1
|
Zhang K, Yuan Z, Wang S, Zhao S, Cui H, Lai Y. The abnormalities of free fatty acid metabolism in patients with hypertrophic cardiomyopathy, a single-center retrospective observational study. BMC Cardiovasc Disord 2024; 24:312. [PMID: 38902636 PMCID: PMC11188237 DOI: 10.1186/s12872-024-03925-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 05/06/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Previous studies have shown the importance of energy deficiency and malfunctioning mitochondria in the pathophysiology of hypertrophic cardiomyopathy (HCM). There has been a little research into the relationship between plasma free fatty acids (FFA), one of the heart's main energy sources, and HCM. We evaluated its clinical importance in HCM to see if there was a link between plasma FFA metabolism and HCM. METHODS In a single-center retrospective observational study, we investigated 420 HCM patients diagnosed at Beijing Anzhen Hospital between January 1, 2018, and December 31, 2022. Meanwhile, 1372 individuals without HCM (non-HCM) were recruited. 391 non-HCM patients were chosen as controls via a propensity score matching (PSM) study with a 1:1 ratio. RESULTS FFA in HCM patients showed statistically significant correlations with creatinine (r = 0.115, p = 0.023), estimated GFR (r=-0.130, p = 0.010), BNP (r = 0.152, p = 0.007), LVEF (r=-0.227, p < 0.001), LVFS (r=-0.160, p = 0.002), and LAD (r = 0.112, p = 0.028). Higher FFA levels were found in HCM patients who had atrial fibrillation and NYHY functional classes III or IV (p = 0.015 and p = 0.022, respectively). In HCM patients, multiple linear regression analysis revealed that BNP and LVEF had independent relationships with increasing FFA (Standardized = 0.139, p = 0.013 and =-0.196, p < 0.001, respectively). CONCLUSIONS Among HCM patients, the plasma FFA concentration was lower, and those with AF and NYHY functional class III or IV had higher FFA levels, and LVEF and BNP were independently associated with increasing FFA. The findings of the study should help inspire future efforts to better understand how energy deficiency contributes to hypertrophic cardiomyopathy (HCM) development.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Chaoyang District, Box: 100011, Beijing, China
- Beijing Anzhen Hospital, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Ministry of Education, Beijing, 100029, China
| | - Zhongyu Yuan
- Beijing Anzhen Hospital, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Ministry of Education, Beijing, 100029, China
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shengwei Wang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Chaoyang District, Box: 100011, Beijing, China
- Beijing Anzhen Hospital, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Ministry of Education, Beijing, 100029, China
| | - Shifeng Zhao
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Chaoyang District, Box: 100011, Beijing, China
- Beijing Anzhen Hospital, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Ministry of Education, Beijing, 100029, China
| | - Hao Cui
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Chaoyang District, Box: 100011, Beijing, China
- Beijing Anzhen Hospital, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Ministry of Education, Beijing, 100029, China
| | - Yongqiang Lai
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Chaoyang District, Box: 100011, Beijing, China.
- Beijing Anzhen Hospital, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Ministry of Education, Beijing, 100029, China.
| |
Collapse
|
2
|
Sequeira V, Maack C, Reil GH, Reil JC. Exploring the Connection Between Relaxed Myosin States and the Anrep Effect. Circ Res 2024; 134:117-134. [PMID: 38175910 DOI: 10.1161/circresaha.123.323173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
The Anrep effect is an adaptive response that increases left ventricular contractility following an acute rise in afterload. Although the mechanistic origin remains undefined, recent findings suggest a two-phase activation of resting myosin for contraction, involving strain-sensitive and posttranslational phases. We propose that this mobilization represents a transition among the relaxed states of myosin-specifically, from the super-relaxed (SRX) to the disordered-relaxed (DRX)-with DRX myosin ready to participate in force generation. This hypothesis offers a unified explanation that connects myosin's SRX-DRX equilibrium and the Anrep effect as parts of a singular phenomenon. We underscore the significance of this equilibrium in modulating contractility, primarily studied in the context of hypertrophic cardiomyopathy, the most common inherited cardiomyopathy associated with diastolic dysfunction, hypercontractility, and left ventricular hypertrophy. As we posit that the cellular basis of the Anrep effect relies on a two-phased transition of myosin from the SRX to the contraction-ready DRX configuration, any dysregulation in this equilibrium may result in the pathological manifestation of the Anrep phenomenon. For instance, in hypertrophic cardiomyopathy, hypercontractility is linked to a considerable shift of myosin to the DRX state, implying a persistent activation of the Anrep effect. These valuable insights call for additional research to uncover a clinical Anrep fingerprint in pathological states. Here, we demonstrate through noninvasive echocardiographic pressure-volume measurements that this fingerprint is evident in 12 patients with hypertrophic obstructive cardiomyopathy before septal myocardial ablation. This unique signature is characterized by enhanced contractility, indicated by a leftward shift and steepening of the end-systolic pressure-volume relationship, and a prolonged systolic ejection time adjusted for heart rate, which reverses post-procedure. The clinical application of this concept has potential implications beyond hypertrophic cardiomyopathy, extending to other genetic cardiomyopathies and even noncongenital heart diseases with complex etiologies across a broad spectrum of left ventricular ejection fractions.
Collapse
Affiliation(s)
- Vasco Sequeira
- Department of Translational Science Universitätsklinikum, Deutsche Zentrum für Herzinsuffizienz (DZHI), Würzburg, Germany (V.S., C.M.)
| | - Christoph Maack
- Department of Translational Science Universitätsklinikum, Deutsche Zentrum für Herzinsuffizienz (DZHI), Würzburg, Germany (V.S., C.M.)
| | - Gert-Hinrich Reil
- Klinik für Kardiologie, Klinikum Oldenburg, Innere Medizin I, Germany (G.-H.R.)
| | - Jan-Christian Reil
- Klinik für Allgemeine und Interventionelle Kardiologie, Herz- und Diabetes-Zentrum Nordrhein-Westphalen, Germany (J.-C.R.)
| |
Collapse
|
3
|
Gupta A. Cardiac 31P MR spectroscopy: development of the past five decades and future vision-will it be of diagnostic use in clinics? Heart Fail Rev 2023; 28:485-532. [PMID: 36427161 DOI: 10.1007/s10741-022-10287-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
In the past five decades, the use of the magnetic resonance (MR) technique for cardiovascular diseases has engendered much attention and raised the opportunity that the technique could be useful for clinical applications. MR has two arrows in its quiver: One is magnetic resonance imaging (MRI), and the other is magnetic resonance spectroscopy (MRS). Non-invasively, highly advanced MRI provides unique and profound information about the anatomical changes of the heart. Excellently developed MRS provides irreplaceable and insightful evidence of the real-time biochemistry of cardiac metabolism of underpinning diseases. Compared to MRI, which has already been successfully applied in routine clinical practice, MRS still has a long way to travel to be incorporated into routine diagnostics. Considering the exceptional potential of 31P MRS to measure the real-time metabolic changes of energetic molecules qualitatively and quantitatively, how far its powerful technique should be waited before a successful transition from "bench-to-bedside" is enticing. The present review highlights the seminal studies on the chronological development of cardiac 31P MRS in the past five decades and the future vision and challenges to incorporating it for routine diagnostics of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Gupta
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow, 226014, India.
| |
Collapse
|
4
|
Marston S, Pinto JR. Suppression of lusitropy as a disease mechanism in cardiomyopathies. Front Cardiovasc Med 2023; 9:1080965. [PMID: 36698941 PMCID: PMC9870330 DOI: 10.3389/fcvm.2022.1080965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
In cardiac muscle the action of adrenaline on β1 receptors of heart muscle cells is essential to adjust cardiac output to the body's needs. Adrenergic activation leads to enhanced contractility (inotropy), faster heart rate (chronotropy) and faster relaxation (lusitropy), mainly through activation of protein kinase A (PKA). Efficient enhancement of heart output under stress requires all of these responses to work together. Lusitropy is essential for shortening the heartbeat when heart rate increases. It therefore follows that, if the lusitropic response is not present, heart function under stress will be compromised. Current literature suggests that lusitropy is primarily achieved due to PKA phosphorylation of troponin I (TnI) and phospholamban (PLB). It has been well documented that PKA-induced phosphorylation of TnI releases Ca2+ from troponin C faster and increases the rate of cardiac muscle relaxation, while phosphorylation of PLB increases SERCA activity, speeding up Ca2+ removal from the cytoplasm. In this review we consider the current scientific evidences for the connection between suppression of lusitropy and cardiac dysfunction in the context of mutations in phospholamban and thin filament proteins that are associated with cardiomyopathies. We will discuss what advances have been made into understanding the physiological mechanism of lusitropy due to TnI and PLB phosphorylation and its suppression by mutations and we will evaluate the evidence whether lack of lusitropy is sufficient to cause cardiomyopathy, and under what circumstances, and consider the range of pathologies associated with loss of lusitropy. Finally, we will discuss whether suppressed lusitropy due to mutations in thin filament proteins can be therapeutically restored.
Collapse
Affiliation(s)
- Steven Marston
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
5
|
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: 32] [Impact Index Per Article: 16.0] [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
|
6
|
Pioner JM, Vitale G, Gentile F, Scellini B, Piroddi N, Cerbai E, Olivotto I, Tardiff J, Coppini R, Tesi C, Poggesi C, Ferrantini C. Genotype-Driven Pathogenesis of Atrial Fibrillation in Hypertrophic Cardiomyopathy: The Case of Different TNNT2 Mutations. Front Physiol 2022; 13:864547. [PMID: 35514357 PMCID: PMC9062294 DOI: 10.3389/fphys.2022.864547] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/28/2022] [Indexed: 11/25/2022] Open
Abstract
Atrial dilation and atrial fibrillation (AF) are common in Hypertrophic CardioMyopathy (HCM) patients and associated with a worsening of prognosis. The pathogenesis of atrial myopathy in HCM remains poorly investigated and no specific association with genotype has been identified. By re-analysis of our cohort of thin-filament HCM patients (Coppini et al. 2014) AF was identified in 10% of patients with sporadic mutations in the cardiac Troponin T gene (TNNT2), while AF occurrence was much higher (25-75%) in patients carrying specific "hot-spot" TNNT2 mutations. To determine the molecular basis of arrhythmia occurrence, two HCM mouse models expressing human TNNT2 variants (a "hot-spot" one, R92Q, and a "sporadic" one, E163R) were selected according to the different pathophysiological pathways previously demonstrated in ventricular tissue. Echocardiography studies showed a significant left atrial dilation in both models, but more pronounced in the R92Q. In E163R atrial trabeculae, in line with what previously observed in ventricular preparations, the energy cost of tension generation was markedly increased. However, no changes of twitch amplitude and kinetics were observed, and there was no atrial arrhythmic propensity. R92Q atrial trabeculae, instead, displayed normal ATP consumption but markedly increased myofilament calcium sensitivity, as previously observed in ventricular preparations. This was associated with reduced inotropic reserve and slower kinetics of twitch contractions and, importantly, with an increased occurrence of spontaneous beats and triggered contractions that represent an intrinsic arrhythmogenic mechanism promoting AF. The association of specific TNNT2 mutations with AF occurrence depends on the mutation-driven pathomechanism (i.e., increased atrial myofilament calcium sensitivity rather than increased myofilament tension cost) and may influence the individual response to treatment.
Collapse
Affiliation(s)
| | - Giulia Vitale
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesca Gentile
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Jil Tardiff
- Department of Medicine and Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
7
|
Harbo MB, Stokke MK, Sjaastad I, Espe EKS. One step closer to myocardial physiology: From PV loop analysis to state-of-the-art myocardial imaging. Acta Physiol (Oxf) 2022; 234:e13759. [PMID: 34978759 DOI: 10.1111/apha.13759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/14/2021] [Accepted: 01/01/2022] [Indexed: 11/29/2022]
Abstract
Recent advances in cardiac imaging have revitalized the assessment of fundamental physiological concepts. In the field of cardiac physiology, invasive measurements with pressure-volume (PV) loops have served as the gold standard methodology for the characterization of left ventricular (LV) function. From PV loop data, fundamental aspects of LV chamber function are derived such as work, efficiency, stiffness and contractility. However, the parametrization of these aspects is limited because of the need for invasive procedures. Through the utilization of recent advances in echocardiography, magnetic resonance imaging and positron emission tomography, it has become increasingly feasible to quantify these fundamental aspects of LV function non-invasively. Importantly, state-of-the-art imaging technology enables direct assessment of myocardial performance, thereby extending functional assessment from the net function of the LV chamber, as is done with PV loops, to the myocardium itself. With a strong coupling to underlying myocardial physiology, imaging measurements of myocardial work, efficiency, stiffness and contractility could represent the next generation of functional parameters. The purpose of this review is to discuss how the new imaging parameters of myocardial work, efficiency, stiffness and contractility can bring cardiac physiologists, researchers and clinicians alike one step closer to underlying myocardial physiology.
