1
|
Taylor J, Lal S, Braet F, McLachlan CS, Li A. The molecular and cellular landscape of hypertrophic cardiomyopathy phenotypes: transition from obstructive to end-stage heart failure. J Mol Med (Berl) 2025; 103:113-123. [PMID: 39774683 DOI: 10.1007/s00109-024-02508-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a myocardial disorder which commonly presents as an obstructive or end-stage disease. This study aims to investigate the transcriptomic changes related to cardiac cell-specific expression profiles that underpin the molecular transition between the HCM phenotypes. This study utilizes bioinformatics meta-analysis to integrate independent datasets to generate a comprehensive gene expression profile of obstructive HCM and end-stage HCM phenotypes compared to donor hearts. Gene set enrichment and cellular deconvolution were applied to identify ontologies and pathways related to each phenotype and to enumerate cell abundances. The intersection between cell lineage genes and meta-genes was identified to explore the cellular contribution to the phenotypic molecular signatures. Meta-analysis revealed, enhanced muscle function and myocardial remodeling, alongside impaired immune and inflammatory processes in obstructive HCM. In contrast, enriched tissue matrix remodeling pathways and altered metabolic and signaling cascades were identified in end-stage HCM, indicating a shift towards cellular dysfunction and loss of homeostasis. These molecular profiles were associated with an altered cellular landscape, with increased cardiomyocytes and lower immune cell populations in obstructive samples but increased fibroblasts and smooth muscle cells in end-stage HCM, implicating extensive tissue remodeling. This study provides novel insights into the cellular contributions of contractile, immune, homeostatic, and vascular alterations underpinning each of the HCM phenotypes. KEY MESSAGES: HCM phenotypes are characterized by distinct molecular and cellular profiles. Obstructive HCM has an enriched contractile profile underpinned by an expanded cardiomyocyte population. End-stage HCM shifts the cellular profile towards extracellular and vascular remodeling.
Collapse
Affiliation(s)
- Jude Taylor
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, 2010, Australia
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Filip Braet
- School of Medical Sciences (Molecular and Cellular Biomedicine), University of Sydney, Sydney, NSW, 2006, Australia
- Australian Centre for Microscopy and Microanalysis, University of Sydney, Sydney, NSW, 2006, Australia
| | - Craig S McLachlan
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, 2010, Australia
| | - Amy Li
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, 2010, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, 3550, Australia.
| |
Collapse
|
2
|
Mohran S, McMillen TS, Mandrycky C, Tu AY, Kooiker KB, Qian W, Neys S, Osegueda B, Moussavi-Harami F, Irving TC, Regnier M, Ma W. Calcium has a direct effect on thick filament activation in porcine myocardium. J Gen Physiol 2024; 156:e202413545. [PMID: 39302315 PMCID: PMC11415303 DOI: 10.1085/jgp.202413545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/03/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
Sarcomere activation in striated muscle requires both thin filament-based and thick filament-based activation mechanisms. Recent studies have shown that myosin heads on the thick filaments undergo OFF to ON structural transitions in response to calcium (Ca2+) in permeabilized porcine myocardium in the presence of a small molecule inhibitor that eliminated active force. The changes in X-ray diffraction signatures of OFF to ON transitions were interpreted as Ca2+ acting to activate the thick filaments. Alternatively, Ca2+ binding to troponin could initiate a Ca2+-dependent crosstalk from the thin filament to the thick filament via interfilament connections such as the myosin binding protein-C. Here, we exchanged native troponin in permeabilized porcine myocardium for troponin containing the cTnC D65A mutation, which disallows the activation of troponin through Ca2+ binding to determine if Ca2+-dependent thick filament activation persists in the absence of thin filament activation. After the exchange protocol, over 95% of the Ca2+-activated force was eliminated. Equatorial intensity ratio increased significantly in both WT and D65A exchanged myocardium with increasing Ca2+ concentration. The degree of helical ordering of the myosin heads decreased by the same amount in WT and D65A myocardium when Ca2+ concentration increased. These results are consistent with a direct effect of Ca2+ in activating the thick filament rather than an indirect effect due to Ca2+-mediated crosstalk between the thick and thin filaments.
Collapse
Affiliation(s)
- Saffie Mohran
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Timothy S. McMillen
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
| | - Christian Mandrycky
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
| | - An-Yue Tu
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kristina B. Kooiker
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Wenjing Qian
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Stephanie Neys
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Brayan Osegueda
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Farid Moussavi-Harami
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Thomas C. Irving
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Center of Translational Muscle Research, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Weikang Ma
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
- Center for Synchrotron Radiation Research and Instrumentation, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
3
|
Guo J, Jiang H, Schuftan D, Moreno JD, Ramahdita G, Aryan L, Bhagavan D, Silva J, Huebsch N. Substrate mechanics unveil early structural and functional pathology in iPSC micro-tissue models of hypertrophic cardiomyopathy. iScience 2024; 27:109954. [PMID: 38827401 PMCID: PMC11141149 DOI: 10.1016/j.isci.2024.109954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Accepted: 05/08/2024] [Indexed: 06/04/2024] Open
Abstract
Hypertension is a major cause of morbidity and mortality in patients with hypertrophic cardiomyopathy (HCM), suggesting a potential role for mechanics in HCM pathogenesis. Here, we developed an in vitro physiological model to investigate how mechanics acts together with HCM-linked myosin binding protein C (MYBPC3) mutations to trigger disease. Micro-heart muscles (μHM) were engineered from induced pluripotent stem cell (iPSC)-derived cardiomyocytes bearing MYBPC3+/- mutations and challenged to contract against substrates of different elasticity. μHMs that worked against substrates with stiffness at or exceeding the stiffness of healthy adult heart muscle exhibited several hallmarks of HCM, including cellular hypertrophy, impaired contractile energetics, and maladaptive calcium handling. Remarkably, we discovered changes in troponin C and T localization in MYBPC3+/- μHM that were entirely absent in 2D culture. Pharmacologic studies suggested that excessive Ca2+ intake through membrane-embedded channels underlie the observed electrophysiological abnormalities. These results illustrate the power of physiologically relevant engineered tissue models to study inherited disease with iPSC technology.
Collapse
Affiliation(s)
- Jingxuan Guo
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Huanzhu Jiang
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - David Schuftan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Jonathan D. Moreno
- Division of Cardiology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Ghiska Ramahdita
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Druv Bhagavan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Jonathan Silva
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| |
Collapse
|
4
|
Menezes Junior ADS, de França-e-Silva ALG, de Oliveira HL, de Lima KBA, Porto IDOP, Pedroso TMA, Silva DDME, Freitas AF. Genetic Mutations and Mitochondrial Redox Signaling as Modulating Factors in Hypertrophic Cardiomyopathy: A Scoping Review. Int J Mol Sci 2024; 25:5855. [PMID: 38892064 PMCID: PMC11173352 DOI: 10.3390/ijms25115855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a heart condition characterized by cellular and metabolic dysfunction, with mitochondrial dysfunction playing a crucial role. Although the direct relationship between genetic mutations and mitochondrial dysfunction remains unclear, targeting mitochondrial dysfunction presents promising opportunities for treatment, as there are currently no effective treatments available for HCM. This review adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews guidelines. Searches were conducted in databases such as PubMed, Embase, and Scopus up to September 2023 using "MESH terms". Bibliographic references from pertinent articles were also included. Hypertrophic cardiomyopathy (HCM) is influenced by ionic homeostasis, cardiac tissue remodeling, metabolic balance, genetic mutations, reactive oxygen species regulation, and mitochondrial dysfunction. The latter is a common factor regardless of the cause and is linked to intracellular calcium handling, energetic and oxidative stress, and HCM-induced hypertrophy. Hypertrophic cardiomyopathy treatments focus on symptom management and complication prevention. Targeted therapeutic approaches, such as improving mitochondrial bioenergetics, are being explored. This includes coenzyme Q and elamipretide therapies and metabolic strategies like therapeutic ketosis. Understanding the biomolecular, genetic, and mitochondrial mechanisms underlying HCM is crucial for developing new therapeutic modalities.
Collapse
Affiliation(s)
- Antonio da Silva Menezes Junior
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Ana Luísa Guedes de França-e-Silva
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Henrique Lima de Oliveira
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Khissya Beatryz Alves de Lima
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Iane de Oliveira Pires Porto
- Faculdade de Medicina, Universidade de Rio Verde (UniRV), Campus Aparecida, Aparecida de Goiânia 74345-030, Brazil; (I.d.O.P.P.); (T.M.A.P.)
| | - Thays Millena Alves Pedroso
- Faculdade de Medicina, Universidade de Rio Verde (UniRV), Campus Aparecida, Aparecida de Goiânia 74345-030, Brazil; (I.d.O.P.P.); (T.M.A.P.)
| | - Daniela de Melo e Silva
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| | - Aguinaldo F. Freitas
- Faculdade de Medicina, Departamento de Clínica Médica, Universidade Federal de Goiás (UFG), Goiânia 74020-020, Brazil; (A.L.G.d.F.-e.-S.); (H.L.d.O.); (K.B.A.d.L.); (D.d.M.e.S.); (A.F.F.J.)
| |
Collapse
|
5
|
Adeniran I, Wadee H, Degens H. An In Silico Cardiomyocyte Reveals the Impact of Changes in CaMKII Signalling on Cardiomyocyte Contraction Kinetics in Hypertrophic Cardiomyopathy. BIOMED RESEARCH INTERNATIONAL 2024; 2024:6160554. [PMID: 38567164 PMCID: PMC10985279 DOI: 10.1155/2024/6160554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 02/27/2024] [Accepted: 03/09/2024] [Indexed: 04/04/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is characterised by asymmetric left ventricular hypertrophy, ventricular arrhythmias, and cardiomyocyte dysfunction that may cause sudden death. HCM is associated with mutations in sarcomeric proteins and is usually transmitted as an autosomal-dominant trait. The aim of this in silico study was to assess the mechanisms that underlie the altered electrophysiological activity, contractility, regulation of energy metabolism, and crossbridge cycling in HCM at the single-cell level. To investigate this, we developed a human ventricular cardiomyocyte model that incorporates electrophysiology, metabolism, and force generation. The model was validated by its ability to reproduce the experimentally observed kinetic properties of human HCM induced by (a) remodelling of several ion channels and Ca2+-handling proteins arising from altered Ca2+/calmodulin kinase II signalling pathways and (b) increased Ca2+ sensitivity of the myofilament proteins. Our simulation showed a decreased phosphocreatine-to-ATP ratio (-9%) suggesting a negative mismatch between energy expenditure and supply. Using a spatial myofilament half-sarcomere model, we also compared the fraction of detached, weakly bound, and strongly bound crossbridges in the control and HCM conditions. Our simulations showed that HCM has more crossbridges in force-producing states than in the control condition. In conclusion, our model reveals that impaired crossbridge kinetics is accompanied by a negative mismatch between the ATP supply and demand ratio. This suggests that improving this ratio may reduce the incidence of sudden death in HCM.
Collapse
Affiliation(s)
- Ismail Adeniran
- Centre for Advanced Computational Science, Manchester Metropolitan University, Manchester M15 6BH, UK
| | - Hafsa Wadee
- Centre for Advanced Computational Science, Manchester Metropolitan University, Manchester M15 6BH, UK
| | - Hans Degens
- Department of Life Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK
- Lithuanian Sports University, Sporto 6, LT-44221 Kaunas, Lithuania
| |
Collapse
|
6
|
Ananthamohan K, Stelzer JE, Sadayappan S. Hypertrophic cardiomyopathy in MYBPC3 carriers in aging. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:9. [PMID: 38406555 PMCID: PMC10883298 DOI: 10.20517/jca.2023.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by abnormal thickening of the myocardium, leading to arrhythmias, heart failure, and elevated risk of sudden cardiac death, particularly among the young. This inherited disease is predominantly caused by mutations in sarcomeric genes, among which those in the cardiac myosin binding protein-C3 (MYBPC3) gene are major contributors. HCM associated with MYBPC3 mutations usually presents in the elderly and ranges from asymptomatic to symptomatic forms, affecting numerous cardiac functions and presenting significant health risks with a spectrum of clinical manifestations. Regulation of MYBPC3 expression involves various transcriptional and translational mechanisms, yet the destiny of mutant MYBPC3 mRNA and protein in late-onset HCM remains unclear. Pathogenesis related to MYBPC3 mutations includes nonsense-mediated decay, alternative splicing, and ubiquitin-proteasome system events, leading to allelic imbalance and haploinsufficiency. Aging further exacerbates the severity of HCM in carriers of MYBPC3 mutations. Advancements in high-throughput omics techniques have identified crucial molecular events and regulatory disruptions in cardiomyocytes expressing MYBPC3 variants. This review assesses the pathogenic mechanisms that promote late-onset HCM through the lens of transcriptional, post-transcriptional, and post-translational modulation of MYBPC3, underscoring its significance in HCM across carriers. The review also evaluates the influence of aging on these processes and MYBPC3 levels during HCM pathogenesis in the elderly. While pinpointing targets for novel medical interventions to conserve cardiac function remains challenging, the emergence of personalized omics offers promising avenues for future HCM treatments, particularly for late-onset cases.
