1
|
Marimon X, Esquinas F, Ferrer M, Cerrolaza M, Portela A, Benítez R. A Novel non-invasive optical framework for simultaneous analysis of contractility and calcium in single-cell cardiomyocytes. J Mech Behav Biomed Mater 2025; 161:106812. [PMID: 39566161 DOI: 10.1016/j.jmbbm.2024.106812] [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: 08/05/2024] [Revised: 10/13/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
The use of a video method based on the Digital Image Correlation (DIC) algorithm from experimental mechanics to estimate the displacements, strain field, and sarcolemma length in a beating single-cell cardiomyocyte is proposed in this work. The obtained deformation is then correlated with the calcium signal, from calcium imaging where fluorescent dyes sensitive to calcium Ca2+ are used. Our proposed video-based method for simultaneous contraction and intracellular calcium analysis results in a low-cost, non-invasive, and label-free method. This technique has shown great advantages in long-term observations because this type of intervention-free measurement neutralizes the possible alteration in the beating cardiomyocyte introduced by other techniques for measuring cell contractility (e.g., Traction Force Microscopy, Atomic Force Microscopy, Microfabrication or Optical tweezers). Three tests were performed with synthetically augmented data from cardiomyocyte images to validate the robustness of the algorithm. First, a simulated rigid translation of a referenced image is applied, then a rotation, and finally a controlled longitudinal deformation of the referenced image, thus simulating a native realistic deformation. Finally, the proposed framework is evaluated with real experimental data. To validate contraction induced by intracellular calcium concentration, this signal is correlated with a new deformation measure proposed in this article, which is independent of cell orientation in the imaging setup. Finally, based on the displacements obtained by the DIC algorithm, the change in sarcolemma length in a contracting cardiomyocyte is calculated and its temporal correlation with the calcium signal is obtained.
Collapse
Affiliation(s)
- Xavier Marimon
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain; Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
| | - Ferran Esquinas
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miquel Ferrer
- Department of Strength of Materials and Structural Engineering, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miguel Cerrolaza
- School of Engineering, Science and Technology, Valencian International University (VIU), Valencia, Spain
| | - Alejandro Portela
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Raúl Benítez
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain
| |
Collapse
|
2
|
Ahmad F, Qaisar R. Nicotinamide riboside kinase 2: A unique target for skeletal muscle and cardiometabolic diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167487. [PMID: 39216649 DOI: 10.1016/j.bbadis.2024.167487] [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: 06/05/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Myopathy leads to skeletal and cardiac muscle degeneration which is a major cause of physical disability and heart failure. Despite the therapeutic advancement the prevalence of particularly cardiac diseases is rising at an alarming rate and novel therapeutic targets are required. Nicotinamide riboside kinase-2 (NRK-2 or NMRK2) is a muscle-specific β1-integrin binding protein abundantly expressed in the skeletal muscle while only a trace amount is detected in the healthy cardiac muscle. The level in cardiac tissue is profoundly upregulated under pathogenic conditions such as ischemia and hypertension. NRK-2 was initially identified to regulate myoblast differentiation and to enhance the levels of NAD+, an important coenzyme that potentiates cellular energy production and stress resilience. Recent advancement has shown that NRK-2 critically regulates numerous cellular and molecular processes under pathogenic conditions to modulate the disease severity. Therefore, given its restricted expression in the cardiac and skeletal muscle, NRK-2 may serve as a unique therapeutic target. In this review, we provided a comprehensive overview of the diverse roles of NRK-2 played in different cardiac and muscular diseases and discussed the underlying molecular mechanisms in detail. Moreover, this review precisely examined how NRK-2 regulates metabolism in cardiac muscle, and how dysfunctional NRK-2 is associated with energetic deficit and impaired muscle function, manifesting various cardiac and skeletal muscle disease conditions.
Collapse
Affiliation(s)
- Firdos Ahmad
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Rizwan Qaisar
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
3
|
Jurgens SJ, Rämö JT, Kramarenko DR, Wijdeveld LFJM, Haas J, Chaffin MD, Garnier S, Gaziano L, Weng LC, Lipov A, Zheng SL, Henry A, Huffman JE, Challa S, Rühle F, Verdugo CD, Krijger Juárez C, Kany S, van Orsouw CA, Biddinger K, Poel E, Elliott AL, Wang X, Francis C, Ruan R, Koyama S, Beekman L, Zimmerman DS, Deleuze JF, Villard E, Trégouët DA, Isnard R, Boomsma DI, de Geus EJC, Tadros R, Pinto YM, Wilde AAM, Hottenga JJ, Sinisalo J, Niiranen T, Walsh R, Schmidt AF, Choi SH, Chang KM, Tsao PS, Matthews PM, Ware JS, Lumbers RT, van der Crabben S, Laukkanen J, Palotie A, Amin AS, Charron P, Meder B, Ellinor PT, Daly M, Aragam KG, Bezzina CR. Genome-wide association study reveals mechanisms underlying dilated cardiomyopathy and myocardial resilience. Nat Genet 2024; 56:2636-2645. [PMID: 39572784 PMCID: PMC11631763 DOI: 10.1038/s41588-024-01975-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 10/08/2024] [Indexed: 12/06/2024]
Abstract
Dilated cardiomyopathy (DCM) is a heart muscle disease that represents an important cause of morbidity and mortality, yet causal mechanisms remain largely elusive. Here, we perform a large-scale genome-wide association study and multitrait analysis for DCM using 9,365 cases and 946,368 controls. We identify 70 genome-wide significant loci, which show broad replication in independent samples and map to 63 prioritized genes. Tissue, cell type and pathway enrichment analyses highlight the central role of the cardiomyocyte and contractile apparatus in DCM pathogenesis. Polygenic risk scores constructed from our genome-wide association study predict DCM across different ancestry groups, show differing contributions to DCM depending on rare pathogenic variant status and associate with systolic heart failure across various clinical settings. Mendelian randomization analyses reveal actionable potential causes of DCM, including higher bodyweight and higher systolic blood pressure. Our findings provide insights into the genetic architecture and mechanisms underlying DCM and myocardial function more broadly.
Collapse
Affiliation(s)
- Sean J Jurgens
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Joel T Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Daria R Kramarenko
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands
| | - Leonoor F J M Wijdeveld
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Physiology, Amsterdam UMC location, Vrije Universiteit, Amsterdam, the Netherlands
| | - Jan Haas
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University Hospital Heidelberg, Heidelberg, Germany
- Site Heidelberg/Mannheim, DZHK, Heidelberg, Germany
| | - Mark D Chaffin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sophie Garnier
- Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics and Pathophysiology of Cardiovascular Disease, Sorbone Université, INSERM, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Liam Gaziano
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Alex Lipov
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Sean L Zheng
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Albert Henry
- Institute of Cardiovascular Science, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Palo Alto Veterans Institute for Research (PAVIR), Palo Alto Health Care System, Palo Alto, CA, USA
- Harvard Medical School, Boston, MA, USA
| | - Saketh Challa
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Frank Rühle
- Bioinformatics Core Facility, Institute of Molecular Biology gGmbH (IMB), Mainz, Germany
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
| | - Carmen Diaz Verdugo
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christian Krijger Juárez
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiology, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Constance A van Orsouw
- Department of Clinical Genetics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Kiran Biddinger
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edwin Poel
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Amanda L Elliott
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Harvard Medical School, Boston, MA, USA
- Department of Psychiatry and Center for Genomic Medicine, Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Xin Wang
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine Francis
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Richard Ruan
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Satoshi Koyama
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Leander Beekman
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Dominic S Zimmerman
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-François Deleuze
- CEA, Centre National de Recherche en Génomique Humaine, Université Paris-Saclay, Evry, France
- Laboratory of Excellence in Medical Genomics, GENMED, Evry, France
- Fondation Jean Dausset, Centre d'Etude du Polymorphisme Humain, Paris, France
| | - Eric Villard
- Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics and Pathophysiology of Cardiovascular Disease, Sorbone Université, INSERM, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - David-Alexandre Trégouët
- Laboratory of Excellence in Medical Genomics, GENMED, Evry, France
- Bordeaux Population Health Research Center, UMR 1219, University of Bordeaux, INSERM, Bordeaux, France
| | - Richard Isnard
- Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics and Pathophysiology of Cardiovascular Disease, Sorbone Université, INSERM, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
- APHP, Cardiology and Genetics Departments, Pitié-Salpêtrière Hospital, Paris, France
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC location, Vrije Universiteit, Amsterdam, the Netherlands
| | - Rafik Tadros
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Arthur A M Wilde
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- The Netherlands Twin Register, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Juha Sinisalo
- Department of Cardiology, Helsinki University Hospital, Helsinki, Finland
- Heart and Lung Center, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Helsinki, Finland
- Division of Medicine, Turku University Hospital, Helsinki, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Roddy Walsh
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Amand F Schmidt
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- University College London British Heart Foundation Research Accelerator, London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Philip S Tsao
- Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul M Matthews
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - R Thomas Lumbers
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research, University College London Hospitals Biomedical Research Centre, University College London, London, UK
| | - Saskia van der Crabben
- Department of Clinical Genetics, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Jari Laukkanen
- Department of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Central Finland Biobank, Central Finland Health Care District, Jyväskylä, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Ahmad S Amin
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands
| | - Philippe Charron
- Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics and Pathophysiology of Cardiovascular Disease, Sorbone Université, INSERM, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
- APHP, Cardiology and Genetics Departments, Pitié-Salpêtrière Hospital, Paris, France
| | - Benjamin Meder
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University Hospital Heidelberg, Heidelberg, Germany
- Site Heidelberg/Mannheim, DZHK, Heidelberg, Germany
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Mark Daly
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
| | - Krishna G Aragam
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location, University of Amsterdam, Amsterdam, the Netherlands.
- European Reference Network for rare low prevalence and complex diseases of the heart: ERN GUARD-Heart, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Mair DB, Tsui JH, Higashi T, Koenig P, Dong Z, Chen JF, Meir JU, Smith AST, Lee PHU, Ahn EH, Countryman S, Sniadecki NJ, Kim DH. Spaceflight-induced contractile and mitochondrial dysfunction in an automated heart-on-a-chip platform. Proc Natl Acad Sci U S A 2024; 121:e2404644121. [PMID: 39312653 PMCID: PMC11459163 DOI: 10.1073/pnas.2404644121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024] Open
Abstract
With current plans for manned missions to Mars and beyond, the need to better understand, prevent, and counteract the harmful effects of long-duration spaceflight on the body is becoming increasingly important. In this study, an automated heart-on-a-chip platform was flown to the International Space Station on a 1-mo mission during which contractile cardiac function was monitored in real-time. Upon return to Earth, engineered human heart tissues (EHTs) were further analyzed with ultrastructural imaging and RNA sequencing to investigate the impact of prolonged microgravity on cardiomyocyte function and health. Spaceflight EHTs exhibited significantly reduced twitch forces, increased incidences of arrhythmias, and increased signs of sarcomere disruption and mitochondrial damage. Transcriptomic analyses showed an up-regulation of genes and pathways associated with metabolic disorders, heart failure, oxidative stress, and inflammation, while genes related to contractility and calcium signaling showed significant down-regulation. Finally, in silico modeling revealed a potential link between oxidative stress and mitochondrial dysfunction that corresponded with RNA sequencing results. This represents an in vitro model to faithfully reproduce the adverse effects of spaceflight on three-dimensional (3D)-engineered heart tissue.
