase power in cardiac myocytes from human failing hearts. Following cTn exchange that included cTnI Ser23/24Asp, power output increased ∼20% in permeabilized cardiac myocyte preparations (n = 6) from the left ventricle of human failing hearts. These results implicate cTnI N-terminal phosphorylation as a molecular regulator of myocyte power and could serve as a regional target for small molecule therapy to unmask myocyte power reserve capacity in human failing hearts.
Collapse
Affiliation(s)
- Laurin M. Hanft
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Joel C. Robinett
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Theodore J. Kalogeris
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Kenneth S. Campbell
- Department of Physiology and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA
| | | | - Kerry S. McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
25
|
Tanner BCW, Awinda PO, Agonias KB, Attili S, Blair CA, Thompson MS, Walker LA, Kampourakis T, Campbell KS. Sarcomere length affects Ca2+ sensitivity of contraction in ischemic but not non-ischemic myocardium. J Gen Physiol 2023; 155:213800. [PMID: 36633584 PMCID: PMC9859763 DOI: 10.1085/jgp.202213200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/18/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
In healthy hearts, myofilaments become more sensitive to Ca2+ as the myocardium is stretched. This effect is known as length-dependent activation and is an important cellular-level component of the Frank-Starling mechanism. Few studies have measured length-dependent activation in the myocardium from failing human hearts. We investigated whether ischemic and non-ischemic heart failure results in different length-dependent activation responses at physiological temperature (37°C). Myocardial strips from the left ventricular free wall were chemically permeabilized and Ca2+-activated at sarcomere lengths (SLs) of 1.9 and 2.3 µm. Data were acquired from 12 hearts that were explanted from patients receiving cardiac transplants; 6 had ischemic heart failure and 6 had non-ischemic heart failure. Another 6 hearts were obtained from organ donors. Maximal Ca2+-activated force increased at longer SL for all groups. Ca2+ sensitivity increased with SL in samples from donors (P < 0.001) and patients with ischemic heart failure (P = 0.003) but did not change with SL in samples from patients with non-ischemic heart failure. Compared with donors, troponin I phosphorylation decreased in ischemic samples and even more so in non-ischemic samples; cardiac myosin binding protein-C (cMyBP-C) phosphorylation also decreased with heart failure. These findings support the idea that troponin I and cMyBP-C phosphorylation promote length-dependent activation and show that length-dependent activation of contraction is blunted, yet extant, in the myocardium from patients with ischemic heart failure and further reduced in the myocardium from patients with non-ischemic heart failure. Patients who have a non-ischemic disease may exhibit a diminished contractile response to increased ventricular filling.
Collapse
Affiliation(s)
- Bertrand C W Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Peter O Awinda
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Keinan B Agonias
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, WA, USA
| | - Seetharamaiah Attili
- Randall Centre for Cell and Molecular Biophysics, King's College London , London, UK
| | - Cheavar A Blair
- Department of Physiology, University of Kentucky , Lexington, KY, USA
| | - Mindy S Thompson
- Department of Physiology, University of Kentucky , Lexington, KY, USA
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus , Aurora, CO, USA
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics, King's College London , London, UK
| | - Kenneth S Campbell
- Department of Physiology, University of Kentucky , Lexington, KY, USA.,Division of Cardiovascular Medicine, University of Kentucky , Lexington, KY, USA
| |
Collapse
|
26
|
Halder SS, Rynkiewicz MJ, Creso JG, Sewanan LR, Howland L, Moore JR, Lehman W, Campbell SG. Mechanisms of pathogenicity in the hypertrophic cardiomyopathy-associated TPM1 variant S215L. PNAS NEXUS 2023; 2:pgad011. [PMID: 36896133 PMCID: PMC9991458 DOI: 10.1093/pnasnexus/pgad011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/12/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disorder often caused by mutations to sarcomeric genes. Many different HCM-associated TPM1 mutations have been identified but they vary in their degrees of severity, prevalence, and rate of disease progression. The pathogenicity of many TPM1 variants detected in the clinical population remains unknown. Our objective was to employ a computational modeling pipeline to assess pathogenicity of one such variant of unknown significance, TPM1 S215L, and validate predictions using experimental methods. Molecular dynamic simulations of tropomyosin on actin suggest that the S215L significantly destabilizes the blocked regulatory state while increasing flexibility of the tropomyosin chain. These changes were quantitatively represented in a Markov model of thin-filament activation to infer the impacts of S215L on myofilament function. Simulations of in vitro motility and isometric twitch force predicted that the mutation would increase Ca2+ sensitivity and twitch force while slowing twitch relaxation. In vitro motility experiments with thin filaments containing TPM1 S215L revealed higher Ca2+ sensitivity compared with wild type. Three-dimensional genetically engineered heart tissues expressing TPM1 S215L exhibited hypercontractility, upregulation of hypertrophic gene markers, and diastolic dysfunction. These data form a mechanistic description of TPM1 S215L pathogenicity that starts with disruption of the mechanical and regulatory properties of tropomyosin, leading thereafter to hypercontractility and finally induction of a hypertrophic phenotype. These simulations and experiments support the classification of S215L as a pathogenic mutation and support the hypothesis that an inability to adequately inhibit actomyosin interactions is the mechanism whereby thin-filament mutations cause HCM.
Collapse
Affiliation(s)
- Saiti S Halder
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | | | - Jenette G Creso
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | - Lorenzo R Sewanan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Department of Internal Medicine, Columbia University, New York, NY 10032
| | - Lindsey Howland
- Department of Biological Sciences, University of Massachusetts Lowell, MA 01854
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts Lowell, MA 01854
| | - William Lehman
- Department of Physiology/Biophysics, Boston University, Boston, MA 02215
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| |
Collapse
|
27
|
Rupert C, López JE, Cortez-Toledo E, De la Cruz Cabrera O, Chesler NC, Simpson PC, Campbell SG, Baker AJ. Increased length-dependent activation of human engineered heart tissue after chronic α 1A-adrenergic agonist treatment: testing a novel heart failure therapy. Am J Physiol Heart Circ Physiol 2023; 324:H293-H304. [PMID: 36637971 PMCID: PMC9886349 DOI: 10.1152/ajpheart.00279.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 12/06/2022] [Accepted: 12/23/2022] [Indexed: 01/14/2023]
Abstract
Chronic stimulation of cardiac α1A-adrenergic receptors (α1A-ARs) improves symptoms in multiple preclinical models of heart failure. However, the translational significance remains unclear. Human engineered heart tissues (EHTs) provide a means of quantifying the effects of chronic α1A-AR stimulation on human cardiomyocyte physiology. EHTs were created from thin slices of decellularized pig myocardium seeded with human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and fibroblasts. With a paired experimental design, EHTs were cultured for 3 wk, mechanically tested, cultured again for 2 wk with α1A-AR agonist A61603 (10 nM) or vehicle control, and retested after drug washout for 24 h. Separate control experiments determined the effects of EHT age (3-5 wk) or repeat mechanical testing. We found that chronic A61603 treatment caused a 25% increase of length-dependent activation (LDA) of contraction compared with vehicle treatment (n = 7/group, P = 0.035). EHT force was not increased after chronic A61603 treatment. However, after vehicle treatment, EHT force was increased by 35% relative to baseline testing (n = 7/group, P = 0.022), suggesting EHT maturation. Control experiments suggested that increased EHT force resulted from repeat mechanical testing, not from EHT aging. RNA-seq analysis confirmed that the α1A-AR is expressed in human EHTs and found chronic A61603 treatment affected gene expression in biological pathways known to be activated by α1A-ARs, including the MAP kinase signaling pathway. In conclusion, increased LDA in human EHT after chronic A61603 treatment raises the possibility that chronic stimulation of the α1A-AR might be beneficial for increasing LDA in human myocardium and might be beneficial for treating human heart failure by restoring LDA.NEW & NOTEWORTHY Chronic stimulation of α1A-adrenergic receptors (α1A-ARs) is known to mediate therapeutic effects in animal heart failure models. To investigate the effects of chronic α1A-AR stimulation in human cardiomyocytes, we tested engineered heart tissue (EHT) created with iPSC-derived cardiomyocytes. RNA-seq analysis confirmed human EHT expressed α1A-ARs. Chronic (2 wk) α1A-AR stimulation with A61603 (10 nM) increased length-dependent activation (LDA) of contraction. Chronic α1A-AR stimulation might be beneficial for treating human heart failure by restoring LDA.