Collapse
Affiliation(s)
- Markus Borge Harbo
- Institute for Experimental Medical Research Oslo University Hospital and University of Oslo Oslo Norway
- K.G. Jebsen Center for Cardiac Research University of Oslo Oslo Norway
| | - Mathis Korseberg Stokke
- Institute for Experimental Medical Research Oslo University Hospital and University of Oslo Oslo Norway
- K.G. Jebsen Center for Cardiac Research University of Oslo Oslo Norway
- Department of Cardiology Oslo University Hospital Rikshospitalet Oslo Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research Oslo University Hospital and University of Oslo Oslo Norway
- K.G. Jebsen Center for Cardiac Research University of Oslo Oslo Norway
| | - Emil Knut Stenersen Espe
- Institute for Experimental Medical Research Oslo University Hospital and University of Oslo Oslo Norway
- K.G. Jebsen Center for Cardiac Research University of Oslo Oslo Norway
| |
Collapse
|
8
|
Ranjbarvaziri S, Kooiker KB, Ellenberger M, Fajardo G, Zhao M, Vander Roest AS, Woldeyes RA, Koyano TT, Fong R, Ma N, Tian L, Traber GM, Chan F, Perrino J, Reddy S, Chiu W, Wu JC, Woo JY, Ruppel KM, Spudich JA, Snyder MP, Contrepois K, Bernstein D. Altered Cardiac Energetics and Mitochondrial Dysfunction in Hypertrophic Cardiomyopathy. Circulation 2021; 144:1714-1731. [PMID: 34672721 PMCID: PMC8608736 DOI: 10.1161/circulationaha.121.053575] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/24/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a complex disease partly explained by the effects of individual gene variants on sarcomeric protein biomechanics. At the cellular level, HCM mutations most commonly enhance force production, leading to higher energy demands. Despite significant advances in elucidating sarcomeric structure-function relationships, there is still much to be learned about the mechanisms that link altered cardiac energetics to HCM phenotypes. In this work, we test the hypothesis that changes in cardiac energetics represent a common pathophysiologic pathway in HCM. METHODS We performed a comprehensive multiomics profile of the molecular (transcripts, metabolites, and complex lipids), ultrastructural, and functional components of HCM energetics using myocardial samples from 27 HCM patients and 13 normal controls (donor hearts). RESULTS Integrated omics analysis revealed alterations in a wide array of biochemical pathways with major dysregulation in fatty acid metabolism, reduction of acylcarnitines, and accumulation of free fatty acids. HCM hearts showed evidence of global energetic decompensation manifested by a decrease in high energy phosphate metabolites (ATP, ADP, and phosphocreatine) and a reduction in mitochondrial genes involved in creatine kinase and ATP synthesis. Accompanying these metabolic derangements, electron microscopy showed an increased fraction of severely damaged mitochondria with reduced cristae density, coinciding with reduced citrate synthase activity and mitochondrial oxidative respiration. These mitochondrial abnormalities were associated with elevated reactive oxygen species and reduced antioxidant defenses. However, despite significant mitochondrial injury, HCM hearts failed to upregulate mitophagic clearance. CONCLUSIONS Overall, our findings suggest that perturbed metabolic signaling and mitochondrial dysfunction are common pathogenic mechanisms in patients with HCM. These results highlight potential new drug targets for attenuation of the clinical disease through improving metabolic function and reducing mitochondrial injury.
Collapse
Affiliation(s)
- Sara Ranjbarvaziri
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristina B. Kooiker
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA, USA
| | - Mathew Ellenberger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Giovanni Fajardo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Mingming Zhao
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Alison Schroer Vander Roest
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Rahel A. Woldeyes
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Robyn Fong
- Department of Cardiothoracic Surgery, Stanford University, CA, USA
| | - Ning Ma
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
| | - Lei Tian
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
| | - Gavin M. Traber
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Frandics Chan
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - John Perrino
- Cell Sciences Imaging Facility, Stanford University, Stanford, CA, USA
| | - Sushma Reddy
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Wah Chiu
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Division of Cryo-EM and Bioimaging, SLAC National Accelerator Laboratory, Stanford University, Stanford, CA, USA
| | - Joseph C. Wu
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
| | - Joseph Y. Woo
- Department of Cardiothoracic Surgery, Stanford University, CA, USA
| | - Kathleen M. Ruppel
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Bernstein
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Research Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
9
|
Ladha FA, Thakar K, Pettinato AM, Legere N, Ghahremani S, Cohn R, Romano R, Meredith E, Chen YS, Hinson JT. Actinin BioID reveals sarcomere crosstalk with oxidative metabolism through interactions with IGF2BP2. Cell Rep 2021; 36:109512. [PMID: 34380038 PMCID: PMC8447243 DOI: 10.1016/j.celrep.2021.109512] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 05/16/2021] [Accepted: 07/21/2021] [Indexed: 01/13/2023] Open
Abstract
Actinins are strain-sensing actin cross-linkers that are ubiquitously expressed and harbor mutations in human diseases. We utilize CRISPR, pluripotent stem cells, and BioID to study actinin interactomes in human cardiomyocytes. We identify 324 actinin proximity partners, including those that are dependent on sarcomere assembly. We confirm 19 known interactors and identify a network of RNA-binding proteins, including those with RNA localization functions. In vivo and biochemical interaction studies support that IGF2BP2 localizes electron transport chain transcripts to actinin neighborhoods through interactions between its K homology (KH) domain and actinin’s rod domain. We combine alanine scanning mutagenesis and metabolic assays to disrupt and functionally interrogate actinin-IGF2BP2 interactions, which reveal an essential role in metabolic responses to pathological sarcomere activation using a hypertrophic cardiomyopathy model. This study expands our functional knowledge of actinin, uncovers sarcomere interaction partners, and reveals sarcomere crosstalk with IGF2BP2 for metabolic adaptation relevant to human disease. Ladha et al. combine BioID, human cardiomyocytes, and CRISPR-Cas9 to interrogate the actinin interactome. This reveals 324 actinin proximity partners, including RNA-binding proteins that bind transcripts encoding ETC functional components. Among these RNA-binding proteins, IGF2BP2 directly binds actinin, and actinin-IGF2BP2 interactions regulate ETC transcript localization and metabolic adaptation to sarcomere function.
Collapse
Affiliation(s)
- Feria A Ladha
- University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Ketan Thakar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | | | - Nicholas Legere
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | | | - Rachel Cohn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Robert Romano
- University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Emily Meredith
- University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Yu-Sheng Chen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - J Travis Hinson
- University of Connecticut Health Center, Farmington, CT 06030, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Cardiology Center, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
10
|
Greenberg MJ, Tardiff JC. Complexity in genetic cardiomyopathies and new approaches for mechanism-based precision medicine. J Gen Physiol 2021; 153:e202012662. [PMID: 33512404 PMCID: PMC7852459 DOI: 10.1085/jgp.202012662] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Genetic cardiomyopathies have been studied for decades, and it has become increasingly clear that these progressive diseases are more complex than originally thought. These complexities can be seen both in the molecular etiologies of these disorders and in the clinical phenotypes observed in patients. While these disorders can be caused by mutations in cardiac genes, including ones encoding sarcomeric proteins, the disease presentation varies depending on the patient mutation, where mutations even within the same gene can cause divergent phenotypes. Moreover, it is challenging to connect the mutation-induced molecular insult that drives the disease pathogenesis with the various compensatory and maladaptive pathways that are activated during the course of the subsequent progressive, pathogenic cardiac remodeling. These inherent complexities have frustrated our ability to understand and develop broadly effective treatments for these disorders. It has been proposed that it might be possible to improve patient outcomes by adopting a precision medicine approach. Here, we lay out a practical framework for such an approach, where patient subpopulations are binned based on common underlying biophysical mechanisms that drive the molecular disease pathogenesis, and we propose that this function-based approach will enable the development of targeted therapeutics that ameliorate these effects. We highlight several mutations to illustrate the need for mechanistic molecular experiments that span organizational and temporal scales, and we describe recent advances in the development of novel therapeutics based on functional targets. Finally, we describe many of the outstanding questions for the field and how fundamental mechanistic studies, informed by our more nuanced understanding of the clinical disorders, will play a central role in realizing the potential of precision medicine for genetic cardiomyopathies.
Collapse
Affiliation(s)
- Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
| | - Jil C. Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ
- Department of Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
11
|
Parbhudayal RY, Harms HJ, Michels M, van Rossum AC, Germans T, van der Velden J. Increased Myocardial Oxygen Consumption Precedes Contractile Dysfunction in Hypertrophic Cardiomyopathy Caused by Pathogenic TNNT2 Gene Variants. J Am Heart Assoc 2020; 9:e015316. [PMID: 32290750 PMCID: PMC7428531 DOI: 10.1161/jaha.119.015316] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Hypertrophic cardiomyopathy is caused by pathogenic sarcomere gene variants. Individuals with a thin‐filament variant present with milder hypertrophy than carriers of thick‐filament variants, although prognosis is poorer. Herein, we defined if decreased energetic status of the heart is an early pathomechanism in TNNT2 (troponin T gene) variant carriers. Methods and Results Fourteen individuals with TNNT2 variants (genotype positive), without left ventricular hypertrophy (G+/LVH−; n=6) and with LVH (G+/LVH+; n=8) and 14 healthy controls were included. All participants underwent cardiac magnetic resonance and [11C]‐acetate positron emission tomography imaging to assess LV myocardial oxygen consumption, contractile parameters and myocardial external efficiency. Cardiac efficiency was significantly reduced compared with controls in G+/LVH− and G+/LVH+. Lower myocardial external efficiency in G+/LVH− is explained by higher global and regional oxygen consumption compared with controls without changes in contractile parameters. Reduced myocardial external efficiency in G+/LVH+ is explained by the increase in LV mass and higher oxygen consumption. Septal oxygen consumption was significantly lower in G+/LVH+ compared with G+/LVH−. Although LV ejection fraction was higher in G+/LVH+, both systolic and diastolic strain parameters were lower compared with controls, which was most evident in the hypertrophied septal wall. Conclusions Using cardiac magnetic resonance and [11C]‐acetate positron emission tomography imaging, we show that G+/LVH− have an initial increase in oxygen consumption preceding contractile dysfunction and cardiac hypertrophy, followed by a decline in oxygen consumption in G+/LVH+. This suggests that high oxygen consumption and reduced myocardial external efficiency characterize the early gene variant–mediated disease mechanisms that may be used for early diagnosis and development of preventive treatments.
Collapse
Affiliation(s)
- Rahana Y Parbhudayal
- Department of Cardiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands.,Department of Physiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands.,The Netherlands Heart Institute Utrecht the Netherlands
| | - Hendrik J Harms
- Department of Nuclear Medicine and PET Center Aarhus University Aarhus Denmark
| | - Michelle Michels
- Department of Cardiology Erasmus Medical Center Rotterdam the Netherlands
| | - Albert C van Rossum
- Department of Cardiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands
| | - Tjeerd Germans
- Department of Cardiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands
| | - Jolanda van der Velden
- Department of Physiology Amsterdam University Medical Center Amsterdam Cardiovascular Sciences Vrije Universiteit University Medical Center Amsterdam Amsterdam the Netherlands.,The Netherlands Heart Institute Utrecht the Netherlands
| |
Collapse
|
12
|
Liu Y, Afzal J, Vakrou S, Greenland GV, Talbot CC, Hebl VB, Guan Y, Karmali R, Tardiff JC, Leinwand LA, Olgin JE, Das S, Fukunaga R, Abraham MR. Differences in microRNA-29 and Pro-fibrotic Gene Expression in Mouse and Human Hypertrophic Cardiomyopathy. Front Cardiovasc Med 2019; 6:170. [PMID: 31921893 PMCID: PMC6928121 DOI: 10.3389/fcvm.2019.00170] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Hypertrophic cardiomyopathy (HCM) is characterized by myocyte hypertrophy and fibrosis. Studies in two mouse models (R92W-TnT/R403Q-MyHC) at early HCM stage revealed upregulation of endothelin (ET1) signaling in both mutants, but TGFβ signaling only in TnT mutants. Dysregulation of miR-29 expression has been implicated in cardiac fibrosis. But it is unknown whether expression of miR-29a/b/c and profibrotic genes is commonly regulated in mouse and human HCM. Methods: In order to understand mechanisms underlying fibrosis in HCM, and examine similarities/differences in expression of miR-29a/b/c and several profibrotic genes in mouse and human HCM, we performed parallel studies in rat cardiac myocyte/fibroblast cultures, examined gene expression in two mouse models of (non-obstructive) HCM (R92W-TnT, R403Q-MyHC)/controls at early (5 weeks) and established (24 weeks) disease stage, and analyzed publicly available mRNA/miRNA expression data from obstructive-HCM patients undergoing septal myectomy/controls (unused donor hearts). Results: Myocyte cultures: ET1 increased superoxide/H2O2, stimulated TGFβ expression/secretion, and suppressed miR-29a expression in myocytes. The effect of ET1 on miR-29 and TGFβ expression/secretion was antagonized by N-acetyl-cysteine, a reactive oxygen species scavenger. Fibroblast cultures: ET1 had no effect on pro-fibrotic gene expression in fibroblasts. TGFβ1/TGFβ2 suppressed miR-29a and increased collagen expression, which was abolished by miR-29a overexpression. Mouse and human HCM: Expression of miR-29a/b/c was lower, and TGFB1/collagen gene expression was higher in TnT mutant-LV at 5 and 24 weeks; no difference was observed in expression of these genes in MyHC mutant-LV and in human myectomy tissue. TGFB2 expression was higher in LV of both mutant mice and human myectomy tissue. ACE2, a negative regulator of the renin-angiotensin-aldosterone system, was the most upregulated transcript in human myectomy tissue. Pathway analysis predicted upregulation of the anti-hypertrophic/anti-fibrotic liver X receptor/retinoid X receptor (LXR/RXR) pathway only in human myectomy tissue. Conclusions: Our in vitro studies suggest that activation of ET1 signaling in cardiac myocytes increases reactive oxygen species and stimulates TGFβ secretion, which downregulates miR-29a and increases collagen in fibroblasts, thus contributing to fibrosis. Our gene expression studies in mouse and human HCM reveal allele-specific differences in miR-29 family/profibrotic gene expression in mouse HCM, and activation of anti-hypertrophic/anti-fibrotic genes and pathways in human HCM.