Collapse
Affiliation(s)
- Devin B. Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jonathan H. Tsui
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Ty Higashi
- Department of Mechanical Engineering, University of Washington, Seattle, WA98195
| | - Paul Koenig
- BioServe Space Technologies, Ann and HJ Smead Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO80303
| | - Zhipeng Dong
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jeffrey F. Chen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jessica U. Meir
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX77058
| | - Alec S. T. Smith
- Department of Physiology and Biophysics, University of Washington, Seattle, WA98195
| | - Peter H. U. Lee
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI02912
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD21205
| | - Stefanie Countryman
- BioServe Space Technologies, Ann and HJ Smead Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO80303
| | - Nathan J. Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA98195
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Center for Cardiovascular Biology, University of Washington, Seattle, WA98109
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA98109
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Center for Cardiovascular Biology, University of Washington, Seattle, WA98109
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA98109
- Department of Medicine, Johns Hopkins University, Baltimore, MD21205
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| |
Collapse
|
5
|
Hansen J, Xiong Y, Siddiq MM, Dhanan P, Hu B, Shewale B, Yadaw AS, Jayaraman G, Tolentino RE, Chen Y, Martinez P, Beaumont KG, Sebra R, Vidovic D, Schürer SC, Goldfarb J, Gallo JM, Birtwistle MR, Sobie EA, Azeloglu EU, Berger SI, Chan A, Schaniel C, Dubois NC, Iyengar R. Multiscale mapping of transcriptomic signatures for cardiotoxic drugs. Nat Commun 2024; 15:7968. [PMID: 39261481 PMCID: PMC11390749 DOI: 10.1038/s41467-024-52145-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
Drug-induced gene expression profiles can identify potential mechanisms of toxicity. We focus on obtaining signatures for cardiotoxicity of FDA-approved tyrosine kinase inhibitors (TKIs) in human induced-pluripotent-stem-cell-derived cardiomyocytes, using bulk transcriptomic profiles. We use singular value decomposition to identify drug-selective patterns across cell lines obtained from multiple healthy human subjects. Cellular pathways affected by cardiotoxic TKIs include energy metabolism, contractile, and extracellular matrix dynamics. Projecting these pathways to published single cell expression profiles indicates that TKI responses can be evoked in both cardiomyocytes and fibroblasts. Integration of transcriptomic outlier analysis with whole genomic sequencing of our six cell lines enables us to correctly reidentify a genomic variant causally linked to anthracycline-induced cardiotoxicity and predict genomic variants potentially associated with TKI-induced cardiotoxicity. We conclude that mRNA expression profiles when integrated with publicly available genomic, pathway, and single cell transcriptomic datasets, provide multiscale signatures for cardiotoxicity that could be used for drug development and patient stratification.
Collapse
Affiliation(s)
- Jens Hansen
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Yuguang Xiong
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mustafa M Siddiq
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Priyanka Dhanan
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Hu
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bhavana Shewale
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arjun S Yadaw
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gomathi Jayaraman
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rosa E Tolentino
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yibang Chen
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pedro Martinez
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kristin G Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dusica Vidovic
- Institute for Data Science and Computing, University of Miami, Coral Gables, FL, 33146, USA
| | - Stephan C Schürer
- Institute for Data Science and Computing, University of Miami, Coral Gables, FL, 33146, USA
| | - Joseph Goldfarb
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - James M Gallo
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- School of Pharmacy and Pharmaceutical Sciences, University of Buffalo SUNY System, Buffalo, NY, 14260, USA
| | - Marc R Birtwistle
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29634, USA
| | - Eric A Sobie
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Evren U Azeloglu
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New, York, NY, 10029, USA
| | - Seth I Berger
- Center for Genetic Medicine Research, Children's National Research Institute, Washington, DC, 20012, USA
| | - Angel Chan
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Cardiology Division, Department of Medicine, Memorial Sloan Kettering Cancer Center New York, New York, NY, 10065, USA
| | - Christoph Schaniel
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicole C Dubois
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Ravi Iyengar
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
6
|
Karpov OA, Stotland A, Raedschelders K, Chazarin B, Ai L, Murray CI, Van Eyk JE. Proteomics of the heart. Physiol Rev 2024; 104:931-982. [PMID: 38300522 PMCID: PMC11381016 DOI: 10.1152/physrev.00026.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Mass spectrometry-based proteomics is a sophisticated identification tool specializing in portraying protein dynamics at a molecular level. Proteomics provides biologists with a snapshot of context-dependent protein and proteoform expression, structural conformations, dynamic turnover, and protein-protein interactions. Cardiac proteomics can offer a broader and deeper understanding of the molecular mechanisms that underscore cardiovascular disease, and it is foundational to the development of future therapeutic interventions. This review encapsulates the evolution, current technologies, and future perspectives of proteomic-based mass spectrometry as it applies to the study of the heart. Key technological advancements have allowed researchers to study proteomes at a single-cell level and employ robot-assisted automation systems for enhanced sample preparation techniques, and the increase in fidelity of the mass spectrometers has allowed for the unambiguous identification of numerous dynamic posttranslational modifications. Animal models of cardiovascular disease, ranging from early animal experiments to current sophisticated models of heart failure with preserved ejection fraction, have provided the tools to study a challenging organ in the laboratory. Further technological development will pave the way for the implementation of proteomics even closer within the clinical setting, allowing not only scientists but also patients to benefit from an understanding of protein interplay as it relates to cardiac disease physiology.
Collapse
Affiliation(s)
- Oleg A Karpov
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Aleksandr Stotland
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Blandine Chazarin
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Lizhuo Ai
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Christopher I Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| |
Collapse
|
7
|
Xie L, Xing Y, Yang J, Liu M, Cai Y. Toxoplasma gondii Reactivation Aggravating Cardiac Function Impairment in Mice. Pathogens 2023; 12:1025. [PMID: 37623985 PMCID: PMC10458591 DOI: 10.3390/pathogens12081025] [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: 06/07/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) reactivation is common, especially among immunocompromised individuals, such as AIDS patients. The cardiac involvement associated with toxoplasmosis, however, is usually obscured by neurological deterioration. The aim of this study was to observe the alterations in cardiac functions in various landmark periods after infection and to assess whether reactivation more seriously damages the heart. METHODS We established three infection models in mice using TgCtwh6, a major strain of T. gondii prevalent in China. The groups included an acute group, chronic latent group, and reactivation group. We evaluated the cardiac function impairment via H & E staining, Masson staining, echocardiography, myocardial enzyme profiles, and cardiac troponin, and detected the expression of inflammatory factors and antioxidant factors with Western blotting. Immunofluorescence was used to detect the expression of the macrophage marker F4/80. RESULTS Our results showed that damage to the heart occurred in the acute and reactivation groups. Impaired cardiac function manifested as a decrease in heart rate and a compensatory increase in left ventricular systolic function. Serum levels of cardiac enzymes also increased dramatically. In the chronic phase, myocardial fibrosis developed, diastolic functions became severely impaired, inflammation persisted, and macrophage expression was slightly reduced. Ultimately, reactivation infection exacerbated damage to cardiac function in mice, potentially leading to diastolic heart failure. Macrophages were strongly activated, and myocardial fibrosis was increased. In addition, the antioxidant capacity of the heart was severely affected by the infection. CONCLUSIONS Taken together, these results suggested that the reactivation of T. gondii infection could aggravate injury to the heart, which could be associated with a host-cell-mediated immune response and strong cytokine production by macrophages, thus representing a novel insight into the pathogenic mechanism of toxoplasmosis.
Collapse
Affiliation(s)
- Linding Xie
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei 230032, China
- Department of Microbiology and Parasitology, the Provincial Laboratory of Pathogen Biology of Anhui, and the Key Laboratory of Zoonoses of Anhui, Anhui Medical University, Hefei 230032, China
| | - Yien Xing
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei 230032, China
- Department of Microbiology and Parasitology, the Provincial Laboratory of Pathogen Biology of Anhui, and the Key Laboratory of Zoonoses of Anhui, Anhui Medical University, Hefei 230032, China
| | - Jun Yang
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei 230032, China
- Department of Microbiology and Parasitology, the Provincial Laboratory of Pathogen Biology of Anhui, and the Key Laboratory of Zoonoses of Anhui, Anhui Medical University, Hefei 230032, China
| | - Min Liu
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei 230032, China
- Department of Microbiology and Parasitology, the Provincial Laboratory of Pathogen Biology of Anhui, and the Key Laboratory of Zoonoses of Anhui, Anhui Medical University, Hefei 230032, China
| | - Yihong Cai
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei 230032, China
- Department of Microbiology and Parasitology, the Provincial Laboratory of Pathogen Biology of Anhui, and the Key Laboratory of Zoonoses of Anhui, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
8
|
Li Y, Li B, Chen WD, Wang YD. Role of G-protein coupled receptors in cardiovascular diseases. Front Cardiovasc Med 2023; 10:1130312. [PMID: 37342437 PMCID: PMC10277692 DOI: 10.3389/fcvm.2023.1130312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/09/2023] [Indexed: 06/22/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally, with CVDs accounting for nearly 30% of deaths worldwide each year. G-protein-coupled receptors (GPCRs) are the most prominent family of receptors on the cell surface, and play an essential regulating cellular physiology and pathology. Some GPCR antagonists, such as β-blockers, are standard therapy for the treatment of CVDs. In addition, nearly one-third of the drugs used to treat CVDs target GPCRs. All the evidence demonstrates the crucial role of GPCRs in CVDs. Over the past decades, studies on the structure and function of GPCRs have identified many targets for the treatment of CVDs. In this review, we summarize and discuss the role of GPCRs in the function of the cardiovascular system from both vascular and heart perspectives, then analyze the complex ways in which multiple GPCRs exert regulatory functions in vascular and heart diseases. We hope to provide new ideas for the treatment of CVDs and the development of novel drugs.
Collapse
Affiliation(s)
- Yuanqiang Li
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Boyu Li
- Department of Gastroenterology and Hematology, The People's Hospital of Hebi, Henan, China
| | - Wei-Dong Chen
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Medicine, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Yan-Dong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
9
|
Moustafa A, Hashemi S, Brar G, Grigull J, Ng SHS, Williams D, Schmitt-Ulms G, McDermott JC. The MEF2A transcription factor interactome in cardiomyocytes. Cell Death Dis 2023; 14:240. [PMID: 37019881 PMCID: PMC10076289 DOI: 10.1038/s41419-023-05665-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 04/07/2023]
Abstract
Transcriptional regulators encoded by the Myocyte Enhancer Factor 2 (MEF2) gene family play a fundamental role in cardiac development, homeostasis and pathology. Previous studies indicate that MEF2A protein-protein interactions serve as a network hub in several cardiomyocyte cellular processes. Based on the idea that interactions with regulatory protein partners underly the diverse roles of MEF2A in cardiomyocyte gene expression, we undertook a systematic unbiased screen of the MEF2A protein interactome in primary cardiomyocytes using an affinity purification-based quantitative mass spectrometry approach. Bioinformatic processing of the MEF2A interactome revealed protein networks involved in the regulation of programmed cell death, inflammatory responses, actin dynamics and stress signaling in primary cardiomyocytes. Further biochemical and functional confirmation of specific protein-protein interactions documented a dynamic interaction between MEF2A and STAT3 proteins. Integration of transcriptome level data from MEF2A and STAT3-depleted cardiomyocytes reveals that the balance between MEF2A and STAT3 activity exerts a level of executive control over the inflammatory response and cardiomyocyte cell survival and experimentally ameliorates Phenylephrine induced cardiomyocyte hypertrophy. Lastly, we identified several MEF2A/STAT3 co-regulated genes, including the MMP9 gene. Herein, we document the cardiomyocyte MEF2A interactome, which furthers our understanding of protein networks involved in the hierarchical control of normal and pathophysiological cardiomyocyte gene expression in the mammalian heart.
Collapse
Affiliation(s)
- Amira Moustafa
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Sara Hashemi
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Seneca College, School of Health Sciences, King City, ON, L7B 1B3, Canada
| | - Gurnoor Brar
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Jörg Grigull
- Department of Mathematics and Statistics, York University, Toronto, ON, M3J1P3, Canada
| | - Siemon H S Ng
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Analytical Development, Notch Therapeutics, Toronto, ON, M5G 1M1, Canada
| | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada.