Collapse
Affiliation(s)
- C. Rupert
- Propria LLC, Branford, Connecticut, United States
| | - J. E. López
- Division of Cardiovascular Medicine, Department of Internal Medicine,
University of California Davis, Davis, California, United States
| | - E. Cortez-Toledo
- Division of Cardiovascular Medicine, Department of Internal Medicine,
University of California Davis, Davis, California, United States
| | | | - N. C. Chesler
- Edwards Lifesciences Foundation Cardiovascular Innovation Research Center, Irvine, California, United States
- Department of Biomedical Engineering, University of California, Irvine, California, United States
| | - P. C. Simpson
- Cardiology Division, Veterans Affairs Medical Center, San Francisco, California, United States
- Department of Medicine, University of California, San Francisco, California, United States
| | - S. G. Campbell
- Departments of Biomedical Engineering and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
| | - A. J. Baker
- Cardiology Division, Veterans Affairs Medical Center, San Francisco, California, United States
- Department of Medicine, University of California, San Francisco, California, United States
| |
Collapse
|
28
|
Nollet EE, Duursma I, Rozenbaum A, Eggelbusch M, Wüst RCI, Schoonvelde SAC, Michels M, Jansen M, van der Wel NN, Bedi KC, Margulies KB, Nirschl J, Kuster DWD, van der Velden J. Mitochondrial dysfunction in human hypertrophic cardiomyopathy is linked to cardiomyocyte architecture disruption and corrected by improving NADH-driven mitochondrial respiration. Eur Heart J 2023; 44:1170-1185. [PMID: 36734059 PMCID: PMC10067466 DOI: 10.1093/eurheartj/ehad028] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 12/19/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023] Open
Abstract
AIMS Genetic hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomere protein-encoding genes (i.e. genotype-positive HCM). In an increasing number of patients, HCM occurs in the absence of a mutation (i.e. genotype-negative HCM). Mitochondrial dysfunction is thought to be a key driver of pathological remodelling in HCM. Reports of mitochondrial respiratory function and specific disease-modifying treatment options in patients with HCM are scarce. METHODS AND RESULTS Respirometry was performed on septal myectomy tissue from patients with HCM (n = 59) to evaluate oxidative phosphorylation and fatty acid oxidation. Mitochondrial dysfunction was most notably reflected by impaired NADH-linked respiration. In genotype-negative patients, but not genotype-positive patients, NADH-linked respiration was markedly depressed in patients with an indexed septal thickness ≥10 compared with <10. Mitochondrial dysfunction was not explained by reduced abundance or fragmentation of mitochondria, as evaluated by transmission electron microscopy. Rather, improper organization of mitochondria relative to myofibrils (expressed as a percentage of disorganized mitochondria) was strongly associated with mitochondrial dysfunction. Pre-incubation with the cardiolipin-stabilizing drug elamipretide and raising mitochondrial NAD+ levels both boosted NADH-linked respiration. CONCLUSION Mitochondrial dysfunction is explained by cardiomyocyte architecture disruption and is linked to septal hypertrophy in genotype-negative HCM. Despite severe myocardial remodelling mitochondria were responsive to treatments aimed at restoring respiratory function, eliciting the mitochondria as a drug target to prevent and ameliorate cardiac disease in HCM. Mitochondria-targeting therapy may particularly benefit genotype-negative patients with HCM, given the tight link between mitochondrial impairment and septal thickening in this subpopulation.
Collapse
Affiliation(s)
- Edgar E Nollet
- Department of Physiology, Amsterdam UMC, Location VUmc, O2 Science building—11W53, De Boelelaan 1108, 1081HZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam UMC, Location VUmc, O2 Science building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Inez Duursma
- Department of Physiology, Amsterdam UMC, Location VUmc, O2 Science building—11W53, De Boelelaan 1108, 1081HZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam UMC, Location VUmc, O2 Science building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Anastasiya Rozenbaum
- Department of Physiology, Amsterdam UMC, Location VUmc, O2 Science building—11W53, De Boelelaan 1108, 1081HZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam UMC, Location VUmc, O2 Science building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Moritz Eggelbusch
- Laboratory for Myology, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Nutrition and Dietetics, Amsterdam UMC, Amsterdam, The Netherlands
- Faculty of Sports and Nutrition, Center of Expertise Urban Vitality, Amsterdam University of Applied Sciences, Amsterdam, The Netherlands
| | - Rob C I Wüst
- Laboratory for Myology, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Michelle Michels
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Mark Jansen
- Division of Genetics, UMC Utrecht, Utrecht, The Netherlands
| | - Nicole N van der Wel
- Department of Medical Biology, Electron Microscopy Centre, Amsterdam UMC, Amsterdam, The Netherlands
| | - Kenneth C Bedi
- Cardiovascular Institute, Perelman School of Medicine, Philadelphia, PA, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeff Nirschl
- Department of Pathology, Stanford University, Stanford, USA
| | - Diederik W D Kuster
- Department of Physiology, Amsterdam UMC, Location VUmc, O2 Science building—11W53, De Boelelaan 1108, 1081HZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam UMC, Location VUmc, O2 Science building, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | | |
Collapse
|
29
|
Sevrieva IR, Ponnam S, Yan Z, Irving M, Kampourakis T, Sun YB. Phosphorylation-dependent interactions of myosin-binding protein C and troponin coordinate the myofilament response to protein kinase A. J Biol Chem 2023; 299:102767. [PMID: 36470422 PMCID: PMC9826837 DOI: 10.1016/j.jbc.2022.102767] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
PKA-mediated phosphorylation of sarcomeric proteins enhances heart muscle performance in response to β-adrenergic stimulation and is associated with accelerated relaxation and increased cardiac output for a given preload. At the cellular level, the latter translates to a greater dependence of Ca2+ sensitivity and maximum force on sarcomere length (SL), that is, enhanced length-dependent activation. However, the mechanisms by which PKA phosphorylation of the most notable sarcomeric PKA targets, troponin I (cTnI) and myosin-binding protein C (cMyBP-C), lead to these effects remain elusive. Here, we specifically altered the phosphorylation level of cTnI in heart muscle cells and characterized the structural and functional effects at different levels of background phosphorylation of cMyBP-C and with two different SLs. We found Ser22/23 bisphosphorylation of cTnI was indispensable for the enhancement of length-dependent activation by PKA, as was cMyBP-C phosphorylation. This high level of coordination between cTnI and cMyBP-C may suggest coupling between their regulatory mechanisms. Further evidence for this was provided by our finding that cardiac troponin (cTn) can directly interact with cMyBP-C in vitro, in a phosphorylation- and Ca2+-dependent manner. In addition, bisphosphorylation at Ser22/Ser23 increased Ca2+ sensitivity at long SL in the presence of endogenously phosphorylated cMyBP-C. When cMyBP-C was dephosphorylated, bisphosphorylation of cTnI increased Ca2+ sensitivity and decreased cooperativity at both SLs, which may translate to deleterious effects in physiological settings. Our results could have clinical relevance for disease pathways, where PKA phosphorylation of cTnI may be functionally uncoupled from cMyBP-C phosphorylation due to mutations or haploinsufficiency.