Collapse
Affiliation(s)
- Yamin Liu
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States.,Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - Junaid Afzal
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States.,Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - Styliani Vakrou
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - Gabriela V Greenland
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States.,Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - C Conover Talbot
- Johns Hopkins School of Medicine, Institute for Basic Biomedical Sciences, Baltimore, MD, United States
| | - Virginia B Hebl
- Intermountain Medical Center, Intermountain Heart Institute, Murray, UT, United States
| | - Yufan Guan
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| | - Rehan Karmali
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States
| | - Jil C Tardiff
- Sarver Heart Center, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Leslie A Leinwand
- Molecular, Cellular and Developmental Biology, Biofrontiers Institute, University of Colorado, Boulder, CO, United States
| | - Jeffrey E Olgin
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States
| | - Samarjit Das
- Department of Anesthesia and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - M Roselle Abraham
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, San Francisco, CA, United States.,Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
13
|
Gannon MP, Link MS. Phenotypic variation and targeted therapy of hypertrophic cardiomyopathy using genetic animal models. Trends Cardiovasc Med 2019; 31:20-31. [PMID: 31862214 DOI: 10.1016/j.tcm.2019.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/14/2019] [Accepted: 11/19/2019] [Indexed: 12/25/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) has a variable clinical presentation due to the diversity of causative genetic mutations. Animal models allow in vivo study of genotypic expression through non-invasive imaging, pathologic sampling, and force analysis. This review focuses on the spontaneous and induced mutations in various animal models affecting mainly sarcomere proteins. The sarcomere is comprised of thick (myosin) filaments and related proteins including myosin heavy chain and myosin binding protein-C; thin (actin) filament proteins and their associated regulators including tropomyosin, troponin I, troponin C, and troponin T. The regulatory milieu including transcription factors and cell signaling also play a significant role. Animal models provide a layered approach of understanding beginning with the causative mutation as a foundation. The functional consequences of protein energy utilization and calcium sensitivity in vivo and ex vivo can be studied. Beyond pathophysiologic disruption of sarcomere function, these models demonstrate the clinical sequalae of diastolic dysfunction, heart failure, and arrhythmogenic death. Through this cascade of understanding the mutation followed by their functional significance, targeted therapies have been developed and are briefly discussed.
Collapse
Affiliation(s)
- Michael P Gannon
- National Heart, Lung and Blood Institute, National Institutes of Health, US Department of Health and Human Services, Bldg 10, Rm B1D416, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Mark S Link
- University of Texas Southwestern Medical Center, USA
| |
Collapse
|
14
|
The relation between sarcomere energetics and the rate of isometric tension relaxation in healthy and diseased cardiac muscle. J Muscle Res Cell Motil 2019; 42:47-57. [PMID: 31745760 PMCID: PMC7932984 DOI: 10.1007/s10974-019-09566-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/13/2019] [Indexed: 12/24/2022]
Abstract
Full muscle relaxation happens when [Ca2+] falls below the threshold for force activation. Several experimental models, from whole muscle organs and intact muscle down to skinned fibers, have been used to explore the cascade of kinetic events leading to mechanical relaxation. The use of single myofibrils together with fast solution switching techniques, has provided new information about the role of cross-bridge (CB) dissociation in the time course of isometric force decay. Myofibril’s relaxation is biphasic starting with a slow seemingly linear phase, with a rate constant, slow kREL, followed by a fast mono-exponential phase. Sarcomeres remain isometric during the slow force decay that reflects CB detachment under isometric conditions while the final fast relaxation phase begins with a sudden give of few sarcomeres and is then dominated by intersarcomere dynamics. Based on a simple two-state model of the CB cycle, myofibril slow kREL represents the apparent forward rate with which CBs leave force generating states (gapp) under isometric conditions and correlates with the energy cost of tension generation (ATPase/tension ratio); in short slow kREL ~ gapp ~ tension cost. The validation of this relationship is obtained by simultaneously measuring maximal isometric force and ATP consumption in skinned myocardial strips that provide an unambiguous determination of the relation between contractile and energetic properties of the sarcomere. Thus, combining kinetic experiments in isolated myofibrils and mechanical and energetic measurements in multicellular cardiac strips, we are able to provide direct evidence for a positive linear correlation between myofibril isometric relaxation kinetics (slow kREL) and the energy cost of force production both measured in preparations from the same cardiac sample. This correlation remains true among different types of muscles with different ATPase activities and also when CB kinetics are altered by cardiomyopathy-related mutations. Sarcomeric mutations associated to hypertrophic cardiomyopathy (HCM), a primary cardiac disorder caused by mutations in genes encoding sarcomeric proteins, have been often found to accelerate CB turnover rate and increase the energy cost of myocardial contraction. Here we review data showing that faster CB detachment results in a proportional increase in the energetic cost of tension generation in heart samples from both HCM patients and mouse models of the disease.
Collapse
|
15
|
Wijnker PJ, Sequeira V, Kuster DW, van der Velden J. Hypertrophic Cardiomyopathy: A Vicious Cycle Triggered by Sarcomere Mutations and Secondary Disease Hits. Antioxid Redox Signal 2019; 31:318-358. [PMID: 29490477 PMCID: PMC6602117 DOI: 10.1089/ars.2017.7236] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 02/23/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
Abstract
Significance: Hypertrophic cardiomyopathy (HCM) is a cardiac genetic disease characterized by left ventricular hypertrophy, diastolic dysfunction, and myocardial disarray. Disease onset occurs between 20 and 50 years of age, thus affecting patients in the prime of their life. HCM is caused by mutations in sarcomere proteins, the contractile building blocks of the heart. Despite increased knowledge of causal mutations, the exact path from genetic defect leading to cardiomyopathy is complex and involves additional disease hits. Recent Advances: Laboratory-based studies indicate that HCM development not only depends on the primary sarcomere impairment caused by the mutation but also on secondary disease-related alterations in the heart. Here we propose a vicious mutation-induced disease cycle, in which a mutation-induced energy depletion alters cellular metabolism with increased mitochondrial work, which triggers secondary disease modifiers that will worsen disease and ultimately lead to end-stage HCM. Critical Issues: Evidence shows excessive cellular reactive oxygen species (ROS) in HCM patients and HCM animal models. Oxidative stress markers are increased in the heart (oxidized proteins, DNA, and lipids) and serum of HCM patients. In addition, increased mitochondrial ROS production and changes in endogenous antioxidants are reported in HCM. Mutant sarcomeric protein may drive excessive levels of cardiac ROS via changes in cardiac efficiency and metabolism, mitochondrial activation and/or dysfunction, impaired protein quality control, and microvascular dysfunction. Future Directions: Interventions restoring metabolism, mitochondrial function, and improved ROS balance may be promising therapeutic approaches. We discuss the effects of current HCM pharmacological therapies and potential future therapies to prevent and reverse HCM. Antioxid. Redox Signal. 31, 318-358.
Collapse
Affiliation(s)
- Paul J.M. Wijnker
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Vasco Sequeira
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Diederik W.D. Kuster
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
16
|
Sequeira V, Bertero E, Maack C. Energetic drain driving hypertrophic cardiomyopathy. FEBS Lett 2019; 593:1616-1626. [PMID: 31209876 DOI: 10.1002/1873-3468.13496] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 01/09/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common form of hereditary cardiomyopathy and is mainly caused by mutations of genes encoding cardiac sarcomeric proteins. HCM is characterized by hypertrophy of the left ventricle, frequently involving the septum, that is not explained solely by loading conditions. HCM has a heterogeneous clinical profile, but diastolic dysfunction and ventricular arrhythmias represent two dominant features of the disease. Preclinical evidence indicates that the enhanced Calcium (Ca2+ ) sensitivity of the myofilaments plays a key role in the pathophysiology of HCM. Notably, this is not always a direct consequence of sarcomeric mutations, but can also result from secondary mutation-driven alterations. Here, we review experimental and clinical evidence indicating that increased myofilament Ca2+ sensitivity lies upstream of numerous cellular derangements which potentially contribute to the progression of HCM toward heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Vasco Sequeira
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Germany
| | - Edoardo Bertero
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Germany
| |
Collapse
|
17
|
Viola HM, Hool LC. Impaired calcium handling and mitochondrial metabolic dysfunction as early markers of hypertrophic cardiomyopathy. Arch Biochem Biophys 2019; 665:166-174. [PMID: 30885674 DOI: 10.1016/j.abb.2019.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 02/07/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a primary myocardial disorder, characterised by myocyte remodeling, disorganisation of sarcomeric proteins, impaired energy metabolism and altered cardiac contractility. Gene mutations encoding cardiac contractile proteins account for 60% of HCM aetiology. Current drug therapy including L-type calcium channel antagonists, are used to manage symptoms in patients with overt HCM, but no treatment exists that can reverse or prevent the cardiomyopathy. Design of effective drug therapy will require a clear understanding of the early pathophysiological mechanisms of the disease. Numerous studies have investigated specific aspects of HCM pathophysiology. This review brings these findings together, in order to develop a holistic understanding of the early pathophysiological mechanisms of the disease. We focus on gene mutations in cardiac myosin binding protein-C, β-cardiac myosin heavy chain, cardiac troponin I, and cardiac troponin T, that comprise the majority of all HCM sarcomeric gene mutations. We find that although some similarities exist, each mutation leads to mutation-specific alterations in calcium handling, myofilament calcium sensitivity and mitochondrial metabolic function. This may contribute to the observed clinical phenotypic variability in sarcomeric-related HCM. An understanding of early mutation-specific mechanisms of the disease may provide useful markers of disease progression, and inform therapeutic design.
Collapse
Affiliation(s)
- Helena M Viola
- School of Human Sciences (Physiology), The University of Western Australia, Crawley, WA, Australia
| | - Livia C Hool
- School of Human Sciences (Physiology), The University of Western Australia, Crawley, WA, Australia; Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
18
|
Moving beyond simple answers to complex disorders in sarcomeric cardiomyopathies: the role of integrated systems. Pflugers Arch 2019; 471:661-671. [PMID: 30848350 DOI: 10.1007/s00424-019-02269-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 12/26/2022]
Abstract
The classic clinical definition of hypertrophic cardiomyopathy (HCM) as originally described by Teare is deceptively simple, "left ventricular hypertrophy in the absence of any identifiable cause." Longitudinal studies, however, including a seminal study performed by Frank and Braunwald in 1968, clearly described the disorder much as we know it today, a complex, progressive, and highly variable cardiomyopathy affecting ~ 1/500 individuals worldwide. Subsequent genetic linkage studies in the early 1990s identified mutations in virtually all of the protein components of the cardiac sarcomere as the primary molecular cause of HCM. In addition, a substantial proportion of inherited dilated cardiomyopathy (DCM) has also been linked to sarcomeric protein mutations. Despite our deep understanding of the overall function of the sarcomere as the primary driver of cardiac contractility, the ability to use genotype in patient management remains elusive. A persistent challenge in the field from both the biophysical and clinical standpoints is how to rigorously link high-resolution protein dynamics and mechanics to the long-term cardiovascular remodeling process that characterizes these complex disorders. In this review, we will explore the depth of the problem from both the standpoint of a multi-subunit, highly conserved and dynamic "machine" to the resultant clinical and structural human phenotype with an emphasis on new, integrative approaches that can be widely applied to identify both novel disease mechanisms and new therapeutic targets for these primary biophysical disorders of the cardiac sarcomere.