Collapse
Affiliation(s)
- Kalyani Ananthamohan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 45267, USA
| | - Sakthivel Sadayappan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
7
|
Coleman JA, Ashkir Z, Raman B, Bueno-Orovio A. Mechanisms and prognostic impact of myocardial ischaemia in hypertrophic cardiomyopathy. Int J Cardiovasc Imaging 2023; 39:1979-1996. [PMID: 37358707 PMCID: PMC10589194 DOI: 10.1007/s10554-023-02894-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/03/2023] [Indexed: 06/27/2023]
Abstract
Despite the progress made in risk stratification, sudden cardiac death and heart failure remain dreaded complications for hypertrophic cardiomyopathy (HCM) patients. Myocardial ischaemia is widely acknowledged as a contributor to cardiovascular events, but the assessment of ischaemia is not yet included in HCM clinical guidelines. This review aims to evaluate the HCM-specific pro-ischaemic mechanisms and the potential prognostic value of imaging for myocardial ischaemia in HCM. A literature review was performed using PubMed to identify studies with non-invasive imaging of ischaemia (cardiovascular magnetic resonance, echocardiography, and nuclear imaging) in HCM, prioritising studies published after the last major review in 2009. Other studies, including invasive ischaemia assessment and post-mortem histology, were also considered for mechanistic or prognostic relevance. Pro-ischaemic mechanisms in HCM reviewed included the effects of sarcomeric mutations, microvascular remodelling, hypertrophy, extravascular compressive forces and left ventricular outflow tract obstruction. The relationship between ischaemia and fibrosis was re-appraised by considering segment-wise analyses in multimodal imaging studies. The prognostic significance of myocardial ischaemia in HCM was evaluated using longitudinal studies with composite endpoints, and reports of ischaemia-arrhythmia associations were further considered. The high prevalence of ischaemia in HCM is explained by several micro- and macrostructural pathological features, alongside mutation-associated energetic impairment. Ischaemia on imaging identifies a subgroup of HCM patients at higher risk of adverse cardiovascular outcomes. Ischaemic HCM phenotypes are a high-risk subgroup associated with more advanced left ventricular remodelling, but further studies are required to evaluate the independent prognostic value of non-invasive imaging for ischaemia.
Collapse
Affiliation(s)
- James A Coleman
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Zakariye Ashkir
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Betty Raman
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | |
Collapse
|
8
|
Wang K, Schriver BJ, Aschar-Sobbi R, Yi AY, Feric NT, Graziano MP. Human engineered cardiac tissue model of hypertrophic cardiomyopathy recapitulates key hallmarks of the disease and the effect of chronic mavacamten treatment. Front Bioeng Biotechnol 2023; 11:1227184. [PMID: 37771571 PMCID: PMC10523579 DOI: 10.3389/fbioe.2023.1227184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Introduction: The development of patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) offers an opportunity to study genotype-phenotype correlation of hypertrophic cardiomyopathy (HCM), one of the most common inherited cardiac diseases. However, immaturity of the iPSC-CMs and the lack of a multicellular composition pose concerns over its faithfulness in disease modeling and its utility in developing mechanism-specific treatment. Methods: The Biowire platform was used to generate 3D engineered cardiac tissues (ECTs) using HCM patient-derived iPSC-CMs carrying a β-myosin mutation (MYH7-R403Q) and its isogenic control (WT), withal ECTs contained healthy human cardiac fibroblasts. ECTs were subjected to electro-mechanical maturation for 6 weeks before being used in HCM phenotype studies. Results: Both WT and R403Q ECTs exhibited mature cardiac phenotypes, including a lack of automaticity and a ventricular-like action potential (AP) with a resting membrane potential < -75 mV. Compared to WT, R403Q ECTs demonstrated many HCM-associated pathological changes including increased tissue size and cell volume, shortened sarcomere length and disorganized sarcomere structure. In functional assays, R403Q ECTs showed increased twitch amplitude, slower contractile kinetics, a less pronounced force-frequency relationship, a smaller post-rest potentiation, prolonged AP durations, and slower Ca2+ transient decay time. Finally, we observed downregulation of calcium handling genes and upregulation of NPPB in R403Q vs. WT ECTs. In an HCM phenotype prevention experiment, ECTs were treated for 5-weeks with 250 nM mavacamten or a vehicle control. We found that chronic mavacamten treatment of R403Q ECTs: (i) shortened relaxation time, (ii) reduced APD90 prolongation, (iii) upregulated ADRB2, ATP2A2, RYR2, and CACNA1C, (iv) decreased B-type natriuretic peptide (BNP) mRNA and protein expression levels, and (v) increased sarcomere length and reduced sarcomere disarray. Discussion: Taken together, we demonstrated R403Q ECTs generated in the Biowire platform recapitulated many cardiac hypertrophy phenotypes and that chronic mavacamten treatment prevented much of the pathology. This demonstrates that the Biowire ECTs are well-suited to phenotypic-based drug discovery in a human-relevant disease model.
Collapse
Affiliation(s)
- Kai Wang
- Valo Health, Inc., Department of Discovery Research, New York, NY, United States
| | | | | | | | | | | |
Collapse
|
9
|
Glavaški M, Velicki L, Vučinić N. Hypertrophic Cardiomyopathy: Genetic Foundations, Outcomes, Interconnections, and Their Modifiers. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1424. [PMID: 37629714 PMCID: PMC10456451 DOI: 10.3390/medicina59081424] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent heritable cardiomyopathy. HCM is considered to be caused by mutations in cardiac sarcomeric protein genes. Recent research suggests that the genetic foundation of HCM is much more complex than originally postulated. The clinical presentations of HCM are very variable. Some mutation carriers remain asymptomatic, while others develop severe HCM, terminal heart failure, or sudden cardiac death. Heterogeneity regarding both genetic mutations and the clinical course of HCM hinders the establishment of universal genotype-phenotype correlations. However, some trends have been identified. The presence of a mutation in some genes encoding sarcomeric proteins is associated with earlier HCM onset, more severe left ventricular hypertrophy, and worse clinical outcomes. There is a diversity in the mechanisms implicated in the pathogenesis of HCM. They may be classified into groups, but they are interrelated. The lack of known supplementary elements that control the progression of HCM indicates that molecular mechanisms that exist between genotype and clinical presentations may be crucial. Secondary molecular changes in pathways implicated in HCM pathogenesis, post-translational protein modifications, and epigenetic factors affect HCM phenotypes. Cardiac loading conditions, exercise, hypertension, diet, alcohol consumption, microbial infection, obstructive sleep apnea, obesity, and environmental factors are non-molecular aspects that change the HCM phenotype. Many mechanisms are implicated in the course of HCM. They are mostly interconnected and contribute to some extent to final outcomes.
Collapse
Affiliation(s)
- Mila Glavaški
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
| | - Lazar Velicki
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
- Institute of Cardiovascular Diseases Vojvodina, Put Doktora Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Nataša Vučinić
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
| |
Collapse
|
10
|
Beltrami M, Fedele E, Fumagalli C, Mazzarotto F, Girolami F, Ferrantini C, Coppini R, Tofani L, Bertaccini B, Poggesi C, Olivotto I. Long-Term Prevalence of Systolic Dysfunction in MYBPC3 Versus MYH7-Related Hypertrophic Cardiomyopathy. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:363-371. [PMID: 37409452 DOI: 10.1161/circgen.122.003832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 05/30/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND The 2 sarcomere genes most commonly associated with hypertrophic cardiomyopathy (HCM), MYBPC3 (myosin-binding protein C3) and MYH7 (β-myosin heavy chain), are indistinguishable at presentation, and genotype-phenotype correlations have been elusive. Based on molecular and pathophysiological differences, however, it is plausible to hypothesize a different behavior in myocardial performance, impacting lifetime changes in left ventricular (LV) function. METHODS We reviewed the initial and final echocardiograms of 402 consecutive HCM patients with pathogenic or likely pathogenic MYBPC3 (n=251) or MYH7 (n=151) mutations, followed over 9±8 years. RESULTS At presentation, MYBPC3 patients were less frequently obstructive (15% versus 26%; P=0.005) and had lower LV ejection fraction compared with MYH7 (66±8% versus 68±8%, respectively; P=0.03). Both HCM patients harboring MYBPC3 and MYH7 mutations exhibited a small but significant decline in LV systolic function during follow-up; however, new onset of severe LV systolic dysfunction (LV ejection fraction, <50%) was greater among MYBPC3 patients (15% versus 5% among MYH7; P=0.013). Prevalence of grade II/III diastolic dysfunction at final evaluation was comparable between MYBPC3 and MYH7 patients (P=0.509). In a Cox multivariable analysis, MYBPC3-positive status (hazard ratio, 2.53 [95% CI, 1.09-5.82]; P=0.029), age (hazard ratio, 1.03 [95% CI, 1.00-1.06]; P=0.027), and atrial fibrillation (hazard ratio, 2.39 [95% CI, 1.14-5.05]; P=0.020) were independent predictors of severe systolic dysfunction. No statistically significant differences occurred with regard to incidence of atrial fibrillation, heart failure, appropriate implanted cardioverter defibrillator shock, or cardiovascular death. CONCLUSIONS MYBPC3-related HCM showed increased long-term prevalence of systolic dysfunction compared with MYH7, in spite of similar outcome. Such observations suggest different pathophysiology of clinical progression in the 2 subsets and may prove relevant for understanding of genotype-phenotype correlations in HCM.
Collapse
Affiliation(s)
- Matteo Beltrami
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (M.B., C.F.)
| | - Elisa Fedele
- Department of Cardiology, Policlinico Casilino, Rome, Italy (E.F.)
| | - Carlo Fumagalli
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (M.B., C.F.)
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania "Luigi Vanvitelli," Naples, Italy (C.F.)
| | - Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Italy (F.M.)
| | | | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine (C.F., C.P., I.O.), University of Florence, Italy
| | - Raffaele Coppini
- Division of Pharmacology, Department of Neuroscience, Psychology, Drug Sciences and Child Health (NeuroFarBa) (R.C.), University of Florence, Italy
| | - Lorenzo Tofani
- Department of Statistics, Computer Science, Applications (L.T., B.B.), University of Florence, Italy
| | - Bruno Bertaccini
- Department of Statistics, Computer Science, Applications (L.T., B.B.), University of Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine (C.F., C.P., I.O.), University of Florence, Italy
| | - Iacopo Olivotto
- Meyer Children's Hospital, IRCSS, Florence, Italy (F.G., I.O.)
- Department of Experimental and Clinical Medicine (C.F., C.P., I.O.), University of Florence, Italy
| |
Collapse
|
11
|
Nag S, Gollapudi SK, Del Rio CL, Spudich JA, McDowell R. Mavacamten, a precision medicine for hypertrophic cardiomyopathy: From a motor protein to patients. SCIENCE ADVANCES 2023; 9:eabo7622. [PMID: 37506209 DOI: 10.1126/sciadv.abo7622] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/23/2023] [Indexed: 07/30/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a primary myocardial disorder characterized by left ventricular hypertrophy, hyperdynamic contraction, and impaired relaxation of the heart. These functional derangements arise directly from altered sarcomeric function due to either mutations in genes encoding sarcomere proteins, or other defects such as abnormal energetics. Current treatment options do not directly address this causal biology but focus on surgical and extra-sarcomeric (sarcolemmal) pharmacological symptomatic relief. Mavacamten (formerly known as MYK-461), is a small molecule designed to regulate cardiac function at the sarcomere level by selectively but reversibly inhibiting the enzymatic activity of myosin, the fundamental motor of the sarcomere. This review summarizes the mechanism and translational progress of mavacamten from proteins to patients, describing how the mechanism of action and pharmacological characteristics, involving both systolic and diastolic effects, can directly target pathophysiological derangements within the cardiac sarcomere to improve cardiac structure and function in HCM. Mavacamten was approved by the Food and Drug Administration in April 2022 for the treatment of obstructive HCM and now goes by the commercial name of Camzyos. Full information about the risks, limitations, and side effects can be found at www.accessdata.fda.gov/drugsatfda_docs/label/2022/214998s000lbl.pdf.