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
10
|
Mora MT, Zaza A, Trenor B. Insights from an electro-mechanical heart failure cell model: Role of SERCA enhancement on arrhythmogenesis and myocyte contraction. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 230:107350. [PMID: 36689807 DOI: 10.1016/j.cmpb.2023.107350] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND OBJECTIVE Structural and electrical remodeling in heart failure predisposes the heart to ventricular arrhythmias. Computer modeling approaches, used to complement experimental results, can provide a more mechanistic knowledge of the biophysical phenomena underlying cardiac pathologies. Indeed, previous in-silico studies have improved the understanding of the electrical correlates of heart failure involved in arrhythmogenesis; however, information on the crosstalk between electrical activity, intracellular Ca2+ and contraction is still incomplete. This study aims to investigate the electro-mechanical behavior of virtual failing human ventricular myocytes to help in the development of therapies, which should ideally target pump failure and arrhythmias at the same time. METHODS We implemented characteristic remodeling of heart failure with reduced ejection fraction by including reported changes in ionic conductances, sarcomere function and cell structure (e.g. T-tubules disarray). Model parametrization was based on published experimental data and the outcome of simulations was validated against experimentally observed patterns. We focused on two aspects of myocardial dysfunction central in heart failure: altered force-frequency relationship and susceptibility to arrhythmogenic early afterdepolarizations. Because biological variability is a major problem in the generalization of in-silico findings based on a unique set of model parameters, we generated and evaluated a population of models. RESULTS The population-based approach is crucial in robust identification of parameters at the core of abnormalities and in generalizing the outcome of their correction. As compared to non-failing ones, failing myocytes had prolonged repolarization, a higher incidence of early afterdepolarizations, reduced contraction and a shallower force-frequency relationship, all features peculiar of heart failure. Component analysis applied to the model population identified reduced SERCA function as a relevant contributor to most of these derangements, which were largely reverted or diminished by restoration of SERCA function alone. CONCLUSIONS These simulated results encourage the development of strategies comprising SERCA stimulation and highlight the need to evaluate both electrical and mechanical outcomes.
Collapse
Affiliation(s)
- Maria Teresa Mora
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain
| | - Antonio Zaza
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi Milano-Bicocca, Italy; Unità di Fisiologia Cardiovascolare, IRCCs Istituto Auxologico Italiano, Italy
| | - Beatriz Trenor
- Centro de Investigación e Innovación en Bioingeniería, Universitat Politècnica de València, Valencia, Spain.
| |
Collapse
|
11
|
Chaulin AM. Diagnostic Role and Methods of Detection of Cardiac Troponins: An Opinion from Historical and Current Points of View. Curr Cardiol Rev 2023; 19:e100622205865. [PMID: 35702796 PMCID: PMC10201884 DOI: 10.2174/1573403x18666220610164946] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 02/08/2023] Open
Abstract
The laboratory methods for the determination of cardiac troponins (cTnI, cTnT) used nowadays are extremely diverse, which has a significant impact on our understanding of the biology and diagnostic the value of cTnI and cTnT as biomarkers. The main classification of methods for the determination of cTnI and cTnT is based on the sensitivity of the immunoassay. Low- and moderately sensitive detection methods are known to be relatively less sensitive, which leads to a relatively late confirmation of cardiomyocyte death. Due to the new highly sensitive methods used to determine cTnI and cTnT, designated as a highly or ultrasensitive immunoassays (hs-TnT and hs-TnT), we received new, revised data about the biology of cardiac troponin molecules. In particular, it became clear that they can be considered products of normal myocardium metabolism since hs-TnT and hs-TnT are detected in almost all healthy patients. It also turned out that hs-TnT and hs-TnT differ by gender (in men, troponin concentration in the blood is higher than in women), age (in elderly patients, the levels of troponins are higher than in young ones) and circadian cycles (morning concentrations of troponins are higher than in the evening). A large variety of methods for determining cTnI and cTnT, differing in their diagnostic capabilities, creates the need for tests to perform an unbiased assessment of the analytical characteristics of each method. This review focuses on the most pressing issues related to the discussion of the biological characteristics of cardiac troponin and the analytical characteristics of troponin immunoassays from a historical and contemporary point of view.
Collapse
Affiliation(s)
- Aleksey Michailovich Chaulin
- Department of Cardiology and Cardiovascular surgery, Medical faculty, Samara State Medical University, Samara, Russia
- Department of Clinical Chemistry, Samara Regional Clinical Cardiological Dispensary, Samara, Russia
| |
Collapse
|
12
|
Renal Denervation Influences Angiotensin II Types 1 and 2 Receptors. Int J Nephrol 2022; 2022:8731357. [PMID: 36262553 PMCID: PMC9576444 DOI: 10.1155/2022/8731357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
The sympathetic and renin-angiotensin systems (RAS) are two critical regulatory systems in the kidney which affect renal hemodynamics and function. These two systems interact with each other so that angiotensin II (Ang II) has the presynaptic effect on the norepinephrine secretion. Another aspect of this interaction is that the sympathetic nervous system affects the function and expression of local RAS receptors, mainly Ang II receptors. Therefore, in many pathological conditions associated with an increased renal sympathetic tone, these receptors' expression changes and renal denervation can normalize these changes and improve the diseases. It seems that the renal sympathectomy can alter Ang II receptors expression and the distribution of RAS receptors in the kidneys, which influence renal functions.
Collapse
|
13
|
Da’as SI, Hasan W, Salem R, Younes N, Abdelrahman D, Mohamed IA, Aldaalis A, Temanni R, Mathew LS, Lorenz S, Yacoub M, Nomikos M, Nasrallah GK, Fakhro KA. Transcriptome Profile Identifies Actin as an Essential Regulator of Cardiac Myosin Binding Protein C3 Hypertrophic Cardiomyopathy in a Zebrafish Model. Int J Mol Sci 2022; 23:ijms23168840. [PMID: 36012114 PMCID: PMC9408294 DOI: 10.3390/ijms23168840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 01/15/2023] Open
Abstract
Variants in cardiac myosin-binding protein C (cMyBP-C) are the leading cause of inherited hypertrophic cardiomyopathy (HCM), demonstrating the key role that cMyBP-C plays in the heart’s contractile machinery. To investigate the c-MYBPC3 HCM-related cardiac impairment, we generated a zebrafish mypbc3-knockout model. These knockout zebrafish displayed significant morphological heart alterations related to a significant decrease in ventricular and atrial diameters at systolic and diastolic states at the larval stages. Immunofluorescence staining revealed significant hyperplasia in the mutant’s total cardiac and ventricular cardiomyocytes. Although cardiac contractility was similar to the wild-type control, the ejection fraction was significantly increased in the mypbc3 mutants. At later stages of larval development, the mutants demonstrated an early cardiac phenotype of myocardium remodeling, concurrent cardiomyocyte hyperplasia, and increased ejection fraction as critical processes in HCM initiation to counteract the increased ventricular myocardial wall stress. The examination of zebrafish adults showed a thickened ventricular cardiac wall with reduced heart rate, swimming speed, and endurance ability in both the mypbc3 heterozygous and homozygous groups. Furthermore, heart transcriptome profiling showed a significant downregulation of the actin-filament-based process, indicating an impaired actin cytoskeleton organization as the main dysregulating factor associated with the early ventricular cardiac hypertrophy in the zebrafish mypbc3 HCM model.
Collapse
Affiliation(s)
- Sahar Isa Da’as
- Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar
- Australian Regenerative Medicine Institute, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
- Correspondence:
| | - Waseem Hasan
- Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Rola Salem
- Health Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Nadine Younes
- Department of Biomedical Sciences, College of Health Science, Member of QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Doua Abdelrahman
- Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Iman A. Mohamed
- Australian Regenerative Medicine Institute, Monash University, Melbourne 3168, Australia
| | - Arwa Aldaalis
- Australian Regenerative Medicine Institute, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| | - Ramzi Temanni
- Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Lisa Sara Mathew
- Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Stephan Lorenz
- Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | | | - Michail Nomikos
- College of Medicine, Member of QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Gheyath K. Nasrallah
- Department of Biomedical Sciences, College of Health Science, Member of QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Khalid A. Fakhro
- Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar
- Australian Regenerative Medicine Institute, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
- Weill Cornell Medical College, Doha P.O. Box 24811, Qatar
| |
Collapse
|
14
|
Mendoza A, Karch J. Keeping the beat against time: Mitochondrial fitness in the aging heart. FRONTIERS IN AGING 2022; 3:951417. [PMID: 35958271 PMCID: PMC9360554 DOI: 10.3389/fragi.2022.951417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/30/2022] [Indexed: 11/21/2022]
Abstract
The process of aging strongly correlates with maladaptive architectural, mechanical, and biochemical alterations that contribute to the decline in cardiac function. Consequently, aging is a major risk factor for the development of heart disease, the leading cause of death in the developed world. In this review, we will summarize the classic and recently uncovered pathological changes within the aged heart with an emphasis on the mitochondria. Specifically, we describe the metabolic changes that occur in the aging heart as well as the loss of mitochondrial fitness and function and how these factors contribute to the decline in cardiomyocyte number. In addition, we highlight recent pharmacological, genetic, or behavioral therapeutic intervention advancements that may alleviate age-related cardiac decline.
Collapse
Affiliation(s)
- Arielys Mendoza
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, United States
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, United States
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
15
|
Saleh ZT, Alraoush AT, Aqel AA, Shawashi TO, Chung M, Lennie TA. Sex Differences in the Association Between Inflammation and Event-Free Survival in Patients With Heart Failure. J Cardiovasc Nurs 2022; 37:386-393. [PMID: 37707972 PMCID: PMC8733054 DOI: 10.1097/jcn.0000000000000831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Heart failure (HF) is associated with chronic inflammation, which is adversely associated with survival. Although sex-related differences in inflammation have been described in patients with HF, whether sex-related differences in inflammation are associated with event-free survival has not been examined. AIM The aim of this study was to determine whether the association between inflammation as indicated by tumor necrosis factor-α and event-free survival differs between men and women with HF after controlling for demographic and clinical variables. METHOD This was a secondary analysis of data from 301 male (age, 61.0 ± 11.4 years) and 137 female (age, 60.3 ± 12.1 years) patients with HF. Serum levels of soluble tumor necrosis factor receptor 1 were used to indicate inflammatory status. Patients were grouped according to median split of soluble tumor necrosis factor receptor 1 level and sex into male with low inflammation (≤1820 pg/mL) (n = 158) or high inflammation (>1820 pg/mL) (n = 143), and female with low inflammation (n = 63) or high inflammation (n = 74). Cox regression models were run separately for men and women to determine whether inflammation contributed to differences in event-free survival between sexes with HF. RESULTS There were 84 male (27.9%) and 27 female (19.7%) patients who had an event. Event-free survival in women did not differ by the severity of inflammation in the Cox regression analysis. In contrast, men with high inflammation had 1.85 times higher risk for an event compared with men with low inflammation. CONCLUSION These data provide evidence that inflammation contributed to differences in event-free survival in men but not women with HF. Clinicians should be aware that men who have higher inflammation may be at a greater risk of HF or cardiac-related events than others with HF.
Collapse
Affiliation(s)
- Zyad T. Saleh
- School of Nursing, The University of Jordan, Queen Rania Street, Amman, Jordan 11942
| | - Ahmad T. Alraoush
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan 22110
| | - Ahmad A. Aqel
- School of Nursing, The University of Jordan, Queen Rania Street, Amman, Jordan 11942
| | - Tagreed O. Shawashi
- School of Nursing, The University of Jordan, Queen Rania Street, Amman, Jordan 11942
| | - Misook Chung
- College of Nursing, University of Kentucky, 315 College of Nursing Building, 751 Rose Street, Lexington, KY 40536
| | - Terry A. Lennie
- College of Nursing, University of Kentucky, 315 College of Nursing Building, 751 Rose Street, Lexington, KY 40536
| |
Collapse
|
16
|
Ahmed R, Moaddab A, Hussain SW, Viriya G, Graham-Hill S. A Rare Case of Dilated Cardiomyopathy, Focal Segmental Glomerulosclerosis, and Bell’s Palsy in a 29-Year-Old Male After Coxsackievirus Infection. Cureus 2022; 14:e26285. [PMID: 35898376 PMCID: PMC9308950 DOI: 10.7759/cureus.26285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a severe myocardial disease with diversified etiologies. Coxsackievirus serotype B (CV-B) is a known cause of infectious myocarditis that leads to DCM. The pathogenesis of CV-B myocarditis is complex and involves a combination of tissue destruction from viral proliferation and host immune response. Diagnosis is based on clinical findings and the presence of post-infection elevated titers of IgM antibodies to CV-B. Echocardiography is an important imaging modality that plays a key role in diagnosing DCM. Rare complications of coxsackievirus infection may include facial paralysis and chronic kidney disease with nephrotic syndrome. Here we present a rare case of a 29-year-old-male with recent Bell’s palsy who presented with new-onset heart failure with left ventricular ejection fraction of 5% and focal segmental glomerulosclerosis nephrotic syndrome in the setting of elevated antibodies to CV-B.