Collapse
|
19
|
Piroddi N, Witjas-Paalberends ER, Ferrara C, Ferrantini C, Vitale G, Scellini B, Wijnker PJM, Sequiera V, Dooijes D, Dos Remedios C, Schlossarek S, Leung MC, Messer A, Ward DG, Biggeri A, Tesi C, Carrier L, Redwood CS, Marston SB, van der Velden J, Poggesi C. The homozygous K280N troponin T mutation alters cross-bridge kinetics and energetics in human HCM. J Gen Physiol 2018; 151:18-29. [PMID: 30578328 PMCID: PMC6314385 DOI: 10.1085/jgp.201812160] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/29/2018] [Indexed: 01/24/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomeric proteins, but the pathogenic mechanism is unclear. Piroddi et al. find impairment of cross-bridge kinetics and energetics in human sarcomeres with a TNNT2 mutation, suggesting that HCM involves inefficient ATP utilization. Hypertrophic cardiomyopathy (HCM) is a genetic form of left ventricular hypertrophy, primarily caused by mutations in sarcomere proteins. The cardiac remodeling that occurs as the disease develops can mask the pathogenic impact of the mutation. Here, to discriminate between mutation-induced and disease-related changes in myofilament function, we investigate the pathogenic mechanisms underlying HCM in a patient carrying a homozygous mutation (K280N) in the cardiac troponin T gene (TNNT2), which results in 100% mutant cardiac troponin T. We examine sarcomere mechanics and energetics in K280N-isolated myofibrils and demembranated muscle strips, before and after replacement of the endogenous troponin. We also compare these data to those of control preparations from donor hearts, aortic stenosis patients (LVHao), and HCM patients negative for sarcomeric protein mutations (HCMsmn). The rate constant of tension generation following maximal Ca2+ activation (kACT) and the rate constant of isometric relaxation (slow kREL) are markedly faster in K280N myofibrils than in all control groups. Simultaneous measurements of maximal isometric ATPase activity and Ca2+-activated tension in demembranated muscle strips also demonstrate that the energy cost of tension generation is higher in the K280N than in all controls. Replacement of mutant protein by exchange with wild-type troponin in the K280N preparations reduces kACT, slow kREL, and tension cost close to control values. In donor myofibrils and HCMsmn demembranated strips, replacement of endogenous troponin with troponin containing the K280N mutant increases kACT, slow kREL, and tension cost. The K280N TNNT2 mutation directly alters the apparent cross-bridge kinetics and impairs sarcomere energetics. This result supports the hypothesis that inefficient ATP utilization by myofilaments plays a central role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Nicoletta Piroddi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - E Rosalie Witjas-Paalberends
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Claudia Ferrara
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Cecilia Ferrantini
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy.,LENS, Sesto Fiorentino (Firenze), Florence, Italy
| | - Giulia Vitale
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Beatrice Scellini
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Paul J M Wijnker
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Vasco Sequiera
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Dennis Dooijes
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Clinical Genetics, University Medical Center, Utrecht, Netherlands
| | - Cristobal Dos Remedios
- Department of Anatomy and Histology, Bosch Institute, The University of Sydney, Sydney, Australia
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Man Ching Leung
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Andrew Messer
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Douglas G Ward
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | - Chiara Tesi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | | | - Steven B Marston
- National Heart and Lung Institute, Imperial College, London, England, UK
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.,Clinical Genetics, University Medical Center, Utrecht, Netherlands.,ICIN-Netherlands, Heart Institute, Utrecht, Netherlands
| | - Corrado Poggesi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Florence, Italy .,LENS, Sesto Fiorentino (Firenze), Florence, Italy
| |
Collapse
|
20
|
Biswas A, Das S, Kapoor M, Shamsudheen KV, Jayarajan R, Verma A, Seth S, Bhargava B, Scaria V, Sivasubbu S, Rao V. Familial Hypertrophic Cardiomyopathy - Identification of cause and risk stratification through exome sequencing. Gene 2018; 660:151-156. [DOI: 10.1016/j.gene.2018.03.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/02/2018] [Accepted: 03/19/2018] [Indexed: 02/01/2023]
|
21
|
Vakrou S, Fukunaga R, Foster DB, Sorensen L, Liu Y, Guan Y, Woldemichael K, Pineda-Reyes R, Liu T, Tardiff JC, Leinwand LA, Tocchetti CG, Abraham TP, O'Rourke B, Aon MA, Abraham MR. Allele-specific differences in transcriptome, miRNome, and mitochondrial function in two hypertrophic cardiomyopathy mouse models. JCI Insight 2018; 3:94493. [PMID: 29563334 DOI: 10.1172/jci.insight.94493] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 02/14/2018] [Indexed: 01/06/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) stems from mutations in sarcomeric proteins that elicit distinct biophysical sequelae, which in turn may yield radically different intracellular signaling and molecular pathologic profiles. These signaling events remain largely unaddressed by clinical trials that have selected patients based on clinical HCM diagnosis, irrespective of genotype. In this study, we determined how two mouse models of HCM differ, with respect to cellular/mitochondrial function and molecular biosignatures, at an early stage of disease. We show that hearts from young R92W-TnT and R403Q-αMyHC mutation-bearing mice differ in their transcriptome, miRNome, intracellular redox environment, mitochondrial antioxidant defense mechanisms, and susceptibility to mitochondrial permeability transition pore opening. Pathway analysis of mRNA-sequencing data and microRNA profiles indicate that R92W-TnT mutants exhibit a biosignature consistent with activation of profibrotic TGF-β signaling. Our results suggest that the oxidative environment and mitochondrial impairment in young R92W-TnT mice promote activation of TGF-β signaling that foreshadows a pernicious phenotype in young individuals. Of the two mutations, R92W-TnT is more likely to benefit from anti-TGF-β signaling effects conferred by angiotensin receptor blockers and may be responsive to mitochondrial antioxidant strategies in the early stage of disease. Molecular and functional profiling may therefore serve as aids to guide precision therapy for HCM.
Collapse
Affiliation(s)
- Styliani Vakrou
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - D Brian Foster
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lars Sorensen
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yamin Liu
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| | - Yufan Guan
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kirubel Woldemichael
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Roberto Pineda-Reyes
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ting Liu
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jill C Tardiff
- Department of Internal Medicine and Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular, and Developmental Biology and the BioFrontiers Institute, University of Colorado, Boulder, Colorado, USA
| | - Carlo G Tocchetti
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Theodore P Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Miguel A Aon
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - M Roselle Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University, Baltimore, Maryland, USA.,Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Division of Cardiology, UCSF, San Francisco, California, USA
| |
Collapse
|
22
|
Rest and Stress Longitudinal Systolic Left Ventricular Mechanics in Hypertrophic Cardiomyopathy: Implications for Prognostication. J Am Soc Echocardiogr 2018; 31:578-586. [PMID: 29426649 DOI: 10.1016/j.echo.2017.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Exercise intolerance is the most common symptom in hypertrophic cardiomyopathy (HCM). We examined whether inability to augment myocardial mechanics during exercise would influence functional performance and clinical outcomes in HCM. METHODS Ninety-five HCM patients (32 nonobstructive, 32 labile-obstructive, 31 obstructive) and 26 controls of similar age and gender distribution were recruited prospectively. They underwent rest and treadmill stress strain echocardiography, and 61 of them underwent magnetic resonance imaging. Mechanical reserve (MRES) was defined as percent change in systolic strain rate (SR) immediately postexercise. RESULTS Global strain and SR were significantly lower in HCM patients at rest (strain: nonobstructive, -15.6 ± 3.0; labile-obstructive, -15.9 ± 3.0; obstructive, -13.8 ± 2.9; control, -17.7% ± 2.1%, P < .001; SR: nonobstructive, -0.92 ± 0.20; labile-obstructive, -0.94 ± 0.17; obstructive, -0.85 ± 0.18; control, -1.04 ± 0.14 s-1, P = .002); and immediately postexercise (strain: nonobstructive, -15.6 ± 3.0; labile-obstructive, -17.6 ± 3.6; obstructive, -15.6 ± 3.6; control, -19.2 ± 3.1%; P = .001; SR: nonobstructive, -1.41 ± 0.37; labile-obstructive, -1.64 ± 0.38; obstructive, -1.32 ± 0.29; control, -1.82 ± 0.29 s-1, P < .001). MRES was lower in nonobstructive and obstructive compared with labile-obstructive and controls (51% ± 29%, 54% ± 31%, 78% ± 38%, 77% ± 30%, P = .001, respectively). Postexercise SR and MRES were associated with exercise capacity (r = 0.47 and 0.42, P < .001 both, respectively). When adjusted for age, gender, body mass index, E/e', and resting peak instantaneous systolic gradient, postexercise SR best predicted exercise capacity (r = 0.74, P = .003). Postexercise SR was correlated with extent of late gadolinium enhancement (r = 0.34, P = .03). By Cox regression, exercise SR and MRES predicted ventricular tachycardia/ventricular fibrillation (VT/VF) even after adjustment for age, gender, family history of sudden cardiac death, septum ≥ 3 cm and abnormal blood pressure response (P = .04 and P = .046, respectively). CONCLUSIONS Nonobstructive and obstructive patients have reduced MRES compared with labile-obstructive and controls. Postexercise SR correlates with LGE and exercise capacity. Exercise SR and MRES predict VT/VF.
Collapse
|
23
|
Ferrantini C, Coppini R, Pioner JM, Gentile F, Tosi B, Mazzoni L, Scellini B, Piroddi N, Laurino A, Santini L, Spinelli V, Sacconi L, De Tombe P, Moore R, Tardiff J, Mugelli A, Olivotto I, Cerbai E, Tesi C, Poggesi C. Pathogenesis of Hypertrophic Cardiomyopathy is Mutation Rather Than Disease Specific: A Comparison of the Cardiac Troponin T E163R and R92Q Mouse Models. J Am Heart Assoc 2017; 6:JAHA.116.005407. [PMID: 28735292 PMCID: PMC5586279 DOI: 10.1161/jaha.116.005407] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background In cardiomyocytes from patients with hypertrophic cardiomyopathy, mechanical dysfunction and arrhythmogenicity are caused by mutation‐driven changes in myofilament function combined with excitation‐contraction (E‐C) coupling abnormalities related to adverse remodeling. Whether myofilament or E‐C coupling alterations are more relevant in disease development is unknown. Here, we aim to investigate whether the relative roles of myofilament dysfunction and E‐C coupling remodeling in determining the hypertrophic cardiomyopathy phenotype are mutation specific. Methods and Results Two hypertrophic cardiomyopathy mouse models carrying the R92Q and the E163R TNNT2 mutations were investigated. Echocardiography showed left ventricular hypertrophy, enhanced contractility, and diastolic dysfunction in both models; however, these phenotypes were more pronounced in the R92Q mice. Both E163R and R92Q trabeculae showed prolonged twitch relaxation and increased occurrence of premature beats. In E163R ventricular myofibrils or skinned trabeculae, relaxation following Ca2+ removal was prolonged; resting tension and resting ATPase were higher; and isometric ATPase at maximal Ca2+ activation, the energy cost of tension generation, and myofilament Ca2+ sensitivity were increased compared with that in wild‐type mice. No sarcomeric changes were observed in R92Q versus wild‐type mice, except for a large increase in myofilament Ca2+ sensitivity. In R92Q myocardium, we found a blunted response to inotropic interventions, slower decay of Ca2+ transients, reduced SERCA function, and increased Ca2+/calmodulin kinase II activity. Contrarily, secondary alterations of E‐C coupling and signaling were minimal in E163R myocardium. Conclusions In E163R models, mutation‐driven myofilament abnormalities directly cause myocardial dysfunction. In R92Q, diastolic dysfunction and arrhythmogenicity are mediated by profound cardiomyocyte signaling and E‐C coupling changes. Similar hypertrophic cardiomyopathy phenotypes can be generated through different pathways, implying different strategies for a precision medicine approach to treatment.
Collapse
MESH Headings
- Animals
- Calcium Signaling
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Cardiomyopathy, Hypertrophic/diagnostic imaging
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/metabolism
- Cardiomyopathy, Hypertrophic/physiopathology
- Disease Models, Animal
- Excitation Contraction Coupling
- Fibrosis
- Genetic Markers
- Genetic Predisposition to Disease
- Hypertrophy, Left Ventricular/diagnostic imaging
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myofibrils/metabolism
- Myofibrils/pathology
- Phenotype
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Troponin T/genetics
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Ventricular Remodeling
Collapse
Affiliation(s)
- Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | - Josè Manuel Pioner
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Francesca Gentile
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Benedetta Tosi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Luca Mazzoni
- Department of NeuroFarBa, University of Florence, Italy
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | | | | | - Leonardo Sacconi
- LENS, University of Florence & National Institute of Optics (INO-CNR), Florence, Italy
| | - Pieter De Tombe
- Loyola University Medical Center Department of Physiology, Chicago, IL
| | | | | | - Alessandro Mugelli
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| |
Collapse
|
24
|
Coppini R, Mazzoni L, Ferrantini C, Gentile F, Pioner JM, Laurino A, Santini L, Bargelli V, Rotellini M, Bartolucci G, Crocini C, Sacconi L, Tesi C, Belardinelli L, Tardiff J, Mugelli A, Olivotto I, Cerbai E, Poggesi C. Ranolazine Prevents Phenotype Development in a Mouse Model of Hypertrophic Cardiomyopathy. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003565. [PMID: 28255011 DOI: 10.1161/circheartfailure.116.003565] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/30/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND Current therapies are ineffective in preventing the development of cardiac phenotype in young carriers of mutations associated with hypertrophic cardiomyopathy (HCM). Ranolazine, a late Na+ current blocker, reduced the electromechanical dysfunction of human HCM myocardium in vitro. METHODS AND RESULTS To test whether long-term treatment prevents cardiomyopathy in vivo, transgenic mice harboring the R92Q troponin-T mutation and wild-type littermates received an oral lifelong treatment with ranolazine and were compared with age-matched vehicle-treated animals. In 12-months-old male R92Q mice, ranolazine at therapeutic plasma concentrations prevented the development of HCM-related cardiac phenotype, including thickening of the interventricular septum, left ventricular volume reduction, left ventricular hypercontractility, diastolic dysfunction, left-atrial enlargement and left ventricular fibrosis, as evaluated in vivo using echocardiography and magnetic resonance. Left ventricular cardiomyocytes from vehicle-treated R92Q mice showed marked excitation-contraction coupling abnormalities, including increased diastolic [Ca2+] and Ca2+ waves, whereas cells from treated mutants were undistinguishable from those from wild-type mice. Intact trabeculae from vehicle-treated mutants displayed inotropic insufficiency, increased diastolic tension, and premature contractions; ranolazine treatment counteracted the development of myocardial mechanical abnormalities. In mutant myocytes, ranolazine inhibited the enhanced late Na+ current and reduced intracellular [Na+] and diastolic [Ca2+], ultimately preventing the pathological increase of calmodulin kinase activity in treated mice. CONCLUSIONS Owing to the sustained reduction of intracellular Ca2+ and calmodulin kinase activity, ranolazine prevented the development of morphological and functional cardiac phenotype in mice carrying a clinically relevant HCM-related mutation. Pharmacological inhibitors of late Na+ current are promising candidates for an early preventive therapy in young phenotype-negative subjects carrying high-risk HCM-related mutations.