Collapse
Affiliation(s)
- Suman Nag
- MyoKardia Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA 94005, USA
| | - Sampath K Gollapudi
- MyoKardia Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA 94005, USA
| | - Carlos L Del Rio
- MyoKardia Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA 94005, USA
- Cardiac Consulting, 1630 S Delaware St. #56426, San Mateo, CA 94403, USA
| | | | - Robert McDowell
- MyoKardia Inc., a wholly owned subsidiary of Bristol Myers Squibb, Brisbane, CA 94005, USA
| |
Collapse
|
12
|
Pirrotta F, Cavati G, Mingiano C, Merlotti D, Nuti R, Gennari L, Palazzuoli A. Vitamin D Deficiency and Cardiovascular Mortality: Retrospective Analysis "Siena Osteoporosis" Cohort. Nutrients 2023; 15:3303. [PMID: 37571241 PMCID: PMC10421091 DOI: 10.3390/nu15153303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/13/2023] Open
Abstract
Vitamin D is a fat-soluble vitamin that plays a key role in bone metabolism, particularly concerning the regulation of calcium and phosphate homeostasis. Cardiovascular disease (CVD) is the main cause of morbidity and mortality in Western countries. Knowledge of the role of vitamin D in CVD arose from evidence of the vitamin D receptor (VDR) inside the cardiovascular system. In this retrospective analysis, we investigated the relationships between vitamin D status and hospitalization for heart failure (HF), overall mortality and cardiovascular mortality. Between 2004 and 2009, age-stratified, random sampling of elderly men and postmenopausal women in the primary care registers of Siena residents was performed. In total, 174 males (mean ± SD, 65.9 ± 6 years) and 975 females (62.5 ± 6 years) were enrolled in the study. We investigated the association between 25OHD status and hospitalization for HF or causes of mortality. A total of 51 subjects (12 males and 39 females) had been hospitalized for acute HF. At the end of the survey, 931 individuals were alive, while 187 had died (43 males and 144 females). A greater proportion of deceased patients showed low 25OHD (particularly patients with levels below 20 ng/mL). A similar trend was observed concerning the prevalence of patients with 25OHD levels below 20 ng/mL who died from stroke (RR = 2.15; 95% CIs 0.98-4.69; p = 0.06). Low 25OHD levels may be predictive of cardiovascular mortality. Whether vitamin deficiency represents a primitive cause or is a simple bystander in increased cardiovascular mortality should be further investigated in prospective large cohort studies specifically designed to assess CVD risk, including a detailed assessment of cardiac dysfunction and the characterization of atherosclerotic lesions.
Collapse
Affiliation(s)
- Filippo Pirrotta
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (F.P.); (G.C.); (C.M.); (D.M.); (R.N.); (L.G.)
| | - Guido Cavati
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (F.P.); (G.C.); (C.M.); (D.M.); (R.N.); (L.G.)
| | - Christian Mingiano
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (F.P.); (G.C.); (C.M.); (D.M.); (R.N.); (L.G.)
| | - Daniela Merlotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (F.P.); (G.C.); (C.M.); (D.M.); (R.N.); (L.G.)
| | - Ranuccio Nuti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (F.P.); (G.C.); (C.M.); (D.M.); (R.N.); (L.G.)
| | - Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (F.P.); (G.C.); (C.M.); (D.M.); (R.N.); (L.G.)
| | - Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio-Thoracic and Vascular Department, Le Scotte Hospital, University of Siena, 53100 Siena, Italy
| |
Collapse
|
13
|
Cserne Szappanos H, Viola HM, Ito DW, Lim S, Mangala M, Holliday M, Barratt Ross S, Semsarian C, Hill A, Dixon RE, Hool LC. Cytoskeletal disarray increases arrhythmogenic vulnerability during sympathetic stimulation in a model of hypertrophic cardiomyopathy. Sci Rep 2023; 13:11296. [PMID: 37438479 PMCID: PMC10338442 DOI: 10.1038/s41598-023-38296-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/06/2023] [Indexed: 07/14/2023] Open
Abstract
Familial hypertrophic cardiomyopathy (FHC) patients are advised to avoid strenuous exercise due to increased risk of arrhythmias. Mice expressing the human FHC-causing mutation R403Q in the myosin heavy chain gene (MYH6) recapitulate the human phenotype, including cytoskeletal disarray and increased arrhythmia susceptibility. Following in vivo administration of isoproterenol, mutant mice exhibited tachyarrhythmias, poor recovery and fatigue. Arrhythmias were attenuated with the β-blocker atenolol and protein kinase A inhibitor PKI. Mutant cardiac myocytes had significantly prolonged action potentials and triggered automaticity due to reduced repolarization reserve and connexin 43 expression. Isoproterenol shortened cycle length, and escalated electrical instability. Surprisingly isoproterenol did not increase CaV1.2 current. We found alterations in CaV1.2-β1 adrenergic receptor colocalization assessed using super-resolution nanoscopy, and increased CaV1.2 phosphorylation in mutant hearts. Our results reveal for the first time that altered ion channel expression, co-localization and β-adrenergic receptor signaling associated with myocyte disarray contribute to electrical instability in the R403Q mutant heart.
Collapse
Affiliation(s)
| | - Helena M Viola
- School of Human Sciences, University of Western Australia, Crawley, WA, Australia
| | - Danica W Ito
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA
| | - Seakcheng Lim
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Melissa Mangala
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Mira Holliday
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Samantha Barratt Ross
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology, Centenary Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Adam Hill
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Rose E Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA
| | - Livia C Hool
- School of Human Sciences, University of Western Australia, Crawley, WA, Australia.
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
14
|
Chou C, Martin GL, Perera G, Awata J, Larson A, Blanton R, Chin MT. A novel αB-crystallin R123W variant drives hypertrophic cardiomyopathy by promoting maladaptive calcium-dependent signal transduction. Front Cardiovasc Med 2023; 10:1223244. [PMID: 37435054 PMCID: PMC10331725 DOI: 10.3389/fcvm.2023.1223244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disorder affecting 1 in 500 people in the general population. Characterized by asymmetric left ventricular hypertrophy, cardiomyocyte disarray and cardiac fibrosis, HCM is a highly complex disease with heterogenous clinical presentation, onset and complication. While mutations in sarcomere genes can account for a substantial proportion of familial cases of HCM, 40%-50% of HCM patients do not carry such sarcomere variants and the causal mutations for their diseases remain elusive. Recently, we identified a novel variant of the alpha-crystallin B chain (CRYABR123W) in a pair of monozygotic twins who developed concordant HCM phenotypes that manifested over a nearly identical time course. Yet, how CRYABR123W promotes the HCM phenotype remains unclear. Here, we generated mice carrying the CryabR123W knock-in allele and demonstrated that hearts from these animals exhibit increased maximal elastance at young age but reduced diastolic function with aging. Upon transverse aortic constriction, mice carrying the CryabR123W allele developed pathogenic left ventricular hypertrophy with substantial cardiac fibrosis and progressively decreased ejection fraction. Crossing of mice with a Mybpc3 frame-shift model of HCM did not potentiate pathological hypertrophy in compound heterozygotes, indicating that the pathological mechanisms in the CryabR123W model are independent of the sarcomere. In contrast to another well-characterized CRYAB variant (R120G) which induced Desmin aggregation, no evidence of protein aggregation was observed in hearts expressing CRYABR123W despite its potent effect on driving cellular hypertrophy. Mechanistically, we uncovered an unexpected protein-protein interaction between CRYAB and calcineurin. Whereas CRYAB suppresses maladaptive calcium signaling in response to pressure-overload, the R123W mutation abolished this effect and instead drove pathologic NFAT activation. Thus, our data establish the CryabR123W allele as a novel genetic model of HCM and unveiled additional sarcomere-independent mechanisms of cardiac pathological hypertrophy.
Collapse
Affiliation(s)
- Chun Chou
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| | - Gregory L. Martin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Gayani Perera
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Junya Awata
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Amy Larson
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Robert Blanton
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Michael T. Chin
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
15
|
McColgan G, Villarroel M, Gehmlich K. Should young athletes be screened for cardiomyopathies to reduce the burden of sudden cardiac death in athletes? Biophys Rev 2023; 15:321-327. [PMID: 37396442 PMCID: PMC10310562 DOI: 10.1007/s12551-023-01085-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/21/2023] [Indexed: 07/04/2023] Open
Abstract
In this correspondence, we highlight the risk of sudden cardiac death associated with undiagnosed cardiomyopathies. Life-threatening arrhythmias, which underlie sudden cardiac death, can be triggered by high-intensity exercise. It raises the question whether, and if so, how athletes should be screened for cardiomyopathies. The example of practice from Italy is discussed. We also briefly discuss novel developments, such as wearable biosensors and machine learning, which could be applied to screening for cardiomyopathies in future.
Collapse
Affiliation(s)
- Grace McColgan
- College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Mauricio Villarroel
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ UK
| | - Katja Gehmlich
- Institute of Cardiovascular Science, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, OX3 9DU UK
| |
Collapse
|
16
|
Liu T, Li X, Wang Y, Zhou M, Liang F. Computational modeling of electromechanical coupling in human cardiomyocyte applied to study hypertrophic cardiomyopathy and its drug response. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 231:107372. [PMID: 36736134 DOI: 10.1016/j.cmpb.2023.107372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/02/2023] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND AND OBJECTIVE Knowledge of electromechanical coupling in cardiomyocyte and how it is influenced by various pathophysiological factors is fundamental to understanding the pathogenesis of myocardial disease and its response to medication, which is however hard to be thoroughly addressed by clinical/experimental studies due to technical limitations. At this point, computational modeling offers an alternative approach. The main objective of the study was to develop a computational model capable of simulating the process of electromechanical coupling and quantifying the roles of various factors in play in the human left ventricular cardiomyocyte. METHODS A new electrophysiological model was firstly built by combining several existing electrophysiological models and incorporating the mechanism of electrophysiological homeostasis, which was subsequently coupled to models representing the cross-bridge dynamics and active force generation during excitation-contraction coupling and the passive mechanical properties of cardiomyocyte to yield an integrative electromechanical model. Model parameters were calibrated or optimized based on a large amount of experimental data. The resulting model was applied to delineate the characteristics of electromechanical coupling and explore underlying determinant factors in hypertrophic cardiomyopathy (HCM) cardiomyocyte, as well as quantify their changes in response to different medications. RESULTS Model predictions captured the major electromechanical characteristics of cardiomyocyte under both normal physiological and HCM conditions. In comparison with normal cardiomyocyte, HCM cardiomyocyte suffered from systemic changes in both electrophysiological and mechanical variables. Numerical simulations of drug response revealed that Mavacamten and Metoprolol could both reduce the active contractility and alleviate calcium overload but had marked differential influences on many other electromechanical variables, which theoretically explained why the two drugs have differential therapeutic effects. In addition, our numerical experiments demonstrated the important role of compensatory ion transport in maintaining electrophysiological homeostasis and regulating cytoplasmic volume. CONCLUSIONS A sophisticated computational model has the advantage of providing quantitative and integrative insights for understanding the pathogenesis and drug responses of HCM or other myocardial diseases at the level of cardiomyocyte, and hence may contribute as a useful complement to clinical/experimental studies. The model may also be coupled to tissue- or organ-level models to strengthen the physiological implications of macro-scale numerical simulations.
Collapse
Affiliation(s)
- Taiwei Liu
- Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Xuanyu Li
- Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Yue Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mi Zhou
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fuyou Liang
- Department of Engineering Mechanics, School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China; State Key Laboratory of Ocean Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow 19991, Russia.
| |
Collapse
|
17
|
Thakkar K, Karajgi AR, Kallamvalappil AM, Avanthika C, Jhaveri S, Shandilya A, Anusheel, Al-Masri R. Sudden cardiac death in childhood hypertrophic cardiomyopathy. Dis Mon 2023; 69:101548. [PMID: 36931945 DOI: 10.1016/j.disamonth.2023.101548] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
The most prevalent cause of mortality in children with hypertrophic cardiomyopathy (HCM) is sudden cardiac death (SCD), which happens more frequently than in adult patients. Risk stratification tactics have generally been drawn from adult practice, however emerging data has revealed significant disparities between children and adult cohorts, implying the need for pediatric-specific risk stratification methodologies. We conducted an all-language literature search on Medline, Cochrane, Embase, and Google Scholar until October 2021. The following search strings and Medical Subject Heading (MeSH) terms were used: "HCM," "SCD," "Sudden Cardiac Death," and "Childhood Onset HCM." We explored the literature on the risk of SCD in HCM for its epidemiology, pathophysiology, the role of various genes and their influence, associated complications leading to SCD and preventive and treatment modalities. Childhood-onset HCM is linked to significant life-long morbidity and mortality, including a higher SCD rate in children than in adults. The present focus is on symptom relief and avoiding illness-related consequences, but the prospect of future disease-modifying medicines offers an intriguing opportunity to alter disease expression and outcomes in these young individuals. Current preventive recommendations promote implantable cardioverter defibrillator placement based on cumulative risk factor thresholds, although they have been demonstrated to have weak discriminating capacity. This article addresses questions and discusses the etiology, risk factors, and method to risk stratification for SCD in children with HCM.
