1
|
Joyce W, He K, Zhang M, Ogunsola S, Wu X, Joseph KT, Bogomolny D, Yu W, Springer MS, Xie J, Signore AV, Campbell KL. Genetic excision of the regulatory cardiac troponin I extension in high-heart rate mammal clades. Science 2024; 385:1466-1471. [PMID: 39325895 DOI: 10.1126/science.adi8146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 01/18/2024] [Accepted: 07/17/2024] [Indexed: 09/28/2024]
Abstract
Mammalian cardiac troponin I (cTnI) contains a highly conserved amino-terminal extension harboring protein kinase A targets [serine-23 and -24 (Ser23/24)] that are phosphorylated during β-adrenergic stimulation to defend diastolic filling by means of an increased cardiomyocyte relaxation rate. In this work, we show that the Ser23/24-encoding exon 3 of TNNI3 was pseudoexonized multiple times in shrews and moles to mimic Ser23/24 phosphorylation without adrenergic stimulation, facilitating the evolution of exceptionally high resting heart rates (~1000 beats per minute). We further reveal alternative exon 3 splicing in distantly related bat families and confirm that both cTnI splice variants are incorporated into cardiac myofibrils. Because exon 3 of human TNNI3 exhibits a relatively low splice strength score, our findings offer an evolutionarily informed strategy to excise this exon to improve diastolic function during heart failure.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology - Zoophysiology, Aarhus University, 8000 Aarhus C, Denmark
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - Kai He
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Mengdie Zhang
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Samuel Ogunsola
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Xini Wu
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Kelvin T Joseph
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - David Bogomolny
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Wenhua Yu
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Mark S Springer
- Department of Evolution, Ecology and Organismal Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Jiuyong Xie
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Anthony V Signore
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Kevin L Campbell
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
2
|
Vogel A, Arnese R, Gudino Carrillo RM, Sehr D, Deszcz L, Bylicki A, Meinhart A, Clausen T. UNC-45 assisted myosin folding depends on a conserved FX 3HY motif implicated in Freeman Sheldon Syndrome. Nat Commun 2024; 15:6272. [PMID: 39054317 PMCID: PMC11272940 DOI: 10.1038/s41467-024-50442-6] [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: 10/24/2023] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Myosin motors are critical for diverse motility functions, ranging from cytokinesis and endocytosis to muscle contraction. The UNC-45 chaperone controls myosin function mediating the folding, assembly, and degradation of the muscle protein. Here, we analyze the molecular mechanism of UNC-45 as a hub in myosin quality control. We show that UNC-45 forms discrete complexes with folded and unfolded myosin, forwarding them to downstream chaperones and E3 ligases. Structural analysis of a minimal chaperone:substrate complex reveals that UNC-45 binds to a conserved FX3HY motif in the myosin motor domain. Disrupting the observed interface by mutagenesis prevents myosin maturation leading to protein aggregation in vivo. We also show that a mutation in the FX3HY motif linked to the Freeman Sheldon Syndrome impairs UNC-45 assisted folding, reducing the level of functional myosin. These findings demonstrate that a faulty myosin quality control is a critical yet unexplored cause of human myopathies.
Collapse
Affiliation(s)
- Antonia Vogel
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Renato Arnese
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Ricardo M Gudino Carrillo
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
- Medical University, Vienna, Austria
| | - Daria Sehr
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Luiza Deszcz
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Andrzej Bylicki
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Anton Meinhart
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Tim Clausen
- Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria.
- Vienna BioCenter Core Facilities, Vienna, Austria.
| |
Collapse
|
3
|
López-Dávila AJ, Lomonte B, Gutiérrez JM. Alterations of the skeletal muscle contractile apparatus in necrosis induced by myotoxic snake venom phospholipases A 2: a mini-review. J Muscle Res Cell Motil 2024; 45:69-77. [PMID: 38063951 PMCID: PMC11096208 DOI: 10.1007/s10974-023-09662-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/07/2023] [Indexed: 05/16/2024]
Abstract
Skeletal muscle necrosis is a common clinical manifestation of snakebite envenoming. The predominant myotoxic components in snake venoms are catalytically-active phospholipases A2 (PLA2) and PLA2 homologs devoid of enzymatic activity, which have been used as models to investigate various aspects of muscle degeneration. This review addresses the changes in the contractile apparatus of skeletal muscle induced by these toxins. Myotoxic components initially disrupt the integrity of sarcolemma, generating a calcium influx that causes various degenerative events, including hypercontraction of myofilaments. There is removal of specific sarcomeric proteins, owing to the hydrolytic action of muscle calpains and proteinases from invading inflammatory cells, causing an initial redistribution followed by widespread degradation of myofibrillar material. Experiments using skinned cardiomyocytes and skeletal muscle fibers show that these myotoxins do not directly affect the contractile apparatus, implying that hypercontraction is due to cytosolic calcium increase secondary to sarcolemmal damage. Such drastic hypercontraction may contribute to muscle damage by generating mechanical stress and further sarcolemmal damage.
Collapse
Affiliation(s)
- Alfredo Jesús López-Dávila
- Institute of Molecular and Cell Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| |
Collapse
|
4
|
Collibee SE, Romero A, Muci AR, Hwee DT, Chuang C, Hartman JJ, Motani AS, Ashcraft L, DeRosier A, Grillo M, Lu Q, Malik FI, Morgan BP. Cardiac Troponin Activator CK-963 Increases Cardiac Contractility in Rats. J Med Chem 2024; 67:7859-7869. [PMID: 38451215 PMCID: PMC11129196 DOI: 10.1021/acs.jmedchem.3c02412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
Novel cardiac troponin activators were identified using a high throughput cardiac myofibril ATPase assay and confirmed using a series of biochemical and biophysical assays. HTS hit 2 increased rat cardiomyocyte fractional shortening without increasing intracellular calcium concentrations, and the biological target of 1 and 2 was determined to be the cardiac thin filament. Subsequent optimization to increase solubility and remove PDE-3 inhibition led to the discovery of CK-963 and enabled pharmacological evaluation of cardiac troponin activation without the competing effects of PDE-3 inhibition. Rat echocardiography studies using CK-963 demonstrated concentration-dependent increases in cardiac fractional shortening up to 95%. Isothermal calorimetry studies confirmed a direct interaction between CK-963 and a cardiac troponin chimera with a dissociation constant of 11.5 ± 3.2 μM. These results provide evidence that direct activation of cardiac troponin without the confounding effects of PDE-3 inhibition may provide benefit for patients with cardiovascular conditions where contractility is reduced.
Collapse
Affiliation(s)
- Scott E. Collibee
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Antonio Romero
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Alexander R. Muci
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Darren T. Hwee
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Chihyuan Chuang
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - James J. Hartman
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Alykhan S. Motani
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Luke Ashcraft
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Andre DeRosier
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Mark Grillo
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Qing Lu
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Fady I. Malik
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Bradley P. Morgan
- Cytokinetics,
Inc., 350 Oyster Point Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
5
|
Chapman EA, Li BH, Krichel B, Chan HJ, Buck KM, Roberts DS, Ge Y. Native Top-Down Mass Spectrometry for Characterizing Sarcomeric Proteins Directly from Cardiac Tissue Lysate. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:738-745. [PMID: 38422011 PMCID: PMC11098619 DOI: 10.1021/jasms.3c00430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Native top-down mass spectrometry (nTDMS) has emerged as a powerful structural biology tool that can localize post-translational modifications (PTMs), explore ligand-binding interactions, and elucidate the three-dimensional structure of proteins and protein complexes in the gas-phase. Fourier-transform ion cyclotron resonance (FTICR) MS offers distinct capabilities for nTDMS, owing to its ultrahigh resolving power, mass accuracy, and robust fragmentation techniques. Previous nTDMS studies using FTICR have mainly been applied to overexpressed recombinant proteins and protein complexes. Here, we report the first nTDMS study that directly analyzes human heart tissue lysate by direct infusion FTICR MS without prior chromatographic separation strategies. We have achieved comprehensive nTDMS characterization of cardiac contractile proteins that play critical roles in heart contraction and relaxation. Specifically, our results reveal structural insights into ventricular myosin light chain 2 (MLC-2v), ventricular myosin light chain 1 (MLC-1v), and alpha-tropomyosin (α-Tpm) in the sarcomere, the basic contractile unit of cardiac muscle. Furthermore, we verified the calcium (Ca2+) binding domain in MLC-2v. In summary, our nTDMS platform extends the application of FTICR MS to directly characterize the structure, PTMs, and metal-binding of endogenous proteins from heart tissue lysate without prior separation methods.