Collapse
Affiliation(s)
- Raffaele Coppini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.).
| | - Luca Mazzoni
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Cecilia Ferrantini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Francesca Gentile
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Josè Manuel Pioner
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Annunziatina Laurino
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Lorenzo Santini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Valentina Bargelli
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Matteo Rotellini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Gianluca Bartolucci
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Claudia Crocini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Leonardo Sacconi
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Chiara Tesi
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Luiz Belardinelli
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Jil Tardiff
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Alessandro Mugelli
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Iacopo Olivotto
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Elisabetta Cerbai
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Corrado Poggesi
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| |
Collapse
|
25
|
Karam CN, Warren CM, Henze M, Banke NH, Lewandowski ED, Solaro RJ. Peroxisome proliferator-activated receptor-α expression induces alterations in cardiac myofilaments in a pressure-overload model of hypertrophy. Am J Physiol Heart Circ Physiol 2017; 312:H681-H690. [PMID: 28130336 DOI: 10.1152/ajpheart.00469.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 01/04/2017] [Accepted: 01/17/2017] [Indexed: 01/22/2023]
Abstract
Although alterations in fatty acid (FA) metabolism have been shown to have a negative impact on contractility of the hypertrophied heart, the targets of action remain elusive. In this study we compared the function of skinned fiber bundles from transgenic (Tg) mice that overexpress a relatively low level of the peroxisome proliferator-activated receptor α (PPARα), and nontransgenic (NTg) littermates. The mice (NTg-T and Tg-T) were stressed by transverse aortic constriction (TAC) and compared with shams (NTg-S and Tg-S). There was an approximate 4-fold increase in PPARα expression in Tg-S compared with NTg-S, but Tg-T hearts showed the same PPARα expression as NTg-T. Expression of PPARα did not alter the hypertrophic response to TAC but did reduce ejection fraction (EF) in Tg-T hearts compared with other groups. The rate of actomyosin ATP hydrolysis was significantly higher in Tg-S skinned fiber bundles compared with all other groups. Tg-T hearts showed an increase in phosphorylation of specific sites on cardiac myosin binding protein-C (cMyBP-C) and β-myosin heavy chain isoform. These results advance our understanding of potential signaling to the myofilaments induced by altered FA metabolism under normal and pathological states. We demonstrate that chronic and transient PPARα activation during pathological stress alters myofilament response to Ca2+ through a mechanism that is possibly mediated by MyBP-C phosphorylation and myosin heavy chain isoforms.NEW & NOTEWORTHY Data presented here demonstrate novel signaling to sarcomeric proteins by chronic alterations in fatty acid metabolism induced by PPARα. The mechanism involves modifications of key myofilament regulatory proteins modifying cross-bridge dynamics with differential effects in controls and hearts stressed by pressure overload.
Collapse
Affiliation(s)
- Chehade N Karam
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois; and
| | - Chad M Warren
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois; and
| | - Marcus Henze
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois; and
| | - Natasha H Banke
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois; and
| | - E Douglas Lewandowski
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois; and.,Sanford Burnham Presbyterian Medical Discovery Institute, Orlando, Florida
| | - R John Solaro
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
26
|
Yang C, Zhang C, Yuan J, Cui J, Liu S, Hu F, Yang W, Bi X, Qiao S. Sex-related differences in the associations between plasma free fatty acid levels and clinical features in patients with hypertrophic cardiomyopathy. Biol Sex Differ 2016; 7:63. [PMID: 27924218 PMCID: PMC5123328 DOI: 10.1186/s13293-016-0118-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/15/2016] [Indexed: 11/25/2022] Open
Abstract
Background Previous studies have indicated that inefficient energy utilization may play a pivotal role in hypertrophic cardiomyopathy (HCM). However, whether plasma free fatty acid (FFA), a main energy substrate of heart, has an effect on HCM remains unclear. Besides, several studies have suggested sex-related differences in HCM features and FFA metabolism. Here, we aimed to explore the association between plasma FFA levels and HCM and potential effects of sex on this relation. Methods A total of 412 patients (age 47.8 ± 12.7 years, 243 males (59.0%)) with HCM were recruited. Complete medical history was collected. Echocardiography and cardiovascular magnetic resonance imaging (CMRI) were performed. Fasting plasma FFA was determined by clinical laboratory. Left ventricular mass (LVM), maximum wall thickness (MWT), and left atrium diameter (LAD) were assessed with CMRI. Results The median FFA levels were 0.38 (interquartile range (IQR) 0.27–0.52) mmol/L in men and 0.40 (IQR 0.30–0.59) mmol/L in women. The FFA levels were significantly lower in men compared with those in women (p = 0.005). Compared with women, men had greater LVM index (LVMI) (96.8 ± 37.6 vs. 78.6 ± 31.5 g/m2, p < 0.001). FFA levels in male patients correlated positively with LVM, LVMI, LAD, cholesterol levels, high-density lipoprotein-cholesterol (HDL-C) levels, heart rate, and systolic blood pressure (SBP). However, none of these variables were significantly associated with sqrt (FFA) in female patients except a borderline correlation of LAD (p = 0.050). Multiple linear regression analysis was performed in male patients and revealed that HDL-C (β = 0.191, p = 0.002), heart rate (β = 0.182, p = 0.004), SBP (β = 0.167, p = 0.007), LVMI (β = 0.132, p = 0.032), and LAD (β = 0.165, p = 0.009) were independently associated with increasing FFA levels. Conclusions In patients with HCM, LVMI, LAD, HDL-C, SBP, and heart rate were independently associated with increasing plasma FFA levels in males, whereas not in females. These results suggest that sex may affect the pathogenesis of HCM through influencing FFA metabolism. And these sex-related differences should be taken into account in therapeutic approaches to influence myocardial FFA metabolism in HCM.
Collapse
Affiliation(s)
- Chengzhi Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Changlin Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Jiansong Yuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Jingang Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Shengwen Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Fenghuan Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Weixian Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Xuanye Bi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| | - Shubin Qiao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037 China
| |
Collapse
|
27
|
Marques MDA, de Oliveira GAP. Cardiac Troponin and Tropomyosin: Structural and Cellular Perspectives to Unveil the Hypertrophic Cardiomyopathy Phenotype. Front Physiol 2016; 7:429. [PMID: 27721798 PMCID: PMC5033975 DOI: 10.3389/fphys.2016.00429] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/09/2016] [Indexed: 12/12/2022] Open
Abstract
Inherited myopathies affect both skeletal and cardiac muscle and are commonly associated with genetic dysfunctions, leading to the production of anomalous proteins. In cardiomyopathies, mutations frequently occur in sarcomeric genes, but the cause-effect scenario between genetic alterations and pathological processes remains elusive. Hypertrophic cardiomyopathy (HCM) was the first cardiac disease associated with a genetic background. Since the discovery of the first mutation in the β-myosin heavy chain, more than 1400 new mutations in 11 sarcomeric genes have been reported, awarding HCM the title of the “disease of the sarcomere.” The most common macroscopic phenotypes are left ventricle and interventricular septal thickening, but because the clinical profile of this disease is quite heterogeneous, these phenotypes are not suitable for an accurate diagnosis. The development of genomic approaches for clinical investigation allows for diagnostic progress and understanding at the molecular level. Meanwhile, the lack of accurate in vivo models to better comprehend the cellular events triggered by this pathology has become a challenge. Notwithstanding, the imbalance of Ca2+ concentrations, altered signaling pathways, induction of apoptotic factors, and heart remodeling leading to abnormal anatomy have already been reported. Of note, a misbalance of signaling biomolecules, such as kinases and tumor suppressors (e.g., Akt and p53), seems to participate in apoptotic and fibrotic events. In HCM, structural and cellular information about defective sarcomeric proteins and their altered interactome is emerging but still represents a bottleneck for developing new concepts in basic research and for future therapeutic interventions. This review focuses on the structural and cellular alterations triggered by HCM-causing mutations in troponin and tropomyosin proteins and how structural biology can aid in the discovery of new platforms for therapeutics. We highlight the importance of a better understanding of allosteric communications within these thin-filament proteins to decipher the HCM pathological state.
Collapse
Affiliation(s)
- Mayra de A Marques
- Programa de Biologia Estrutural, Centro Nacional de Ressonância Magnética Nuclear Jiri Jonas, Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Guilherme A P de Oliveira
- Programa de Biologia Estrutural, Centro Nacional de Ressonância Magnética Nuclear Jiri Jonas, Instituto de Bioquímica Médica Leopoldo de Meis, Instituto Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
28
|
In Vivo Cannulation Methods for Cardiomyocytes Isolation from Heart Disease Models. PLoS One 2016; 11:e0160605. [PMID: 27500929 PMCID: PMC4976940 DOI: 10.1371/journal.pone.0160605] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/21/2016] [Indexed: 12/13/2022] Open
Abstract
Isolation of high quality cardiomyocytes is critically important for achieving successful experiments in many cellular and molecular cardiology studies. Methods for isolating cardiomyocytes from the murine heart generally are time-sensitive and experience-dependent, and often fail to produce high quality cells. Major technical difficulties can be related to the surgical procedures needed to explant the heart and to cannulate the vessel to mount onto the Langendorff system before in vitro reperfusion can begin. During this period, transient hypoxia and ischemia may damage the heart, resulting in low yield and poor quality of cells, especially for heart disease models that have fragile cells. We have developed novel in vivo cannulation methods to minimize hypoxia and ischemia, and fine-tuned the entire protocol to produce high quality ventricular myocytes. The high cell quality has been confirmed using important structural and functional criteria such as morphology, t-tubule structure, action potential morphology, Ca2+ signaling, responsiveness to beta-adrenergic agonist, and ability to have robust contraction under mechanically loaded condition. Together these assessments show the preservation of the cardiac excitation–contraction machinery in cells isolated using this technique. The in vivo cannulation method enables consistent isolation of high-quality cardiomyocytes, even from heart disease models that were notoriously difficult for cell isolation using traditional methods.
Collapse
|
29
|
|
30
|
Liver Kinase B1 complex acts as a novel modifier of myofilament function and localizes to the Z-disk in cardiac myocytes. Arch Biochem Biophys 2016; 601:32-41. [PMID: 26971467 DOI: 10.1016/j.abb.2016.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/09/2016] [Accepted: 03/08/2016] [Indexed: 01/15/2023]
Abstract
Contractile perturbations downstream of Ca(2+) binding to troponin C, the so-called sarcomere-controlled mechanisms, represent the earliest indicators of energy remodeling in the diseased heart [1]. Central to cellular energy "sensing" is the adenosine monophosphate-activated kinase (AMPK) pathway, which is known to directly target myofilament proteins and alter contractility [2-6]. We previously showed that the upstream AMPK kinase, LKB1/MO25/STRAD, impacts myofilament function independently of AMPK [5]. Therefore, we hypothesized that the LKB1 complex associated with myofilament proteins and that alterations in energy signaling modulated targeting or localization of the LKB1 complex to the myofilament. Using an integrated strategy of myofilament mechanics, immunoblot analysis, co-immunoprecipitation, mass spectroscopy, and immunofluorescence, we showed that 1) LKB1 and MO25 associated with myofibrillar proteins, 2) cellular energy stress re-distributed AMPK/LKB1 complex proteins within the sarcomere, and 3) the LKB1 complex localized to the Z-Disk and interacted with cytoskeletal and energy-regulating proteins, including vinculin and ATP Synthase (Complex V). These data represent a novel role for LKB1 complex proteins in myofilament function and myocellular "energy" sensing in the heart.