Collapse
|
17
|
Liu Z, Song YN, Chen KY, Gao WL, Chen HJ, Liang GY. Bioinformatics prediction of potential mechanisms and biomarkers underlying dilated cardiomyopathy. World J Cardiol 2022; 14:282-296. [PMID: 35702326 PMCID: PMC9157606 DOI: 10.4330/wjc.v14.i5.282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/19/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Heart failure is a health burden responsible for high morbidity and mortality worldwide, and dilated cardiomyopathy (DCM) is one of the most common causes of heart failure. DCM is a disease of the heart muscle and is characterized by enlargement and dilation of at least one ventricle alongside impaired contractility with left ventricular ejection fraction < 40%. It is also associated with abnormalities in cytoskeletal proteins, mitochondrial ATP transporter, microvasculature, and fibrosis. However, the pathogenesis and potential biomarkers of DCM remain to be investigated. AIM To investigate the candidate genes and pathways involved in DCM patients. METHODS Two expression datasets (GSE3585 and GSE5406) were downloaded from the Gene Expression Omnibus database. The differentially expressed genes (DEGs) between the DCM patients and healthy individuals were identified using the R package "linear models for microarray data." The pathways with common DEGs were analyzed via Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analyses. Moreover, a protein-protein interaction network (PPI) was constructed to identify the hub genes and modules. The MicroRNA Database was applied to predict the microRNAs (miRNAs) targeting the hub genes. Additionally, immune cell infiltration in DCM was analyzed using CIBERSORT. RESULTS In total, 97 DEGs (47 upregulated and 50 downregulated) were identified. GO analysis showed that the DEGs were mainly enriched in "response to growth factor," "extracellular matrix," and "extracellular matrix structural constituent." KEGG pathway analysis indicated that the DEGs were mainly enriched in "protein digestion and absorption" and "interleukin 17 (IL-17) signaling pathway." The PPI network suggested that collagen type III alpha 1 chain (COL3A1) and COL1A2 contribute to the pathogenesis of DCM. Additionally, visualization of the interactions between miRNAs and the hub genes revealed that hsa-miR-5682 and hsa-miR-4500 interacted with both COL3A1 and COL1A2, and thus these miRNAs might play roles in DCM. Immune cell infiltration analysis revealed that DCM patients had more infiltrated plasma cells and fewer infiltrated B memory cells, T follicular helper cells, and resting dendritic cells. CONCLUSION COL1A2 and COL3A1 and their targeting miRNAs, hsa-miR-5682 and hsa-miR-4500, may play critical roles in the pathogenesis of DCM, which are closely related to the IL-17 signaling pathway and acute inflammatory response. These results may provide useful clues for the diagnosis and treatment of DCM.
Collapse
Affiliation(s)
- Zhou Liu
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Ying-Nan Song
- Translational Medicine Research Center, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Guiyang 510000, Guizhou Province, China
| | - Kai-Yuan Chen
- Translational Medicine Research Center, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Wei-Long Gao
- Translational Medicine Research Center, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
| | - Hong-Jin Chen
- Translational Medicine Research Center, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Guiyang 510000, Guizhou Province, China
| | - Gui-You Liang
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang 550025, Guizhou Province, China
- Department of Cardiovascular Surgery, the Affiliated Hospital of Guizhou Medical University, Guiyang 510000, Guizhou Province, China.
| |
Collapse
|
18
|
Suay-Corredera C, Alegre-Cebollada J. The mechanics of the heart: zooming in on hypertrophic cardiomyopathy and cMyBP-C. FEBS Lett 2022; 596:703-746. [PMID: 35224729 DOI: 10.1002/1873-3468.14301] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Hypertrophic cardiomyopathy (HCM), a disease characterized by cardiac muscle hypertrophy and hypercontractility, is the most frequently inherited disorder of the heart. HCM is mainly caused by variants in genes encoding proteins of the sarcomere, the basic contractile unit of cardiomyocytes. The most frequently mutated among them is MYBPC3, which encodes cardiac myosin-binding protein C (cMyBP-C), a key regulator of sarcomere contraction. In this review, we summarize clinical and genetic aspects of HCM and provide updated information on the function of the healthy and HCM sarcomere, as well as on emerging therapeutic options targeting sarcomere mechanical activity. Building on what is known about cMyBP-C activity, we examine different pathogenicity drivers by which MYBPC3 variants can cause disease, focussing on protein haploinsufficiency as a common pathomechanism also in nontruncating variants. Finally, we discuss recent evidence correlating altered cMyBP-C mechanical properties with HCM development.
Collapse
|
19
|
Becker E, Francino A, Pich A, Perrot A, Kraft T, Radocaj A. AQUA Mutant Protein Quantification of Endomyocardial Biopsy-Sized Samples From a Patient With Hypertrophic Cardiomyopathy. Front Cardiovasc Med 2022; 9:816330. [PMID: 35265683 PMCID: PMC8899185 DOI: 10.3389/fcvm.2022.816330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
In genetic diseases like hypertrophic cardiomyopathy, reliable quantification of the expression level of mutant protein can play an important role in disease research, diagnosis, treatment and prognosis. For heterozygous β-myosin heavy chain (β-MyHC) mutations it has been shown that disease severity is related to the fraction of mutant protein in the myocardium. Yet, heart tissue from patients with genetically characterized diseases is scarce. Here we asked, if even in the case of small endomyocardial biopsies, single quantifications produce reliable results. Myocardial samples were taken from four different regions of an explanted heart of a patient with hypertrophic cardiomyopathy carrying point mutation p.Gly716Arg in β-MyHC. From both, large samples (15 mg) and small, endomyocardial biopsy-sized samples (≤ 1 mg) myosin was extracted and enzymatically digested to yield a specific peptide of interest that allowed to distinguish mutant and wild-type β-MyHC. Absolute quantification by mass spectrometry (AQUA) of the peptide of interest was performed repeatedly for both sample sizes to determine the fraction of mutant β-MyHC. Fractions of mutant β-MyHC (32% on average) showed only small differences between the four cardiac regions and for large and small samples. The standard deviations were smaller than five percentage points for all cardiac regions. The two quantification methods (large and small sample size) produce results with comparable accuracy and precision. Consequently, with our method even small endomyocardial biopsies allow reliable protein quantification for potential diagnostic purposes.
Collapse
Affiliation(s)
- Edgar Becker
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Antonio Francino
- Cardiology Department, Hospital Clinic/IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Andreas Pich
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Andreas Perrot
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Theresia Kraft
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Ante Radocaj
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- *Correspondence: Ante Radocaj
| |
Collapse
|
20
|
Ušaj M, Moretto L, Månsson A. Critical Evaluation of Current Hypotheses for the Pathogenesis of Hypertrophic Cardiomyopathy. Int J Mol Sci 2022; 23:2195. [PMID: 35216312 PMCID: PMC8880276 DOI: 10.3390/ijms23042195] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Hereditary hypertrophic cardiomyopathy (HCM), due to mutations in sarcomere proteins, occurs in more than 1/500 individuals and is the leading cause of sudden cardiac death in young people. The clinical course exhibits appreciable variability. However, typically, heart morphology and function are normal at birth, with pathological remodeling developing over years to decades, leading to a phenotype characterized by asymmetric ventricular hypertrophy, scattered fibrosis and myofibrillar/cellular disarray with ultimate mechanical heart failure and/or severe arrhythmias. The identity of the primary mutation-induced changes in sarcomere function and how they trigger debilitating remodeling are poorly understood. Support for the importance of mutation-induced hypercontractility, e.g., increased calcium sensitivity and/or increased power output, has been strengthened in recent years. However, other ideas that mutation-induced hypocontractility or non-uniformities with contractile instabilities, instead, constitute primary triggers cannot yet be discarded. Here, we review evidence for and criticism against the mentioned hypotheses. In this process, we find support for previous ideas that inefficient energy usage and a blunted Frank-Starling mechanism have central roles in pathogenesis, although presumably representing effects secondary to the primary mutation-induced changes. While first trying to reconcile apparently diverging evidence for the different hypotheses in one unified model, we also identify key remaining questions and suggest how experimental systems that are built around isolated primarily expressed proteins could be useful.
Collapse
Affiliation(s)
| | | | - Alf Månsson
- Department of Chemistry and Biomedical Sciences, Faculty of Health and Life Sciences, Linnaeus University, SE-39182 Kalmar, Sweden; (M.U.); (L.M.)
| |
Collapse
|
21
|
Petersen CE, Tripoli BA, Schoborg TA, Smyth JT. Analysis of Drosophila cardiac hypertrophy by microcomputerized tomography for genetic dissection of heart growth mechanisms. Am J Physiol Heart Circ Physiol 2022; 322:H296-H309. [PMID: 34951542 PMCID: PMC8782661 DOI: 10.1152/ajpheart.00387.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Heart failure is often preceded by pathological cardiac hypertrophy, a thickening of the heart musculature driven by complex gene regulatory and signaling processes. The Drosophila heart has great potential as a genetic model for deciphering the underlying mechanisms of cardiac hypertrophy. However, current methods for evaluating hypertrophy of the Drosophila heart are laborious and difficult to carry out reproducibly. Here, we demonstrate that microcomputerized tomography (microCT) is an accessible, highly reproducible method for nondestructive, quantitative analysis of Drosophila heart morphology and size. To validate our microCT approach for analyzing Drosophila cardiac hypertrophy, we show that expression of constitutively active Ras (Ras85DV12), previously shown to cause hypertrophy of the fly heart, results in significant thickening of both adult and larval heart walls when measured from microCT images. We then show using microCT analysis that genetic upregulation of store-operated Ca2+ entry (SOCE) driven by expression of constitutively active Stim (StimCA) or Orai (OraiCA) proteins also results in significant hypertrophy of the Drosophila heart, through a process that specifically depends on Orai Ca2+ influx channels. Intravital imaging of heart contractility revealed significantly reduced end-diastolic and end-systolic dimensions in StimCA- and OraiCA-expressing hearts, consistent with the hypertrophic phenotype. These results demonstrate that increased SOCE activity is an important driver of hypertrophic cardiomyocyte growth, and demonstrate how microCT analysis combined with tractable genetic tools in Drosophila can be used to delineate molecular signaling processes that underlie cardiac hypertrophy and heart failure.NEW & NOTEWORTHY Genetic analysis of Drosophila cardiac hypertrophy holds immense potential for the discovery of new therapeutic targets to prevent and treat heart failure. This potential has been hindered by a lack of rapid and effective methods for analyzing heart size in flies. Here, we demonstrate that analysis of the Drosophila heart with microcomputerized tomography yields accurate and highly reproducible heart size measurements that can be used to analyze heart growth and cardiac hypertrophy in Drosophila.