Collapse
Affiliation(s)
- Keval Thakkar
- G.M.E.R.S. Medical College and General Hospital, Gandhinagar, India
| | | | | | - Chaithanya Avanthika
- Karnataka Institute of Medical /Sciences, PB Rd, Vidya Nagar, Hubli, Karnataka, India.
| | | | | | - Anusheel
- Ryazan State I P Pavlov Medical Institute, Ryazan, Russia
| | - Rayan Al-Masri
- Jordan University of Science and technology, Irbid, Jordan
| |
Collapse
|
18
|
Halder SS, Rynkiewicz MJ, Creso JG, Sewanan LR, Howland L, Moore JR, Lehman W, Campbell SG. Mechanisms of pathogenicity in the hypertrophic cardiomyopathy-associated TPM1 variant S215L. PNAS NEXUS 2023; 2:pgad011. [PMID: 36896133 PMCID: PMC9991458 DOI: 10.1093/pnasnexus/pgad011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/12/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disorder often caused by mutations to sarcomeric genes. Many different HCM-associated TPM1 mutations have been identified but they vary in their degrees of severity, prevalence, and rate of disease progression. The pathogenicity of many TPM1 variants detected in the clinical population remains unknown. Our objective was to employ a computational modeling pipeline to assess pathogenicity of one such variant of unknown significance, TPM1 S215L, and validate predictions using experimental methods. Molecular dynamic simulations of tropomyosin on actin suggest that the S215L significantly destabilizes the blocked regulatory state while increasing flexibility of the tropomyosin chain. These changes were quantitatively represented in a Markov model of thin-filament activation to infer the impacts of S215L on myofilament function. Simulations of in vitro motility and isometric twitch force predicted that the mutation would increase Ca2+ sensitivity and twitch force while slowing twitch relaxation. In vitro motility experiments with thin filaments containing TPM1 S215L revealed higher Ca2+ sensitivity compared with wild type. Three-dimensional genetically engineered heart tissues expressing TPM1 S215L exhibited hypercontractility, upregulation of hypertrophic gene markers, and diastolic dysfunction. These data form a mechanistic description of TPM1 S215L pathogenicity that starts with disruption of the mechanical and regulatory properties of tropomyosin, leading thereafter to hypercontractility and finally induction of a hypertrophic phenotype. These simulations and experiments support the classification of S215L as a pathogenic mutation and support the hypothesis that an inability to adequately inhibit actomyosin interactions is the mechanism whereby thin-filament mutations cause HCM.
Collapse
Affiliation(s)
- Saiti S Halder
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | | | - Jenette G Creso
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| | - Lorenzo R Sewanan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Department of Internal Medicine, Columbia University, New York, NY 10032
| | - Lindsey Howland
- Department of Biological Sciences, University of Massachusetts Lowell, MA 01854
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts Lowell, MA 01854
| | - William Lehman
- Department of Physiology/Biophysics, Boston University, Boston, MA 02215
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| |
Collapse
|
19
|
Moore J, Ewoldt J, Venturini G, Pereira AC, Padilha K, Lawton M, Lin W, Goel R, Luptak I, Perissi V, Seidman CE, Seidman J, Chin MT, Chen C, Emili A. Multi-Omics Profiling of Hypertrophic Cardiomyopathy Reveals Altered Mechanisms in Mitochondrial Dynamics and Excitation-Contraction Coupling. Int J Mol Sci 2023; 24:4724. [PMID: 36902152 PMCID: PMC10002553 DOI: 10.3390/ijms24054724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Hypertrophic cardiomyopathy is one of the most common inherited cardiomyopathies and a leading cause of sudden cardiac death in young adults. Despite profound insights into the genetics, there is imperfect correlation between mutation and clinical prognosis, suggesting complex molecular cascades driving pathogenesis. To investigate this, we performed an integrated quantitative multi-omics (proteomic, phosphoproteomic, and metabolomic) analysis to illuminate the early and direct consequences of mutations in myosin heavy chain in engineered human induced pluripotent stem-cell-derived cardiomyocytes relative to late-stage disease using patient myectomies. We captured hundreds of differential features, which map to distinct molecular mechanisms modulating mitochondrial homeostasis at the earliest stages of pathobiology, as well as stage-specific metabolic and excitation-coupling maladaptation. Collectively, this study fills in gaps from previous studies by expanding knowledge of the initial responses to mutations that protect cells against the early stress prior to contractile dysfunction and overt disease.
Collapse
Affiliation(s)
- Jarrod Moore
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jourdan Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02218, USA
| | | | | | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Clinical Hospital, Faculty of Medicine, University of São Paulo, Sao Paulo 05508-000, Brazil
| | - Matthew Lawton
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Weiwei Lin
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Raghuveera Goel
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ivan Luptak
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA 02118, USA
| | - Valentina Perissi
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Jonathan Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02145, USA
| | - Christopher Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02218, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Andrew Emili
- Center for Network Systems Biology, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
20
|
Rao SJ, Iqbal SB, Kanwal AS, Aronow WS, Naidu SS. Multi-modality management of hypertrophic cardiomyopathy. Hosp Pract (1995) 2023; 51:2-11. [PMID: 36598161 DOI: 10.1080/21548331.2022.2162297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is an autosomal dominant inherited condition defined by left ventricular wall thickness greater than 15 mm in the absence of other conditions that could explain that degree of hypertrophy. Obstructive HCM associated with left ventricular outflow tract obstruction is defined by an intraventricular systolic pressure gradient greater than or equal to 30 mm Hg. Over the past couple of decades, there has been an expansion of both invasive and pharmacotherapeutic options for patients with HCM, with recent guidelines calling for a melody of invasive and non-invasive treatment strategies. There are several invasive therapies including proven therapies such as alcohol septal ablation and septal myectomy. Novel invasive therapies such as MitraClip, radiofrequency septal ablation and SESAME procedure have more recently been promoted. Pharmacological therapy has also dramatically evolved and includes conventional medications such as beta-blockers, calcium channel blockers, and disopyramide. Mavacamten, a novel cardiac myosin inhibitor, may significantly change management. Other myosin inhibitors and modulators are also being developed and tested in large clinical trials. Given significant phenotypical variability in patients with HCM, clinical management can be challenging, and often requires an individualized approach with a combination of invasive and non-invasive options.
Collapse
Affiliation(s)
- Shiavax J Rao
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, USA
| | - Shaikh B Iqbal
- Department of Medicine, MedStar Union Memorial Hospital, Baltimore, USA
| | - Arjun S Kanwal
- Department of Cardiology, Westchester Medical Center, Valhalla, USA
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center and Department of Medicine, New York Medical College, Valhalla, USA
| | - Srihari S Naidu
- Hypertrophic Cardiomyopathy Center, Cardiac Catheterization Laboratory, Department of Cardiology, Westchester Medical Center and Department of Medicine, New York Medical College, Valhalla, USA
| |
Collapse
|
21
|
Ho BX, Pang JKS, Chen Y, Loh YH, An O, Yang HH, Seshachalam VP, Koh JLY, Chan WK, Ng SY, Soh BS. Robust generation of human-chambered cardiac organoids from pluripotent stem cells for improved modelling of cardiovascular diseases. Stem Cell Res Ther 2022; 13:529. [PMID: 36544188 PMCID: PMC9773542 DOI: 10.1186/s13287-022-03215-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Tissue organoids generated from human pluripotent stem cells are valuable tools for disease modelling and to understand developmental processes. While recent progress in human cardiac organoids revealed the ability of these stem cell-derived organoids to self-organize and intrinsically formed chamber-like structure containing a central cavity, it remained unclear the processes involved that enabled such chamber formation. METHODS Chambered cardiac organoids (CCOs) differentiated from human embryonic stem cells (H7) were generated by modulation of Wnt/ß-catenin signalling under fully defined conditions, and several growth factors essential for cardiac progenitor expansion. Transcriptomic profiling of day 8, day 14 and day 21 CCOs was performed by quantitative PCR and single-cell RNA sequencing. Endothelin-1 (EDN1) known to induce oxidative stress in cardiomyocytes was used to induce cardiac hypertrophy in CCOs in vitro. Functional characterization of cardiomyocyte contractile machinery was performed by immunofluorescence staining and analysis of brightfield and fluorescent video recordings. Quantitative PCR values between groups were compared using two-tailed Student's t tests. Cardiac organoid parameters comparison between groups was performed using two-tailed Mann-Whitney U test when sample size is small; otherwise, Welch's t test was used. Comparison of calcium kinetics parameters derived from the fluorescent data was performed using two-tailed Student's t tests. RESULTS Importantly, we demonstrated that a threshold number of cardiac progenitor was essential to line the circumference of the inner cavity to ensure proper formation of a chamber within the organoid. Single-cell RNA sequencing revealed improved maturation over a time course, as evidenced from increased mRNA expression of cardiomyocyte maturation genes, ion channel genes and a metabolic shift from glycolysis to fatty acid ß-oxidation. Functionally, CCOs recapitulated clinical cardiac hypertrophy by exhibiting thickened chamber walls, reduced fractional shortening, and increased myofibrillar disarray upon treatment with EDN1. Furthermore, electrophysiological assessment of calcium transients displayed tachyarrhythmic phenotype observed as a consequence of rapid depolarization occurring prior to a complete repolarization. CONCLUSIONS Our findings shed novel insights into the role of progenitors in CCO formation and pave the way for the robust generation of cardiac organoids, as a platform for future applications in disease modelling and drug screening in vitro.
Collapse
Affiliation(s)
- Beatrice Xuan Ho
- grid.418812.60000 0004 0620 9243Disease Modelling and Therapeutics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore ,grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore
| | - Jeremy Kah Sheng Pang
- grid.418812.60000 0004 0620 9243Disease Modelling and Therapeutics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore ,grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore
| | - Ying Chen
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore ,grid.4280.e0000 0001 2180 6431Integrative Sciences and Engineering Programme, National University of Singapore, 21 Lower Kent Ridge Road, Singapore, 119077 Singapore ,grid.418812.60000 0004 0620 9243Epigenetics and Cell Fates Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore
| | - Yuin-Han Loh
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore ,grid.418812.60000 0004 0620 9243Epigenetics and Cell Fates Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore
| | - Omer An
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Singapore
| | - Henry He Yang
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Singapore
| | - Veerabrahma Pratap Seshachalam
- grid.510300.7Computational Phenomics Group, Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, 138670 Singapore
| | - Judice L. Y. Koh
- grid.510300.7Computational Phenomics Group, Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, 138670 Singapore
| | - Woon-Khiong Chan
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore
| | - Shi Yan Ng
- grid.418812.60000 0004 0620 9243Neurotherapeutics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore ,grid.4280.e0000 0001 2180 6431Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117456 Singapore ,grid.276809.20000 0004 0636 696XNational Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Boon Seng Soh
- grid.418812.60000 0004 0620 9243Disease Modelling and Therapeutics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673 Singapore ,grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117543 Singapore
| |
Collapse
|
22
|
Subramanian M, Atreya AR, Yalagudri SD, Shekar PV, Saggu DK, Narasimhan C. Catheter Ablation for Ventricular Arrhythmias in Hypertrophic Cardiomyopathy. Card Electrophysiol Clin 2022; 14:693-699. [PMID: 36396186 DOI: 10.1016/j.ccep.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Implantable cardioverter-defibrillators are the mainstay of therapy for prevention of sudden cardiac death in high-risk patients with hypertrophic cardiomyopathy (HCM). Catheter ablation is a useful option for patients with recurrent, drug refractory monomorphic ventricular tachycardia (VT), and device therapy. Compared with other nonischemic substrates, there are limited data on the role and outcomes of catheter ablation in HCM. The challenges of VT ablation in HCM patients include deep intramural and epicardial substrates, suboptimal power delivery, and higher recurrence due to progression of disease. Patient selection, using cardiac MRI scar localization, and optimizing ablation techniques can improve outcomes in these patients.
Collapse
Affiliation(s)
- Muthiah Subramanian
- Electrophysiology Section, AIG Hospitals Institute of Cardiac Sciences and Research, Mindspace Road, Gachibowli, Hyderabad 500032, India
| | - Auras R Atreya
- Electrophysiology Section, AIG Hospitals Institute of Cardiac Sciences and Research, Mindspace Road, Gachibowli, Hyderabad 500032, India; Division of Cardiovascular Medicine, Electrophysiology Section, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sachin D Yalagudri
- Electrophysiology Section, AIG Hospitals Institute of Cardiac Sciences and Research, Mindspace Road, Gachibowli, Hyderabad 500032, India
| | - P Vijay Shekar
- Electrophysiology Section, AIG Hospitals Institute of Cardiac Sciences and Research, Mindspace Road, Gachibowli, Hyderabad 500032, India
| | - Daljeet Kaur Saggu
- Electrophysiology Section, AIG Hospitals Institute of Cardiac Sciences and Research, Mindspace Road, Gachibowli, Hyderabad 500032, India
| | - Calambur Narasimhan
- Electrophysiology Section, AIG Hospitals Institute of Cardiac Sciences and Research, Mindspace Road, Gachibowli, Hyderabad 500032, India.
| |
Collapse
|
23
|
Doste R, Coppini R, Bueno-Orovio A. Remodelling of potassium currents underlies arrhythmic action potential prolongation under beta-adrenergic stimulation in hypertrophic cardiomyopathy. J Mol Cell Cardiol 2022; 172:120-131. [PMID: 36058298 DOI: 10.1016/j.yjmcc.2022.08.361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/15/2022] [Accepted: 08/27/2022] [Indexed: 12/14/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) patients often present an enhanced arrhythmogenicity that can lead to lethal arrhythmias, especially during exercise. Recent studies have indicated an abnormal response of HCM cardiomyocytes to β-adrenergic receptor stimulation (β-ARS), with prolongation of their action potential rather than shortening. The mechanisms underlying this aberrant response to sympathetic stimulation and its possible proarrhythmic role remain unknown. The aims of this study are to investigate the key ionic mechanisms underlying the HCM abnormal response to β-ARS and the resultant repolarisation abnormalities using human-based experimental and computational methodologies. We integrated and calibrated the latest models of human ventricular electrophysiology and β-ARS using experimental measurements of human adult cardiomyocytes from control and HCM patients. Our major findings include: (1) the developed in silico models of β-ARS capture the behaviour observed in the experimental data, including the aberrant response of HCM cardiomyocytes to β-ARS; (2) the reduced increase of potassium currents under β-ARS was identified as the main mechanism of action potential prolongation in HCM, rather than a more sustained inward calcium current; (3) action potential duration differences between healthy and HCM cardiomyocytes were increased upon β-ARS, while endocardial to epicardial differences in HCM cardiomyocytes were reduced; (4) models presenting repolarisation abnormalities were characterised by downregulation of the rapid delayed rectifier potassium current and the sodium‑potassium pump, while inward currents were upregulated. In conclusion, our results identify causal relationships between the HCM phenotype and its arrhythmogenic response to β-ARS through the downregulation of potassium currents.