Collapse
Affiliation(s)
- Ivanka R Sevrieva
- Randall Centre for Cell and Molecular Biophysics, and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom.
| | - Saraswathi Ponnam
- Randall Centre for Cell and Molecular Biophysics, and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Ziqian Yan
- Randall Centre for Cell and Molecular Biophysics, and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Malcolm Irving
- Randall Centre for Cell and Molecular Biophysics, and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics, and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Yin-Biao Sun
- Randall Centre for Cell and Molecular Biophysics, and British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| |
Collapse
|
30
|
Nefedova VV, Kopylova GV, Shchepkin DV, Kochurova AM, Kechko OI, Borzova VA, Ryabkova NS, Katrukha IA, Mitkevich VA, Bershitsky SY, Levitsky DI, Matyushenko AM. Impact of Troponin in Cardiomyopathy Development Caused by Mutations in Tropomyosin. Int J Mol Sci 2022; 23:ijms232415723. [PMID: 36555368 PMCID: PMC9779223 DOI: 10.3390/ijms232415723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Tropomyosin (Tpm) mutations cause inherited cardiac diseases such as hypertrophic and dilated cardiomyopathies. We applied various approaches to investigate the role of cardiac troponin (Tn) and especially the troponin T (TnT) in the pathogenic effects of Tpm cardiomyopathy-associated mutations M8R, K15N, A277V, M281T, and I284V located in the overlap junction of neighboring Tpm dimers. Using co-sedimentation assay and viscosity measurements, we showed that TnT1 (fragment of TnT) stabilizes the overlap junction of Tpm WT and all Tpm mutants studied except Tpm M8R. However, isothermal titration calorimetry (ITC) indicated that TnT1 binds Tpm WT and all Tpm mutants similarly. By using ITC, we measured the direct KD of the Tpm overlap region, N-end, and C-end binding to TnT1. The ITC data revealed that the Tpm C-end binds to TnT1 independently from the N-end, while N-end does not bind. Therefore, we suppose that Tpm M8R binds to TnT1 without forming the overlap junction. We also demonstrated the possible role of Tn isoform composition in the cardiomyopathy development caused by M8R mutation. TnT1 dose-dependently reduced the velocity of F-actin-Tpm filaments containing Tpm WT, Tpm A277V, and Tpm M281T mutants in an in vitro motility assay. All mutations impaired the calcium regulation of the actin-myosin interaction. The M281T and I284V mutations increased the calcium sensitivity, while the K15N and A277V mutations reduced it. The Tpm M8R, M281T, and I284V mutations under-inhibited the velocity at low calcium concentrations. Our results demonstrate that Tpm mutations likely implement their pathogenic effects through Tpm interaction with Tn, cardiac myosin, or other protein partners.
Collapse
Affiliation(s)
- Victoria V. Nefedova
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
- Correspondence:
| | - Galina V. Kopylova
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia
| | - Daniil V. Shchepkin
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia
| | - Anastasia M. Kochurova
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia
| | - Olga I. Kechko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - Vera A. Borzova
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Natalia S. Ryabkova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- HyTest Ltd., 20520 Turku, Finland
| | - Ivan A. Katrukha
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- HyTest Ltd., 20520 Turku, Finland
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia
| | - Sergey Y. Bershitsky
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia
| | - Dmitrii I. Levitsky
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | | |
Collapse
|
31
|
Jones MR, Tran C, Singh J, Dawson JF. A gradient of force generation at rest differentiates cardiomyopathy outcomes with variants of actin located at the same residue. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 2:100023. [PMID: 39802493 PMCID: PMC11708414 DOI: 10.1016/j.jmccpl.2022.100023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/21/2022] [Accepted: 11/27/2022] [Indexed: 01/16/2025]
Abstract
The calcium sensitivity hypothesis helps explain the development of different forms of cardiomyopathy: increased sensitivity to calcium in cardiac sarcomeres leads to hypertrophic cardiomyopathy (HCM) and decreased sensitivity results in dilated cardiomyopathy (DCM). This hypothesis has driven the development of next generation drugs targeting sarcomere proteins to correct the amount of force generated as a result of changes in calcium sensitivity (e.g. mavacamten decreases cardiac myosin activity to treat HCM). Characterization of variants of cardiac actin (ACTC) found in patients with HCM or DCM has generally supported the calcium sensitivity hypothesis. Of interest are two different substitution mutations at R312 on ACTC: R312H leads to DCM, while R312C was found in patients with HCM. To determine how changes in the same codon on the same gene lead to different disease phenotypes, we characterized recombinant R312H- and R312C-ACTC variant proteins. Both variants exhibited the same change in calcium sensitivity, suggesting that a factor other than calcium sensitivity is responsible for disease differentiation. We observed a gradient of increased residual myosin activity with R312-ACTC variant proteins under relaxing conditions which may trigger different disease development. Our findings suggest that factors other than calcium sensitivity may contribute to cardiomyopathy development and should be considered when planning treatments.
Collapse
Affiliation(s)
- Michael R. Jones
- Corresponding author at: Department of Molecular & Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | | | | | - John F. Dawson
- Department of Molecular & Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
- Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
32
|
Xu M, Bermea KC, Ayati M, Kim HB, Yang X, Medina A, Fu Z, Heravi A, Zhang X, Na CH, Everett AD, Gabrielson K, Foster DB, Paolocci N, Murphy AM, Ramirez-Correa GA. Alteration in tyrosine phosphorylation of cardiac proteome and EGFR pathway contribute to hypertrophic cardiomyopathy. Commun Biol 2022; 5:1251. [PMID: 36380187 PMCID: PMC9666710 DOI: 10.1038/s42003-022-04021-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Alterations of serine/threonine phosphorylation of the cardiac proteome are a hallmark of heart failure. However, the contribution of tyrosine phosphorylation (pTyr) to the pathogenesis of cardiac hypertrophy remains unclear. We use global mapping to discover and quantify site-specific pTyr in two cardiac hypertrophic mouse models, i.e., cardiac overexpression of ErbB2 (TgErbB2) and α myosin heavy chain R403Q (R403Q-αMyHC Tg), compared to control hearts. From this, there are significant phosphoproteomic alterations in TgErbB2 mice in right ventricular cardiomyopathy, hypertrophic cardiomyopathy (HCM), and dilated cardiomyopathy (DCM) pathways. On the other hand, R403Q-αMyHC Tg mice indicated that the EGFR1 pathway is central for cardiac hypertrophy, along with angiopoietin, ErbB, growth hormone, and chemokine signaling pathways activation. Surprisingly, most myofilament proteins have downregulation of pTyr rather than upregulation. Kinase-substrate enrichment analysis (KSEA) shows a marked downregulation of MAPK pathway activity downstream of k-Ras in TgErbB2 mice and activation of EGFR, focal adhesion, PDGFR, and actin cytoskeleton pathways. In vivo ErbB2 inhibition by AG-825 decreases cardiomyocyte disarray. Serine/threonine and tyrosine phosphoproteome confirm the above-described pathways and the effectiveness of AG-825 Treatment. Thus, altered pTyr may play a regulatory role in cardiac hypertrophic models.