Collapse
|
31
|
Cheng Y, Regnier M. Cardiac troponin structure-function and the influence of hypertrophic cardiomyopathy associated mutations on modulation of contractility. Arch Biochem Biophys 2016; 601:11-21. [PMID: 26851561 PMCID: PMC4899195 DOI: 10.1016/j.abb.2016.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 11/29/2022]
Abstract
Cardiac troponin (cTn) acts as a pivotal regulator of muscle contraction and relaxation and is composed of three distinct subunits (cTnC: a highly conserved Ca(2+) binding subunit, cTnI: an actomyosin ATPase inhibitory subunit, and cTnT: a tropomyosin binding subunit). In this mini-review, we briefly summarize the structure-function relationship of cTn and its subunits, its modulation by PKA-mediated phosphorylation of cTnI, and what is known about how these properties are altered by hypertrophic cardiomyopathy (HCM) associated mutations of cTnI. This includes recent work using computational modeling approaches to understand the atomic-based structural level basis of disease-associated mutations. We propose a viewpoint that it is alteration of cTnC-cTnI interaction (rather than the Ca(2+) binding properties of cTn) per se that disrupt the ability of PKA-mediated phosphorylation at cTnI Ser-23/24 to alter contraction and relaxation in at least some HCM-associated mutations. The combination of state of the art biophysical approaches can provide new insight on the structure-function mechanisms of contractile dysfunction resulting cTnI mutations and exciting new avenues for the diagnosis, prevention, and even treatment of heart diseases.
Collapse
Affiliation(s)
- Yuanhua Cheng
- University of Washington, Department of Bioengineering, Seattle, WA, USA
| | - Michael Regnier
- University of Washington, Department of Bioengineering, Seattle, WA, USA.
| |
Collapse
|
32
|
ADP-stimulated contraction: A predictor of thin-filament activation in cardiac disease. Proc Natl Acad Sci U S A 2015; 112:E7003-12. [PMID: 26621701 DOI: 10.1073/pnas.1513843112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Diastolic dysfunction is general to all idiopathic dilated (IDCM) and hypertrophic cardiomyopathy (HCM) patients. Relaxation deficits may result from increased actin-myosin formation during diastole due to altered tropomyosin position, which blocks myosin binding to actin in the absence of Ca(2+). We investigated whether ADP-stimulated force development (without Ca(2+)) can be used to reveal changes in actin-myosin blockade in human cardiomyopathy cardiomyocytes. Cardiac samples from HCM patients, harboring thick-filament (MYH7mut, MYBPC3mut) and thin-filament (TNNT2mut, TNNI3mut) mutations, and IDCM were compared with sarcomere mutation-negative HCM (HCMsmn) and nonfailing donors. Myofilament ADP sensitivity was higher in IDCM and HCM compared with donors, whereas it was lower for MYBPC3. Increased ADP sensitivity in IDCM, HCMsmn, and MYH7mut was caused by low phosphorylation of myofilament proteins, as it was normalized to donors by protein kinase A (PKA) treatment. Troponin exchange experiments in a TNNT2mut sample corrected the abnormal actin-myosin blockade. In MYBPC3trunc samples, ADP sensitivity highly correlated with cardiac myosin-binding protein-C (cMyBP-C) protein level. Incubation of cardiomyocytes with cMyBP-C antibody against the actin-binding N-terminal region reduced ADP sensitivity, indicative of cMyBP-C's role in actin-myosin regulation. In the presence of Ca(2+), ADP increased myofilament force development and sarcomere stiffness. Enhanced sarcomere stiffness in sarcomere mutation-positive HCM samples was irrespective of the phosphorylation background. In conclusion, ADP-stimulated contraction can be used as a tool to study how protein phosphorylation and mutant proteins alter accessibility of myosin binding on actin. In the presence of Ca(2+), pathologic [ADP] and low PKA-phosphorylation, high actin-myosin formation could contribute to the impaired myocardial relaxation observed in cardiomyopathies.
Collapse
|
33
|
Coppini R, Ho CY, Ashley E, Day S, Ferrantini C, Girolami F, Tomberli B, Bardi S, Torricelli F, Cecchi F, Mugelli A, Poggesi C, Tardiff J, Olivotto I. Clinical phenotype and outcome of hypertrophic cardiomyopathy associated with thin-filament gene mutations. J Am Coll Cardiol 2015; 64:2589-2600. [PMID: 25524337 PMCID: PMC4270453 DOI: 10.1016/j.jacc.2014.09.059] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 09/03/2014] [Accepted: 09/16/2014] [Indexed: 11/30/2022]
Abstract
Background Mild hypertrophy but increased arrhythmic risk characterizes the stereotypic phenotype proposed for hypertrophic cardiomyopathy (HCM) caused by thin-filament mutations. However, whether such clinical profile is different from more prevalent thick-filament–associated disease is unresolved. Objectives This study aimed to assess clinical features and outcomes in a large cohort of patients with HCM associated with thin-filament mutations compared with thick-filament HCM. Methods Adult HCM patients (age >18 years), 80 with thin-filament and 150 with thick-filament mutations, were followed for an average of 4.5 years. Results Compared with thick-filament HCM, patients with thin-filament mutations showed: 1) milder and atypically distributed left ventricular (LV) hypertrophy (maximal wall thickness 18 ± 5 mm vs. 24 ± 6 mm; p < 0.001) and less prevalent outflow tract obstruction (19% vs. 34%; p = 0.015); 2) higher rate of progression to New York Heart Association functional class III or IV (15% vs. 5%; p = 0.013); 3) higher prevalence of systolic dysfunction or restrictive LV filling at last evaluation (20% vs. 9%; p = 0.038); 4) 2.4-fold increase in prevalence of triphasic LV filling pattern (26% vs. 11%; p = 0.002); and 5) similar rates of malignant ventricular arrhythmias and sudden cardiac death (p = 0.593). Conclusions In adult HCM patients, thin-filament mutations are associated with increased likelihood of advanced LV dysfunction and heart failure compared with thick-filament disease, whereas arrhythmic risk in both subsets is comparable. Triphasic LV filling is particularly common in thin-filament HCM, reflecting profound diastolic dysfunction.
Collapse
Affiliation(s)
- Raffaele Coppini
- Center of Molecular Medicine (CIMMBA), University of Florence, Florence, Italy.
| | - Carolyn Y Ho
- Brigham and Women's Hospital, Boston, Massachusetts
| | - Euan Ashley
- Cardiovascular Medicine, Stanford Medical Center, Stanford, California
| | - Sharlene Day
- University of Michigan Medical Center, Ann Arbor, Michigan
| | - Cecilia Ferrantini
- Center of Molecular Medicine (CIMMBA), University of Florence, Florence, Italy
| | | | - Benedetta Tomberli
- Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy
| | - Sara Bardi
- Genetics Unit; Careggi University Hospital, Florence, Italy
| | | | - Franco Cecchi
- Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy
| | - Alessandro Mugelli
- Center of Molecular Medicine (CIMMBA), University of Florence, Florence, Italy
| | - Corrado Poggesi
- Center of Molecular Medicine (CIMMBA), University of Florence, Florence, Italy
| | - Jil Tardiff
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Iacopo Olivotto
- Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy
| |
Collapse
|
34
|
Chen Y, Zhang Z, Hu F, Yang W, Yuan J, Cui J, Hao S, Hu J, Zhou Y, Qiao S. 17β-estradiol prevents cardiac diastolic dysfunction by stimulating mitochondrial function: a preclinical study in a mouse model of a human hypertrophic cardiomyopathy mutation. J Steroid Biochem Mol Biol 2015; 147:92-102. [PMID: 25541436 DOI: 10.1016/j.jsbmb.2014.12.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/01/2014] [Accepted: 12/18/2014] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We investigated the effect of ovariectomy (OVX) and 17β-estradiol (E2) replacement on both mitochondrial and myocardial function in cTnT-Q92 transgenic mice generated by cardiac-restricted expression of a human hypertrophic cardiomyopathy (HCM) mutation. METHODS The cTnT-Q92 mice were ovariectomized at twenty weeks of age and were treated with either placebo (OVX group) or E2 (OVX+E2 group) for twelve weeks before being sacrificed. Wild-type and cTnT-Q92 female mice receiving sham operation were used as controls. Indices of diastolic function such as mitral early (E) and late (A) inflow as well as isovolumic relaxation time (IVRT) were measured by echocardiography. A Clark-type electrode was used to detect respiratory control, and ATP levels were determined at the mitochondrial level using HPLC. Key components related to mitochondrial energy metabolism, such as peroxisome proliferator-activated receptor α (PPARα), PPARγ coactivator 1α (PGC-1α) and nuclear respiratory factor-1 (NRF-1), were also analyzed using Western blot and RT-PCR. The levels of oxidative stress markers were determined by measuring malondialdehyde (MDA) using the thiobarbituric acid assay. RESULTS The cTnT-Q92 mice had impaired diastolic function compared with wild-type mice (E/A ratio, 1.39 ± 0.04 vs. 1.21 ± 0.01, p<0.001; IVRT, 19.17 ± 0.85 vs. 22.15 ± 1.43 ms, p=0.028). In response to ovariectomy, cardiac function further decreased compared with that observed in cTnT-Q92 mice that received the sham operation (E/A ratio, 1.15 ± 0.04 vs. 1.21 ± 0.01, p<0.001; IVRT, 28.31 ± 0.39 vs. 22.15 ± 1.43 ms, p=0.002). Myocardial energy metabolism, as determined by ATP levels (3.49 ± 0.31 vs. 5.07 ± 0.47 μmol/g, p<0.001), and the mitochondrial respiratory ratio (2.04 ± 0.10 vs. 2.63 ± 0.11, p=0.01) also decreased significantly. By contrast, myocardial concentrations of MDA increased significantly in the OVX group, and PGC-1α, PPARα and NRF-1decreased significantly. E2 supplementation significantly elevated myocardial ATP levels (4.55 ± 0.21 vs. 3.49 ± 0.31 μmol/g, p=0.003) and mitochondrial respiratory function (3.93 ± 0.05 vs. 2.63 ± 0.11, p=0.001); however, it reduced the MDA level (0.21 ± 0.02 vs. 0.36 ± 0.03 nmol/g, p<0.001), which subsequently improved diastolic function (E/A ratio, 1.35 ± 0.06 vs. 1.15 ± 0.04, p<0.001; IVRT, 18.22 ± 1.16 vs. 28.31 ± 0.39 ms, p=0.007). CONCLUSIONS Our study has shown that 17β-estradiol improved myocardial diastolic function, prevented myocardial energy dysregulation, and reduced myocardial oxidative stress in cTnT-Q92 mice.
Collapse
Affiliation(s)
- Youzhou Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Zhuoli Zhang
- Department of Radiology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, 737 N Michigan Ave., 16th Floor, Chicago, USA
| | - Fenghuan Hu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Weixian Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Jiansong Yuan
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Jingang Cui
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Shujing Hao
- Clinical Laboratory of Zhongke, Beijing, China
| | - Jie Hu
- Clinical Laboratory of Zhongke, Beijing, China
| | - Ying Zhou
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Shubin Qiao
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| |
Collapse
|
35
|
Duncker DJ, Bakkers J, Brundel BJ, Robbins J, Tardiff JC, Carrier L. Animal and in silico models for the study of sarcomeric cardiomyopathies. Cardiovasc Res 2015; 105:439-48. [PMID: 25600962 DOI: 10.1093/cvr/cvv006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Over the past decade, our understanding of cardiomyopathies has improved dramatically, due to improvements in screening and detection of gene defects in the human genome as well as a variety of novel animal models (mouse, zebrafish, and drosophila) and in silico computational models. These novel experimental tools have created a platform that is highly complementary to the naturally occurring cardiomyopathies in cats and dogs that had been available for some time. A fully integrative approach, which incorporates all these modalities, is likely required for significant steps forward in understanding the molecular underpinnings and pathogenesis of cardiomyopathies. Finally, novel technologies, including CRISPR/Cas9, which have already been proved to work in zebrafish, are currently being employed to engineer sarcomeric cardiomyopathy in larger animals, including pigs and non-human primates. In the mouse, the increased speed with which these techniques can be employed to engineer precise 'knock-in' models that previously took years to make via multiple rounds of homologous recombination-based gene targeting promises multiple and precise models of human cardiac disease for future study. Such novel genetically engineered animal models recapitulating human sarcomeric protein defects will help bridging the gap to translate therapeutic targets from small animal and in silico models to the human patient with sarcomeric cardiomyopathy.