Collapse
Affiliation(s)
- Courtney E. Petersen
- 1Graduate Program in Molecular and Cell Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Benjamin A. Tripoli
- 1Graduate Program in Molecular and Cell Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Todd A. Schoborg
- 2Department of Molecular Biology, University of Wyoming, Laramie, Wyoming
| | - Jeremy T. Smyth
- 3Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
22
|
Bourque K, Hawey C, Jiang A, Mazarura GR, Hébert TE. Biosensor-based profiling to track cellular signalling in patient-derived models of dilated cardiomyopathy. Cell Signal 2022; 91:110239. [PMID: 34990783 DOI: 10.1016/j.cellsig.2021.110239] [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] [Received: 09/30/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathies (DCM) represent a diverse group of cardiovascular diseases impacting the structure and function of the myocardium. To better treat these diseases, we need to understand the impact of such cardiomyopathies on critical signalling pathways that drive disease progression downstream of receptors we often target therapeutically. Our understanding of cellular signalling events has progressed substantially in the last few years, in large part due to the design, validation and use of biosensor-based approaches to studying such events in cells, tissues and in some cases, living animals. Another transformative development has been the use of human induced pluripotent stem cells (hiPSCs) to generate disease-relevant models from individual patients. We highlight the importance of going beyond monocellular cultures to incorporate the influence of paracrine signalling mediators. Finally, we discuss the recent coalition of these approaches in the context of DCM. We discuss recent work in generating patient-derived models of cardiomyopathies and the utility of using signalling biosensors to track disease progression and test potential therapeutic strategies that can be later used to inform treatment options in patients.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Alyson Jiang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Grace R Mazarura
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
23
|
Camman M, Joanne P, Agbulut O, Hélary C. 3D models of dilated cardiomyopathy: Shaping the chemical, physical and topographical properties of biomaterials to mimic the cardiac extracellular matrix. Bioact Mater 2022; 7:275-291. [PMID: 34466733 PMCID: PMC8379361 DOI: 10.1016/j.bioactmat.2021.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
The pathophysiology of dilated cardiomyopathy (DCM), one major cause of heart failure, is characterized by the dilation of the heart but remains poorly understood because of the lack of adequate in vitro models. Current 2D models do not allow for the 3D organotypic organization of cardiomyocytes and do not reproduce the ECM perturbations. In this review, the different strategies to mimic the chemical, physical and topographical properties of the cardiac tissue affected by DCM are presented. The advantages and drawbacks of techniques generating anisotropy required for the cardiomyocytes alignment are discussed. In addition, the different methods creating macroporosity and favoring organotypic organization are compared. Besides, the advances in the induced pluripotent stem cells technology to generate cardiac cells from healthy or DCM patients will be described. Thanks to the biomaterial design, some features of the DCM extracellular matrix such as stiffness, porosity, topography or chemical changes can impact the cardiomyocytes function in vitro and increase their maturation. By mimicking the affected heart, both at the cellular and at the tissue level, 3D models will enable a better understanding of the pathology and favor the discovery of novel therapies.
Collapse
Affiliation(s)
- Marie Camman
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Pierre Joanne
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
| |
Collapse
|
24
|
Derrick CJ, Pollitt EJG, Sanchez Sevilla Uruchurtu A, Hussein F, Grierson AJ, Noël ES. Lamb1a regulates atrial growth by limiting second heart field addition during zebrafish heart development. Development 2021; 148:272294. [PMID: 34568948 DOI: 10.1242/dev.199691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
During early vertebrate heart development, the heart transitions from a linear tube to a complex asymmetric structure, a morphogenetic process that occurs simultaneously with growth of the heart. Cardiac growth during early heart morphogenesis is driven by deployment of cells from the second heart field (SHF) into both poles of the heart. Laminin is a core component of the extracellular matrix and, although mutations in laminin subunits are linked with cardiac abnormalities, no role for laminin has been identified in early vertebrate heart morphogenesis. We identified tissue-specific expression of laminin genes in the developing zebrafish heart, supporting a role for laminins in heart morphogenesis. Analysis of heart development in lamb1a zebrafish mutant embryos reveals mild morphogenetic defects and progressive cardiomegaly, and that Lamb1a functions to limit heart size during cardiac development by restricting SHF addition. lamb1a mutants exhibit hallmarks of altered haemodynamics, and blocking cardiac contractility in lamb1a mutants rescues heart size and atrial SHF addition. Together, these results suggest that laminin mediates interactions between SHF deployment and cardiac biomechanics during heart morphogenesis and growth in the developing embryo.
Collapse
Affiliation(s)
| | - Eric J G Pollitt
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | | | - Farah Hussein
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Andrew J Grierson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Emily S Noël
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
25
|
Duan Y, Glazier R, Bazrafshan A, Hu Y, Rashid SA, Petrich BG, Ke Y, Salaita K. Mechanically Triggered Hybridization Chain Reaction. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202107660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Yuxin Duan
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | - Roxanne Glazier
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| | | | - Yuesong Hu
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | - Sk Aysha Rashid
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | | | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| | - Khalid Salaita
- Department of Chemistry Emory University Atlanta GA 30322 USA
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| |
Collapse
|
26
|
Duan Y, Glazier R, Bazrafshan A, Hu Y, Rashid SA, Petrich BG, Ke Y, Salaita K. Mechanically Triggered Hybridization Chain Reaction. Angew Chem Int Ed Engl 2021; 60:19974-19981. [PMID: 34242462 PMCID: PMC8390435 DOI: 10.1002/anie.202107660] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Indexed: 01/16/2023]
Abstract
Cells transmit piconewton forces to receptors to mediate processes such as migration and immune recognition. A major challenge in quantifying such forces is the sparsity of cell mechanical events. Accordingly, molecular tension is typically quantified with high resolution fluorescence microscopy, which hinders widespread adoption and application. Here, we report a mechanically triggered hybridization chain reaction (mechano-HCR) that allows chemical amplification of mechanical events. The amplification is triggered when a cell receptor mechanically denatures a duplex revealing a cryptic initiator to activate the HCR reaction in situ. Importantly, mechano-HCR enables direct readout of pN forces using a plate reader. We leverage this capability and measured mechano-IC50 for aspirin, Y-27632, and eptifibatide. Given that cell mechanical phenotypes are of clinical importance, mechano-HCR may offer a convenient route for drug discovery, personalized medicine, and disease diagnosis.
Collapse
Affiliation(s)
- Yuxin Duan
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Roxanne Glazier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | | | - Yuesong Hu
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Sk Aysha Rashid
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Brian G Petrich
- Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
27
|
Modeling Cardiomyopathies in a Dish: State-of-the-Art and Novel Perspectives on hiPSC-Derived Cardiomyocytes Maturation. BIOLOGY 2021; 10:biology10080730. [PMID: 34439963 PMCID: PMC8389603 DOI: 10.3390/biology10080730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/23/2022]
Abstract
The stem cell technology and the induced pluripotent stem cells (iPSCs) production represent an excellent alternative tool to study cardiomyopathies, which overcome the limitations associated with primary cardiomyocytes (CMs) access and manipulation. CMs from human iPSCs (hiPSC-CMs) are genetically identical to patient primary cells of origin, with the main electrophysiological and mechanical features of CMs. The key issue to be solved is to achieve a degree of structural and functional maturity typical of adult CMs. In this perspective, we will focus on the main differences between fetal-like hiPSC-CMs and adult CMs. A viewpoint is given on the different approaches used to improve hiPSC-CMs maturity, spanning from long-term culture to complex engineered heart tissue. Further, we outline limitations and future developments needed in cardiomyopathy disease modeling.
Collapse
|
28
|
Chen YJ, Chien CS, Chiang CE, Chen CH, Cheng HM. From Genetic Mutations to Molecular Basis of Heart Failure Treatment: An Overview of the Mechanism and Implication of the Novel Modulators for Cardiac Myosin. Int J Mol Sci 2021; 22:6617. [PMID: 34205587 PMCID: PMC8234187 DOI: 10.3390/ijms22126617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a syndrome encompassing several important etiologies that lead to the imbalance between oxygen demand and supply. Despite the usage of guideline-directed medical therapy for HF has shown better outcomes, novel therapeutic strategies are desirable, especially for patients with preserved or mildly reduced left ventricular ejection fraction. In this regard, understanding the molecular basis for cardiomyopathies is expected to fill in the knowledge gap and generate new therapies to improve prognosis for HF. This review discusses an evolutionary mechanism designed to regulate cardiac contraction and relaxation through the most often genetically determined cardiomyopathies associated with HF. In addition, both the myosin inhibitor and myosin activator are promising new treatments for cardiomyopathies. A comprehensive review from genetic mutations to the molecular basis of direct sarcomere modulators will help shed light on future studies for a better characterization of HF etiologies and potential therapeutic targets.
Collapse
Affiliation(s)
- Yu-Jen Chen
- Department of Internal Medicine, Division of Cardiovascular Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan;
- Department of Internal Medicine, Division of Cardiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chian-Shiu Chien
- Innovative Cellular Therapy Center, Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
| | - Chern-En Chiang
- General Clinical Research Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- Department of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei 112304, Taiwan
| | - Chen-Huan Chen
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- College of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Hao-Min Cheng
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei 112304, Taiwan
- Center for Evidence-Based Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| |
Collapse
|
29
|
The Impact of Chronic Heart Failure on Retinal Vessel Density Assessed by Optical Coherence Tomography Angiography in Children with Dilated Cardiomyopathy. J Clin Med 2021; 10:jcm10122659. [PMID: 34208770 PMCID: PMC8235508 DOI: 10.3390/jcm10122659] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 01/23/2023] Open
Abstract
(1) Introduction: The aim of this study is to assess retinal vessel density (VD) in the superficial capillary plexus layer (SP) and deep capillary plexus layer (DP) in children with chronic heart failure (CHF) in the course of dilated cardiomyopathy (DCM) using optical coherence tomography angiography (OCTA). (2) Methods: Thirty children with CHF due to DCM lasting more than six months, with an enlarged left ventricle and impaired left ventricular systolic function (left ventricular ejection fraction (LVEF) ≤ 55%), were enrolled to have both their eyes assessed for this study. Mean age of the children was 9.9 ± 3.57 years. The control group consisted of an additional 30 children without CHF (mean age 11.27 ± 3.33 years) matched for age and gender against the study group. All participants underwent transthoracic echocardiography to measure LVEF using Simpson method. Blood serum was tested for N-terminal-pro-brain natriuretic peptide (NT-proBNP) marker value. All children underwent OCTA with evaluation of the foveal avascular zone (FAZ), whole superficial vessel density (wsVD), foveal superficial vessel density (fsVD), parafoveal superficial vessel density (psVD), whole deep vessel density (wdVD), foveal deep vessel density (fdVD), parafoveal deep vessel density (pdVD), whole thickness (WT), foveal thickness (FT), and parafoveal thickness (PFT). (3) Results: Retinal VD in SP was significantly lower in children with CHF as compared to the controls. The following SP parameters in the study group were statistically significantly lower than these same measurements for the control group. Details, with study group findings quantified first, include wsVD (46.2% vs. 49.83%, p < 0.05), fsVD (18.07% vs. 24.15%, p < 0.05), and psVD (49.24% vs. 52.51%, p < 0.05). The WT (311.03 micrometers (μm) vs. 323.55 μm, p < 0.05), FT (244.57 μm vs. 256.98 μm, p < 0.05), and PFT (320.63 μm vs. 332.02 μm, p < 0.05). No significant differences in DP retinal VD were found between the two groups. No statistically significant differences in the FAZ were found. The fsVD and FT were correlated with biometry and the age of the study participants. There was a correlation between FAZ and FT (p < 0.001). There were no correlations between retinal VD in both plexuses and refractive error, sex, NT-proBNP, and LVEF. (4) Conclusions: In children with CHF in the course of DCM as compared to the control group, significantly decreased retinal VD in SP was observed. The results of our study indicate that measurements of the OCTA may be a useful diagnostic method in children with chronic heart failure, but it is necessary to conduct further studies in larger groups of participants and long-term observation of these patients.