Collapse
Affiliation(s)
- Mingguo Xu
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,Department of Pediatrics, The Third People’s Hospital of Longgang District, Shenzhen, 518115 China
| | - Kevin C. Bermea
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Marzieh Ayati
- grid.449717.80000 0004 5374 269XDeparment of Computer Science/College of Engineering and Computer Science, University of Texas Rio Grande Valley School of Medicine, Edinburgh, Texas USA
| | - Han Byeol Kim
- grid.21107.350000 0001 2171 9311Department of Neurology/Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xiaomei Yang
- grid.27255.370000 0004 1761 1174Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Andres Medina
- Department of Molecular Science/UT Health Rio Grande Valley, McAllen, TX USA
| | - Zongming Fu
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Amir Heravi
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xinyu Zhang
- grid.27255.370000 0004 1761 1174Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Chan Hyun Na
- grid.21107.350000 0001 2171 9311Department of Neurology/Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Biological Chemistry/McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Allen D. Everett
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Kathleen Gabrielson
- grid.21107.350000 0001 2171 9311Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - D. Brian Foster
- grid.21107.350000 0001 2171 9311Department of Medicine/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Nazareno Paolocci
- grid.21107.350000 0001 2171 9311Department of Medicine/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Anne M. Murphy
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Genaro A. Ramirez-Correa
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,Department of Molecular Science/UT Health Rio Grande Valley, McAllen, TX USA
| |
Collapse
|
33
|
Hool LC. Elucidating the role of the L-type calcium channel in excitability and energetics in the heart: The ISHR 2020 Research Achievement Award Lecture. J Mol Cell Cardiol 2022; 172:100-108. [PMID: 36041287 DOI: 10.1016/j.yjmcc.2022.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease continues to be the leading health burden worldwide and with the rising rates in obesity and type II diabetes and ongoing effects of long COVID, it is anticipated that the burden of cardiovascular morbidity and mortality will increase. Calcium is essential to cardiac excitation and contraction. The main route for Ca2+ influx is the L-type Ca2+ channel (Cav1.2) and embryos that are homozygous null for the Cav1.2 gene are lethal at day 14 postcoitum. Acute changes in Ca2+ influx through the channel contribute to arrhythmia and sudden death, and chronic increases in intracellular Ca2+ contribute to pathological hypertrophy and heart failure. We use a multidisciplinary approach to study the regulation of the channel from the molecular level through to in vivo CRISPR mutant animal models. Here we describe some examples of our work from over 2 decades studying the role of the channel under physiological and pathological conditions. Our single channel analysis of purified human Cav1.2 protein in proteoliposomes has contributed to understanding direct molecular regulation of the channel including identifying the critical serine involved in the "fight or flight" response. Using the same approach we identified the cysteine responsible for altered function during oxidative stress. Chronic activation of the L-type Ca2+ channel during oxidative stress occurs as a result of persistent glutathionylation of the channel that contributes to the development of hypertrophy. We describe for the first time that activation of the channel alters mitochondrial function (and energetics) on a beat-to-beat basis via movement of cytoskeletal proteins. In translational studies we have used this response to "report" mitochondrial function in models of cardiomyopathy and to test efficacy of novel therapies to prevent cardiomyopathy.
Collapse
Affiliation(s)
- Livia C Hool
- School of Human Sciences, University of Western Australia, Crawley, WA, Australia; Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
34
|
Sewanan LR, Shimada YJ. Prospects for remodeling the hypertrophic heart with myosin modulators. Front Cardiovasc Med 2022; 9:1051564. [PMID: 36330009 PMCID: PMC9622926 DOI: 10.3389/fcvm.2022.1051564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/06/2022] [Indexed: 09/25/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a complex but relatively common genetic disease that usually arises from pathogenic variants that disrupt sarcomere function and lead to variable structural, hypertrophic, and fibrotic remodeling of the heart which result in substantial adverse clinical outcomes including arrhythmias, heart failure, and sudden cardiac death. HCM has had few effective treatments with the potential to ameliorate disease progression until the recent advent of inhibitory myosin modulators like mavacamten. Preclinical investigations and clinical trials utilizing this treatment targeted to this specific pathophysiological mechanism of sarcomere hypercontractility in HCM have confirmed that myosin modulators can alter disease expression and attenuate hypertrophic remodeling. Here, we summarize the state of hypertrophic remodeling and consider the arguments for and against salutary HCM disease modification using targeted myosin modulators. Further, we consider critical unanswered questions for future investigative and therapeutic avenues in HCM disease modification. We are at the precipice of a new era in understanding and treating HCM, with the potential to target agents toward modifying disease expression and natural history of this most common inherited disease of the heart.
Collapse
Affiliation(s)
- Lorenzo R. Sewanan
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Yuichi J. Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
35
|
Sequeira V, Wang L, Wijnker PJ, Kim K, Pinto JR, dos Remedios C, Redwood C, Knollmann BC, van der Velden J. Low expression of the K280N TNNT2 mutation is sufficient to increase basal myofilament activation in human hypertrophy cardiomyopathy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 1:100007. [PMID: 37159677 PMCID: PMC10160007 DOI: 10.1016/j.jmccpl.2022.100007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 05/11/2023]
Abstract
Background Hypertrophic cardiomyopathy (HCM) is an autosomal dominant genetic disorder with patients typically showing heterozygous inheritance of a pathogenic variant in a gene encoding a contractile protein. Here, we study the contractile effects of a rare homozygous mutation using explanted tissue and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to gain insight into how the balance between mutant and WT protein expression affects cardiomyocyte function. Methods Force measurements were performed in cardiomyocytes isolated from a HCM patient carrying a homozygous troponin T mutation (cTnT-K280N) and healthy donors. To discriminate between mutation-mediated and phosphorylation-related effects on Ca2+-sensitivity, cardiomyocytes were treated with alkaline phosphatase (AP) or protein kinase A (PKA). Troponin exchange experiments characterized the relation between mutant levels and myofilament function. To define mutation-mediated effects on Ca2+-dynamics we used CRISPR/Cas9 to generate hiPSC-CMs harbouring heterozygous and homozygous TnT-K280N mutations. Ca2+-transient and cell shortening experiments compared these lines against isogenic controls. Results Myofilament Ca2+-sensitivity was higher in homozygous cTnT-K280N cardiomyocytes and was not corrected by AP- and PKA-treatment. In cTnT-K280N cells exchanged with cTnT-WT, a low level (14%) of cTnT-K280N mutation elevated Ca2+-sensitivity. Similarly, exchange of donor cells with 45 ± 2% cTnT-K280N increased Ca2+-sensitivity and was not corrected by PKA. cTnT-K280N hiPSC-CMs show elevated diastolic Ca2+ and increases in cell shortening. Impaired cardiomyocyte relaxation was only evident in homozygous cTnT-K280N hiPSC-CMs. Conclusions The cTnT-K280N mutation increases myofilament Ca2+-sensitivity, elevates diastolic Ca2+, enhances contractility and impairs cellular relaxation. A low level (14%) of the cTnT-K280N sensitizes myofilaments to Ca2+, a universal finding of human HCM.