Collapse
Affiliation(s)
- Ruben Doste
- Department of Computer Science, BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | | | - Alfonso Bueno-Orovio
- Department of Computer Science, BHF Centre of Research Excellence, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
24
|
Li XQ, Lu S, Xia L, Shan XL, Zhao WX, Chen HH, Zhang C, Guo W, Xu M, Lu R, Zhao P. Stachydrine hydrochloride ameliorates cardiac hypertrophy through CaMKII/HDAC4/MEF2C signal pathway. Am J Transl Res 2022; 14:3840-3853. [PMID: 35836883 PMCID: PMC9274579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
Stachydrine hydrochloride (Sta), an activated alkaloid, is isolated from traditional Chinese medicine Yimucao. In previous studies, the cardioprotective effects of Sta were found in our laboratory. However, the underling mechanisms of Sta is not fully elucidated. The aim of this study was to provide a detailed account of the anti-hypertrophic effects of Sta on transcriptional regulation. In vivo, C57BL/6J mice were subjected to transverse aortic constriction (TAC) and were orally treated with Sta. Morphological assessments, echocardiographic parameters, histological analyses and immunofluorescence were used to evaluate cardiac hypertrophy. In vitro, cardiomyocytes were stimulated by phenylephrine (PE), and cell surface and hypertrophy markers were tested by immunofluorescence and real-time polymerase chain reaction (RT-PCR). Moreover, western blotting, RT-PCR and luciferase reporter genes were used to assess the expression of proteins, mRNA and the activity of the CaMKII/HDAC4/MEF2C signal pathway in vivo and in vitro. We found that Sta blocked cardiac hypertrophy induced by pressure overload. We also demonstrated that Sta inhibited nuclear export or promoted nuclear import of HDAC4 through regulation of p-CaMKII, and it further improved the repression of MEF2C. Taken together, our findings demonstrated that Sta ameliorates cardiac hypertrophy through CaMKII/HDAC4/MEF2C signal pathway.
Collapse
Affiliation(s)
- Xue-Qin Li
- School of Basic Medical Science, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Shuang Lu
- School of Basic Medical Science, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Lei Xia
- Yueyang Hospital of Integrative Chinese and Western Medicine, Shanghai University of Traditional Chinese MedicineChina
| | - Xiao-Li Shan
- Public Laboratory Platform, School of Basic Medical Science, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Wen-Xia Zhao
- School of Basic Medical Science, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Hui-Hua Chen
- School of Basic Medical Science, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Chen Zhang
- Department of Pathology, Shanghai University of Traditional Chinese MedicineChina
| | - Wei Guo
- Department of Pathology, Shanghai University of Traditional Chinese MedicineChina
| | - Ming Xu
- Department of Physiology, Shanghai University of Traditional Chinese MedicineChina
| | - Rong Lu
- School of Basic Medical Science, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Pei Zhao
- Public Laboratory Platform, School of Basic Medical Science, Shanghai University of Traditional Chinese MedicineShanghai, China
| |
Collapse
|
25
|
Abstract
Hypertrophic cardiomyopathy (HCM), the most common inherited heart disease, is still orphan of a specific drug treatment. The erroneous consideration of HCM as a rare disease has hampered the design and conduct of large, randomized trials in the last 50 years, and most of the indications in the current guidelines are derived from small non-randomized studies, case series, or simply from the consensus of experts. Guideline-directed therapy of HCM includes non-selective drugs such as disopyramide, non-dihydropyridine calcium channel blockers, or β-adrenergic receptor blockers, mainly used in patients with symptomatic obstruction of the outflow tract. Following promising preclinical studies, several drugs acting on potential HCM-specific targets were tested in patients. Despite the huge efforts, none of these studies was able to change clinical practice for HCM patients, because tested drugs were proven to be scarcely effective or hardly tolerated in patients. However, novel compounds have been developed in recent years specifically for HCM, addressing myocardial hypercontractility and altered energetics in a direct manner, through allosteric inhibition of myosin. In this paper, we will critically review the use of different classes of drugs in HCM patients, starting from "old" established agents up to novel selective drugs that have been recently trialed in patients.
Collapse
|
26
|
Pioner JM, Vitale G, Gentile F, Scellini B, Piroddi N, Cerbai E, Olivotto I, Tardiff J, Coppini R, Tesi C, Poggesi C, Ferrantini C. Genotype-Driven Pathogenesis of Atrial Fibrillation in Hypertrophic Cardiomyopathy: The Case of Different TNNT2 Mutations. Front Physiol 2022; 13:864547. [PMID: 35514357 PMCID: PMC9062294 DOI: 10.3389/fphys.2022.864547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/28/2022] [Indexed: 11/25/2022] Open
Abstract
Atrial dilation and atrial fibrillation (AF) are common in Hypertrophic CardioMyopathy (HCM) patients and associated with a worsening of prognosis. The pathogenesis of atrial myopathy in HCM remains poorly investigated and no specific association with genotype has been identified. By re-analysis of our cohort of thin-filament HCM patients (Coppini et al. 2014) AF was identified in 10% of patients with sporadic mutations in the cardiac Troponin T gene (TNNT2), while AF occurrence was much higher (25-75%) in patients carrying specific "hot-spot" TNNT2 mutations. To determine the molecular basis of arrhythmia occurrence, two HCM mouse models expressing human TNNT2 variants (a "hot-spot" one, R92Q, and a "sporadic" one, E163R) were selected according to the different pathophysiological pathways previously demonstrated in ventricular tissue. Echocardiography studies showed a significant left atrial dilation in both models, but more pronounced in the R92Q. In E163R atrial trabeculae, in line with what previously observed in ventricular preparations, the energy cost of tension generation was markedly increased. However, no changes of twitch amplitude and kinetics were observed, and there was no atrial arrhythmic propensity. R92Q atrial trabeculae, instead, displayed normal ATP consumption but markedly increased myofilament calcium sensitivity, as previously observed in ventricular preparations. This was associated with reduced inotropic reserve and slower kinetics of twitch contractions and, importantly, with an increased occurrence of spontaneous beats and triggered contractions that represent an intrinsic arrhythmogenic mechanism promoting AF. The association of specific TNNT2 mutations with AF occurrence depends on the mutation-driven pathomechanism (i.e., increased atrial myofilament calcium sensitivity rather than increased myofilament tension cost) and may influence the individual response to treatment.
Collapse
Affiliation(s)
| | - Giulia Vitale
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesca Gentile
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Jil Tardiff
- Department of Medicine and Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
27
|
Occurrence of early afterdepolarization under healthy or hypertrophic cardiomyopathy conditions in the human ventricular endocardial myocyte: In silico study using 109 torsadogenic or non-torsadogenic compounds. Toxicol Appl Pharmacol 2022; 438:115914. [DOI: 10.1016/j.taap.2022.115914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
|
28
|
Ramachandra CJA, Kp MMJ, Chua J, Hernandez-Resendiz S, Liehn EA, Knöll R, Gan LM, Michaëlsson E, Jonsson MKB, Ryden-Markinhuhta K, Bhat RV, Fritsche-Danielson R, Lin YH, Sadayappan S, Tang HC, Wong P, Shim W, Hausenloy DJ. Inhibiting cardiac myeloperoxidase alleviates the relaxation defect in hypertrophic cardiomyocytes. Cardiovasc Res 2022; 118:517-530. [PMID: 33705529 PMCID: PMC8803077 DOI: 10.1093/cvr/cvab077] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/12/2020] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is characterized by cardiomyocyte hypertrophy and disarray, and myocardial stiffness due to interstitial fibrosis, which result in impaired left ventricular filling and diastolic dysfunction. The latter manifests as exercise intolerance, angina, and dyspnoea. There is currently no specific treatment for improving diastolic function in HCM. Here, we investigated whether myeloperoxidase (MPO) is expressed in cardiomyocytes and provides a novel therapeutic target for alleviating diastolic dysfunction in HCM. METHODS AND RESULTS Human cardiomyocytes derived from control-induced pluripotent stem cells (iPSC-CMs) were shown to express MPO, with MPO levels being increased in iPSC-CMs generated from two HCM patients harbouring sarcomeric mutations in the MYBPC3 and MYH7 genes. The presence of cardiomyocyte MPO was associated with higher chlorination and peroxidation activity, increased levels of 3-chlorotyrosine-modified cardiac myosin binding protein-C (MYBPC3), attenuated phosphorylation of MYBPC3 at Ser-282, perturbed calcium signalling, and impaired cardiomyocyte relaxation. Interestingly, treatment with the MPO inhibitor, AZD5904, reduced 3-chlorotyrosine-modified MYBPC3 levels, restored MYBPC3 phosphorylation, and alleviated the calcium signalling and relaxation defects. Finally, we found that MPO protein was expressed in healthy adult murine and human cardiomyocytes, and MPO levels were increased in diseased hearts with left ventricular hypertrophy. CONCLUSION This study demonstrates that MPO inhibition alleviates the relaxation defect in hypertrophic iPSC-CMs through MYBPC3 phosphorylation. These findings highlight cardiomyocyte MPO as a novel therapeutic target for improving myocardial relaxation associated with HCM, a treatment strategy which can be readily investigated in the clinical setting, given that MPO inhibitors are already available for clinical testing.
Collapse
MESH Headings
- Animals
- Cardiac Myosins/genetics
- Cardiac Myosins/metabolism
- Cardiomyopathy, Hypertrophic/drug therapy
- Cardiomyopathy, Hypertrophic/enzymology
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/physiopathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line
- Disease Models, Animal
- Enzyme Inhibitors/pharmacology
- Humans
- Hypertrophy, Left Ventricular/drug therapy
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/physiopathology
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/enzymology
- Induced Pluripotent Stem Cells/pathology
- Male
- Mice, Inbred C57BL
- Mutation, Missense
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Peroxidase/antagonists & inhibitors
- Peroxidase/metabolism
- Phosphorylation
- Reactive Oxygen Species/metabolism
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
- Ventricular Function, Left/drug effects
- Mice
Collapse
Affiliation(s)
- Chrishan J A Ramachandra
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
| | - Myu Mai Ja Kp
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
| | - Jasper Chua
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Faculty of Science, National University of Singapore, Singapore,
Singapore
| | - Sauri Hernandez-Resendiz
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
| | - Elisa A Liehn
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
| | - Ralph Knöll
- Bioscience, Cardiovascular, Renal & Metabolism, BioPharmaceuticals
R&D, AstraZeneca, Gothenburg, Sweden
- Department of Medicine (MedH), Integrated Cardio Metabolic Centre
(ICMC), Heart and Vascular Theme, Karolinska Institute, Stockholm SE-171 77,
Sweden
| | - Li-Ming Gan
- Early Clinical Development, Research and Early Development Cardiovascular,
Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca,
Gothenburg, Sweden
| | - Erik Michaëlsson
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal
and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg,
Sweden
| | - Malin K B Jonsson
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal
and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg,
Sweden
| | - Katarina Ryden-Markinhuhta
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal
and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg,
Sweden
| | - Ratan V Bhat
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM),
BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Regina Fritsche-Danielson
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM),
BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ying-Hsi Lin
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine,
Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati,
OH, USA
| | - Hak Chiaw Tang
- Department of Cardiology, National Heart Centre Singapore,
Singapore, Singapore
| | - Philip Wong
- Department of Cardiology, National Heart Centre Singapore,
Singapore, Singapore
| | - Winston Shim
- Health and Social Sciences Cluster, Singapore Institute of
Technology, Singapore, Singapore
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre
Singapore, Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of
Singapore Medical School, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of
Singapore, Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London,
London, UK
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia
University, Taichung, Taiwan
| |
Collapse
|
29
|
Moore J, Emili A. Mass-Spectrometry-Based Functional Proteomic and Phosphoproteomic Technologies and Their Application for Analyzing Ex Vivo and In Vitro Models of Hypertrophic Cardiomyopathy. Int J Mol Sci 2021; 22:13644. [PMID: 34948439 PMCID: PMC8709159 DOI: 10.3390/ijms222413644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an autosomal dominant disease thought to be principally caused by mutations in sarcomeric proteins. Despite extensive genetic analysis, there are no comprehensive molecular frameworks for how single mutations in contractile proteins result in the diverse assortment of cellular, phenotypic, and pathobiological cascades seen in HCM. Molecular profiling and system biology approaches are powerful tools for elucidating, quantifying, and interpreting dynamic signaling pathways and differential macromolecule expression profiles for a wide range of sample types, including cardiomyopathy. Cutting-edge approaches combine high-performance analytical instrumentation (e.g., mass spectrometry) with computational methods (e.g., bioinformatics) to study the comparative activity of biochemical pathways based on relative abundances of functionally linked proteins of interest. Cardiac research is poised to benefit enormously from the application of this toolkit to cardiac tissue models, which recapitulate key aspects of pathogenesis. In this review, we evaluate state-of-the-art mass-spectrometry-based proteomic and phosphoproteomic technologies and their application to in vitro and ex vivo models of HCM for global mapping of macromolecular alterations driving disease progression, emphasizing their potential for defining the components of basic biological systems, the fundamental mechanistic basis of HCM pathogenesis, and treating the ensuing varied clinical outcomes seen among affected patient cohorts.