Collapse
Affiliation(s)
- Emily A. Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Brad H. Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Boris Krichel
- School of Life Sciences, University of Siegen, 57076, Germany
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Hsin-Ju Chan
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Kevin M. Buck
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - David S. Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| |
Collapse
|
6
|
Kondacs L, Parijat P, Cobb AJA, Kampourakis T. Synthesis and Biophysical Characterization of Fingolimod Derivatives as Cardiac Troponin Antagonists. ACS Med Chem Lett 2024; 15:413-417. [PMID: 38505838 PMCID: PMC10945792 DOI: 10.1021/acsmedchemlett.3c00511] [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: 11/13/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 03/21/2024] Open
Abstract
Calcium binding to cardiac troponin C (cTnC) in the thin filaments acts as a trigger for cardiac muscle contraction. The N-lobe of cTnC (NcTnC) undergoes a conformational change in the presence of calcium that allows for interaction with the switch region of cardiac troponin I (cTnISP), releasing its inhibitory effect on the thin filament structure. The small molecule fingolimod inhibits cTnC-cTnISP interactions via electrostatic repulsion between its positively charged tail and positively charged residues in cTnISP and acts as a calcium desensitizer of the contractile myofilaments. Here we investigate the structure-activity relationship of the fingolimod hydrophobic headgroup and show that increasing the alkyl chain length increases both its affinity for NcTnC and its inhibitory effect on the NcTnC-cTnISP interaction and that decreasing flexibility completely abolishes these effects. Strikingly, the longer derivatives have no effect on the calcium affinity of cTnC, suggesting that they act as better inhibitors.
Collapse
Affiliation(s)
- Laszlo Kondacs
- Department
of Chemistry, King’s College London, Britannia House, London SE1 1DB, United Kingdom
| | - Priyanka Parijat
- Randall
Centre for Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s
College London, London SE1 1UL, United Kingdom
| | - Alexander J. A. Cobb
- Department
of Chemistry, King’s College London, Britannia House, London SE1 1DB, United Kingdom
| | - Thomas Kampourakis
- Randall
Centre for Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s
College London, London SE1 1UL, United Kingdom
| |
Collapse
|
7
|
Caudal A, Snyder MP, Wu JC. Harnessing human genetics and stem cells for precision cardiovascular medicine. CELL GENOMICS 2024; 4:100445. [PMID: 38359791 PMCID: PMC10879032 DOI: 10.1016/j.xgen.2023.100445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/22/2023] [Accepted: 10/25/2023] [Indexed: 02/17/2024]
Abstract
Human induced pluripotent stem cell (iPSC) platforms are valuable for biomedical and pharmaceutical research by providing tissue-specific human cells that retain patients' genetic integrity and display disease phenotypes in a dish. Looking forward, combining iPSC phenotyping platforms with genomic and screening technologies will continue to pave new directions for precision medicine, including genetic prediction, visualization, and treatment of heart disease. This review summarizes the recent use of iPSC technology to unpack the influence of genetic variants in cardiovascular pathology. We focus on various state-of-the-art genomic tools for cardiovascular therapies-including the expansion of genetic toolkits for molecular interrogation, in vitro population studies, and function-based drug screening-and their current applications in patient- and genome-edited iPSC platforms that are heralding new avenues for cardiovascular research.
Collapse
Affiliation(s)
- Arianne Caudal
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Greenstone Biosciences, Palo Alto, CA 94304, USA.
| |
Collapse
|
8
|
Rogers HT, Melby JA, Ehlers LE, Fischer MS, Larson EJ, Gao Z, Rossler KJ, Wang D, Alpert AJ, Ge Y. Small-Scale Serial Size Exclusion Chromatography (s 3SEC) for High Sensitivity Top-Down Proteomics of Large Proteoforms. Anal Chem 2024:10.1021/acs.analchem.3c05733. [PMID: 38315630 PMCID: PMC11298573 DOI: 10.1021/acs.analchem.3c05733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Top-down-mass spectrometry (MS)-based proteomics has emerged as a premier technology to examine proteins at the proteoform level, enabling characterization of genetic mutations, alternative splicing, and post-translational modifications. However, significant challenges that remain in top-down proteomics include the analysis of large proteoforms and the sensitivity required to examine proteoforms from minimal amounts of sample. To address these challenges, we have developed a new method termed "small-scale serial Size Exclusion Chromatography" (s3SEC), which incorporates a small-scale protein extraction (1 mg of tissue) and serial SEC without postfractionation sample handling, coupled with online high sensitivity capillary reversed-phase liquid chromatography tandem MS (RPLC-MS/MS) for analysis of large proteoforms. The s3SEC-RPLC-MS/MS method significantly enhanced the sensitivity and reduced the proteome complexity across the fractions, enabling the detection of high MW proteoforms previously undetected in one-dimensional (1D)-RPLC analysis. Importantly, we observed a drastic improvement in the signal intensity of high MW proteoforms in early fractions when using the s3SEC-RPLC method. Moreover, we demonstrate that this s3SEC-RPLC-MS/MS method also allows the analysis of lower MW proteoforms in subsequent fractions without significant alteration in proteoform abundance and equivalent or improved fragmentation efficiency to that of the 1D-RPLC approach. Although this study focuses on the use of cardiac tissue, the s3SEC-RPLC-MS/MS method could be broadly applicable to other systems with limited sample inputs.
Collapse
Affiliation(s)
- Holden T. Rogers
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Jake A. Melby
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Lauren E. Ehlers
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Matthew S. Fischer
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Eli J. Larson
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Zhan Gao
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Kalina J. Rossler
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | | | | | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| |
Collapse
|
9
|
Rodero C, Baptiste TMG, Barrows RK, Lewalle A, Niederer SA, Strocchi M. Advancing clinical translation of cardiac biomechanics models: a comprehensive review, applications and future pathways. FRONTIERS IN PHYSICS 2023; 11:1306210. [PMID: 38500690 PMCID: PMC7615748 DOI: 10.3389/fphy.2023.1306210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Cardiac mechanics models are developed to represent a high level of detail, including refined anatomies, accurate cell mechanics models, and platforms to link microscale physiology to whole-organ function. However, cardiac biomechanics models still have limited clinical translation. In this review, we provide a picture of cardiac mechanics models, focusing on their clinical translation. We review the main experimental and clinical data used in cardiac models, as well as the steps followed in the literature to generate anatomical meshes ready for simulations. We describe the main models in active and passive mechanics and the different lumped parameter models to represent the circulatory system. Lastly, we provide a summary of the state-of-the-art in terms of ventricular, atrial, and four-chamber cardiac biomechanics models. We discuss the steps that may facilitate clinical translation of the biomechanics models we describe. A well-established software to simulate cardiac biomechanics is lacking, with all available platforms involving different levels of documentation, learning curves, accessibility, and cost. Furthermore, there is no regulatory framework that clearly outlines the verification and validation requirements a model has to satisfy in order to be reliably used in applications. Finally, better integration with increasingly rich clinical and/or experimental datasets as well as machine learning techniques to reduce computational costs might increase model reliability at feasible resources. Cardiac biomechanics models provide excellent opportunities to be integrated into clinical workflows, but more refinement and careful validation against clinical data are needed to improve their credibility. In addition, in each context of use, model complexity must be balanced with the associated high computational cost of running these models.