Collapse
Affiliation(s)
- Vasco Sequeira
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
- Division of Clinical Pharmacology, Vanderbilt School of Medicine, Nashville, United States
- Comprehensive Heart Failure Center (CHFC) University Clinic Würzburg, Würzburg, Germany
| | - Lili Wang
- Division of Clinical Pharmacology, Vanderbilt School of Medicine, Nashville, United States
| | - Paul J.M. Wijnker
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Kyungsoo Kim
- Division of Clinical Pharmacology, Vanderbilt School of Medicine, Nashville, United States
| | - Jose R. Pinto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Cris dos Remedios
- Muscle Research Unit, Discipline of Anatomy & Histology, Bosch Institute, The University of Sydney, Sydney, Australia
| | | | - Bjorn C. Knollmann
- Division of Clinical Pharmacology, Vanderbilt School of Medicine, Nashville, United States
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| |
Collapse
|
36
|
Singh RR, Slater RE, Wang J, Wang C, Guo Q, Motani AS, Hartman JJ, Sadayappan S, Ason BL. Distinct Mechanisms for Increased Cardiac Contraction Through Selective Alteration of Either Myosin or Troponin Activity. JACC Basic Transl Sci 2022; 7:1021-1037. [DOI: 10.1016/j.jacbts.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 10/14/2022]
|
37
|
Burkart V, Kowalski K, Aldag-Niebling D, Beck J, Frick DA, Holler T, Radocaj A, Piep B, Zeug A, Hilfiker-Kleiner D, dos Remedios CG, van der Velden J, Montag J, Kraft T. Transcriptional bursts and heterogeneity among cardiomyocytes in hypertrophic cardiomyopathy. Front Cardiovasc Med 2022; 9:987889. [PMID: 36082122 PMCID: PMC9445301 DOI: 10.3389/fcvm.2022.987889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/02/2022] [Indexed: 12/01/2022] Open
Abstract
Transcriptional bursting is a common expression mode for most genes where independent transcription of alleles leads to different ratios of allelic mRNA from cell to cell. Here we investigated burst-like transcription and its consequences in cardiac tissue from Hypertrophic Cardiomyopathy (HCM) patients with heterozygous mutations in the sarcomeric proteins cardiac myosin binding protein C (cMyBP-C, MYBPC3) and cardiac troponin I (cTnI, TNNI3). Using fluorescence in situ hybridization (RNA-FISH) we found that both, MYBPC3 and TNNI3 are transcribed burst-like. Along with that, we show unequal allelic ratios of TNNI3-mRNA among single cardiomyocytes and unequally distributed wildtype cMyBP-C protein across tissue sections from heterozygous HCM-patients. The mutations led to opposing functional alterations, namely increasing (cMyBP-Cc.927−2A>G) or decreasing (cTnIR145W) calcium sensitivity. Regardless, all patients revealed highly variable calcium-dependent force generation between individual cardiomyocytes, indicating contractile imbalance, which appears widespread in HCM-patients. Altogether, we provide strong evidence that burst-like transcription of sarcomeric genes can lead to an allelic mosaic among neighboring cardiomyocytes at mRNA and protein level. In HCM-patients, this presumably induces the observed contractile imbalance among individual cardiomyocytes and promotes HCM-development.
Collapse
Affiliation(s)
- Valentin Burkart
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Valentin Burkart
| | - Kathrin Kowalski
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - David Aldag-Niebling
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Julia Beck
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Dirk Alexander Frick
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Tim Holler
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Ante Radocaj
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Birgit Piep
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Andre Zeug
- Institute for Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | | | - Cristobal G. dos Remedios
- Mechanosensory Biophysics Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | | | - Judith Montag
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- *Correspondence: Judith Montag
| | - Theresia Kraft
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
38
|
Chevalier C, Wendner M, Suling A, Cavus E, Muellerleile K, Lund G, Kirchhof P, Patten M. Association of NT-proBNP and hs-cTnT with Imaging Markers of Diastolic Dysfunction and Focal Myocardial Fibrosis in Hypertrophic Cardiomyopathy. Life (Basel) 2022; 12:1241. [PMID: 36013420 PMCID: PMC9410236 DOI: 10.3390/life12081241] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Serum biomarkers such as N-terminal prohormone of the brain natriuretic peptide (NT-proBNP) and cardiac troponins are elevated in patients with hypertrophic cardiomyopathy (HCM). At present, it is not clear if these markers are associated with distinct clinical alterations in HCM, such as left ventricular hypertrophy, outflow tract obstruction, myocardial fibrosis and/or diastolic dysfunction (DD), which are associated with adverse cardiovascular outcome. Here we evaluate the association of NT-proBNP and high sensitivity cardiac troponin T (hs-cTnT) to a variety of cardiac imaging parameters in HCM patients in a multivariable regression analysis. This retrospective cross-sectional study included 366 HCM patients who underwent transthoracic echocardiography (TTE), 218 of whom also obtained cardiovascular magnetic resonance (CMR) to assess focal myocardial fibrosis by LGE. Multivariable regression analyses revealed the strongest association of the DD parameters E/E′ mean and E/E′ septal with NT-proBNP (b = 0.06, 95%-CI [0.05−0.07], p < 0.001, R2 = 0.28; b = 0.08, 95%-CI [0.06−0.1], p < 0.001, R2 = 0.25) and LGE size showed the strongest association with hs-cTnT (b = 0.20, 95%-CI [0.15−0.24], p < 0.001, R2 = 0.28). This study indicates that NT-proBNP and hs-cTnT are associated with structural and functional alterations in HCM. NT-proBNP is a stronger predictor for DD, while hs-cTnT is associated with the extent of focal myocardial fibrosis. Both biomarkers might be useful in the diagnostic procedure in addition to imaging parameters.
Collapse
Affiliation(s)
- Céleste Chevalier
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Miriam Wendner
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Anna Suling
- Institute of Medical Biometry and Epidemiology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Christoph-Probst-Weg 1, 20246 Hamburg, Germany
| | - Ersin Cavus
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Kai Muellerleile
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Gunnar Lund
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Paulus Kirchhof
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Monica Patten
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
39
|
Sultan A, Adeghate E, Emerald BS, Qureshi MA, Minhas ST, Howarth FC. Effects of Obesity and Diabesity on Ventricular Muscle Structure and Function in the Zucker Rat. Life (Basel) 2022; 12:1221. [PMID: 36013400 PMCID: PMC9410105 DOI: 10.3390/life12081221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023] Open
Abstract
(1) Background: Cardiovascular complications are a leading cause of morbidity and mortality in diabetic patients. The effects of obesity and diabesity on the function and structure of ventricular myocytes in the Zucker fatty (ZF) rat and the Zucker diabetic fatty (ZDF) rat compared to Zucker lean (ZL) control rats have been investigated. (2) Methods: Shortening and intracellular Ca2+ were simultaneously measured with cell imaging and fluorescence photometry, respectively. Ventricular muscle protein expression and structure were investigated with Western blot and electron microscopy, respectively. (3) Results: The amplitude of shortening was increased in ZF compared to ZL but not compared to ZDF myocytes. Resting Ca2+ was increased in ZDF compared to ZL myocytes. Time to half decay of the Ca2+ transient was prolonged in ZDF compared to ZL and was reduced in ZF compared to ZL myocytes. Changes in expression of proteins associated with cardiac muscle contraction are presented. Structurally, there were reductions in sarcomere length in ZDF and ZF compared to ZL and reductions in mitochondria count in ZF compared to ZDF and ZL myocytes. (4) Conclusions: Alterations in ventricular muscle proteins and structure may partly underlie the defects observed in Ca2+ signaling in ZDF and ZF compared to ZL rat hearts.