Collapse
|
30
|
Müller D, Klamt T, Gentemann L, Heisterkamp A, Kalies SMK. Evaluation of laser induced sarcomere micro-damage: Role of damage extent and location in cardiomyocytes. PLoS One 2021; 16:e0252346. [PMID: 34086732 PMCID: PMC8177425 DOI: 10.1371/journal.pone.0252346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/12/2021] [Indexed: 11/18/2022] Open
Abstract
Whereas it is evident that a well aligned and regular sarcomeric structure in cardiomyocytes is vital for heart function, considerably less is known about the contribution of individual elements to the mechanics of the entire cell. For instance, it is unclear whether altered Z-disc elements are the reason or the outcome of related cardiomyopathies. Therefore, it is crucial to gain more insight into this cellular organization. This study utilizes femtosecond laser-based nanosurgery to better understand sarcomeres and their repair upon damage. We investigated the influence of the extent and the location of the Z-disc damage. A single, three, five or ten Z-disc ablations were performed in neonatal rat cardiomyocytes. We employed image-based analysis using a self-written software together with different already published algorithms. We observed that cardiomyocyte survival associated with the damage extent, but not with the cell area or the total number of Z-discs per cell. The cell survival is independent of the damage position and can be compensated. However, the sarcomere alignment/orientation is changing over time after ablation. The contraction time is also independent of the extent of damage for the tested parameters. Additionally, we observed shortening rates between 6–7% of the initial sarcomere length in laser treated cardiomyocytes. This rate is an important indicator for force generation in myocytes. In conclusion, femtosecond laser-based nanosurgery together with image-based sarcomere tracking is a powerful tool to better understand the Z-disc complex and its force propagation function and role in cellular mechanisms.
Collapse
Affiliation(s)
- Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
- * E-mail:
| | - Thorben Klamt
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Lara Gentemann
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Stefan Michael Klaus Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
31
|
Rakusiewicz K, Kanigowska K, Hautz W, Ziółkowska L. Choroidal thickness changes in children with chronic heart failure due to dilated cardiomyopathy. Int Ophthalmol 2021; 41:2167-2177. [PMID: 33966146 PMCID: PMC8172512 DOI: 10.1007/s10792-021-01774-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/05/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE To evaluate choroidal thickness (CTh) in children with chronic heart failure (CHF) secondary to dilated cardiomyopathy (DCM) using spectral domain optical coherence tomography (SD-OCT) and to compare their values to those of healthy children. METHODS Sixty eyes of thirty children (mean age 9.9 ± 3.57 years) with chronic heart failure (left ventricular ejection fraction, LVEF ≤ 55%) due to DCM lasting for over 6 months were prospectively enrolled. The control group consisted of 30 age- (mean age 10.16 ± 3.42 years) and sex-matched healthy children. All participants underwent transthoracic echocardiography with LVEF measured using the Simpson method and had the blood serum level of N-terminal-pro-brain natriuretic peptide marker (NT-proBNP) determined. All children underwent SD-OCT and had subfoveal choroidal thickness (SFCTh) and CTh measured at 1500 µm (μm) nasally, temporally, superiorly and inferiorly from the fovea in both eyes by two investigators. RESULTS CTh at all locations was statistically significantly lower in children with DCM compared to the control group. Mean CTh in the group with CHF compared to the control group were (304.03 vs. 369.72 μm, p < 0.05) at the subfoveal location, (245.87 vs. 284 μm, p < 0.05) 1500 μm nasally from the fovea, (291.5 vs. 355.95 μm, p < 0.05) 1500 μm temporally from the fovea, (303.98 vs. 357.58 μm, p < 0.05) 1500 μm superiorly from the fovea and (290.92 vs. 344.96 μm, p < 0.05) 1500 μm inferiorly from the fovea. The average difference CTh between the study groups ranged from 38.13 to 65.69 μm at individual locations. In both groups, CTh was the thickest at subfoveal location (304.03 vs. 369.72 μm, p < 0.05) and the thinnest was 1500 μm nasally from the fovea (262.37 vs. 336.87 μm, p < 0.05). There was no correlation between CTh and age, gender, biometry and refractive error. No correlation was found between CTh and LVEF and NT-proBNP. CONCLUSION Patients with CHF due to DCM had a thinner CTh at all measured locations. The results of our research indicate that CHF affects CTh and this parameter may be very helpful in monitoring the clinical course of the disease in children with DCM.
Collapse
Affiliation(s)
- Klaudia Rakusiewicz
- Department of Ophthalmology, Children's Memorial Health Institute, Warsaw, Poland.
| | - Krystyna Kanigowska
- Department of Ophthalmology, Children's Memorial Health Institute, Warsaw, Poland
| | - Wojciech Hautz
- Department of Ophthalmology, Children's Memorial Health Institute, Warsaw, Poland
| | - Lidia Ziółkowska
- Department of Cardiology, Children's Memorial Health Institute, Warsaw, Poland
| |
Collapse
|
32
|
Sun K, Li YY, Jin J. A double-edged sword of immuno-microenvironment in cardiac homeostasis and injury repair. Signal Transduct Target Ther 2021; 6:79. [PMID: 33612829 PMCID: PMC7897720 DOI: 10.1038/s41392-020-00455-6] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/14/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023] Open
Abstract
The response of immune cells in cardiac injury is divided into three continuous phases: inflammation, proliferation and maturation. The kinetics of the inflammatory and proliferation phases directly influence the tissue repair. In cardiac homeostasis, cardiac tissue resident macrophages (cTMs) phagocytose bacteria and apoptotic cells. Meanwhile, NK cells prevent the maturation and transport of inflammatory cells. After cardiac injury, cTMs phagocytose the dead cardiomyocytes (CMs), regulate the proliferation and angiogenesis of cardiac progenitor cells. NK cells prevent the cardiac fibrosis, and promote vascularization and angiogenesis. Type 1 macrophages trigger the cardioprotective responses and promote tissue fibrosis in the early stage. Reversely, type 2 macrophages promote cardiac remodeling and angiogenesis in the late stage. Circulating macrophages and neutrophils firstly lead to chronic inflammation by secreting proinflammatory cytokines, and then release anti-inflammatory cytokines and growth factors, which regulate cardiac remodeling. In this process, dendritic cells (DCs) mediate the regulation of monocyte and macrophage recruitment. Recruited eosinophils and Mast cells (MCs) release some mediators which contribute to coronary vasoconstriction, leukocyte recruitment, formation of new blood vessels, scar formation. In adaptive immunity, effector T cells, especially Th17 cells, lead to the pathogenesis of cardiac fibrosis, including the distal fibrosis and scar formation. CMs protectors, Treg cells, inhibit reduce the inflammatory response, then directly trigger the regeneration of local progenitor cell via IL-10. B cells reduce myocardial injury by preserving cardiac function during the resolution of inflammation.
Collapse
Affiliation(s)
- Kang Sun
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Yi-Yuan Li
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Jin Jin
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
- Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
33
|
Piroddi N, Pesce P, Scellini B, Manzini S, Ganzetti GS, Badi I, Menegollo M, Cora V, Tiso S, Cinquetti R, Monti L, Chiesa G, Bleyl SB, Busnelli M, Dellera F, Bruno D, Caicci F, Grimaldi A, Taramelli R, Manni L, Sacerdoti D, Tesi C, Poggesi C, Ausoni S, Acquati F, Campione M. Myocardial overexpression of ANKRD1 causes sinus venosus defects and progressive diastolic dysfunction. Cardiovasc Res 2021; 116:1458-1472. [PMID: 31688894 DOI: 10.1093/cvr/cvz291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 09/26/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
AIMS Increased Ankyrin Repeat Domain 1 (ANKRD1) levels linked to gain of function mutations have been associated to total anomalous pulmonary venous return and adult cardiomyopathy occurrence in humans. The link between increased ANKRD1 level and cardiac structural and functional disease is not understood. To get insight into this problem, we have generated a gain of function ANKRD1 mouse model by overexpressing ANKRD1 in the myocardium. METHODS AND RESULTS Ankrd1 is expressed non-homogeneously in the embryonic myocardium, with a dynamic nucleo-sarcomeric localization in developing cardiomyocytes. ANKRD1 transgenic mice present sinus venosus defect, which originates during development by impaired remodelling of early embryonic heart. Adult transgenic hearts develop diastolic dysfunction with preserved ejection fraction, which progressively evolves into heart failure, as shown histologically and haemodynamically. Transgenic cardiomyocyte structure, sarcomeric assembly, and stability are progressively impaired from embryonic to adult life. Postnatal transgenic myofibrils also present characteristic functional alterations: impaired compliance at neonatal stage and impaired lusitropism in adult hearts. Altogether, our combined analyses suggest that impaired embryonic remodelling and adult heart dysfunction in ANKRD1 transgenic mice present a common ground of initial cardiomyocyte defects, which are exacerbated postnatally. Molecular analysis showed transient activation of GATA4-Nkx2.5 transcription in early transgenic embryos and subsequent dynamic transcriptional modulation within titin gene. CONCLUSIONS ANKRD1 is a fine mediator of cardiomyocyte response to haemodynamic load in the developing and adult heart. Increased ANKRD1 levels are sufficient to initiate an altered cellular phenotype, which is progressively exacerbated into a pathological organ response by the high ventricular workload during postnatal life. Our study defines for the first time a unifying picture for ANKRD1 role in heart development and disease and provides the first mechanistic link between ANKRD1 overexpression and cardiac disease onset.
Collapse
Affiliation(s)
- Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Paola Pesce
- Department of Medicine, University of Padua, 35121 Padua, Italy
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Giulia S Ganzetti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Ileana Badi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy.,Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Michela Menegollo
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Virginia Cora
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Simone Tiso
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Raffaella Cinquetti
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Laura Monti
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Steven B Bleyl
- Department of Pediatrics, University of Utah, Salt Lake City, 84132 UT, USA
| | - Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Federica Dellera
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Daniele Bruno
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Federico Caicci
- Department of Biology, University of Padua, 35121 Padua, Italy
| | - Annalisa Grimaldi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Roberto Taramelli
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Lucia Manni
- Department of Biology, University of Padua, 35121 Padua, Italy
| | - David Sacerdoti
- Department of Medicine, University of Padua, 35121 Padua, Italy
| | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Simonetta Ausoni
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Francesco Acquati
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Marina Campione
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy.,CNR-Neuroscience Institute, 35121 Padua, Italy
| |
Collapse
|
34
|
Yadav S, Ta HT, Nguyen N. Mechanobiology in cardiology: Micro‐ and nanotechnologies to probe mechanosignaling. VIEW 2021. [DOI: 10.1002/viw.20200080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Sharda Yadav
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
| | - Hang T. Ta
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
- School of Environment and Science Griffith University Nathan Queensland Australia
| | - Nam‐Trung Nguyen
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
| |
Collapse
|
35
|
Liu R, Li D, Sun F, Rampoldi A, Maxwell JT, Wu R, Fischbach P, Castellino SM, Du Y, Fu H, Mandawat A, Xu C. Melphalan induces cardiotoxicity through oxidative stress in cardiomyocytes derived from human induced pluripotent stem cells. Stem Cell Res Ther 2020; 11:470. [PMID: 33153480 PMCID: PMC7643439 DOI: 10.1186/s13287-020-01984-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/20/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Treatment-induced cardiotoxicity is a leading noncancer-related cause of acute and late onset morbidity and mortality in cancer patients on antineoplastic drugs such as melphalan-increasing clinical case reports have documented that it could induce cardiotoxicity including severe arrhythmias and heart failure. As the mechanism by which melphalan impairs cardiac cells remains poorly understood, here, we aimed to use cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) to investigate the cellular and molecular mechanisms of melphalan-induced cardiotoxicity. METHODS hiPSC-CMs were generated and treated with clinically relevant doses of melphalan. To characterize melphalan-induced cardiotoxicity, cell viability and apoptosis were quantified at various treatment durations. Ca2+ transient and contractility analyses were used to examine the alterations of hiPSC-CM function. Proteomic analysis, reactive oxygen species detection, and RNA-Sequencing were conducted to investigate underlying mechanisms. RESULTS Melphalan treatment of hiPSC-CMs induced oxidative stress, caused Ca2+ handling defects and dysfunctional contractility, altered global transcriptomic and proteomic profiles, and resulted in apoptosis and cell death. The antioxidant N-acetyl-L-cysteine attenuated these genomic, cellular, and functional alterations. In addition, several other signaling pathways including the p53 and transforming growth factor-β signaling pathways were also implicated in melphalan-induced cardiotoxicity according to the proteomic and transcriptomic analyses. CONCLUSIONS Melphalan induces cardiotoxicity through the oxidative stress pathway. This study provides a unique resource of the global transcriptomic and proteomic datasets for melphalan-induced cardiotoxicity and can potentially open up new clinical mechanism-based targets to prevent and treat melphalan-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rui Liu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Dong Li
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Fangxu Sun
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Antonio Rampoldi
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Joshua T Maxwell
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Peter Fischbach
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Sharon M Castellino
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Yuhong Du
- Emory Chemical Biology Discovery Center and the Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haian Fu
- Emory Chemical Biology Discovery Center and the Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Anant Mandawat
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Cardio-Oncology Program, Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
36
|
Vikhorev PG, Vikhoreva NN, Yeung W, Li A, Lal S, dos Remedios CG, Blair CA, Guglin M, Campbell KS, Yacoub MH, de Tombe P, Marston SB. Titin-truncating mutations associated with dilated cardiomyopathy alter length-dependent activation and its modulation via phosphorylation. Cardiovasc Res 2020; 118:241-253. [PMID: 33135063 PMCID: PMC8752363 DOI: 10.1093/cvr/cvaa316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
Abstract
Aims
Dilated cardiomyopathy (DCM) is associated with mutations in many genes encoding sarcomere proteins. Truncating mutations in the titin gene TTN are the most frequent. Proteomic and functional characterizations are required to elucidate the origin of the disease and the pathogenic mechanisms of TTN-truncating variants.