Collapse
Affiliation(s)
- Cristobal Rodero
- Cardiac Electro-Mechanics Research Group (CEMRG), National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Tiffany M. G. Baptiste
- Cardiac Electro-Mechanics Research Group (CEMRG), National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Rosie K. Barrows
- Cardiac Electro-Mechanics Research Group (CEMRG), National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Alexandre Lewalle
- Cardiac Electro-Mechanics Research Group (CEMRG), National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Steven A. Niederer
- Cardiac Electro-Mechanics Research Group (CEMRG), National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
- Turing Research and Innovation Cluster in Digital Twins (TRIC: DT), The Alan Turing Institute, London, United Kingdom
| | - Marina Strocchi
- Cardiac Electro-Mechanics Research Group (CEMRG), National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
10
|
Hitsumoto T, Tsukamoto O, Matsuoka K, Li J, Liu L, Kuramoto Y, Higo S, Ogawa S, Fujino N, Yoshida S, Kioka H, Kato H, Hakui H, Saito Y, Okamoto C, Inoue H, Hyejin J, Ueda K, Segawa T, Nishimura S, Asano Y, Asanuma H, Tani A, Imamura R, Komagawa S, Kanai T, Takamura M, Sakata Y, Kitakaze M, Haruta JI, Takashima S. Restoration of Cardiac Myosin Light Chain Kinase Ameliorates Systolic Dysfunction by Reducing Superrelaxed Myosin. Circulation 2023; 147:1902-1918. [PMID: 37128901 PMCID: PMC10270284 DOI: 10.1161/circulationaha.122.062885] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 04/05/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Cardiac-specific myosin light chain kinase (cMLCK), encoded by MYLK3, regulates cardiac contractility through phosphorylation of ventricular myosin regulatory light chain. However, the pathophysiological and therapeutic implications of cMLCK in human heart failure remain unclear. We aimed to investigate whether cMLCK dysregulation causes cardiac dysfunction and whether the restoration of cMLCK could be a novel myotropic therapy for systolic heart failure. METHODS We generated the knock-in mice (Mylk3+/fs and Mylk3fs/fs) with a familial dilated cardiomyopathy-associated MYLK3 frameshift mutation (MYLK3+/fs) that had been identified previously by us (c.1951-1G>T; p.P639Vfs*15) and the human induced pluripotent stem cell-derived cardiomyocytes from the carrier of the mutation. We also developed a new small-molecule activator of cMLCK (LEUO-1154). RESULTS Both mice (Mylk3+/fs and Mylk3fs/fs) showed reduced cMLCK expression due to nonsense-mediated messenger RNA decay, reduced MLC2v (ventricular myosin regulatory light chain) phosphorylation in the myocardium, and systolic dysfunction in a cMLCK dose-dependent manner. Consistent with this result, myocardium from the mutant mice showed an increased ratio of cardiac superrelaxation/disordered relaxation states that may contribute to impaired cardiac contractility. The phenotypes observed in the knock-in mice were rescued by cMLCK replenishment through the AAV9_MYLK3 vector. Human induced pluripotent stem cell-derived cardiomyocytes with MYLK3+/fs mutation reduced cMLCK expression by 50% and contractile dysfunction, accompanied by an increased superrelaxation/disordered relaxation ratio. CRISPR-mediated gene correction, or cMLCK replenishment by AAV9_MYLK3 vector, successfully recovered cMLCK expression, the superrelaxation/disordered relaxation ratio, and contractile dysfunction. LEUO-1154 increased human cMLCK activity ≈2-fold in the Vmax for ventricular myosin regulatory light chain phosphorylation without affecting the Km. LEUO-1154 treatment of human induced pluripotent stem cell-derived cardiomyocytes with MYLK3+/fs mutation restored the ventricular myosin regulatory light chain phosphorylation level and superrelaxation/disordered relaxation ratio and improved cardiac contractility without affecting calcium transients, indicating that the cMLCK activator acts as a myotrope. Finally, human myocardium from advanced heart failure with a wide variety of causes had a significantly lower MYLK3/PPP1R12B messenger RNA expression ratio than control hearts, suggesting an altered balance between myosin regulatory light chain kinase and phosphatase in the failing myocardium, irrespective of the causes. CONCLUSIONS cMLCK dysregulation contributes to the development of cardiac systolic dysfunction in humans. Our strategy to restore cMLCK activity could form the basis of a novel myotropic therapy for advanced systolic heart failure.
Collapse
Affiliation(s)
- Tatsuro Hitsumoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Ken Matsuoka
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Junjun Li
- Department of Cardiovascular Surgery (J.L., L.L.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Li Liu
- Department of Cardiovascular Surgery (J.L., L.L.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Yuki Kuramoto
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Shuichiro Higo
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Shou Ogawa
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Noboru Fujino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University. Kanazawa, Ishikawa, Japan (N.F., S.Y., M.T.)
| | - Shohei Yoshida
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University. Kanazawa, Ishikawa, Japan (N.F., S.Y., M.T.)
| | - Hidetaka Kioka
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Hideyuki Hakui
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Yuki Saito
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Chisato Okamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Hijiri Inoue
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Jo Hyejin
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Kyoko Ueda
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Takatsugu Segawa
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Shunsuke Nishimura
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Yoshihiro Asano
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
- Department of Genomic Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (Y.A.)
| | - Hiroshi Asanuma
- Department of Internal Medicine, Meiji University of Integrative Medicine, Nantan, Kyoto, Japan (H.A.)
| | - Akiyoshi Tani
- Compound Library Screening Center (A.T.), Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Riyo Imamura
- Drug Discovery Initiative, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan (R.I.)
| | - Shinsuke Komagawa
- Lead Explorating Units (S.K., T.K., J.-i.H.), Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Toshio Kanai
- Lead Explorating Units (S.K., T.K., J.-i.H.), Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University. Kanazawa, Ishikawa, Japan (N.F., S.Y., M.T.)
| | - Yasushi Sakata
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | | | - Jun-ichi Haruta
- Lead Explorating Units (S.K., T.K., J.-i.H.), Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| |
Collapse
|
11
|
Melby JA, Brown KA, Gregorich ZR, Roberts DS, Chapman EA, Ehlers LE, Gao Z, Larson EJ, Jin Y, Lopez JR, Hartung J, Zhu Y, McIlwain SJ, Wang D, Guo W, Diffee GM, Ge Y. High sensitivity top-down proteomics captures single muscle cell heterogeneity in large proteoforms. Proc Natl Acad Sci U S A 2023; 120:e2222081120. [PMID: 37126723 PMCID: PMC10175728 DOI: 10.1073/pnas.2222081120] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/05/2023] [Indexed: 05/03/2023] Open
Abstract
Single-cell proteomics has emerged as a powerful method to characterize cellular phenotypic heterogeneity and the cell-specific functional networks underlying biological processes. However, significant challenges remain in single-cell proteomics for the analysis of proteoforms arising from genetic mutations, alternative splicing, and post-translational modifications. Herein, we have developed a highly sensitive functionally integrated top-down proteomics method for the comprehensive analysis of proteoforms from single cells. We applied this method to single muscle fibers (SMFs) to resolve their heterogeneous functional and proteomic properties at the single-cell level. Notably, we have detected single-cell heterogeneity in large proteoforms (>200 kDa) from the SMFs. Using SMFs obtained from three functionally distinct muscles, we found fiber-to-fiber heterogeneity among the sarcomeric proteoforms which can be related to the functional heterogeneity. Importantly, we detected multiple isoforms of myosin heavy chain (~223 kDa), a motor protein that drives muscle contraction, with high reproducibility to enable the classification of individual fiber types. This study reveals single muscle cell heterogeneity in large proteoforms and establishes a direct relationship between sarcomeric proteoforms and muscle fiber types, highlighting the potential of top-down proteomics for uncovering the molecular underpinnings of cell-to-cell variation in complex systems.
Collapse
Affiliation(s)
- Jake A. Melby
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Kyle A. Brown
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Zachery R. Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI53706
| | - David S. Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Emily A. Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Lauren E. Ehlers
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Zhan Gao
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705
| | - Eli J. Larson
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Yutong Jin
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Justin R. Lopez
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI53706
| | - Jared Hartung
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI53706
| | - Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI53705
| | - Sean J. McIlwain
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI53705
| | | | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI53706
| | - Gary M. Diffee
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI53706
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI53705
| |
Collapse
|
12
|
Parijat P, Ponnam S, Attili S, Campbell KS, El-Mezgueldi M, Pfuhl M, Kampourakis T. Discovery of novel cardiac troponin activators using fluorescence polarization-based high throughput screening assays. Sci Rep 2023; 13:5216. [PMID: 36997544 PMCID: PMC10063609 DOI: 10.1038/s41598-023-32476-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
The large unmet demand for new heart failure therapeutics is widely acknowledged. Over the last decades the contractile myofilaments themselves have emerged as an attractive target for the development of new therapeutics for both systolic and diastolic heart failure. However, the clinical use of myofilament-directed drugs has been limited, and further progress has been hampered by incomplete understanding of myofilament function on the molecular level and screening technologies for small molecules that accurately reproduce this function in vitro. In this study we have designed, validated and characterized new high throughput screening platforms for small molecule effectors targeting the interactions between the troponin C and troponin I subunits of the cardiac troponin complex. Fluorescence polarization-based assays were used to screen commercially available compound libraries, and hits were validated using secondary screens and orthogonal assays. Hit compound-troponin interactions were characterized using isothermal titration calorimetry and NMR spectroscopy. We identified NS5806 as novel calcium sensitizer that stabilizes active troponin. In good agreement, NS5806 greatly increased the calcium sensitivity and maximal isometric force of demembranated human donor myocardium. Our results suggest that sarcomeric protein-directed screening platforms are suitable for the development of compounds that modulate cardiac myofilament function.