Collapse
Affiliation(s)
- Ahmed Sultan
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine & Health Sciences, UAE University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine & Health Sciences, UAE University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Muhammad A. Qureshi
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Saeed Tariq Minhas
- Department of Anatomy, College of Medicine & Health Sciences, UAE University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Frank Christopher Howarth
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain P.O. Box 17666, United Arab Emirates
| |
Collapse
|
40
|
Previs MJ, O’Leary TS, Morley MP, Palmer B, LeWinter M, Yob J, Pagani FD, Petucci C, Kim MS, Margulies KB, Arany Z, Kelly DP, Day SM. Defects in the Proteome and Metabolome in Human Hypertrophic Cardiomyopathy. Circ Heart Fail 2022; 15:e009521. [PMID: 35543134 PMCID: PMC9708114 DOI: 10.1161/circheartfailure.121.009521] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Defects in energetics are thought to be central to the pathophysiology of hypertrophic cardiomyopathy (HCM); yet, the determinants of ATP availability are not known. The purpose of this study is to ascertain the nature and extent of metabolic reprogramming in human HCM, and its potential impact on contractile function. METHODS We conducted proteomic and targeted, quantitative metabolomic analyses on heart tissue from patients with HCM and from nonfailing control human hearts. RESULTS In the proteomic analysis, the greatest differences observed in HCM samples compared with controls were increased abundances of extracellular matrix and intermediate filament proteins and decreased abundances of muscle creatine kinase and mitochondrial proteins involved in fatty acid oxidation. These differences in protein abundance were coupled with marked reductions in acyl carnitines, byproducts of fatty acid oxidation, in HCM samples. Conversely, the ketone body 3-hydroxybutyrate, branched chain amino acids, and their breakdown products, were all significantly increased in HCM hearts. ATP content, phosphocreatine, nicotinamide adenine dinucleotide and its phosphate derivatives, NADP and NADPH, and acetyl CoA were also severely reduced in HCM compared with control hearts. Functional assays performed on human skinned myocardial fibers demonstrated that the magnitude of observed reduction in ATP content in the HCM samples would be expected to decrease the rate of cross-bridge detachment. Moreover, left atrial size, an indicator of diastolic compliance, was inversely correlated with ATP content in hearts from patients with HCM. CONCLUSIONS HCM hearts display profound deficits in nucleotide availability with markedly reduced capacity for fatty acid oxidation and increases in ketone bodies and branched chain amino acids. These results have important therapeutic implications for the future design of metabolic modulators to treat HCM.
Collapse
Affiliation(s)
- Michael J. Previs
- Department of Molecular Physiology and Biophysics, University of Vermont, Larner College of Medicine
| | - Thomas S. O’Leary
- Department of Molecular Physiology and Biophysics, University of Vermont, Larner College of Medicine
| | - Michael P. Morley
- Division of Cardiovascular Medicine and the Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania
| | - Brad Palmer
- Department of Molecular Physiology and Biophysics, University of Vermont, Larner College of Medicine
| | - Martin LeWinter
- Department of Molecular Physiology and Biophysics, University of Vermont, Larner College of Medicine
| | - Jaime Yob
- Division of Cardiovascular Medicine and the Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania
| | - Francis D. Pagani
- Department of Cardiothoracic Surgery, University of Michigan School of Medicine
| | - Christopher Petucci
- Division of Cardiovascular Medicine and the Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania
| | - Min-Soo Kim
- Division of Cardiovascular Medicine and the Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania
| | - Kenneth B. Margulies
- Division of Cardiovascular Medicine and the Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania
| | - Zoltan Arany
- Division of Cardiovascular Medicine and the Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania
| | - Daniel P. Kelly
- Division of Cardiovascular Medicine and the Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania
| | - Sharlene M. Day
- Division of Cardiovascular Medicine and the Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
41
|
Stachowski-Doll MJ, Papadaki M, Martin TG, Ma W, Gong HM, Shao S, Shen S, Muntu NA, Kumar M, Perez E, Martin JL, Moravec CS, Sadayappan S, Campbell SG, Irving T, Kirk JA. GSK-3β Localizes to the Cardiac Z-Disc to Maintain Length Dependent Activation. Circ Res 2022; 130:871-886. [PMID: 35168370 PMCID: PMC8930626 DOI: 10.1161/circresaha.121.319491] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 02/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Altered kinase localization is gaining appreciation as a mechanism of cardiovascular disease. Previous work suggests GSK-3β (glycogen synthase kinase 3β) localizes to and regulates contractile function of the myofilament. We aimed to discover GSK-3β's in vivo role in regulating myofilament function, the mechanisms involved, and the translational relevance. METHODS Inducible cardiomyocyte-specific GSK-3β knockout mice and left ventricular myocardium from nonfailing and failing human hearts were studied. RESULTS Skinned cardiomyocytes from knockout mice failed to exhibit calcium sensitization with stretch indicating a loss of length-dependent activation (LDA), the mechanism underlying the Frank-Starling Law. Titin acts as a length sensor for LDA, and knockout mice had decreased titin stiffness compared with control mice, explaining the lack of LDA. Knockout mice exhibited no changes in titin isoforms, titin phosphorylation, or other thin filament phosphorylation sites known to affect passive tension or LDA. Mass spectrometry identified several z-disc proteins as myofilament phospho-substrates of GSK-3β. Agreeing with the localization of its targets, GSK-3β that is phosphorylated at Y216 binds to the z-disc. We showed pY216 was necessary and sufficient for z-disc binding using adenoviruses for wild-type, Y216F, and Y216E GSK-3β in neonatal rat ventricular cardiomyocytes. One of GSK-3β's z-disc targets, abLIM-1 (actin-binding LIM protein 1), binds to the z-disc domains of titin that are important for maintaining passive tension. Genetic knockdown of abLIM-1 via siRNA in human engineered heart tissues resulted in enhancement of LDA, indicating abLIM-1 may act as a negative regulator that is modulated by GSK-3β. Last, GSK-3β myofilament localization was reduced in left ventricular myocardium from failing human hearts, which correlated with depressed LDA. CONCLUSIONS We identified a novel mechanism by which GSK-3β localizes to the myofilament to modulate LDA. Importantly, z-disc GSK-3β levels were reduced in patients with heart failure, indicating z-disc localized GSK-3β is a possible therapeutic target to restore the Frank-Starling mechanism in patients with heart failure.