Collapse
Affiliation(s)
- Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bianca J Brundel
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jeff Robbins
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Jil C Tardiff
- Department of Medicine and Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
36
|
Messer AE, Marston SB. Investigating the role of uncoupling of troponin I phosphorylation from changes in myofibrillar Ca(2+)-sensitivity in the pathogenesis of cardiomyopathy. Front Physiol 2014; 5:315. [PMID: 25202278 PMCID: PMC4142463 DOI: 10.3389/fphys.2014.00315] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/02/2014] [Indexed: 12/12/2022] Open
Abstract
Contraction in the mammalian heart is controlled by the intracellular Ca(2+) concentration as it is in all striated muscle, but the heart has an additional signaling system that comes into play to increase heart rate and cardiac output during exercise or stress. β-adrenergic stimulation of heart muscle cells leads to release of cyclic-AMP and the activation of protein kinase A which phosphorylates key proteins in the sarcolemma, sarcoplasmic reticulum and contractile apparatus. Troponin I (TnI) and Myosin Binding Protein C (MyBP-C) are the prime targets in the myofilaments. TnI phosphorylation lowers myofibrillar Ca(2+)-sensitivity and increases the speed of Ca(2+)-dissociation and relaxation (lusitropic effect). Recent studies have shown that this relationship between Ca(2+)-sensitivity and TnI phosphorylation may be unstable. In familial cardiomyopathies, both dilated and hypertrophic (DCM and HCM), a mutation in one of the proteins of the thin filament often results in the loss of the relationship (uncoupling) and blunting of the lusitropic response. For familial dilated cardiomyopathy in thin filament proteins it has been proposed that this uncoupling is causative of the phenotype. Uncoupling has also been found in human heart tissue from patients with hypertrophic obstructive cardiomyopathy as a secondary effect. Recently, it has been found that Ca(2+)-sensitizing drugs can promote uncoupling, whilst one Ca(2+)-desensitizing drug Epigallocatechin 3-Gallate (EGCG) can reverse uncoupling. We will discuss recent findings about the role of uncoupling in the development of cardiomyopathies and the molecular mechanism of the process.
Collapse
Affiliation(s)
- Andrew E. Messer
- National Heart & Lung Institute, Imperial College LondonLondon, UK
| | | |
Collapse
|
37
|
Vakrou S, Abraham MR. Hypertrophic cardiomyopathy: a heart in need of an energy bar? Front Physiol 2014; 5:309. [PMID: 25191275 PMCID: PMC4137386 DOI: 10.3389/fphys.2014.00309] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/30/2014] [Indexed: 01/08/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) has been recently recognized as the most common inherited cardiovascular disorder, affecting 1 in 500 adults worldwide. HCM is characterized by myocyte hypertrophy resulting in thickening of the ventricular wall, myocyte disarray, interstitial and/or replacement fibrosis, decreased ventricular cavity volume and diastolic dysfunction. HCM is also the most common cause of sudden death in the young. A large proportion of patients diagnosed with HCM have mutations in sarcomeric proteins. However, it is unclear how these mutations lead to the cardiac phenotype, which is variable even in patients carrying the same causal mutation. Abnormalities in calcium cycling, oxidative stress, mitochondrial dysfunction and energetic deficiency have been described constituting the basis of therapies in experimental models of HCM and HCM patients. This review focuses on evidence supporting the role of cellular metabolism and mitochondria in HCM.
Collapse
Affiliation(s)
- Styliani Vakrou
- Division of Cardiology, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| | - M Roselle Abraham
- Division of Cardiology, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
38
|
Witjas-Paalberends ER, Güçlü A, Germans T, Knaapen P, Harms HJ, Vermeer AMC, Christiaans I, Wilde AAM, Dos Remedios C, Lammertsma AA, van Rossum AC, Stienen GJM, van Slegtenhorst M, Schinkel AF, Michels M, Ho CY, Poggesi C, van der Velden J. Gene-specific increase in the energetic cost of contraction in hypertrophic cardiomyopathy caused by thick filament mutations. Cardiovasc Res 2014; 103:248-57. [PMID: 24835277 DOI: 10.1093/cvr/cvu127] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Disease mechanisms regarding hypertrophic cardiomyopathy (HCM) are largely unknown and disease onset varies. Sarcomere mutations might induce energy depletion for which until now there is no direct evidence at sarcomere level in human HCM. This study investigated if mutations in genes encoding myosin-binding protein C (MYBPC3) and myosin heavy chain (MYH7) underlie changes in the energetic cost of contraction in the development of human HCM disease. METHODS AND RESULTS Energetic cost of contraction was studied in vitro by measurements of force development and ATPase activity in cardiac muscle strips from 26 manifest HCM patients (11 MYBPC3mut, 9 MYH7mut, and 6 sarcomere mutation-negative, HCMsmn). In addition, in vivo, the ratio between external work (EW) and myocardial oxygen consumption (MVO2) to obtain myocardial external efficiency (MEE) was determined in 28 pre-hypertrophic mutation carriers (14 MYBPC3mut and 14 MYH7mut) and 14 healthy controls using [(11)C]-acetate positron emission tomography and cardiovascular magnetic resonance imaging. Tension cost (TC), i.e. ATPase activity during force development, was higher in MYBPC3mut and MYH7mut compared with HCMsmn at saturating [Ca(2+)]. TC was also significantly higher in MYH7mut at submaximal, more physiological [Ca(2+)]. EW was significantly lower in both mutation carrier groups, while MVO2 did not differ. MEE was significantly lower in both mutation carrier groups compared with controls, showing the lowest efficiency in MYH7 mutation carriers. CONCLUSION We provide direct evidence that sarcomere mutations perturb the energetic cost of cardiac contraction. Gene-specific severity of cardiac abnormalities may underlie differences in disease onset and suggests that early initiation of metabolic treatment may be beneficial, in particular, in MYH7 mutation carriers.
Collapse
Affiliation(s)
- E Rosalie Witjas-Paalberends
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Ahmet Güçlü
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands ICIN Netherlands Heart Institute, Utrecht, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Paul Knaapen
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Hendrik J Harms
- Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Alexa M C Vermeer
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Imke Christiaans
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Arthur A M Wilde
- Department of Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Cris Dos Remedios
- Institute for Biomedical Research, Muscle Research Unit, University of Sydney, Sydney, Australia
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Albert C van Rossum
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Ger J M Stienen
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands Department of Physics and Astronomy, VU University, Amsterdam, The Netherlands
| | | | - Arend F Schinkel
- Thorax Center, Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Michelle Michels
- Thorax Center, Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carolyn Y Ho
- Brigham and Women's Hospital, Cardiology, Boston, MA, USA
| | - Corrado Poggesi
- Department of Physiology, University of Florence, Florence, Italy
| | - Jolanda van der Velden
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands ICIN Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
39
|
Poggesi C, Ho CY. Muscle dysfunction in hypertrophic cardiomyopathy: what is needed to move to translation? J Muscle Res Cell Motil 2014; 35:37-45. [PMID: 24493262 DOI: 10.1007/s10974-014-9374-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 01/02/2014] [Indexed: 02/04/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomere genes. As such, HCM provides remarkable opportunities to study how changes to the heart's molecular motor apparatus may influence cardiac structure and function. Although the genetic basis of HCM is well-described, there is much more limited understanding of the precise consequences of sarcomere mutations--how they remodel the heart, and how these changes lead to the dramatic clinical consequences associated with HCM. More precise characterization of the mechanisms leading from sarcomere mutation to altered cardiac muscle function is critical to gain insight into fundamental disease biology and phenotypic evolution. Such knowledge will help foster development of novel treatment strategies aimed at correcting and preventing disease development in HCM.
Collapse
Affiliation(s)
- Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Morgagni 63, 50134, Florence, Italy,
| | | |
Collapse
|
40
|
Lu D, Dong W, Zhang X, Quan X, Bao D, Lu Y, Zhang L. WIF1 causes dysfunction of heart in transgenic mice. Transgenic Res 2013; 22:1179-89. [PMID: 23921644 PMCID: PMC3835953 DOI: 10.1007/s11248-013-9738-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/28/2013] [Indexed: 11/29/2022]
Abstract
Wnt activity is a key regulator of cardiac progenitor cell self-renewal, differentiation and morphogenesis. However, Wnt inhibitory factor 1 (WIF1), a antagonists of Wnt signaling activity, its potential effects on heart development has not yet been approached by either in vivo or in vitro studies. Here, the expression of WIF1 was regulated in a different way in the dilated and hypertrophic cardiomyopathy heart from transgenic mice by mutations in cardiac troponin T, cTnT(R141W) and cTnT(R92Q). The heart tissue specific transgenic mice of WIF1 was studied using M-mode echocardiography and histologic analyses. Production levels of an array of effectors and transcription factors that impact cellular organization and tissue morphology were measured. The effects of WIF1 on β-catenin pathway could be reversed by LiCl regarding signaling pathways and effector and respondent molecules in H9c2 cells, consistent with the expression levels of c-myc, natriuretic peptide precursor type B and skeletal muscle actin α1. Among the most noteworthy findings were that WIF1 impaired the function and structure of heart, and the effects on β-catenin pathway maybe the course of the former. It is anticipated that our findings will contribute to expansion of our understanding of WIF1 biological function on heart development and possible modes of treatment of heart diseases.
Collapse
Affiliation(s)
- Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Xu Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiongzhi Quan
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Dan Bao
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Yingdong Lu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of Human Disease Animal Model, State Administration of Traditional Chinese Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing, 100021 People’s Republic of China
| |
Collapse
|
41
|
Güçlü A, Germans T, Witjas-Paalberends ER, Stienen GJM, Brouwer WP, Harms HJ, Marcus JT, Vonk ABA, Stooker W, Yilmaz A, Klein P, Ten Berg JM, Kluin J, Asselbergs FW, Lammertsma AA, Knaapen P, van Rossum AC, van der Velden J. ENerGetIcs in hypertrophic cardiomyopathy: traNslation between MRI, PET and cardiac myofilament function (ENGINE study). Neth Heart J 2013; 21:567-71. [PMID: 24114686 PMCID: PMC3833912 DOI: 10.1007/s12471-013-0478-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Introduction Hypertrophic cardiomyopathy (HCM) is an autosomal dominant heart disease mostly due to mutations in genes encoding sarcomeric proteins. HCM is characterised by asymmetric hypertrophy of the left ventricle (LV) in the absence of another cardiac or systemic disease. At present it lacks specific treatment to prevent or reverse cardiac dysfunction and hypertrophy in mutation carriers and HCM patients. Previous studies have indicated that sarcomere mutations increase energetic costs of cardiac contraction and cause myocardial dysfunction and hypertrophy. By using a translational approach, we aim to determine to what extent disturbances of myocardial energy metabolism underlie disease progression in HCM. Methods Hypertrophic obstructive cardiomyopathy (HOCM) patients and aortic valve stenosis (AVS) patients will undergo a positron emission tomography (PET) with acetate and cardiovascular magnetic resonance imaging (CMR) with tissue tagging before and 4 months after myectomy surgery or aortic valve replacement + septal biopsy. Myectomy tissue or septal biopsy will be used to determine efficiency of sarcomere contraction in-vitro, and results will be compared with in-vivo cardiac performance. Healthy subjects and non-hypertrophic HCM mutation carriers will serve as a control group. Endpoints Our study will reveal whether perturbations in cardiac energetics deteriorate during disease progression in HCM and whether these changes are attributed to cardiac remodelling or the presence of a sarcomere mutation per se. In-vitro studies in hypertrophied cardiac muscle from HOCM and AVS patients will establish whether sarcomere mutations increase ATP consumption of sarcomeres in human myocardium. Our follow-up imaging study in HOCM and AVS patients will reveal whether impaired cardiac energetics are restored by cardiac surgery.
Collapse
Affiliation(s)
- A Güçlü
- Department of Cardiology, Institute for Cardiovascular Research (ICaR-VU, VU University Medical Center, ZH 5F-13, PO Box 7057, 1007MB, Amsterdam, the Netherlands,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Song W, Vikhorev PG, Kashyap MN, Rowlands C, Ferenczi MA, Woledge RC, MacLeod K, Marston S, Curtin NA. Mechanical and energetic properties of papillary muscle from ACTC E99K transgenic mouse models of hypertrophic cardiomyopathy. Am J Physiol Heart Circ Physiol 2013; 304:H1513-24. [PMID: 23604709 DOI: 10.1152/ajpheart.00951.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We compared the contractile performance of papillary muscle from a mouse model of hypertrophic cardiomyopathy [α-cardiac actin (ACTC) E99K mutation] with nontransgenic (non-TG) littermates. In isometric twitches, ACTC E99K papillary muscle produced three to four times greater force than non-TG muscle under the same conditions independent of stimulation frequency and temperature, whereas maximum isometric force in myofibrils from these muscles was not significantly different. ACTC E99K muscle relaxed slower than non-TG muscle in both papillary muscle (1.4×) and myofibrils (1.7×), whereas the rate of force development after stimulation was the same as non-TG muscle for both electrical stimulation in intact muscle and after a Ca²⁺ jump in myofibrils. The EC₅₀ for Ca²⁺ activation of force in myofibrils was 0.39 ± 0.33 μmol/l in ACTC E99K myofibrils and 0.80 ± 0.11 μmol/l in non-TG myofibrils. There were no significant differences in the amplitude and time course of the Ca²⁺ transient in myocytes from ACTC E99K and non-TG mice. We conclude that hypercontractility is caused by higher myofibrillar Ca²⁺ sensitivity in ACTC E99K muscles. Measurement of the energy (work + heat) released in actively cycling heart muscle showed that for both genotypes, the amount of energy turnover increased with work done but with decreasing efficiency as energy turnover increased. Thus, ACTC E99K mouse heart muscle produced on average 3.3-fold more work than non-TG muscle, and the cost in terms of energy turnover was disproportionately higher than in non-TG muscles. Efficiency for ACTC E99K muscle was in the range of 11-16% and for non-TG muscle was 15-18%.