Collapse
Affiliation(s)
- Priyanka Parijat
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
- British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, UK
| | - Saraswathi Ponnam
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
- British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, UK
| | - Seetharamaiah Attili
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
- British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, UK
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine and Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Mohammed El-Mezgueldi
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
- British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, UK
| | - Mark Pfuhl
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
- British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, UK
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK.
- British Heart Foundation Centre of Research Excellence, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
13
|
Dvornikov AV, Bunch TA, Lepak VC, Colson BA. Fluorescence lifetime-based assay reports structural changes in cardiac muscle mediated by effectors of contractile regulation. J Gen Physiol 2023; 155:e202113054. [PMID: 36633587 PMCID: PMC9859762 DOI: 10.1085/jgp.202113054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 09/23/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
Cardiac muscle contraction is regulated by Ca2+-induced structural changes of the thin filaments to permit myosin cross-bridge cycling driven by ATP hydrolysis in the sarcomere. In congestive heart failure, contraction is weakened, and thus targeting the contractile proteins of the sarcomere is a promising approach to therapy. However, development of novel therapeutic interventions has been challenging due to a lack of precise discovery tools. We have developed a fluorescence lifetime-based assay using an existing site-directed probe, N,N'-dimethyl-N-(iodoacetyl)-N'-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamine (IANBD) attached to human cardiac troponin C (cTnC) mutant cTnCT53C, exchanged into porcine cardiac myofibrils. We hypothesized that IANBD-cTnCT53C fluorescence lifetime measurements provide insight into the activation state of the thin filament. The sensitivity and precision of detecting structural changes in cTnC due to physiological and therapeutic modulators of thick and thin filament functions were determined. The effects of Ca2+ binding to cTnC and myosin binding to the thin filament were readily detected by this assay in mock high-throughput screen tests using a fluorescence lifetime plate reader. We then evaluated known effectors of altered cTnC-Ca2+ binding, W7 and pimobendan, and myosin-binding drugs, mavacamten and omecamtiv mecarbil, used to treat cardiac diseases. Screening assays were determined to be of high quality as indicated by the Z' factor. We conclude that cTnC lifetime-based probes allow for precise evaluation of the thin filament activation in functioning myofibrils that can be used in future high-throughput screens of small-molecule modulators of function of the thin and thick filaments.
Collapse
Affiliation(s)
- Alexey V. Dvornikov
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Thomas A. Bunch
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Victoria C. Lepak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Brett A. Colson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
14
|
Chapman EA, Aballo TJ, Melby JA, Zhou T, Price SJ, Rossler KJ, Lei I, Tang PC, Ge Y. Defining the Sarcomeric Proteoform Landscape in Ischemic Cardiomyopathy by Top-Down Proteomics. J Proteome Res 2023; 22:931-941. [PMID: 36800490 PMCID: PMC10115148 DOI: 10.1021/acs.jproteome.2c00729] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Ischemic cardiomyopathy (ICM) is a prominent form of heart failure, but the molecular mechanisms underlying ICM remain relatively understudied due to marked phenotypic heterogeneity. Alterations in post-translational modifications (PTMs) and isoform switches in sarcomeric proteins play important roles in cardiac pathophysiology. Thus, it is essential to define sarcomeric proteoform landscape to better understand ICM. Herein, we have implemented a top-down liquid chromatography (LC)-mass spectrometry (MS)-based proteomics method for the identification and quantification of sarcomeric proteoforms in the myocardia of donors without heart diseases (n = 16) compared to end-stage ICM patients (n = 16). Importantly, quantification of post-translational modifications (PTMs) and expression reveal significant changes in various sarcomeric proteins extracted from ICM tissues. Changes include altered phosphorylation and expression of cardiac troponin I (cTnI) and enigma homologue 2 (ENH2) as well as an increase in muscle LIM protein (MLP) and calsarcin-1 (Cal-1) phosphorylation in ICM hearts. Our results imply that the contractile apparatus of the sarcomere is severely dysregulated during ICM. Thus, this is the first study to uncover significant molecular changes to multiple sarcomeric proteins in the LV myocardia of the end-stage ICM patients using liquid chromatography-mass spectrometry (LC-MS)-based top-down proteomics. Raw data are available via the PRIDE repository with identifier PXD038066.
Collapse
Affiliation(s)
- Emily A. Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Timothy J. Aballo
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Jake A. Melby
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Tianhua Zhou
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Scott J. Price
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Kalina J. Rossler
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Paul C. Tang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| |
Collapse
|
15
|
Titin-truncating variants in hiPSC cardiomyocytes induce pathogenic proteinopathy and sarcomere defects with preserved core contractile machinery. Stem Cell Reports 2022; 18:220-236. [PMID: 36525964 PMCID: PMC9860080 DOI: 10.1016/j.stemcr.2022.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 12/23/2022] Open
Abstract
Titin-truncating variants (TTNtv) are the single largest genetic cause of dilated cardiomyopathy (DCM). In this study we modeled disease phenotypes of A-band TTNtv-induced DCM in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using genome editing and tissue engineering technologies. Transcriptomic, cellular, and micro-tissue studies revealed that A-band TTNtv hiPSC-CMs exhibit pathogenic proteinopathy, sarcomere defects, aberrant Na+ channel activities, and contractile dysfunction. These phenotypes establish a dual mechanism of poison peptide effect and haploinsufficiency that collectively contribute to DCM pathogenesis. However, TTNtv cellular defects did not interfere with the function of the core contractile machinery, the actin-myosin-troponin-Ca2+ complex, and preserved the therapeutic mechanism of sarcomere modulators. Treatment of TTNtv cardiac micro-tissues with investigational sarcomere modulators augmented contractility and resulted in sustained transcriptomic changes that promote reversal of DCM disease signatures. Together, our findings elucidate the underlying pathogenic mechanisms of A-band TTNtv-induced DCM and demonstrate the validity of sarcomere modulators as potential therapeutics.
Collapse
|
16
|
Reitz CJ, Rasouli M, Alibhai FJ, Khatua TN, Pyle WG, Martino TA. A brief morning rest period benefits cardiac repair in pressure overload hypertrophy and postmyocardial infarction. JCI Insight 2022; 7:164700. [PMID: 36256456 DOI: 10.1172/jci.insight.164700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/12/2022] [Indexed: 12/15/2022] Open
Abstract
Rest has long been considered beneficial to patient healing; however, remarkably, there are no evidence-based experimental models determining how it benefits disease outcomes. Here, we created an experimental rest model in mice that briefly extends the morning rest period. We found in 2 major cardiovascular disease conditions (cardiac hypertrophy, myocardial infarction) that imposing a short, extended period of morning rest each day limited cardiac remodeling compared with controls. Mechanistically, rest mitigates autonomic-mediated hemodynamic stress on the cardiovascular system, relaxes myofilament contractility, and attenuates cardiac remodeling genes, consistent with the benefits on cardiac structure and function. These same rest-responsive gene pathways underlie the pathophysiology of many major human cardiovascular conditions, as demonstrated by interrogating open-source transcriptomic data; thus, patients with other conditions may also benefit from a morning rest period in a similar manner. Our findings implicate rest as a key driver of physiology, creating a potentially new field - as broad and important as diet, sleep, or exercise - and provide a strong rationale for investigation of rest-based therapy for major clinical diseases.