Collapse
Affiliation(s)
- Marisa J Stachowski-Doll
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Maria Papadaki
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Thomas G Martin
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Weikang Ma
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Henry M Gong
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Stephanie Shao
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
| | - Shi Shen
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
| | - Nitha Aima Muntu
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung, and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Edith Perez
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Jody L Martin
- Department of Pharmacology, Cardiovascular Research Institute, UC Davis School of Medicine, CA (J.L.M.)
| | - Christine S Moravec
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, OH (C.S.M.)
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung, and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Stuart G Campbell
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT (S.G.C.)
| | - Thomas Irving
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| |
Collapse
|
42
|
Ma W, Irving TC. Small Angle X-ray Diffraction as a Tool for Structural Characterization of Muscle Disease. Int J Mol Sci 2022; 23:3052. [PMID: 35328477 PMCID: PMC8949570 DOI: 10.3390/ijms23063052] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023] Open
Abstract
Small angle X-ray fiber diffraction is the method of choice for obtaining molecular level structural information from striated muscle fibers under hydrated physiological conditions. For many decades this technique had been used primarily for investigating basic biophysical questions regarding muscle contraction and regulation and its use confined to a relatively small group of expert practitioners. Over the last 20 years, however, X-ray diffraction has emerged as an important tool for investigating the structural consequences of cardiac and skeletal myopathies. In this review we show how simple and straightforward measurements, accessible to non-experts, can be used to extract biophysical parameters that can help explain and characterize the physiology and pathology of a given experimental system. We provide a comprehensive guide to the range of the kinds of measurements that can be made and illustrate how they have been used to provide insights into the structural basis of pathology in a comprehensive review of the literature. We also show how these kinds of measurements can inform current controversies and indicate some future directions.
Collapse
Affiliation(s)
- Weikang Ma
- The Biophysics Collaborative Access Team (BioCAT), Center for Synchrotron Radiation Research and Instrumentation (CSSRI), Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Thomas C. Irving
- The Biophysics Collaborative Access Team (BioCAT), Center for Synchrotron Radiation Research and Instrumentation (CSSRI), Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
43
|
Emrahi L, Hosseinzadeh H, Tabrizi MT. Two rare variants in the MYBPC3 gene associated with familial hypertrophic cardiomyopathy. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2021.101471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
44
|
Suay-Corredera C, Alegre-Cebollada J. The mechanics of the heart: zooming in on hypertrophic cardiomyopathy and cMyBP-C. FEBS Lett 2022; 596:703-746. [PMID: 35224729 DOI: 10.1002/1873-3468.14301] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Hypertrophic cardiomyopathy (HCM), a disease characterized by cardiac muscle hypertrophy and hypercontractility, is the most frequently inherited disorder of the heart. HCM is mainly caused by variants in genes encoding proteins of the sarcomere, the basic contractile unit of cardiomyocytes. The most frequently mutated among them is MYBPC3, which encodes cardiac myosin-binding protein C (cMyBP-C), a key regulator of sarcomere contraction. In this review, we summarize clinical and genetic aspects of HCM and provide updated information on the function of the healthy and HCM sarcomere, as well as on emerging therapeutic options targeting sarcomere mechanical activity. Building on what is known about cMyBP-C activity, we examine different pathogenicity drivers by which MYBPC3 variants can cause disease, focussing on protein haploinsufficiency as a common pathomechanism also in nontruncating variants. Finally, we discuss recent evidence correlating altered cMyBP-C mechanical properties with HCM development.
Collapse
|
45
|
Ušaj M, Moretto L, Månsson A. Critical Evaluation of Current Hypotheses for the Pathogenesis of Hypertrophic Cardiomyopathy. Int J Mol Sci 2022; 23:2195. [PMID: 35216312 PMCID: PMC8880276 DOI: 10.3390/ijms23042195] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Hereditary hypertrophic cardiomyopathy (HCM), due to mutations in sarcomere proteins, occurs in more than 1/500 individuals and is the leading cause of sudden cardiac death in young people. The clinical course exhibits appreciable variability. However, typically, heart morphology and function are normal at birth, with pathological remodeling developing over years to decades, leading to a phenotype characterized by asymmetric ventricular hypertrophy, scattered fibrosis and myofibrillar/cellular disarray with ultimate mechanical heart failure and/or severe arrhythmias. The identity of the primary mutation-induced changes in sarcomere function and how they trigger debilitating remodeling are poorly understood. Support for the importance of mutation-induced hypercontractility, e.g., increased calcium sensitivity and/or increased power output, has been strengthened in recent years. However, other ideas that mutation-induced hypocontractility or non-uniformities with contractile instabilities, instead, constitute primary triggers cannot yet be discarded. Here, we review evidence for and criticism against the mentioned hypotheses. In this process, we find support for previous ideas that inefficient energy usage and a blunted Frank-Starling mechanism have central roles in pathogenesis, although presumably representing effects secondary to the primary mutation-induced changes. While first trying to reconcile apparently diverging evidence for the different hypotheses in one unified model, we also identify key remaining questions and suggest how experimental systems that are built around isolated primarily expressed proteins could be useful.
Collapse
Affiliation(s)
| | | | - Alf Månsson
- Department of Chemistry and Biomedical Sciences, Faculty of Health and Life Sciences, Linnaeus University, SE-39182 Kalmar, Sweden; (M.U.); (L.M.)
| |
Collapse
|
46
|
Hassoun R, Budde H, Zhazykbayeva S, Herwig M, Sieme M, Delalat S, Mostafi N, Gömöri K, Tangos M, Jarkas M, Pabel S, Bruckmüller S, Skrygan M, Lódi M, Jaquet K, Sequeira V, Gambichler T, Remedios CD, Kovács Á, Mannherz HG, Mügge A, Sossalla S, Hamdani N. Stress activated signalling impaired protein quality control pathways in human hypertrophic cardiomyopathy. Int J Cardiol 2021; 344:160-169. [PMID: 34517018 DOI: 10.1016/j.ijcard.2021.09.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 01/09/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a complex myocardial disorder with no well-established disease-modifying therapy so far. Our study aimed to investigate how autophagy, oxidative stress, inflammation, stress signalling pathways, and apoptosis are hallmark of HCM and their contribution to the cardiac dysfunction. Demembranated cardiomyocytes from patients with HCM display increased titin-based stiffness (Fpassive), which was corrected upon antioxidant treatment. Titin as a main determinant of Fpassive was S-glutathionylated and highly ubiquitinated in HCM patients. This was associated with a shift in the balance of reduced and oxidized forms of glutathione (GSH and GSSG, respectively). Both heat shock proteins (HSP27 and α-ß crystalline) were upregulated and S-glutathionylated in HCM. Administration of HSPs in vitro significantly reduced HCM cardiomyocyte stiffness. High levels of the phosphorylated monomeric superoxide anion-generating endothelial nitric oxide synthase (eNOS), decreased nitric oxide (NO) bioavailability, decreased soluble guanylyl cyclase (sGC) activity, and high levels of 3-nitrotyrosine were observed in HCM. Many regulators of signal transduction pathways that are involved in autophagy, apoptosis, cardiac contractility, and growth including the mitogen-activated protein kinase (MAPK), protein kinase B (AKT), glycogen synthase kinase 3ß (GSK-3ß), mammalian target of rapamycin (mTOR), forkhead box O transcription factor (FOXO), c-Jun N-terminal protein kinase (JNK), and extracellular-signal-regulated kinase (ERK1/2) were modified in HCM. The apoptotic factors cathepsin, procaspase 3, procaspase 9 and caspase 12, but not caspase 9, were elevated in HCM hearts and associated with increased proinflammatory cytokines (Interleukin 6 (IL-6), interleukin 18 (IL-18), intercellular cell adhesion molecule-1 (ICAM1), vascular cell adhesion molecule-1 (VCAM1), the Toll-like receptors 2 (TLR2) and the Toll-like receptors 4 (TLR4)) and oxidative stress (3-nitrotyrosine and hydrogen peroxide (H2O2)). Here we reveal stress signalling and impaired PQS as potential mechanisms underlying the HCM phenotype. Our data suggest that reducing oxidative stress can be a viable therapeutic approach to attenuating the severity of cardiac dysfunction in heart failure and potentially in HCM and prevent its progression.