Collapse
Affiliation(s)
- Weihua Song
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Akki A, Gupta A, Weiss RG. Magnetic resonance imaging and spectroscopy of the murine cardiovascular system. Am J Physiol Heart Circ Physiol 2013; 304:H633-48. [PMID: 23292717 DOI: 10.1152/ajpheart.00771.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Magnetic resonance imaging (MRI) has emerged as a powerful and reliable tool to noninvasively study the cardiovascular system in clinical practice. Because transgenic mouse models have assumed a critical role in cardiovascular research, technological advances in MRI have been extended to mice over the last decade. These have provided critical insights into cardiac and vascular morphology, function, and physiology/pathophysiology in many murine models of heart disease. Furthermore, magnetic resonance spectroscopy (MRS) has allowed the nondestructive study of myocardial metabolism in both isolated hearts and in intact mice. This article reviews the current techniques and important pathophysiological insights from the application of MRI/MRS technology to murine models of cardiovascular disease.
Collapse
Affiliation(s)
- Ashwin Akki
- Division of Cardiology, Department of Medicine, and Division of Magnetic Resonance Research, Department of Radiology, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
44
|
Coppini R, Ferrantini C, Yao L, Fan P, Del Lungo M, Stillitano F, Sartiani L, Tosi B, Suffredini S, Tesi C, Yacoub M, Olivotto I, Belardinelli L, Poggesi C, Cerbai E, Mugelli A. Late sodium current inhibition reverses electromechanical dysfunction in human hypertrophic cardiomyopathy. Circulation 2012; 127:575-84. [PMID: 23271797 DOI: 10.1161/circulationaha.112.134932] [Citation(s) in RCA: 292] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM), the most common mendelian heart disorder, remains an orphan of disease-specific pharmacological treatment because of the limited understanding of cellular mechanisms underlying arrhythmogenicity and diastolic dysfunction. METHODS AND RESULTS We assessed the electromechanical profile of cardiomyocytes from 26 HCM patients undergoing myectomy compared with those from nonfailing nonhypertrophic surgical patients by performing patch-clamp and intracellular Ca(2+) (Ca(2+)(i)) studies. Compared with controls, HCM cardiomyocytes showed prolonged action potential related to increased late Na(+) (I(NaL)) and Ca(2+) (I(CaL)) currents and decreased repolarizing K(+) currents, increased occurrence of cellular arrhythmias, prolonged Ca(2+)(i) transients, and higher diastolic Ca(2+)(i). Such changes were related to enhanced Ca(2+)/calmodulin kinase II (CaMKII) activity and increased phosphorylation of its targets. Ranolazine at therapeutic concentrations partially reversed the HCM-related cellular abnormalities via I(NaL) inhibition, with negligible effects in controls. By shortening the action potential duration in HCM cardiomyocytes, ranolazine reduced the occurrence of early and delayed afterdepolarizations. Finally, as a result of the faster kinetics of Ca(2+)(i) transients and the lower diastolic Ca(2+)(i), ranolazine accelerated the contraction-relaxation cycle of HCM trabeculae, ameliorating diastolic function. CONCLUSIONS We highlighted a specific set of functional changes in human HCM myocardium that stem from a complex remodeling process involving alterations of CaMKII-dependent signaling, rather than being a direct consequence of the causal sarcomeric mutations. Among the several ion channel and Ca(2+)(i) handling proteins changes identified, an enhanced I(NaL) seems to be a major contributor to the electrophysiological and Ca(2+)(i) dynamic abnormalities of ventricular myocytes and trabeculae from patients with HCM, suggesting potential therapeutic implications of I(NaL) inhibition.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department of Preclinical and Clinical Pharmacology, University of Florence, V. le G. Pieraccini 6, 50139 Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
He H, Hoyer K, Tao H, Rice R, Jimenez J, Tardiff JC, Ingwall JS. Myosin-driven rescue of contractile reserve and energetics in mouse hearts bearing familial hypertrophic cardiomyopathy-associated mutant troponin T is mutation-specific. J Physiol 2012; 590:5371-88. [PMID: 22907055 DOI: 10.1113/jphysiol.2012.234252] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The thin filament protein troponin T (TnT) is a regulator of sarcomere function. Whole heart energetics and contractile reserve are compromised in transgenic mice bearing missense mutations at R92 within the tropomyosin-binding domain of cTnT, despite being distal to the ATP hydrolysis domain of myosin. These mutations are associated with familial hypertrophic cardiomyopathy (FHC). Here we test the hypothesis that genetically replacing murine αα-MyHC with murine ββ-MyHC in hearts bearing the R92Q cTnT mutation, a particularly lethal FHC-associated mutation, leads to sufficiently large perturbations in sarcomere function to rescue whole heart energetics and decrease the cost of contraction. By comparing R92Q cTnT and R92L cTnT mutant hearts, we also test whether any rescue is mutation-specific. We defined the energetic state of the isolated perfused heart using (31)P-NMR spectroscopy while simultaneously measuring contractile performance at four work states. We found that the cost of increasing contraction in intact mouse hearts with R92Q cTnT depends on the type of myosin present in the thick filament. We also found that the salutary effect of this manoeuvre is mutation-specific, demonstrating the major regulatory role of cTnT on sarcomere function at the whole heart level.
Collapse
Affiliation(s)
- Huamei He
- NMR Laboratory for Physiological Chemistry, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Hypertrophic cardiomyopathy (HCM) is the most-common monogenically inherited form of heart disease, characterized by thickening of the left ventricular wall, contractile dysfunction, and potentially fatal arrhythmias. HCM is also the most-common cause of sudden cardiac death in individuals younger than 35 years of age. Much progress has been made in the elucidation of the genetic basis of HCM, resulting in the identification of more than 900 individual mutations in over 20 genes. Interestingly, most of these genes encode sarcomeric proteins, such as myosin-7 (also known as cardiac muscle β-myosin heavy chain; MYH7), cardiac myosin-binding protein C (MYBPC3), and cardiac muscle troponin T (TNNT2). However, the molecular events that ultimately lead to the clinical phenotype of HCM are still unclear. We discuss several potential pathways, which include altered calcium cycling and sarcomeric calcium sensitivity, increased fibrosis, disturbed biomechanical stress sensing, and impaired cardiac energy homeostasis. An improved understanding of the pathological mechanisms involved will result in greater specificity and success of therapies for patients with HCM.
Collapse
Affiliation(s)
- Norbert Frey
- Department of Cardiology and Angiology, University of Kiel, Schittenhelmstrasse 12, 24105 Kiel, Germany
| | | | | |
Collapse
|
47
|
Abstract
Sixteen years ago, mutations in cardiac troponin (Tn)T and α-tropomyosin were linked to familial hypertrophic cardiomyopathy, thus transforming the disorder from a disease of the β-myosin heavy chain to a disease of the cardiac sarcomere. From the outset, studies suggested that mutations in the regulatory thin filament caused a complex, heterogeneous pattern of ventricular remodeling with wide variations in clinical expression. To date, the clinical heterogeneity is well matched by an extensive array of nearly 100 independent mutations in all components of the cardiac thin filament. Significant advances in our understanding of the biophysics of myofilament activation, coupled to the emerging evidence that thin filament linked cardiomyopathies are progressive, suggests that a renewed focus on the most proximal events in both the molecular and clinical pathogenesis of the disease will be necessary to achieve the central goal of using genotype information to manage affected patients. In this review, we examine the existing biophysical and clinical evidence in support of a more proximal definition of thin filament cardiomyopathies. In addition, new high-resolution, integrated approaches are presented to help define the way forward as the field works toward developing a more robust link between genotype and phenotype in this complex disorder.
Collapse
Affiliation(s)
- Jil C Tardiff
- Department of Physiology and Biophysics, Department of Internal Medicine, Division of Adult Cardiology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
48
|
Pinz I, Tian R, Belke D, Swanson E, Dillmann W, Ingwall JS. Compromised myocardial energetics in hypertrophied mouse hearts diminish the beneficial effect of overexpressing SERCA2a. J Biol Chem 2011; 286:10163-8. [PMID: 21278384 DOI: 10.1074/jbc.m110.210757] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The sarcoplasmic reticulum calcium ATPase (SERCA) plays a central role in regulating intracellular Ca(2+) homeostasis and myocardial contractility. Several studies show that improving Ca(2+) handling in hypertrophied rodent hearts by increasing SERCA activity results in enhanced contractile function. This suggests that SERCA is a potential target for gene therapy in cardiac hypertrophy and failure. However, it raises the issue of increased energy cost resulting from a higher ATPase activity. In this study, we determined whether SERCA overexpression alters the energy cost of increasing myocardial contraction in mouse hearts with pressure-overload hypertrophy using (31)P NMR spectroscopy. We isolated and perfused mouse hearts from wild-type (WT) and transgenic (TG) mice overexpressing the cardiac isoform of SERCA (SERCA2a) 8 weeks after ascending aortic constriction (left ventricular hypertrophy (LVH)) or sham operation. We found that overexpressing SERCA2a enhances myocardial contraction and relaxation in normal mouse hearts during inotropic stimulation with isoproterenol. Energy consumption was proportionate to the increase in contractile function. Thus, increasing SERCA2a expression in the normal heart allows an enhanced inotropic response with no compromise in energy supply and demand. However, this advantage was not sustained in LVH hearts in which the energetic status was compromised. Although the overexpression of SERCA2a prevented the down-regulation of SERCA protein in LVH hearts, TG-LVH hearts showed no increase in inotropic response when compared with WT-LVH hearts. Our results suggest that energy supply may be a limiting factor for the benefit of SERCA overexpression in hypertrophied hearts. Thus, strategies combining energetic support with increasing SERCA activity may improve the therapeutic effectiveness for heart failure.
Collapse
Affiliation(s)
- Ilka Pinz
- NMR Laboratory for Physiological Chemistry, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Jimenez J, Tardiff JC. Abnormal heart rate regulation in murine hearts with familial hypertrophic cardiomyopathy-related cardiac troponin T mutations. Am J Physiol Heart Circ Physiol 2010; 300:H627-35. [PMID: 21131475 DOI: 10.1152/ajpheart.00247.2010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mutations in cardiac troponin T (cTnT), Δ160E and R92Q, have been linked to familial hypertrophic cardiomyopathy (FHC), and some studies have indicated that these mutations can lead to a high incidence of sudden cardiac death in the relative absence of significant ventricular hypertrophy. Alterations in autonomic function have been documented in patients with hypertrophic cardiomyopathy. We hypothesize that alterations in autonomic function may contribute to mutation-specific clinical phenotypes in cTnT-related FHC. Heart rate (HR) variability (HRV) has been used to assess autonomic function from an electrocardiograph. Nontransgenic, Δ160E, or R92Q mice were implanted with radiofrequency transmitters to obtain continuous electrocardiograph recordings during 24-h baseline and 30-min recordings after β-adrenergic receptor drug injections. Although Δ160E mice did not differ from nontransgenic mice for any 24-h HRV measurements, R92Q mice had impaired HR regulation, as measured by a decrease in the SD of the R-R interval, a decrease in the low frequency-to-high frequency ratio, a decrease in normalized low frequency, and an increase in normalized high frequency. β-Adrenergic receptor density measurements and HRV analysis after drug injections did not reveal any significant differences for Δ160E or R92Q mice versus nontransgenic mice. Arrhythmia analysis revealed both an increased incidence of heart block in R92Q mice at baseline and frequency of premature ventricular contractions after isoproterenol injections in Δ160E and R92Q mice. In addition, Δ160E and R92Q mice exhibited a prolonged P duration after drug injections. Therefore, between two independent and clinically severe cTnT mutations within the same functional domain, only R92Q mice exhibited altered autonomic function, whereas both mutations demonstrated abnormalities in conduction and ventricular ectopy.
Collapse
Affiliation(s)
- Jesus Jimenez
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Ullmann Bldg. 316, Bronx, NY 10803, USA
| | | |
Collapse
|
50
|
Abstract
The aim of this study was to make cellular-level measurements of the mechanical efficiency of mouse cardiac muscle and to use these measurements to determine (1) the work performed by a cross-bridge in one ATP-splitting cycle and (2) the fraction of the free energy available in metabolic substrates that is transferred by oxidative phosphorylation to free energy in ATP (i.e. mitochondrial thermodynamic efficiency). Experiments were performed using isolated left ventricular mouse papillary muscles (n = 9; studied at 27°C) and the myothermic technique. The production of work and heat was measured during and after 40 contractions at a contraction frequency of 2 Hz. Each contraction consisted of a brief isometric period followed by isovelocity shortening. Work output, heat output and enthalpy output were all independent of shortening velocity. Maximum initial mechanical efficiency (mean ± SEM) was 31.1 ± 1.3% and maximum net mechanical efficiency 16.9 ± 1.5%. It was calculated that the maximum work per cross-bridge cycle was 20 zJ, comparable to values for mouse skeletal muscle, and that mitochondrial thermodynamic efficiency was 72%. Analysis of data in the literature suggests that mitochondrial efficiency of cardiac muscle from other species is also likely to be between 70 and 80%.
Collapse
|