Collapse
|
17
|
Zhao Y, Ding S, Todoh M. Validate the force-velocity relation of the Hill's muscle model from a molecular perspective. Front Bioeng Biotechnol 2022; 10:1006571. [PMID: 36312549 PMCID: PMC9614041 DOI: 10.3389/fbioe.2022.1006571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/30/2022] [Indexed: 07/30/2023] Open
Affiliation(s)
- Yongkun Zhao
- Division of Human Mechanical Systems and Design, Graduate School of Engineering, Hokkaido University, Sapporo, Japan
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Shihang Ding
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Masahiro Todoh
- Division of Mechanical and Aerospace Engineering, Faculty of Engineering, Hokkaido University, Sapporo, Japan
| |
Collapse
|
18
|
Mahmud Z, Tikunova S, Belevych N, Wagg CS, Zhabyeyev P, Liu PB, Rasicci DV, Yengo CM, Oudit GY, Lopaschuk GD, Reiser PJ, Davis JP, Hwang PM. Small Molecule RPI-194 Stabilizes Activated Troponin to Increase the Calcium Sensitivity of Striated Muscle Contraction. Front Physiol 2022; 13:892979. [PMID: 35755445 PMCID: PMC9213791 DOI: 10.3389/fphys.2022.892979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Small molecule cardiac troponin activators could potentially enhance cardiac muscle contraction in the treatment of systolic heart failure. We designed a small molecule, RPI-194, to bind cardiac/slow skeletal muscle troponin (Cardiac muscle and slow skeletal muscle share a common isoform of the troponin C subunit.) Using solution NMR and stopped flow fluorescence spectroscopy, we determined that RPI-194 binds to cardiac troponin with a dissociation constant KD of 6-24 μM, stabilizing the activated complex between troponin C and the switch region of troponin I. The interaction between RPI-194 and troponin C is weak (KD 311 μM) in the absence of the switch region. RPI-194 acts as a calcium sensitizer, shifting the pCa50 of isometric contraction from 6.28 to 6.99 in mouse slow skeletal muscle fibers and from 5.68 to 5.96 in skinned cardiac trabeculae at 100 μM concentration. There is also some cross-reactivity with fast skeletal muscle fibers (pCa50 increases from 6.27 to 6.52). In the slack test performed on the same skinned skeletal muscle fibers, RPI-194 slowed the velocity of unloaded shortening at saturating calcium concentrations, suggesting that it slows the rate of actin-myosin cross-bridge cycling under these conditions. However, RPI-194 had no effect on the ATPase activity of purified actin-myosin. In isolated unloaded mouse cardiomyocytes, RPI-194 markedly decreased the velocity and amplitude of contractions. In contrast, cardiac function was preserved in mouse isolated perfused working hearts. In summary, the novel troponin activator RPI-194 acts as a calcium sensitizer in all striated muscle types. Surprisingly, it also slows the velocity of unloaded contraction, but the cause and significance of this is uncertain at this time. RPI-194 represents a new class of non-specific troponin activator that could potentially be used either to enhance cardiac muscle contractility in the setting of systolic heart failure or to enhance skeletal muscle contraction in neuromuscular disorders.
Collapse
Affiliation(s)
- Zabed Mahmud
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Svetlana Tikunova
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States
| | - Natalya Belevych
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Cory S Wagg
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Pavel Zhabyeyev
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Philip B Liu
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - David V Rasicci
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, University Park, PA, United States
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, University Park, PA, United States
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Gary D Lopaschuk
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Peter J Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States
| | - Peter M Hwang
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
19
|
Liang D, Chen C, Huang S, Liu S, Fu L, Niu Y. Alterations of Lysine Acetylation Profile in Murine Skeletal Muscles Upon Exercise. Front Aging Neurosci 2022; 14:859313. [PMID: 35592697 PMCID: PMC9110802 DOI: 10.3389/fnagi.2022.859313] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022] Open
Abstract
Objective Regular exercise is a powerful tool that enhances skeletal muscle mass and strength. Lysine acetylation is an important post-translational modification (PTM) involved in a broad array of cellular functions. Skeletal muscle protein contains a considerable number of lysine-acetylated (Kac) sites, so we aimed to investigate the effects of exercise-induced lysine acetylation on skeletal muscle proteins. Methods We randomly divided 20 male C57BL/6 mice into exercise and control groups. After 6 weeks of treadmill exercise, a lysine acetylation proteomics analysis of the gastrocnemius muscles of mice was performed. Results A total of 2,254 lysine acetylation sites in 693 protein groups were identified, among which 1,916 sites in 528 proteins were quantified. The enrichment analysis suggested that protein acetylation could influence both structural and functional muscle protein properties. Moreover, molecular docking revealed that mimicking protein deacetylation primarily influenced the interaction between substrates and enzymes. Conclusion Exercise-induced lysine acetylation appears to be a crucial contributor to the alteration of skeletal muscle protein binding free energy, suggesting that its modulation is a potential approach for improving exercise performance.
Collapse
Affiliation(s)
- Dehuan Liang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Cheng Chen
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Song Huang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Sujuan Liu
- Department of Anatomy and Histology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Li Fu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Yanmei Niu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
20
|
Acharya A, Nemade H, Rajendra Prasad K, Khan K, Hescheler J, Blackburn N, Hemmersbach R, Papadopoulos S, Sachinidis A. Live-Cell Imaging of the Contractile Velocity and Transient Intracellular Ca 2+ Fluctuations in Human Stem Cell-Derived Cardiomyocytes. Cells 2022; 11:1280. [PMID: 35455960 PMCID: PMC9031802 DOI: 10.3390/cells11081280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/25/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Live-cell imaging techniques are essential for acquiring vital physiological and pathophysiological knowledge to understand and treat heart disease. For live-cell imaging of transient alterations of [Ca2+]i in human cardiomyocytes, we engineered human-induced pluripotent stem cells carrying a genetically-encoded Ca2+-indicator (GECI). To monitor sarcomere shortening and relaxation in cardiomyocytes in real-time, we generated a α-cardiac actinin (ACTN2)-copepod (cop) green fluorescent protein (GFP+)-human-induced pluripotent stem cell line by using the CRISPR-Cas9 and a homology directed recombination approach. The engineered human-induced pluripotent stem cells were differentiated in transgenic GECI-enhanced GFP+-cardiomyocytes and ACTN2-copGFP+-cardiomyocytes, allowing real-time imaging of [Ca2+]i transients and live recordings of the sarcomere shortening velocity of ACTN2-copGFP+-cardiomyocytes. We developed a video analysis software tool to quantify various parameters of sarcoplasmic Ca2+ fluctuations recorded during contraction of cardiomyocytes and to calculate the contraction velocity of cardiomyocytes in the presence and absence of different drugs affecting cardiac function. Our cellular and software tool not only proved the positive and negative inotropic and lusitropic effects of the tested cardioactive drugs but also quantified the expected effects precisely. Our platform will offer a human-relevant in vitro alternative for high-throughput drug screenings, as well as a model to explore the underlying mechanisms of cardiac diseases.
Collapse
Affiliation(s)
- Aviseka Acharya
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Harshal Nemade
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Krishna Rajendra Prasad
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Khadija Khan
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Jürgen Hescheler
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Nick Blackburn
- Bioras Company, Kaarsbergsvej 2, 8400 Ebeltoft, Denmark;
| | - Ruth Hemmersbach
- German Aerospace Center, Institute of Aerospace Medicine, Gravitational Biology, Linder Hoehe, 51147 Cologne, Germany;
| | - Symeon Papadopoulos
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
| | - Agapios Sachinidis
- Working Group Sachinidis, Center for Physiology, Faculty of Medicine and University Hospital Cologne, The University of Cologne, 50931 Cologne, Germany; (A.A.); (H.N.); (K.R.P.); (K.K.); (J.H.); (S.P.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
21
|
Poppe L, Hartman JJ, Romero A, Reagan JD. Structural and Thermodynamic Model for the Activation of Cardiac Troponin. Biochemistry 2022; 61:741-748. [PMID: 35349258 DOI: 10.1021/acs.biochem.2c00084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cardiac troponin is a regulatory protein complex located on the sarcomere that regulates the engagement of myosin on actin filaments. Low-molecular weight modulators of troponin that bind allosterically with the calcium ion have the potential to improve cardiac contractility in patients with reduced cardiac function. Here we propose an approach to the rational design of troponin modulators through the combined use of solution nuclear magnetic resonance and isothermal titration calorimetry methods. In contrast to traditional approaches limited to calcium and activator-bound troponin structures, here we analyzed the structural and thermodynamic impact of an activator in the context of the troponin functional cycle. This led us to propose a rationale for developing an efficacious troponin activator.