Collapse
Affiliation(s)
- Jarrod Moore
- Center for Network Systems Biology, Boston University School of Medicine, Boston, MA 02118, USA;
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
- MD-PhD Program, Boston University School of Medicine, Boston, MA 02118, USA
| | - Andrew Emili
- Center for Network Systems Biology, Boston University School of Medicine, Boston, MA 02118, USA;
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
30
|
Korotkov SM, Sobol KV, Shemarova IV, Nesterov VP. Effect of Sodium Ions on Calcium-Loaded Rat Heart Mitochondria and Frog Myocardium. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021060041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Giardini F, Lazzeri E, Vitale G, Ferrantini C, Costantini I, Pavone FS, Poggesi C, Bocchi L, Sacconi L. Quantification of Myocyte Disarray in Human Cardiac Tissue. Front Physiol 2021; 12:750364. [PMID: 34867455 PMCID: PMC8635020 DOI: 10.3389/fphys.2021.750364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/24/2021] [Indexed: 11/13/2022] Open
Abstract
Proper three-dimensional (3D)-cardiomyocyte orientation is important for an effective tension production in cardiac muscle. Cardiac diseases can cause severe remodeling processes in the heart, such as cellular misalignment, that can affect both the electrical and mechanical functions of the organ. To date, a proven methodology to map and quantify myocytes disarray in massive samples is missing. In this study, we present an experimental pipeline to reconstruct and analyze the 3D cardiomyocyte architecture in massive samples. We employed tissue clearing, staining, and advanced microscopy techniques to detect sarcomeres in relatively large human myocardial strips with micrometric resolution. Z-bands periodicity was exploited in a frequency analysis approach to extract the 3D myofilament orientation, providing an orientation map used to characterize the tissue organization at different spatial scales. As a proof-of-principle, we applied the proposed method to healthy and pathologically remodeled human cardiac tissue strips. Preliminary results suggest the reliability of the method: strips from a healthy donor are characterized by a well-organized tissue, where the local disarray is log-normally distributed and slightly depends on the spatial scale of analysis; on the contrary, pathological strips show pronounced tissue disorganization, characterized by local disarray significantly dependent on the spatial scale of analysis. A virtual sample generator is developed to link this multi-scale disarray analysis with the underlying cellular architecture. This approach allowed us to quantitatively assess tissue organization in terms of 3D myocyte angular dispersion and may pave the way for developing novel predictive models based on structural data at cellular resolution.
Collapse
Affiliation(s)
- Francesco Giardini
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Erica Lazzeri
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Giulia Vitale
- Division of Physiology, Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,Division of Physiology, Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Irene Costantini
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, University of Florence, Florence, Italy.,Department of Biology, University of Florence, Florence, Italy
| | - Francesco S Pavone
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, University of Florence, Florence, Italy.,Department of Physics, University of Florence, Florence, Italy
| | - Corrado Poggesi
- Division of Physiology, Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Leonardo Bocchi
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,Department of Information Engineering, University of Florence, Florence, Italy
| | - Leonardo Sacconi
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, University of Florence, Florence, Italy.,Faculty of Medicine, Institute for Experimental Cardiovascular Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
32
|
Russo D, Sclafani M, Tini G, Musumeci MB, Arcari L, Limite LR, Francia P, Autore C. Prognostic implications of different clinical profiles in hypertrophic cardiomyopathy. Minerva Cardiol Angiol 2021; 70:189-206. [PMID: 34713676 DOI: 10.23736/s2724-5683.21.05752-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a myocardial genetic disease relatively common in the general population with heterogenous clinical presentation, natural history and prognosis. About 60% of HCM patients have a stable clinical course, while others may experience a variety of HCMrelated complications which follows relatively independent pathways, and that can be distinguished in different subgroups. These subgroups are represented by patients with left ventricular outflow tract obstruction; patients with end-stage disease and reduced or preserved systolic function; patients with apical hypertrophy; patients with apical aneurysm; patients with atrial fibrillation, patients at high risk of sudden death and patients with pre-clinical HCM. The purpose of this review is to describe each of these clinical profiles with its prognostic implications.
Collapse
Affiliation(s)
- Domitilla Russo
- Cardiology, Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Matteo Sclafani
- Cardiology, Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Giacomo Tini
- Cardiology, Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Maria B Musumeci
- Cardiology, Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Luca Arcari
- Cardiology Unit, Madre Giuseppina Vannini Hospital, Rome, Italy
| | - Luca R Limite
- Arrhythmia Unit and Electrophysiology Laboratories, Department of Cardiology and Cardiothoracic Surgery, San Raffaele Hospital, Milan, Italy
| | - Pietro Francia
- Cardiology, Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Camillo Autore
- Cardiology, Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy -
| |
Collapse
|
33
|
Ion Channel Impairment and Myofilament Ca 2+ Sensitization: Two Parallel Mechanisms Underlying Arrhythmogenesis in Hypertrophic Cardiomyopathy. Cells 2021; 10:cells10102789. [PMID: 34685769 PMCID: PMC8534456 DOI: 10.3390/cells10102789] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
Life-threatening ventricular arrhythmias are the main clinical burden in patients with hypertrophic cardiomyopathy (HCM), and frequently occur in young patients with mild structural disease. While massive hypertrophy, fibrosis and microvascular ischemia are the main mechanisms underlying sustained reentry-based ventricular arrhythmias in advanced HCM, cardiomyocyte-based functional arrhythmogenic mechanisms are likely prevalent at earlier stages of the disease. In this review, we will describe studies conducted in human surgical samples from HCM patients, transgenic animal models and human cultured cell lines derived from induced pluripotent stem cells. Current pieces of evidence concur to attribute the increased risk of ventricular arrhythmias in early HCM to different cellular mechanisms. The increase of late sodium current and L-type calcium current is an early observation in HCM, which follows post-translation channel modifications and increases the occurrence of early and delayed afterdepolarizations. Increased myofilament Ca2+ sensitivity, commonly observed in HCM, may promote afterdepolarizations and reentry arrhythmias with direct mechanisms. Decrease of K+-currents due to transcriptional regulation occurs in the advanced disease and contributes to reducing the repolarization-reserve and increasing the early afterdepolarizations (EADs). The presented evidence supports the idea that patients with early-stage HCM should be considered and managed as subjects with an acquired channelopathy rather than with a structural cardiac disease.
Collapse
|
34
|
Guo J, Jiang H, Oguntuyo K, Rios B, Boodram Z, Huebsch N. Interplay of Genotype and Substrate Stiffness in Driving the Hypertrophic Cardiomyopathy Phenotype in iPSC-Micro-Heart Muscle Arrays. Cell Mol Bioeng 2021; 14:409-425. [PMID: 34777601 PMCID: PMC8548480 DOI: 10.1007/s12195-021-00684-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/04/2021] [Indexed: 10/21/2022] Open
Abstract
INTRODUCTION In clinical and animal studies, Hypertrophic Cardiomyopathy (HCM) shares many similarities with non-inherited cardiac hypertrophy induced by pressure overload (hypertension). This suggests a potential role for mechanical stress in priming tissues with mutation-induced changes in the sarcomere to develop phenotypes associated with HCM, including hypercontractility and aberrant calcium handling. Here, we tested the hypothesis that heterozygous loss of function of Myosin Binding Protein C (MYBCP3 +/- , mutations in which account for almost 50% of inherited HCM) combines with environmental stiffness to drive HCM phenotypes. METHODS We differentiated isogenic control (WTC) and MYBPC3 +/- iPSC into cardiomyocytes using small molecule manipulation of Wnt signaling, and then purified them using lactate media. The purified cardiomyocytes were seeded into "dog bone" shaped stencil molds to form micro-heart muscle arrays (μHM). To mimic changes in myocardial stiffness stemming from pressure overload, we varied the rigidity of the substrates μHM contract against. Stiffness levels ranged from those corresponding to fetal (5 kPa), healthy (15 kPa), pre-fibrotic (30 kPa) to fibrotic (65 kPa) myocardium. Substrates were embedded with a thin layer of fluorescent beads to track contractile force, and parent iPSC were engineered to express the genetic calcium indicator, GCaMP6f. High speed video microscopy and image analysis were used to quantify calcium handling and contractility of μHM. RESULTS Substrate rigidity triggered physiological adaptation for both genotypes. However, MYBPC3 +/- μHM showed a lower tolerance to substrate stiffness with the peak traction on 15 kPa, while WTC μHM had peak traction on 30 kPa. MYBPC3 +/- μHM exhibited hypercontractility, which was exaggerated by substrate rigidity. MYBPC3 +/- μHM hypercontractility was associated with longer rise times for calcium uptake and force development, along with higher overall Ca2+ intake. CONCLUSION We found MYBPC3 +/- mutations cause iPSC-μHM to exhibit hypercontractility, and also a lower tolerance for mechanical stiffness. Understanding how genetics work in combination with mechanical stiffness to trigger and/or exacerbate pathophysiology may lead to more effective therapies for HCM. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at (10.1007/s12195-021-00684-x).
Collapse
Affiliation(s)
- Jingxuan Guo
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, USA
| | - Huanzhu Jiang
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Kasoorelope Oguntuyo
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Brandon Rios
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Zoë Boodram
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
- NSF Science and Technology Center for Engineering Mechanobiology, McKelvey School of Engineering, Saint Louis, USA
- Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, Saint Louis, USA
| |
Collapse
|
35
|
Yigit G, Wollnik B. Cellular models and therapeutic perspectives in hypertrophic cardiomyopathy. MED GENET-BERLIN 2021; 33:235-243. [PMID: 38835701 PMCID: PMC11006313 DOI: 10.1515/medgen-2021-2094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/28/2021] [Indexed: 06/06/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is a clinically heterogeneous cardiac disease that is mainly characterized by left ventricular hypertrophy in the absence of any additional cardiac or systemic disease. HCM is genetically heterogeneous, inherited mainly in an autosomal dominant pattern, and so far pathogenic variants have been identified in more than 20 genes, mostly encoding proteins of the cardiac sarcomere. Based on its variable penetrance and expressivity, pathogenicity of newly identified variants often remains unsolved, underlining the importance of cellular and tissue-based models that help to uncover causative genetic alterations and, additionally, provide appropriate systems for the analysis of disease hallmarks as well as for the design and application of new therapeutic strategies like drug screenings and genome/base editing approaches. Here, we review the current state of cellular and tissue-engineered models and provide future perspectives for personalized therapeutic strategies of HCM.