Methods and results
We isolated myofibrils from DCM hearts carrying truncating TTN mutations and measured the Ca2+ sensitivity of force and its length dependence. Simultaneous measurement of force and adenosine triphosphate (ATP) consumption in skinned cardiomyocytes was also performed. Phosphorylation levels of troponin I (TnI) and myosin binding protein-C (MyBP-C) were manipulated using protein kinase A and λ phosphatase. mRNA sequencing was employed to overview gene expression profiles. We found that Ca2+ sensitivity of myofibrils carrying TTN mutations was significantly higher than in myofibrils from donor hearts. The length dependence of the Ca2+ sensitivity was absent in DCM myofibrils with TTN-truncating variants. No significant difference was found in the expression level of TTN mRNA between the DCM and donor groups. TTN exon usage and splicing were also similar. However, we identified down-regulation of genes encoding Z-disk proteins, while the atrial-specific regulatory myosin light chain gene, MYL7, was up-regulated in DCM patients with TTN-truncating variants.
Conclusion
Titin-truncating mutations lead to decreased length-dependent activation and increased elasticity of myofibrils. Phosphorylation levels of TnI and MyBP-C seen in the left ventricles are essential for the length-dependent changes in Ca2+ sensitivity in healthy donors, but they are reduced in DCM patients with TTN-truncating variants. A decrease in expression of Z-disk proteins may explain the observed decrease in myofibril passive stiffness and length-dependent activation.
Collapse
Affiliation(s)
- Petr G Vikhorev
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Natalia N Vikhoreva
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK
| | - WaiChun Yeung
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Amy Li
- Department of Pharmacy and Biomedical Sciences, La Trobe University, Bendigo, VIC 3550, Australia
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Cristobal G dos Remedios
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Cheavar A Blair
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Maya Guglin
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Magdi H Yacoub
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Pieter de Tombe
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Steven B Marston
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
37
|
Investigating Ganglion Cell Complex Thickness in Children with Chronic Heart Failure due to Dilated Cardiomyopathy. J Clin Med 2020; 9:jcm9092882. [PMID: 32906583 PMCID: PMC7563704 DOI: 10.3390/jcm9092882] [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: 07/27/2020] [Revised: 08/23/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To assess ganglion cell complex (GCC) thickness in children with chronic heart failure (CHF) due to dilated cardiomyopathy (DCM) using optical coherence tomography (OCT). METHODS Sixty eyes of 30 patients with chronic heart failure (CHF) due to dilated cardiomyopathy (DCM) and 60 eyes of 30 age- and sex-matched healthy volunteers (control group) were enrolled. The mean age of the patients and controls was 9.9 ± 3.57 (range 5-17) years and 10.08 ± 3.41 (range 4-16) years, respectively. All patients underwent a complete ophthalmic assessment and OCT imaging using RTVue XR Avanti (Optovue). The following OCT-based parameters were analysed: average ganglion cell complex thickness (avgGCC), superior ganglion cell complex thickness (supGCC), inferior ganglion cell complex thickness (infGCC), global loss of volume (GLV) and focal loss of volume (FLV). RESULTS There were no significant differences in avgGCC (98.13 μm vs. 99.96 μm, p = 0.21), supGCC (97.17 μm vs. 99.29 μm, p = 0.13), infGCC (99.03 μm vs. 100.71 μm, p = 0.25), FVL (0.49% vs. 0.4%, p = 0.25) and GVL (2.1% vs. 1.3%, p = 0.09) between patients with chronic heart failure due to dilated cardiomyopathy and healthy children. There was no correlation between avgGCC, supGCC, infGCC, FLV, GLV and ocular biometry, refractive errors or age. There was no correlation between avgGCC, supGCC, infGCC, FLV, GLV and NT-proBNP or LVEF. There were no significant differences in the studied parameters between the sexes. There were no significant differences in the studied parameters between the left and right eye. CONCLUSION Our study seems to be the first to analyse ganglion cell complex in paediatric patients with dilated cardiomyopathy. We have demonstrated no changes in the ganglion cell complex thickness parameters in children with chronic heart failure due dilated cardiomyopathy, as compared to their healthy peers.
Collapse
|
38
|
Cassani M, Fernandes S, Vrbsky J, Ergir E, Cavalieri F, Forte G. Combining Nanomaterials and Developmental Pathways to Design New Treatments for Cardiac Regeneration: The Pulsing Heart of Advanced Therapies. Front Bioeng Biotechnol 2020; 8:323. [PMID: 32391340 PMCID: PMC7193099 DOI: 10.3389/fbioe.2020.00323] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
The research for heart therapies is challenged by the limited intrinsic regenerative capacity of the adult heart. Moreover, it has been hampered by the poor results obtained by tissue engineering and regenerative medicine attempts at generating functional beating constructs able to integrate with the host tissue. For this reason, organ transplantation remains the elective treatment for end-stage heart failure, while novel strategies aiming to promote cardiac regeneration or repair lag behind. The recent discovery that adult cardiomyocytes can be ectopically induced to enter the cell cycle and proliferate by a combination of microRNAs and cardioprotective drugs, like anti-oxidant, anti-inflammatory, anti-coagulants and anti-platelets agents, fueled the quest for new strategies suited to foster cardiac repair. While proposing a revolutionary approach for heart regeneration, these studies raised serious issues regarding the efficient controlled delivery of the therapeutic cargo, as well as its timely removal or metabolic inactivation from the site of action. Especially, there is need for innovative treatment because of evidence of severe side effects caused by pleiotropic drugs. Biocompatible nanoparticles possess unique physico-chemical properties that have been extensively exploited for overcoming the limitations of standard medical therapies. Researchers have put great efforts into the optimization of the nanoparticles synthesis and functionalization, to control their interactions with the biological milieu and use as a viable alternative to traditional approaches. Nanoparticles can be used for diagnosis and deliver therapies in a personalized and targeted fashion. Regarding the treatment of cardiovascular diseases, nanoparticles-based strategies have provided very promising outcomes, in preclinical studies, during the last years. Efficient encapsulation of a large variety of cargos, specific release at the desired site and improvement of cardiac function are some of the main achievements reached so far by nanoparticle-based treatments in animal models. This work offers an overview on the recent nanomedical applications for cardiac regeneration and highlights how the versatility of nanomaterials can be combined with the newest molecular biology discoveries to advance cardiac regeneration therapies.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Soraia Fernandes
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Jan Vrbsky
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| | - Ece Ergir
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
| | - Francesca Cavalieri
- School of Science, RMIT University, Melbourne, VIC, Australia
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma “Tor Vergata”, Via Della Ricerca Scientifica, Rome, Italy
| | - Giancarlo Forte
- International Clinical Research Center, St Anne’s University Hospital, Brno, Czechia
| |
Collapse
|
39
|
Blair CA, Pruitt BL. Mechanobiology Assays with Applications in Cardiomyocyte Biology and Cardiotoxicity. Adv Healthc Mater 2020; 9:e1901656. [PMID: 32270928 PMCID: PMC7480481 DOI: 10.1002/adhm.201901656] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/19/2022]
Abstract
Cardiomyocytes are the motor units that drive the contraction and relaxation of the heart. Traditionally, testing of drugs for cardiotoxic effects has relied on primary cardiomyocytes from animal models and focused on short-term, electrophysiological, and arrhythmogenic effects. However, primary cardiomyocytes present challenges arising from their limited viability in culture, and tissue from animal models suffers from a mismatch in their physiology to that of human heart muscle. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can address these challenges. They also offer the potential to study not only electrophysiological effects but also changes in cardiomyocyte contractile and mechanical function in response to cardiotoxic drugs. With growing recognition of the long-term cardiotoxic effects of some drugs on subcellular structure and function, there is increasing interest in using hiPSC-CMs for in vitro cardiotoxicity studies. This review provides a brief overview of techniques that can be used to quantify changes in the active force that cardiomyocytes generate and variations in their inherent stiffness in response to cardiotoxic drugs. It concludes by discussing the application of these tools in understanding how cardiotoxic drugs directly impact the mechanobiology of cardiomyocytes and how cardiomyocytes sense and respond to mechanical load at the cellular level.
Collapse
Affiliation(s)
- Cheavar A. Blair
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Beth L. Pruitt
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
40
|
Williams JL, Paudyal A, Awad S, Nicholson J, Grzesik D, Botta J, Meimaridou E, Maharaj AV, Stewart M, Tinker A, Cox RD, Metherell LA. Mylk3 null C57BL/6N mice develop cardiomyopathy, whereas Nnt null C57BL/6J mice do not. Life Sci Alliance 2020; 3:3/4/e201900593. [PMID: 32213617 PMCID: PMC7103425 DOI: 10.26508/lsa.201900593] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/30/2022] Open
Abstract
The C57BL/6J and C57BL/6N mice have well-documented phenotypic and genotypic differences, including the infamous nicotinamide nucleotide transhydrogenase (Nnt) null mutation in the C57BL/6J substrain, which has been linked to cardiovascular traits in mice and cardiomyopathy in humans. To assess whether Nnt loss alone causes a cardiovascular phenotype, we investigated the C57BL/6N, C57BL/6J mice and a C57BL/6J-BAC transgenic rescuing NNT expression, at 3, 12, and 18 mo. We identified a modest dilated cardiomyopathy in the C57BL/6N mice, absent in the two B6J substrains. Immunofluorescent staining of cardiomyocytes revealed eccentric hypertrophy in these mice, with defects in sarcomere organisation. RNAseq analysis identified differential expression of a number of cardiac remodelling genes commonly associated with cardiac disease segregating with the phenotype. Variant calling from RNAseq data identified a myosin light chain kinase 3 (Mylk3) mutation in C57BL/6N mice, which abolishes MYLK3 protein expression. These results indicate the C57BL/6J Nnt-null mice do not develop cardiomyopathy; however, we identified a null mutation in Mylk3 as a credible cause of the cardiomyopathy phenotype in the C57BL/6N.