Collapse
Affiliation(s)
- Leszek Poppe
- Amgen, Inc., Thousand Oaks, California 91320, United States
| | - James J Hartman
- Cytokinetics, Inc., South San Francisco, California 94080, United States
| | - Antonio Romero
- Cytokinetics, Inc., South San Francisco, California 94080, United States
| | - Jeffrey D Reagan
- Amgen, Inc., South San Francisco, California 94080, United States
| |
Collapse
|
22
|
Yu HL, Hwang SPL. Zebrafish integrin a3b is required for cardiac contractility and cardiomyocyte proliferation. Biochem Biophys Res Commun 2022; 595:89-95. [PMID: 35121232 DOI: 10.1016/j.bbrc.2022.01.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/23/2022] [Indexed: 01/11/2023]
Abstract
In cardiac muscle cells, heterodimeric integrin transmembrane receptors are known to serve as mechanotransducers, translating mechanical force to biochemical signaling. However, the roles of many individual integrins have still not been delineated. In this report, we demonstrate that Itga3b is localized to the sarcolemma of cardiomyocytes from 24 to 96 hpf. We further show that heterozygous and homozygous itga3b/bdf mutant embryos display a cardiomyopathy phenotype, with decreased cardiac contractility and reduced cardiomyocyte number. Correspondingly, proliferation of ventricular and atrial cardiomyoctyes and ventricular epicardial cells is decreased in itga3b mutant hearts. The contractile dysfunction of itga3b mutants can be attributed to cardiomyocyte sarcomeric disorganization, including thin myofilaments with blurred and shortened Z-discs. Together, our results reveal that Itga3b localizes to the myocardium sarcolemma, and it is required for cardiac contractility and cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Hsiang-Ling Yu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202301, Taiwan
| | - Sheng-Ping L Hwang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 202301, Taiwan; Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
23
|
Glaser M, Bruce NJ, Han SB, Wade RC. Simulation of the Positive Inotropic Peptide S100A1ct in Aqueous Environment by Gaussian Accelerated Molecular Dynamics. J Phys Chem B 2021; 125:4654-4666. [PMID: 33944558 DOI: 10.1021/acs.jpcb.1c00902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The S100A1ct peptide, consisting of the C-terminal 20 residues of the S100A1 protein fused to an N-terminal 6-residue hydrophilic tag, has been found to exert a positive inotropic effect, resulting in improved contractile performance of failing cardiac and skeletal muscle without arrhythmic side-effects. The S100A1ct peptide thus has high potential for the treatment of acute heart failure. As a step toward understanding its molecular mechanism of action, and to provide a basis for peptidomimetic design to optimize its properties, we here describe de novo structure predictions and molecular dynamics simulations to characterize the conformational landscape of S100A1ct in aqueous environment. In S100A1, the C-terminal 20 residues form an α-helix, but de novo peptide structure predictions indicate that other conformations are also possible. Conventional molecular dynamics simulations in implicit and explicit solvent corroborated this finding. To ensure adequate sampling, we performed simulations of a tagged 10-residue segment of S100A1ct, and we carried out Gaussian accelerated molecular dynamics simulations of the peptides. These simulations showed that although the helical conformation of S100A1ct was the most energetically stable, the peptide can adopt a range of kinked conformations, suggesting that its activity may be related to its ability to act as a conformational switch.
Collapse
Affiliation(s)
- Manuel Glaser
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Informatics for Life, Heidelberg, Germany
| | - Neil J Bruce
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Informatics for Life, Heidelberg, Germany
| | - Sungho Bosco Han
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Rebecca C Wade
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Informatics for Life, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.,Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance and Interdisciplinary Center for Scientific Computing (IWR), 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Sponga A, Arolas JL, Schwarz TC, Jeffries CM, Rodriguez Chamorro A, Kostan J, Ghisleni A, Drepper F, Polyansky A, De Almeida Ribeiro E, Pedron M, Zawadzka-Kazimierczuk A, Mlynek G, Peterbauer T, Doto P, Schreiner C, Hollerl E, Mateos B, Geist L, Faulkner G, Kozminski W, Svergun DI, Warscheid B, Zagrovic B, Gautel M, Konrat R, Djinović-Carugo K. Order from disorder in the sarcomere: FATZ forms a fuzzy but tight complex and phase-separated condensates with α-actinin. SCIENCE ADVANCES 2021; 7:eabg7653. [PMID: 34049882 PMCID: PMC8163081 DOI: 10.1126/sciadv.abg7653] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/13/2021] [Indexed: 05/03/2023]
Abstract
In sarcomeres, α-actinin cross-links actin filaments and anchors them to the Z-disk. FATZ (filamin-, α-actinin-, and telethonin-binding protein of the Z-disk) proteins interact with α-actinin and other core Z-disk proteins, contributing to myofibril assembly and maintenance. Here, we report the first structure and its cellular validation of α-actinin-2 in complex with a Z-disk partner, FATZ-1, which is best described as a conformational ensemble. We show that FATZ-1 forms a tight fuzzy complex with α-actinin-2 and propose an interaction mechanism via main molecular recognition elements and secondary binding sites. The obtained integrative model reveals a polar architecture of the complex which, in combination with FATZ-1 multivalent scaffold function, might organize interaction partners and stabilize α-actinin-2 preferential orientation in Z-disk. Last, we uncover FATZ-1 ability to phase-separate and form biomolecular condensates with α-actinin-2, raising the question whether FATZ proteins can create an interaction hub for Z-disk proteins through membraneless compartmentalization during myofibrillogenesis.
Collapse
Affiliation(s)
- Antonio Sponga
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Joan L Arolas
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Thomas C Schwarz
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Cy M Jeffries
- European Molecular Biology Laboratory (EMBL), Hamburg Unit, Hamburg, Germany
| | - Ariadna Rodriguez Chamorro
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Julius Kostan
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Andrea Ghisleni
- King's College London BHF Centre for Research Excellence, Randall Centre for Cell and Molecular Biophysics, London SE1 1UL, UK
| | - Friedel Drepper
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Anton Polyansky
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
- National Research University Higher School of Economics, Moscow 101000, Russia
| | - Euripedes De Almeida Ribeiro
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Miriam Pedron
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Anna Zawadzka-Kazimierczuk
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland
| | - Georg Mlynek
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Thomas Peterbauer
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Dr. BohrGasse 9, A-1030 Vienna, Austria
| | - Pierantonio Doto
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Claudia Schreiner
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Eneda Hollerl
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Borja Mateos
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Leonhard Geist
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | | | - Wiktor Kozminski
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, Zwirki i Wigury 101, 02-089 Warsaw, Poland
| | - Dmitri I Svergun
- King's College London BHF Centre for Research Excellence, Randall Centre for Cell and Molecular Biophysics, London SE1 1UL, UK
| | - Bettina Warscheid
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Bojan Zagrovic
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Mathias Gautel
- King's College London BHF Centre for Research Excellence, Randall Centre for Cell and Molecular Biophysics, London SE1 1UL, UK
| | - Robert Konrat
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria
| | - Kristina Djinović-Carugo
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Campus Vienna Biocenter 5, A-1030 Vienna, Austria.
- Department of Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
25
|
Jiao S, Xu R, Du S. Smyd1 is essential for myosin expression and sarcomere organization in craniofacial, extraocular, and cardiac muscles. J Genet Genomics 2021; 48:208-218. [PMID: 33958316 PMCID: PMC9234968 DOI: 10.1016/j.jgg.2021.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 01/02/2023]
Abstract
Skeletal and cardiac muscles are striated myofibers that contain highly organized sarcomeres for muscle contraction. Recent studies revealed that Smyd1, a lysine methyltransferase, plays a key role in sarcomere assembly in heart and trunk skeletal muscles. However, Smyd1 expression and function in craniofacial muscles are not known. Here, we analyze the developmental expression and function of two smyd1 paralogous genes, smyd1a and smyd1b, in craniofacial and cardiac muscles of zebrafish embryos. Our data show that loss of smyd1a (smyd1amb5) or smyd1b (smyd1bsa15678) has no visible effects on myogenic commitment and expression of myod and myosin heavy-chain mRNA transcripts in craniofacial muscles. However, myosin heavy-chain protein accumulation and sarcomere organization are dramatically reduced in smyd1bsa15678 single mutant, and almost completely diminish in smyd1amb5; smyd1bsa15678 double mutant, but not in smyd1amb5 mutant. Similar defects are also observed in cardiac muscles of smyd1bsa15678 mutant. Defective craniofacial and cardiac muscle formation is associated with an upregulation of hsp90α1 and unc45b mRNA expression in smyd1bsa15678 and smyd1amb5; smyd1bsa15678 mutants. Together, our studies indicate that Smyd1b, but not Smyd1a, plays a key role in myosin heavy-chain protein expression and sarcomere organization in craniofacial and cardiac muscles. Loss of smyd1b results in muscle-specific stress response.