Collapse
Affiliation(s)
- Gökhan Yigit
- Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
36
|
Coppini R, Santini L, Olivotto I, Ackerman MJ, Cerbai E. Abnormalities in sodium current and calcium homoeostasis as drivers of arrhythmogenesis in hypertrophic cardiomyopathy. Cardiovasc Res 2021; 116:1585-1599. [PMID: 32365196 DOI: 10.1093/cvr/cvaa124] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/06/2020] [Accepted: 04/24/2020] [Indexed: 12/28/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common inherited monogenic disease with a prevalence of 1/500 in the general population, representing an important cause of arrhythmic sudden cardiac death (SCD), heart failure, and atrial fibrillation in the young. HCM is a global condition, diagnosed in >50 countries and in all continents. HCM affects people of both sexes and various ethnic and racial origins, with similar clinical course and phenotypic expression. The most unpredictable and devastating consequence of HCM is represented by arrhythmic SCD, most commonly caused by sustained ventricular tachycardia or ventricular fibrillation. Indeed, HCM represents one of the main causes of arrhythmic SCD in the young, with a marked preference for children and adults <30 years. SCD is most prevalent in patients with paediatric onset of HCM but may occur at any age. However, risk is substantially lower after 60 years, suggesting that the potential for ventricular tachyarrhythmias is mitigated by ageing. SCD had been linked originally to sports and vigorous activity in HCM patients. However, it is increasingly clear that the majority of events occurs at rest or during routine daily occupations, suggesting that triggers are far from consistent. In general, the pathophysiology of SCD in HCM remains unresolved. While the pathologic and physiologic substrates abound and have been described in detail, specific factors precipitating ventricular tachyarrhythmias are still unknown. SCD is a rare phenomenon in HCM cohorts (<1%/year) and attempts to identify patients at risk, while generating clinically useful algorithms for primary prevention, remain very inaccurate on an individual basis. One of the reasons for our limited understanding of these phenomena is that limited translational research exists in the field, while most efforts have focused on clinical markers of risk derived from pathology, instrumental patient evaluation, and imaging. Specifically, few studies conducted in animal models and human samples have focused on targeting the cellular mechanisms of arrhythmogenesis in HCM, despite potential implications for therapeutic innovation and SCD prevention. These studies found that altered intracellular Ca2+ homoeostasis and increased late Na+ current, leading to an increased likelihood of early and delayed after-depolarizations, contribute to generate arrhythmic events in diseased cardiomyocytes. As an array of novel experimental opportunities have emerged to investigate these mechanisms, including novel 'disease-in-the-dish' cellular models with patient-specific induced pluripotent stem cell-derived cardiomyocytes, important gaps in knowledge remain. Accordingly, the aim of the present review is to provide a contemporary reappraisal of the cellular basis of SCD-predisposing arrhythmias in patients with HCM and discuss the implications for risk stratification and management.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department of Neurosciences, Psychiatry, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Lorenzo Santini
- Department of Neurosciences, Psychiatry, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Iacopo Olivotto
- Department of Clinical and Experimental Medicine, University of Florence, Largo Brambilla, 3 - 50134 Florence, Italy.,Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Michael J Ackerman
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, USA.,Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, USA.,Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, USA
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychiatry, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy.,Laboratory of Non-Linear Spectroscopy (LENS), Via Nello Carrara 1, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
37
|
Marian AJ, Asatryan B, Wehrens XHT. Genetic basis and molecular biology of cardiac arrhythmias in cardiomyopathies. Cardiovasc Res 2021; 116:1600-1619. [PMID: 32348453 DOI: 10.1093/cvr/cvaa116] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/09/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias are common, often the first, and sometimes the life-threatening manifestations of hereditary cardiomyopathies. Pathogenic variants in several genes known to cause hereditary cardiac arrhythmias have also been identified in the sporadic cases and small families with cardiomyopathies. These findings suggest a shared genetic aetiology of a subset of hereditary cardiomyopathies and cardiac arrhythmias. The concept of a shared genetic aetiology is in accord with the complex and exquisite interplays that exist between the ion currents and cardiac mechanical function. However, neither the causal role of cardiac arrhythmias genes in cardiomyopathies is well established nor the causal role of cardiomyopathy genes in arrhythmias. On the contrary, secondary changes in ion currents, such as post-translational modifications, are common and contributors to the pathogenesis of arrhythmias in cardiomyopathies through altering biophysical and functional properties of the ion channels. Moreover, structural changes, such as cardiac hypertrophy, dilatation, and fibrosis provide a pro-arrhythmic substrate in hereditary cardiomyopathies. Genetic basis and molecular biology of cardiac arrhythmias in hereditary cardiomyopathies are discussed.
Collapse
Affiliation(s)
- Ali J Marian
- Department of Medicine, Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xander H T Wehrens
- Department of Biophysics and Molecular Physiology, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
38
|
Chou C, Chin MT. Pathogenic Mechanisms of Hypertrophic Cardiomyopathy beyond Sarcomere Dysfunction. Int J Mol Sci 2021; 22:ijms22168933. [PMID: 34445638 PMCID: PMC8396307 DOI: 10.3390/ijms22168933] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 01/23/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disorder, affecting 1 in 500 people in the general population. Although characterized by asymmetric left ventricular hypertrophy, cardiomyocyte disarray, and cardiac fibrosis, HCM is in fact a highly complex disease with heterogenous clinical presentation, onset, and complications. While HCM is generally accepted as a disease of the sarcomere, variable penetrance in families with identical genetic mutations challenges the monogenic origin of HCM and instead implies a multifactorial cause. Furthermore, large-scale genome sequencing studies revealed that many genes previously reported as causative of HCM in fact have little or no evidence of disease association. These findings thus call for a re-evaluation of the sarcomere-centered view of HCM pathogenesis. Here, we summarize our current understanding of sarcomere-independent mechanisms of cardiomyocyte hypertrophy, highlight the role of extracellular signals in cardiac fibrosis, and propose an alternative but integrated model of HCM pathogenesis.
Collapse
Affiliation(s)
- Chun Chou
- Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Michael T. Chin
- Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA;
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
- Correspondence: ; Tel.: +1-617-636-8776
| |
Collapse
|
39
|
Glavaški M, Velicki L. Shared Molecular Mechanisms of Hypertrophic Cardiomyopathy and Its Clinical Presentations: Automated Molecular Mechanisms Extraction Approach. Life (Basel) 2021; 11:life11080785. [PMID: 34440529 PMCID: PMC8398249 DOI: 10.3390/life11080785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 12/30/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disease with a prevalence of 1 in 500 people and varying clinical presentations. Although there is much research on HCM, underlying molecular mechanisms are poorly understood, and research on the molecular mechanisms of its specific clinical presentations is scarce. Our aim was to explore the molecular mechanisms shared by HCM and its clinical presentations through the automated extraction of molecular mechanisms. Molecular mechanisms were congregated by a query of the INDRA database, which aggregates knowledge from pathway databases and combines it with molecular mechanisms extracted from abstracts and open-access full articles by multiple machine-reading systems. The molecular mechanisms were extracted from 230,072 articles on HCM and 19 HCM clinical presentations, and their intersections were found. Shared molecular mechanisms of HCM and its clinical presentations were represented as networks; the most important elements in the intersections’ networks were found, centrality scores for each element of each network calculated, networks with reduced level of noise generated, and cooperatively working elements detected in each intersection network. The identified shared molecular mechanisms represent possible mechanisms underlying different HCM clinical presentations. Applied methodology produced results consistent with the information in the scientific literature.
Collapse
Affiliation(s)
- Mila Glavaški
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Correspondence: or
| | - Lazar Velicki
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Institute of Cardiovascular Diseases Vojvodina, Put Doktora Goldmana 4, 21204 Sremska Kamenica, Serbia
| |
Collapse
|
40
|
Vakrou S, Liu Y, Zhu L, Greenland GV, Simsek B, Hebl VB, Guan Y, Woldemichael K, Talbot CC, Aon MA, Fukunaga R, Abraham MR. Differences in molecular phenotype in mouse and human hypertrophic cardiomyopathy. Sci Rep 2021; 11:13163. [PMID: 34162896 PMCID: PMC8222321 DOI: 10.1038/s41598-021-89451-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/23/2021] [Indexed: 11/09/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by phenotypic heterogeneity. We investigated the molecular basis of the cardiac phenotype in two mouse models at established disease stage (mouse-HCM), and human myectomy tissue (human-HCM). We analyzed the transcriptome in 2 mouse models with non-obstructive HCM (R403Q-MyHC, R92W-TnT)/littermate-control hearts at 24 weeks of age, and in myectomy tissue of patients with obstructive HCM/control hearts (GSE36961, GSE36946). Additionally, we examined myocyte redox, cardiac mitochondrial DNA copy number (mtDNA-CN), mt-respiration, mt-ROS generation/scavenging and mt-Ca2+ handling in mice. We identified distinct allele-specific gene expression in mouse-HCM, and marked differences between mouse-HCM and human-HCM. Only two genes (CASQ1, GPT1) were similarly dysregulated in both mutant mice and human-HCM. No signaling pathway or transcription factor was predicted to be similarly dysregulated (by Ingenuity Pathway Analysis) in both mutant mice and human-HCM. Losartan was a predicted therapy only in TnT-mutant mice. KEGG pathway analysis revealed enrichment for several metabolic pathways, but only pyruvate metabolism was enriched in both mutant mice and human-HCM. Both mutant mouse myocytes demonstrated evidence of an oxidized redox environment. Mitochondrial complex I RCR was lower in both mutant mice compared to controls. MyHC-mutant mice had similar mtDNA-CN and mt-Ca2+ handling, but TnT-mutant mice exhibited lower mtDNA-CN and impaired mt-Ca2+ handling, compared to littermate-controls. Molecular profiling reveals differences in gene expression, transcriptional regulation, intracellular signaling and mt-number/function in 2 mouse models at established disease stage. Further studies are needed to confirm differences in gene expression between mouse and human-HCM, and to examine whether cardiac phenotype, genotype and/or species differences underlie the divergence in molecular profiles.
Collapse
Affiliation(s)
- Styliani Vakrou
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Yamin Liu
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Li Zhu
- Department of Biological Chemistry, Johns Hopkins School of Medicine, 725 N. Wolfe St, 521A Physiology, Baltimore, MD, 21205, USA
| | - Gabriela V Greenland
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Bahadir Simsek
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Virginia B Hebl
- Intermountain Medical Center, Intermountain Heart Institute, Murray, UT, USA
| | - Yufan Guan
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kirubel Woldemichael
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Conover C Talbot
- Johns Hopkins School of Medicine, Institute for Basic Biomedical Sciences, Baltimore, MD, USA
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Ryuya Fukunaga
- Department of Biological Chemistry, Johns Hopkins School of Medicine, 725 N. Wolfe St, 521A Physiology, Baltimore, MD, 21205, USA.
| | - M Roselle Abraham
- Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Division of Cardiology, Hypertrophic Cardiomyopathy Center of Excellence, University of California San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
41
|
Zampieri M, Berteotti M, Ferrantini C, Tassetti L, Gabriele M, Tomberli B, Castelli G, Cappelli F, Stefàno P, Marchionni N, Coppini R, Olivotto I. Pathophysiology and Treatment of Hypertrophic Cardiomyopathy: New Perspectives. Curr Heart Fail Rep 2021; 18:169-179. [PMID: 34148184 DOI: 10.1007/s11897-021-00523-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW We provide a state of the art of therapeutic options in hypertrophic cardiomyopathy (HCM), focusing on recent advances in our understanding of the pathophysiology of sarcomeric disease. RECENT FINDINGS A wealth of novel information regarding the molecular mechanisms associated with the clinical phenotype and natural history of HCM have been developed over the last two decades. Such advances have only recently led to a number of controlled randomized studies, often limited in size and fortune. Recently, however, the allosteric inhibitors of cardiac myosin adenosine triphosphatase, countering the main pathophysiological abnormality associated with HCM-causing mutations, i.e. hypercontractility, have opened new management perspectives. Mavacamten is the first drug specifically developed for HCM used in a successful phase 3 trial, with the promise to reach symptomatic obstructive patients in the near future. In addition, the fine characterization of cardiomyocyte electrophysiological remodelling has recently highlighted relevant therapeutic targets. Current therapies for HCM focus on late disease manifestations without addressing the intrinsic pathological mechanisms. However, novel evidence-based approaches have opened the way for agents targeting HCM molecular substrates. The impact of these targeted interventions will hopefully alter the natural history of the disease in the near future.
Collapse
Affiliation(s)
- Mattia Zampieri
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy.
| | - Martina Berteotti
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luigi Tassetti
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Martina Gabriele
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Benedetta Tomberli
- Division of Interventional Structural Cardiology, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | - Gabriele Castelli
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Francesco Cappelli
- Division of Interventional Structural Cardiology, Cardiothoracovascular Department, Careggi University Hospital, Florence, Italy
| | - Pierluigi Stefàno
- Division of Cardiac Surgery, Careggi University Hospital, Florence, Italy
| | - Niccolò Marchionni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Division of General Cardiology, Careggi University Hospital, Florence, Italy
| | | | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
42
|
Ma K, Ma G, Guo Z, Liu G, Liang W. Regulatory mechanism of calcium/calmodulin-dependent protein kinase II in the occurrence and development of ventricular arrhythmia (Review). Exp Ther Med 2021; 21:656. [PMID: 33968186 PMCID: PMC8097202 DOI: 10.3892/etm.2021.10088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/05/2021] [Indexed: 11/24/2022] Open
Abstract
Ventricular arrhythmia (VA) is a highly fatal arrhythmia that involves multiple ion channels. Of all sudden cardiac death events, ~85% result from VAs, including ventricular tachycardia and ventricular fibrillation. Calcium/calmodulin-dependent pro-tein kinase II (CaMKII) is an important ion channel regulator that participates in the excitation-contraction coupling of the heart, and as such is important for regulating its electrophysiological function. CaMKII can be activated in a Ca2+/calmodulin (CaM)-dependent or Ca2+/CaM-independent manner, serving a key role in the occurrence and development of VA. The present review aimed to determine whether activated CaMKII induces early afterdepolarizations and delayed afterdepolarizations that result in VA by regulating sodium, potassium and calcium ions. Assessing VA mechanisms based on the CaMKII pathway is of great significance to the clinical treatment of VA and the de-velopment of effective drugs for use in clinical practice.