Collapse
Affiliation(s)
- Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Saltanat Zhazykbayeva
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Melissa Herwig
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Marcel Sieme
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Simin Delalat
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Nusratul Mostafi
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Kamilla Gömöri
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Melina Tangos
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Muhammad Jarkas
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Steffen Pabel
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany.
| | - Stefanie Bruckmüller
- Department of Dermatology, Skin Cancer Center, Ruhr University Bochum, Bochum, Germany.
| | - Marina Skrygan
- Department of Dermatology, Skin Cancer Center, Ruhr University Bochum, Bochum, Germany.
| | - Mária Lódi
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany.
| | - Kornelia Jaquet
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Vasco Sequeira
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Germany.
| | - Thilo Gambichler
- Department of Dermatology, Skin Cancer Center, Ruhr University Bochum, Bochum, Germany.
| | - Cris Dos Remedios
- Molecular Biophysics, Victor Chang Cardiac Research Institute, Faculty of Medicine and Health, Darlinghurst, Australia.
| | - Árpád Kovács
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Hans Georg Mannherz
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Anatomy and Molecular Embryology, Ruhr University, Bochum, Germany.
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany; Clinic for Cardiology & Pneumology, Georg-August University Goettingen, and DZHK (German Centre for Cardiovascular Research), partner site Goettingen, Germany.
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
47
|
Distinct Metabolomic Signatures in Preclinical and Obstructive Hypertrophic Cardiomyopathy. Cells 2021; 10:cells10112950. [PMID: 34831173 PMCID: PMC8616419 DOI: 10.3390/cells10112950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
Hypertrophic Cardiomyopathy (HCM) is a common inherited heart disease with poor risk prediction due to incomplete penetrance and a lack of clear genotype–phenotype correlations. Advanced imaging techniques have shown altered myocardial energetics already in preclinical gene variant carriers. To determine whether disturbed myocardial energetics with the potential to serve as biomarkers are also reflected in the serum metabolome, we analyzed the serum metabolome of asymptomatic carriers in comparison to healthy controls and obstructive HCM patients (HOCM). We performed non-quantitative direct-infusion high-resolution mass spectrometry-based untargeted metabolomics on serum from fasted asymptomatic gene variant carriers, symptomatic HOCM patients and healthy controls (n = 31, 14 and 9, respectively). Biomarker panels that discriminated the groups were identified by performing multivariate modeling with gradient-boosting classifiers. For all three group-wise comparisons we identified a panel of 30 serum metabolites that best discriminated the groups. These metabolite panels performed equally well as advanced cardiac imaging modalities in distinguishing the groups. Seven metabolites were found to be predictive in two different comparisons and may play an important role in defining the disease stage. This study reveals unique metabolic signatures in serum of preclinical carriers and HOCM patients that may potentially be used for HCM risk stratification and precision therapeutics.
Collapse
|
48
|
Ion Channel Impairment and Myofilament Ca 2+ Sensitization: Two Parallel Mechanisms Underlying Arrhythmogenesis in Hypertrophic Cardiomyopathy. Cells 2021; 10:cells10102789. [PMID: 34685769 PMCID: PMC8534456 DOI: 10.3390/cells10102789] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
Life-threatening ventricular arrhythmias are the main clinical burden in patients with hypertrophic cardiomyopathy (HCM), and frequently occur in young patients with mild structural disease. While massive hypertrophy, fibrosis and microvascular ischemia are the main mechanisms underlying sustained reentry-based ventricular arrhythmias in advanced HCM, cardiomyocyte-based functional arrhythmogenic mechanisms are likely prevalent at earlier stages of the disease. In this review, we will describe studies conducted in human surgical samples from HCM patients, transgenic animal models and human cultured cell lines derived from induced pluripotent stem cells. Current pieces of evidence concur to attribute the increased risk of ventricular arrhythmias in early HCM to different cellular mechanisms. The increase of late sodium current and L-type calcium current is an early observation in HCM, which follows post-translation channel modifications and increases the occurrence of early and delayed afterdepolarizations. Increased myofilament Ca2+ sensitivity, commonly observed in HCM, may promote afterdepolarizations and reentry arrhythmias with direct mechanisms. Decrease of K+-currents due to transcriptional regulation occurs in the advanced disease and contributes to reducing the repolarization-reserve and increasing the early afterdepolarizations (EADs). The presented evidence supports the idea that patients with early-stage HCM should be considered and managed as subjects with an acquired channelopathy rather than with a structural cardiac disease.
Collapse
|
49
|
Hassoun R, Budde H, Mügge A, Hamdani N. Cardiomyocyte Dysfunction in Inherited Cardiomyopathies. Int J Mol Sci 2021; 22:11154. [PMID: 34681814 PMCID: PMC8541428 DOI: 10.3390/ijms222011154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 01/10/2023] Open
Abstract
Inherited cardiomyopathies form a heterogenous group of disorders that affect the structure and function of the heart. Defects in the genes encoding sarcomeric proteins are associated with various perturbations that induce contractile dysfunction and promote disease development. In this review we aimed to outline the functional consequences of the major inherited cardiomyopathies in terms of myocardial contraction and kinetics, and to highlight the structural and functional alterations in some sarcomeric variants that have been demonstrated to be involved in the pathogenesis of the inherited cardiomyopathies. A particular focus was made on mutation-induced alterations in cardiomyocyte mechanics. Since no disease-specific treatments for familial cardiomyopathies exist, several novel agents have been developed to modulate sarcomere contractility. Understanding the molecular basis of the disease opens new avenues for the development of new therapies. Furthermore, the earlier the awareness of the genetic defect, the better the clinical prognostication would be for patients and the better the prevention of development of the disease.
Collapse
Affiliation(s)
- Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
50
|
Ma W, Henze M, Anderson RL, Gong H, Wong FL, Del Rio CL, Irving T. The Super-Relaxed State and Length Dependent Activation in Porcine Myocardium. Circ Res 2021; 129:617-630. [PMID: 34365814 DOI: 10.1161/circresaha.120.318647] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Weikang Ma
- BioCAT, Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.G., T.I.)
| | - Marcus Henze
- MyoKardia Inc, Brisbane, CA (M.H., R.L.A., F.L.W., C.L.d.R.)
| | | | - Henry Gong
- BioCAT, Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.G., T.I.)
| | - Fiona L Wong
- MyoKardia Inc, Brisbane, CA (M.H., R.L.A., F.L.W., C.L.d.R.)
| | | | - Thomas Irving
- BioCAT, Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.G., T.I.)
| |
Collapse
|