Collapse
Affiliation(s)
- Shuang Jiao
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD 21202, USA; Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Rui Xu
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD 21202, USA
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD 21202, USA.
| |
Collapse
|
26
|
Li MX, Mercier P, Hartman JJ, Sykes BD. Structural Basis of Tirasemtiv Activation of Fast Skeletal Muscle. J Med Chem 2021; 64:3026-3034. [PMID: 33703886 DOI: 10.1021/acs.jmedchem.0c01412] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Troponin regulates the calcium-mediated activation of skeletal muscle. Muscle weakness in diseases such as amyotrophic lateral sclerosis and spinal muscular atrophy occurs from diminished neuromuscular output. The first direct fast skeletal troponin activator, tirasemtiv, amplifies the response of muscle to neuromuscular input. Tirasemtiv binds selectively and strongly to fast skeletal troponin, slowing the rate of calcium release and sensitizing muscle to calcium. We report the solution NMR structure of tirasemtiv bound to a fast skeletal troponin C-troponin I chimera. The structure reveals that tirasemtiv binds in a hydrophobic pocket between the regulatory domain of troponin C and the switch region of troponin I, which overlaps with that of Anapoe in the X-ray structure of skeletal troponin. Multiple interactions stabilize the troponin C-troponin I interface, increase the affinity of troponin C for the switch region of fast skeletal troponin I, and drive the equilibrium toward the active state.
Collapse
Affiliation(s)
- Monica X Li
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Pascal Mercier
- National High Field NMR Centre, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - James J Hartman
- Cytokinetics, Inc., South San Francisco, California 94080, United States
| | - Brian D Sykes
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
27
|
Parijat P, Kondacs L, Alexandrovich A, Gautel M, Cobb AJA, Kampourakis T. High Throughput Screen Identifies Small Molecule Effectors That Modulate Thin Filament Activation in Cardiac Muscle. ACS Chem Biol 2021; 16:225-235. [PMID: 33315370 DOI: 10.1021/acschembio.0c00908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Current therapeutic interventions for both heart disease and heart failure are largely insufficient and associated with undesired side effects. Biomedical research has emphasized the role of sarcomeric protein function for the normal performance and energy efficiency of the heart, suggesting that directly targeting the contractile myofilaments themselves using small molecule effectors has therapeutic potential and will likely result in greater drug efficacy and selectivity. In this study, we developed a robust and highly reproducible fluorescence polarization-based high throughput screening (HTS) assay that directly targets the calcium-dependent interaction between cardiac troponin C (cTnC) and the switch region of cardiac troponin I (cTnISP), with the aim of identifying small molecule effectors of the cardiac thin filament activation pathway. We screened a commercially available small molecule library and identified several hit compounds with both inhibitory and activating effects. We used a range of biophysical and biochemical methods to characterize hit compounds and identified fingolimod, a sphingosin-1-phosphate receptor modulator, as a new troponin-based small molecule effector. Fingolimod decreased the ATPase activity and calcium sensitivity of demembranated cardiac muscle fibers in a dose-dependent manner, suggesting that the compound acts as a calcium desensitizer. We investigated fingolimod's mechanism of action using a combination of computational studies, biophysical methods, and synthetic chemistry, showing that fingolimod bound to cTnC repels cTnISP via mainly electrostatic repulsion of its positively charged tail. These results suggest that fingolimod is a potential new lead compound/scaffold for the development of troponin-directed heart failure therapeutics.
Collapse
Affiliation(s)
- Priyanka Parijat
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Laszlo Kondacs
- Department of Chemistry, King’s College London, 7 Trinity Street, London, SE1 1DB, United Kingdom
| | - Alexander Alexandrovich
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| | - Alexander J. A. Cobb
- Department of Chemistry, King’s College London, 7 Trinity Street, London, SE1 1DB, United Kingdom
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics, King’s College London, and British Heart Foundation Centre of Research Excellence, London SE1 1UL, United Kingdom
| |
Collapse
|
28
|
Ezeani M, Prabhu S. Pathophysiology and therapeutic relevance of PI3K(p110α) protein in atrial fibrillation: A non-interventional molecular therapy strategy. Pharmacol Res 2021; 165:105415. [PMID: 33412279 DOI: 10.1016/j.phrs.2020.105415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/04/2020] [Accepted: 12/23/2020] [Indexed: 10/22/2022]
Abstract
Genetically modified animal studies have revealed specific expression patterns and unequivocal roles of class I PI3K isoenzymes. PI3K(p110α), a catalytic subunit of class I PI3Ks is ubiquitously expressed and is well characterised in the cardiovascular system. Given that genetic inhibition of PI3K(p110α) causes lethal phenotype embryonically, the catalytic subunit is critically important in housekeeping and biological processes. A growing number of studies underpin crucial roles of PI3K(p110α) in cell survival, proliferation, hypertrophy and arrhythmogenesis. While the studies provide great insights, the precise mechanisms involved in PI3K(p110α) hypofunction and atrial fibrillation (AF) are not fully known. AF is a well recognised clinical problem with significant management limitations. In this translational review, we attempted a narration of PI3K(p110α) hypofunction in the molecular basis of AF pathophysiology. We sought to cautiously highlight the relevance of this molecule in the therapeutic approaches for AF management per se (i.e without conditions associate with cell proliferation, like cancer), and in mitigating effects of clinical risk factors in atrial substrate formation leading to AF progression. We also considered PI3K(p110α) in AF gene association, with the aim of identifying mechanistic links between the ever increasingly well-defined genetic loci (regions and genes) and AF. Such mechanisms will aid in identifying new drug targets for arrhythmogenic substrate and AF.
Collapse
Affiliation(s)
- Martin Ezeani
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia.
| | - Sandeep Prabhu
- The Alfred, and Baker Heart and Diabetes Institute, Melbourne, Australia; University of Melbourne, Melbourne, Australia
| |
Collapse
|
29
|
Optimal loading of omecamtiv mecarbil by chitosan: A comprehensive and comparative molecular dynamics study. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.114908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Hashem S, Davies WG, Fornili A. Heart Failure Drug Modifies the Intrinsic Dynamics of the Pre-Power Stroke State of Cardiac Myosin. J Chem Inf Model 2020; 60:6438-6446. [PMID: 33283509 DOI: 10.1021/acs.jcim.0c00953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Omecamtiv mecarbil (OM), currently investigated for the treatment of heart failure, is the first example of a new class of drugs (cardiac myotropes) that can modify muscle contractility by directly targeting sarcomeric proteins. Using atomistic molecular dynamics simulations, we show that the binding of OM to the pre-power stroke state of cardiac myosin inhibits the functional motions of the protein and potentially affects Pi release from the nucleotide binding site. We also show that the changes in myosin ATPase activity induced by a set of OM analogues can be predicted from their relative affinity to the pre-power stroke state compared to the near rigor one, indicating that conformational selectivity plays an important role in determining the activity of these compounds.
Collapse
Affiliation(s)
- Shaima Hashem
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, United Kingdom
| | - William George Davies
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Arianna Fornili
- School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, United Kingdom.,The Thomas Young Centre for Theory and Simulation of Materials, London WC1E 6BN, United Kingdom
| |
Collapse
|
31
|
An insight on Drosophila myogenesis and its assessment techniques. Mol Biol Rep 2020; 47:9849-9863. [PMID: 33263930 DOI: 10.1007/s11033-020-06006-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 11/16/2020] [Indexed: 10/22/2022]
Abstract
Movement assisted by muscles forms the basis of various behavioural traits seen in Drosophila. Myogenesis involves developmental processes like cellular specification, differentiation, migration, fusion, adherence to tendons and neuronal innervation in a series of coordinated event well defined in body space and time. Gene regulatory networks are switched on-off, fine tuning at the right developmental stage to assist each cellular event. Drosophila is a holometabolous organism that undergoes myogenesis waves at two developmental stages, and is ideal for comparative analysis of the role of genes and genetic pathways conserved across phyla. In this review we have summarized myogenic events from the embryo to adult focussing on the somatic muscle development during the early embryonic stage and then on indirect flight muscles (IFM) formation required for adult life, emphasizing on recent trends of analysing muscle mutants and advances in Drosophila muscle biology.