Collapse
Affiliation(s)
- Kexin Ma
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Guoping Ma
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zijing Guo
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Gang Liu
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Wenjie Liang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
43
|
Mechanisms underlying pathological Ca 2+ handling in diseases of the heart. Pflugers Arch 2021; 473:331-347. [PMID: 33399957 PMCID: PMC10070045 DOI: 10.1007/s00424-020-02504-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/01/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
Cardiomyocyte contraction relies on precisely regulated intracellular Ca2+ signaling through various Ca2+ channels and transporters. In this article, we will review the physiological regulation of Ca2+ handling and its role in maintaining normal cardiac rhythm and contractility. We discuss how inherited variants or acquired defects in Ca2+ channel subunits contribute to the development or progression of diseases of the heart. Moreover, we highlight recent insights into the role of protein phosphatase subunits and striated muscle preferentially expressed protein kinase (SPEG) in atrial fibrillation, heart failure, and cardiomyopathies. Finally, this review summarizes current drug therapies and new advances in genome editing as therapeutic strategies for the cardiac diseases caused by aberrant intracellular Ca2+ signaling.
Collapse
|
44
|
Jiang SJ, Wang W. Research progress on the role of CaMKII in heart disease. Am J Transl Res 2020; 12:7625-7639. [PMID: 33437349 PMCID: PMC7791482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/21/2020] [Indexed: 06/12/2023]
Abstract
In the heart, Ca2+ participates in electrical activity and myocardial contraction, which is closely related to the generation of action potential and excitation contraction coupling (ECC) and plays an important role in various signal cascades and regulates different physiological processes. In the Ca2+ related physiological activities, CaMKII is a key downstream regulator, involving autophosphorylation and post-translational modification, and plays an important role in the excitation contraction coupling and relaxation events of cardiomyocytes. This paper reviews the relationship between CaMKII and various substances in the pathological process of myocardial apoptosis and necrosis, myocardial hypertrophy and arrhythmia, and what roles it plays in the development of disease in complex networks. This paper also introduces the drugs targeting at CaMKII to treat heart disease.
Collapse
Affiliation(s)
- Shi-Jun Jiang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Wei Wang
- Department of Cardiology, Affiliated Taihe Hospital of Hubei University of MedicineShiyan 442000, Hubei, China
| |
Collapse
|
45
|
T-tubule remodeling in human hypertrophic cardiomyopathy. J Muscle Res Cell Motil 2020; 42:305-322. [PMID: 33222034 PMCID: PMC8332592 DOI: 10.1007/s10974-020-09591-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/22/2020] [Indexed: 11/17/2022]
Abstract
The highly organized transverse T-tubule membrane system represents the ultrastructural substrate for excitation–contraction coupling in ventricular myocytes. While the architecture and function of T-tubules have been well described in animal models, there is limited morpho-functional data on T-tubules in human myocardium. Hypertrophic cardiomyopathy (HCM) is a primary disease of the heart muscle, characterized by different clinical presentations at the various stages of its progression. Most HCM patients, indeed, show a compensated hypertrophic disease (“non-failing hypertrophic phase”), with preserved left ventricular function, and only a small subset of individuals evolves into heart failure (“end stage HCM”). In terms of T-tubule remodeling, the “end-stage” disease does not differ from other forms of heart failure. In this review we aim to recapitulate the main structural features of T-tubules during the “non-failing hypertrophic stage” of human HCM by revisiting data obtained from human myectomy samples. Moreover, by comparing pathological changes observed in myectomy samples with those introduced by acute (experimentally induced) detubulation, we discuss the role of T-tubular disruption as a part of the complex excitation–contraction coupling remodeling process that occurs during disease progression. Lastly, we highlight how T-tubule morpho-functional changes may be related to patient genotype and we discuss the possibility of a primitive remodeling of the T-tubule system in rare HCM forms associated with genes coding for proteins implicated in T-tubule structural integrity, formation and maintenance.
Collapse
|
46
|
Spath NB, Singh T, Papanastasiou G, Kershaw L, Baker AH, Janiczek RL, Gulsin GS, Dweck MR, McCann G, Newby DE, Semple SI. Manganese-enhanced magnetic resonance imaging in dilated cardiomyopathy and hypertrophic cardiomyopathy. Eur Heart J Cardiovasc Imaging 2020:jeaa273. [PMID: 33200175 DOI: 10.1093/ehjci/jeaa273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/17/2020] [Indexed: 01/07/2023] Open
Abstract
AIMS The aim of this study is to quantify altered myocardial calcium handling in non-ischaemic cardiomyopathy using magnetic resonance imaging. METHODS AND RESULTS Patients with dilated cardiomyopathy (n = 10) or hypertrophic cardiomyopathy (n = 17) underwent both gadolinium and manganese contrast-enhanced magnetic resonance imaging and were compared with healthy volunteers (n = 20). Differential manganese uptake (Ki) was assessed using a two-compartment Patlak model. Compared with healthy volunteers, reduction in T1 with manganese-enhanced magnetic resonance imaging was lower in patients with dilated cardiomyopathy [mean reduction 257 ± 45 (21%) vs. 288 ± 34 (26%) ms, P < 0.001], with higher T1 at 40 min (948 ± 57 vs. 834 ± 28 ms, P < 0.0001). In patients with hypertrophic cardiomyopathy, reductions in T1 were less than healthy volunteers [mean reduction 251 ± 86 (18%) and 277 ± 34 (23%) vs. 288 ± 34 (26%) ms, with and without fibrosis respectively, P < 0.001]. Myocardial manganese uptake was modelled, rate of uptake was reduced in both dilated and hypertrophic cardiomyopathy in comparison with healthy volunteers (mean Ki 19 ± 4, 19 ± 3, and 23 ± 4 mL/100 g/min, respectively; P = 0.0068). In patients with dilated cardiomyopathy, manganese uptake rate correlated with left ventricular ejection fraction (r2 = 0.61, P = 0.009). Rate of myocardial manganese uptake demonstrated stepwise reductions across healthy myocardium, hypertrophic cardiomyopathy without fibrosis and hypertrophic cardiomyopathy with fibrosis providing absolute discrimination between the healthy myocardium and fibrosed myocardium (mean Ki 23 ± 4, 19 ± 3, and 13 ± 4 mL/100 g/min, respectively; P < 0.0001). CONCLUSION The rate of manganese uptake in both dilated and hypertrophic cardiomyopathy provides a measure of altered myocardial calcium handling. This holds major promise for the detection and monitoring of dysfunctional myocardium, with the potential for early intervention and prognostication.
Collapse
Affiliation(s)
- N B Spath
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4SA, UK
- Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, EH16 4SB, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - T Singh
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4SA, UK
- Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, EH16 4SB, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - G Papanastasiou
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4SA, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - L Kershaw
- Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - A H Baker
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4SA, UK
| | - R L Janiczek
- Department of Clinical Imaging, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - G S Gulsin
- Department of Cardiovascular Sciences, University of Leicester, NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - M R Dweck
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4SA, UK
- Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, EH16 4SB, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - G McCann
- Department of Cardiovascular Sciences, University of Leicester, NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - D E Newby
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4SA, UK
- Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, EH16 4SB, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - S I Semple
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4SA, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
47
|
Varró A, Tomek J, Nagy N, Virág L, Passini E, Rodriguez B, Baczkó I. Cardiac transmembrane ion channels and action potentials: cellular physiology and arrhythmogenic behavior. Physiol Rev 2020; 101:1083-1176. [PMID: 33118864 DOI: 10.1152/physrev.00024.2019] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrhythmias are among the leading causes of mortality. They often arise from alterations in the electrophysiological properties of cardiac cells and their underlying ionic mechanisms. It is therefore critical to further unravel the pathophysiology of the ionic basis of human cardiac electrophysiology in health and disease. In the first part of this review, current knowledge on the differences in ion channel expression and properties of the ionic processes that determine the morphology and properties of cardiac action potentials and calcium dynamics from cardiomyocytes in different regions of the heart are described. Then the cellular mechanisms promoting arrhythmias in congenital or acquired conditions of ion channel function (electrical remodeling) are discussed. The focus is on human-relevant findings obtained with clinical, experimental, and computational studies, given that interspecies differences make the extrapolation from animal experiments to human clinical settings difficult. Deepening the understanding of the diverse pathophysiology of human cellular electrophysiology will help in developing novel and effective antiarrhythmic strategies for specific subpopulations and disease conditions.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
48
|
Santini L, Palandri C, Nediani C, Cerbai E, Coppini R. Modelling genetic diseases for drug development: Hypertrophic cardiomyopathy. Pharmacol Res 2020; 160:105176. [DOI: 10.1016/j.phrs.2020.105176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/16/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
|
49
|
Shoykhet M, Trenz S, Kempf E, Williams T, Gerull B, Schinner C, Yeruva S, Waschke J. Cardiomyocyte adhesion and hyperadhesion differentially require ERK1/2 and plakoglobin. JCI Insight 2020; 5:140066. [PMID: 32841221 PMCID: PMC7526536 DOI: 10.1172/jci.insight.140066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/12/2020] [Indexed: 12/23/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a heart disease often caused by mutations in genes coding for desmosomal proteins, including desmoglein-2 (DSG2), plakoglobin (PG), and desmoplakin (DP). Therapy is based on symptoms and limiting arrhythmia, because the mechanisms by which desmosomal components control cardiomyocyte function are largely unknown. A new paradigm could be to stabilize desmosomal cardiomyocyte adhesion and hyperadhesion, which renders desmosomal adhesion independent from Ca2+. Here, we further characterized the mechanisms behind enhanced cardiomyocyte adhesion and hyperadhesion. Dissociation assays performed in HL-1 cells and murine ventricular cardiac slice cultures allowed us to define a set of signaling pathways regulating cardiomyocyte adhesion under basal and hyperadhesive conditions. Adrenergic signaling, activation of PKC, and inhibition of p38MAPK enhanced cardiomyocyte adhesion, referred to as positive adhesiotropy, and induced hyperadhesion. Activation of ERK1/2 paralleled positive adhesiotropy, whereas adrenergic signaling induced PG phosphorylation at S665 under both basal and hyperadhesive conditions. Adrenergic signaling and p38MAPK inhibition recruited DSG2 to cell junctions. In PG-deficient mice with an AC phenotype, only PKC activation and p38MAPK inhibition enhanced cardiomyocyte adhesion. Our results demonstrate that cardiomyocyte adhesion can be stabilized by different signaling mechanisms, which are in part offset in PG-deficient AC. Desmosome mediated cardiomyocyte adhesion, crucial in the pathology of arrhythmogenic cardiomyopathy, is differentially regulated by multiple signaling mechanisms that depend either on ERK1/2 or plakoglobin.
Collapse
Affiliation(s)
- Maria Shoykhet
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sebastian Trenz
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ellen Kempf
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center and Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Brenda Gerull
- Comprehensive Heart Failure Center and Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Camilla Schinner
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sunil Yeruva
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
50
|
Pioner JM, Fornaro A, Coppini R, Ceschia N, Sacconi L, Donati MA, Favilli S, Poggesi C, Olivotto I, Ferrantini C. Advances in Stem Cell Modeling of Dystrophin-Associated Disease: Implications for the Wider World of Dilated Cardiomyopathy. Front Physiol 2020; 11:368. [PMID: 32477154 PMCID: PMC7235370 DOI: 10.3389/fphys.2020.00368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
Familial dilated cardiomyopathy (DCM) is mostly caused by mutations in genes encoding cytoskeletal and sarcomeric proteins. In the pediatric population, DCM is the predominant type of primitive myocardial disease. A severe form of DCM is associated with mutations in the DMD gene encoding dystrophin, which are the cause of Duchenne Muscular Dystrophy (DMD). DMD-associated cardiomyopathy is still poorly understood and orphan of a specific therapy. In the last 5 years, a rise of interest in disease models using human induced pluripotent stem cells (hiPSCs) has led to more than 50 original studies on DCM models. In this review paper, we provide a comprehensive overview on the advances in DMD cardiomyopathy disease modeling and highlight the most remarkable findings obtained from cardiomyocytes differentiated from hiPSCs of DMD patients. We will also describe how hiPSCs based studies have contributed to the identification of specific myocardial disease mechanisms that may be relevant in the pathogenesis of DCM, representing novel potential therapeutic targets.
Collapse
Affiliation(s)
- Josè Manuel Pioner
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | | | - Raffaele Coppini
- Department of NeuroFarBa, Università degli Studi di Firenze, Florence, Italy
| | - Nicole Ceschia
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Leonardo Sacconi
- LENS, Università degli Studi di Firenze and National Institute of Optics (INO-CNR), Florence, Italy
| | | | - Silvia Favilli
- Pediatric Cardiology, Meyer Children's Hospital, Florence, Italy
| | - Corrado Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Cecilia Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|