Collapse
Affiliation(s)
- Jack L Williams
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anju Paudyal
- Medical Research Council Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - Sherine Awad
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dominika Grzesik
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Joaquin Botta
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eirini Meimaridou
- School of Human Sciences, London Metropolitan University, London, UK
| | - Avinaash V Maharaj
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michelle Stewart
- Medical Research Council Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - Andrew Tinker
- William Harvey Heart Centre, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Roger D Cox
- Medical Research Council Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, UK
| | - Lou A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
41
|
Lin YH, Schmidt W, Fritz KS, Jeong MY, Cammarato A, Foster DB, Biesiadecki BJ, McKinsey TA, Woulfe KC. Site-specific acetyl-mimetic modification of cardiac troponin I modulates myofilament relaxation and calcium sensitivity. J Mol Cell Cardiol 2020; 139:135-147. [PMID: 31981571 DOI: 10.1016/j.yjmcc.2020.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Cardiac troponin I (cTnI) is an essential physiological and pathological regulator of cardiac relaxation. Significant to this regulation, the post-translational modification of cTnI through phosphorylation functions as a key mechanism to accelerate myofibril relaxation. Similar to phosphorylation, post-translational modification by acetylation alters amino acid charge and protein function. Recent studies have demonstrated that the acetylation of cardiac myofibril proteins accelerates relaxation and that cTnI is acetylated in the heart. These findings highlight the potential significance of myofilament acetylation; however, it is not known if site-specific acetylation of cTnI can lead to changes in myofilament, myofibril, and/or cellular mechanics. The objective of this study was to determine the effects of mimicking acetylation at a single site of cTnI (lysine-132; K132) on myofilament, myofibril, and cellular mechanics and elucidate its influence on molecular function. METHODS To determine if pseudo-acetylation of cTnI at 132 modulates thin filament regulation of the acto-myosin interaction, we reconstituted thin filaments containing WT or K132Q (to mimic acetylation) cTnI and assessed in vitro motility. To test if mimicking acetylation at K132 alters cellular relaxation, adult rat ventricular cardiomyocytes were infected with adenoviral constructs expressing either cTnI K132Q or K132 replaced with arginine (K132R; to prevent acetylation) and cell shortening and isolated myofibril mechanics were measured. Finally, to confirm that changes in cell shortening and myofibril mechanics were directly due to pseudo-acetylation of cTnI at K132, we exchanged troponin containing WT or K132Q cTnI into isolated myofibrils and measured myofibril mechanical properties. RESULTS Reconstituted thin filaments containing K132Q cTnI exhibited decreased calcium sensitivity compared to thin filaments reconstituted with WT cTnI. Cardiomyocytes expressing K132Q cTnI had faster relengthening and myofibrils isolated from these cells had faster relaxation along with decreased calcium sensitivity compared to cardiomyocytes expressing WT or K132R cTnI. Myofibrils exchanged with K132Q cTnI ex vivo demonstrated faster relaxation and decreased calcium sensitivity. CONCLUSIONS Our results indicate for the first time that mimicking acetylation of a specific cTnI lysine accelerates myofilament, myofibril, and myocyte relaxation. This work underscores the importance of understanding how acetylation of specific sarcomeric proteins affects cardiac homeostasis and disease and suggests that modulation of myofilament lysine acetylation may represent a novel therapeutic target to alter cardiac relaxation.
Collapse
Affiliation(s)
- Ying H Lin
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - William Schmidt
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Kristofer S Fritz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Mark Y Jeong
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Anthony Cammarato
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - D Brian Foster
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Brandon J Biesiadecki
- Department of Physiology and Cell Biology, The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States of America
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America; Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America.
| | - Kathleen C Woulfe
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America.
| |
Collapse
|
42
|
Tsukamoto O. Direct Sarcomere Modulators Are Promising New Treatments for Cardiomyopathies. Int J Mol Sci 2019; 21:ijms21010226. [PMID: 31905684 PMCID: PMC6982115 DOI: 10.3390/ijms21010226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/24/2019] [Accepted: 12/25/2019] [Indexed: 01/10/2023] Open
Abstract
Mutations in sarcomere genes can cause both hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). However, the complex genotype-phenotype relationships in pathophysiology of cardiomyopathies by gene or mutation location are not fully understood. In addition, it is still unclear how mutations within same molecule result in different clinical phenotypes such as HCM and DCM. To clarify how the initial functional insult caused by a subtle change in one protein component of the sarcomere with a given mutation is critical for the development of proper effective treatments for cardiomyopathies. Fortunately, recent technological advances and the development of direct sarcomere modulators have provided a more detailed understanding of the molecular mechanisms that govern the effects of specific mutations. The direct inhibition of sarcomere contractility may be able to suppress the development and progression of HCM with hypercontractile mutations and improve clinical parameters in patients with HCM. On the other hand, direct activation of sarcomere contractility appears to exert unexpected beneficial effects such as reverse remodeling and lower heart rate without increasing adverse cardiovascular events in patients with systolic heart failure due to DCM. Direct sarcomere modulators that can positively influence the natural history of cardiomyopathies represent promising treatment options.
Collapse
Affiliation(s)
- Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan; ; Tel.: +81-6-6879-3492
- Department of Medical Biochemistry, Graduate School of Frontier Bioscience, Osaka University, 1-1 Yamadaoka, Suita 565-0871, Japan
| |
Collapse
|
43
|
Special Issue: The Actin-Myosin Interaction in Muscle: Background and Overview. Int J Mol Sci 2019; 20:ijms20225715. [PMID: 31739584 PMCID: PMC6887992 DOI: 10.3390/ijms20225715] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022] Open
Abstract
Muscular contraction is a fundamental phenomenon in all animals; without it life as we know it would be impossible. The basic mechanism in muscle, including heart muscle, involves the interaction of the protein filaments myosin and actin. Motility in all cells is also partly based on similar interactions of actin filaments with non-muscle myosins. Early studies of muscle contraction have informed later studies of these cellular actin-myosin systems. In muscles, projections on the myosin filaments, the so-called myosin heads or cross-bridges, interact with the nearby actin filaments and, in a mechanism powered by ATP-hydrolysis, they move the actin filaments past them in a kind of cyclic rowing action to produce the macroscopic muscular movements of which we are all aware. In this special issue the papers and reviews address different aspects of the actin-myosin interaction in muscle as studied by a plethora of complementary techniques. The present overview provides a brief and elementary introduction to muscle structure and function and the techniques used to study it. It goes on to give more detailed descriptions of what is known about muscle components and the cross-bridge cycle using structural biology techniques, particularly protein crystallography, electron microscopy and X-ray diffraction. It then has a quick look at muscle mechanics and it summarises what can be learnt about how muscle works based on the other studies covered in the different papers in the special issue. A picture emerges of the main molecular steps involved in the force-producing process; steps that are also likely to be seen in non-muscle myosin interactions with cellular actin filaments. Finally, the remarkable advances made in studying the effects of mutations in the contractile assembly in causing specific muscle diseases, particularly those in heart muscle, are outlined and discussed.
Collapse
|
44
|
Lin YH, Yap J, Ramachandra CJ, Hausenloy DJ. New insights provided by myofibril mechanics in inherited cardiomyopathies. CONDITIONING MEDICINE 2019; 2:213-224. [PMID: 32133438 PMCID: PMC7055865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cardiomyopathies represent a heterogeneous group of cardiac disorders that perturb cardiac contraction and/or relaxation, and can result in arrhythmias, heart failure, and sudden cardiac death. Based on morphological and functional differences, cardiomyopathies have been classified into hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), and restrictive cardiomyopathy (RCM). It has been well documented that mutations in genes encoding sarcomeric proteins are associated with the onset of inherited cardiomyopathies. However, correlating patient genotype to the clinical phenotype has been challenging because of the complex genetic backgrounds, environmental influences, and lifestyles of individuals. Thus, "scaling down" the focus to the basic contractile unit of heart muscle using isolated single myofibril function techniques is of great importance and may be used to understand the molecular basis of disease-causing sarcomeric mutations. Single myofibril bundles harvested from diseased human or experimental animal hearts, as well as cultured adult cardiomyocytes or human cardiomyocytes derived from induced pluripotent stem cells, can be used, thereby providing an ideal multi-level, cross-species platform to dissect sarcomeric function in cardiomyopathies. Here, we will review the myofibril function technique, and discuss alterations in myofibril mechanics, which are known to occur in sarcomeric genetic mutations linked to inherited HCM, DCM, and RCM, and describe the therapeutic potential for future target identification.
Collapse
Affiliation(s)
- Ying-Hsi Lin
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, USA
| | - Chrishan J.A. Ramachandra
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Derek J. Hausenloy
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
- The National Institute of Health Research University College London Hospitals
- Biomedical Research Centre, Research & Development, London, UK
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico
| |
Collapse
|
45
|
Role of Inflammatory Cell Subtypes in Heart Failure. J Immunol Res 2019; 2019:2164017. [PMID: 31565659 PMCID: PMC6745095 DOI: 10.1155/2019/2164017] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 07/25/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammation is a well-known feature of heart failure. Studies have shown that while some inflammation is required for repair during injury and is protective, prolonged inflammation leads to myocardial remodeling and apoptosis of cardiac myocytes. Various types of immune cells are implicated in myocardial inflammation and include neutrophils, macrophages, eosinophils, mast cells, natural killer cells, T cells, and B cells. Recent clinical trials have targeted inflammatory cascades as therapy for heart failure with limited success. A better understanding of the temporal course of the infiltration of the different immune cells and their contribution to the inflammatory process may improve the success for therapy. This brief review outlines the major cell types involved in heart failure, and some of their actions are summarized in the supplementary figure.
Collapse
|
46
|
Mosqueira D, Smith JGW, Bhagwan JR, Denning C. Modeling Hypertrophic Cardiomyopathy: Mechanistic Insights and Pharmacological Intervention. Trends Mol Med 2019; 25:775-790. [PMID: 31324451 DOI: 10.1016/j.molmed.2019.06.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/12/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a prevalent and complex cardiovascular disease where cardiac dysfunction often associates with mutations in sarcomeric genes. Various models based on tissue explants, isolated cardiomyocytes, skinned myofibrils, and purified actin/myosin preparations have uncovered disease hallmarks, enabling the development of putative therapeutics, with some reaching clinical trials. Newly developed human pluripotent stem cell (hPSC)-based models could be complementary by overcoming some of the inconsistencies of earlier systems, whilst challenging and/or clarifying previous findings. In this article we compare recent progress in unveiling multiple HCM mechanisms in different models, highlighting similarities and discrepancies. We explore how insight is facilitating the design of new HCM therapeutics, including those that regulate metabolism, contraction and heart rhythm, providing a future perspective for treatment of HCM.
Collapse
Affiliation(s)
- Diogo Mosqueira
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK.
| | - James G W Smith
- Faculty of Medicine and Health Sciences, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Jamie R Bhagwan
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Chris Denning
- Department of Stem Cell Biology, Centre of Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
47
|
Li CJ, Chen CS, Yiang GT, Tsai APY, Liao WT, Wu MY. Advanced Evolution of Pathogenesis Concepts in Cardiomyopathies. J Clin Med 2019; 8:jcm8040520. [PMID: 30995779 PMCID: PMC6518034 DOI: 10.3390/jcm8040520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/12/2019] [Accepted: 04/12/2019] [Indexed: 12/15/2022] Open
Abstract
Cardiomyopathy is a group of heterogeneous cardiac diseases that impair systolic and diastolic function, and can induce chronic heart failure and sudden cardiac death. Cardiomyopathy is prevalent in the general population, with high morbidity and mortality rates, and contributes to nearly 20% of sudden cardiac deaths in younger individuals. Genetic mutations associated with cardiomyopathy play a key role in disease formation, especially the mutation of sarcomere encoding genes and ATP kinase genes, such as titin, lamin A/C, myosin heavy chain 7, and troponin T1. Pathogenesis of cardiomyopathy occurs by multiple complex steps involving several pathways, including the Ras-Raf-mitogen-activated protein kinase-extracellular signal-activated kinase pathway, G-protein signaling, mechanotransduction pathway, and protein kinase B/phosphoinositide 3-kinase signaling. Excess biomechanical stress induces apoptosis signaling in cardiomyocytes, leading to cell loss, which can induce myocardial fibrosis and remodeling. The clinical features and pathophysiology of cardiomyopathy are discussed. Although several basic and clinical studies have investigated the mechanism of cardiomyopathy, the detailed pathophysiology remains unclear. This review summarizes current concepts and focuses on the molecular mechanisms of cardiomyopathy, especially in the signaling from mutation to clinical phenotype, with the aim of informing the development of therapeutic interventions.
Collapse
Affiliation(s)
- Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Chien-Sheng Chen
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Andy Po-Yi Tsai
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan.
| | - Wan-Ting Liao
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan.
- Chinese Medicine Department, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| |
Collapse
|