Collapse
|
32
|
Teng GZ, Dawson JF. The Dark Side of Actin: Cardiac actin variants highlight the role of allostery in disease development. Arch Biochem Biophys 2020; 695:108624. [PMID: 33049292 DOI: 10.1016/j.abb.2020.108624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/24/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022]
Abstract
Mutations in the α-cardiac actin ACTC1 gene cause dilated or hypertrophic cardiomyopathy. These diseases are the result of changes in protein interactions between ACTC protein and force-generating β-myosin or the calcium-dependent cardiac-tropomyosin (cTm) and cardiac troponin (cTn) regulatory complex, altering the overall contractile force. The T126I and S271F ACTC variants possess amino acid substitutions on the other side of actin relative to the myosin or regulatory protein binding sites on what we call the "dark side" of actin. The T126I change results in hyposensitivity to calcium, in accordance with the calcium sensitivity pathway of cardiomyopathy development while the S271F change alters the maximum in vitro motility sliding speed, reflecting a change in maximum force. These results demonstrate the role of actin allostery in the cardiac disease development.
Collapse
Affiliation(s)
- Grace Zi Teng
- Department of Molecular & Cellular Biology and Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - John F Dawson
- Department of Molecular & Cellular Biology and Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
33
|
Tucholski T, Cai W, Gregorich ZR, Bayne EF, Mitchell SD, McIlwain SJ, de Lange WJ, Wrobbel M, Karp H, Hite Z, Vikhorev PG, Marston SB, Lal S, Li A, Dos Remedios C, Kohmoto T, Hermsen J, Ralphe JC, Kamp TJ, Moss RL, Ge Y. Distinct hypertrophic cardiomyopathy genotypes result in convergent sarcomeric proteoform profiles revealed by top-down proteomics. Proc Natl Acad Sci U S A 2020; 117:24691-24700. [PMID: 32968017 PMCID: PMC7547245 DOI: 10.1073/pnas.2006764117] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heritable heart disease. Although the genetic cause of HCM has been linked to mutations in genes encoding sarcomeric proteins, the ability to predict clinical outcomes based on specific mutations in HCM patients is limited. Moreover, how mutations in different sarcomeric proteins can result in highly similar clinical phenotypes remains unknown. Posttranslational modifications (PTMs) and alternative splicing regulate the function of sarcomeric proteins; hence, it is critical to study HCM at the level of proteoforms to gain insights into the mechanisms underlying HCM. Herein, we employed high-resolution mass spectrometry-based top-down proteomics to comprehensively characterize sarcomeric proteoforms in septal myectomy tissues from HCM patients exhibiting severe outflow track obstruction (n = 16) compared to nonfailing donor hearts (n = 16). We observed a complex landscape of sarcomeric proteoforms arising from combinatorial PTMs, alternative splicing, and genetic variation in HCM. A coordinated decrease of phosphorylation in important myofilament and Z-disk proteins with a linear correlation suggests PTM cross-talk in the sarcomere and dysregulation of protein kinase A pathways in HCM. Strikingly, we discovered that the sarcomeric proteoform alterations in the myocardium of HCM patients undergoing septal myectomy were remarkably consistent, regardless of the underlying HCM-causing mutations. This study suggests that the manifestation of severe HCM coalesces at the proteoform level despite distinct genotype, which underscores the importance of molecular characterization of HCM phenotype and presents an opportunity to identify broad-spectrum treatments to mitigate the most severe manifestations of this genetically heterogenous disease.
Collapse
Affiliation(s)
- Trisha Tucholski
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Wenxuan Cai
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Zachery R Gregorich
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Elizabeth F Bayne
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Stanford D Mitchell
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Sean J McIlwain
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705
| | - Willem J de Lange
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53705
| | - Max Wrobbel
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Hannah Karp
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Zachary Hite
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Petr G Vikhorev
- National Heart & Lung Institute, Imperial College London, London W12 0NN, United Kingdom
| | - Steven B Marston
- National Heart & Lung Institute, Imperial College London, London W12 0NN, United Kingdom
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Camperdown, NSW 2006,Australia
| | - Amy Li
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Camperdown, NSW 2006,Australia
- Department of Pharmacy & Biomedical Sciences, La Trobe University, Bundoora, VIC 3086, Australia
| | - Cristobal Dos Remedios
- School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Camperdown, NSW 2006,Australia
- Department of Molecular Cardiology & Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Takushi Kohmoto
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706
| | - Joshua Hermsen
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706
| | - J Carter Ralphe
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53705
| | - Timothy J Kamp
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Richard L Moss
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706;
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705
- Human Proteomics Program, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
34
|
Sarcomere integrated biosensor detects myofilament-activating ligands in real time during twitch contractions in live cardiac muscle. J Mol Cell Cardiol 2020; 147:49-61. [PMID: 32791214 DOI: 10.1016/j.yjmcc.2020.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 11/24/2022]
Abstract
The sarcomere is the functional unit of cardiac muscle, essential for normal heart function. To date, it has not been possible to study, in real time, thin filament-based activation dynamics in live cardiac muscle. We report here results from a cardiac troponin C (TnC) FRET-based biosensor integrated into the cardiac sarcomere via stoichiometric replacement of endogenous TnC. The TnC biosensor provides, for the first time, evidence of multiple thin filament activating ligands, including troponin I interfacing with TnC and cycling myosin, during a cardiac twitch. Results show that the TnC FRET biosensor transient significantly precedes that of peak twitch force. Using small molecules and genetic modifiers known to alter sarcomere activation, independently of the intracellular Ca2+ transient, the data show that the TnC biosensor detects significant effects of the troponin I switch domain as a sarcomere-activating ligand. Interestingly, the TnC biosensor also detected the effects of load-dependent altered myosin cycling, as shown by a significant delay in TnC biosensor transient inactivation during the isometric twitch. In addition, the TnC biosensor detected the effects of myosin as an activating ligand during the twitch by using a small molecule that directly alters cross-bridge cycling, independently of the intracellular Ca2+ transient. Collectively, these results aid in illuminating the basis of cardiac muscle contractile activation with implications for gene, protein, and small molecule-based strategies designed to target the sarcomere in regulating beat-to-beat heart performance in health and disease.
Collapse
|
35
|
Guhathakurta P, Phung LA, Prochniewicz E, Lichtenberger S, Wilson A, Thomas DD. Actin-binding compounds, previously discovered by FRET-based high-throughput screening, differentially affect skeletal and cardiac muscle. J Biol Chem 2020; 295:14100-14110. [PMID: 32788211 DOI: 10.1074/jbc.ra120.014445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/06/2020] [Indexed: 01/21/2023] Open
Abstract
Actin's interactions with myosin and other actin-binding proteins are essential for cellular viability in numerous cell types, including muscle. In a previous high-throughput time-resolved FRET (TR-FRET) screen, we identified a class of compounds that bind to actin and affect actomyosin structure and function. For clinical utility, it is highly desirable to identify compounds that affect skeletal and cardiac muscle differently. Because actin is more highly conserved than myosin and most other muscle proteins, most such efforts have not targeted actin. Nevertheless, in the current study, we tested the specificity of the previously discovered actin-binding compounds for effects on skeletal and cardiac α-actins as well as on skeletal and cardiac myofibrils. We found that a majority of these compounds affected the transition of monomeric G-actin to filamentous F-actin, and that several of these effects were different for skeletal and cardiac actin isoforms. We also found that several of these compounds affected ATPase activity differently in skeletal and cardiac myofibrils. We conclude that these structural and biochemical assays can be used to identify actin-binding compounds that differentially affect skeletal and cardiac muscles. The results of this study set the stage for screening of large chemical libraries for discovery of novel compounds that act therapeutically and specifically on cardiac or skeletal muscle.
Collapse
Affiliation(s)
- Piyali Guhathakurta
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lien A Phung
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ewa Prochniewicz
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sarah Lichtenberger
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anna Wilson
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA .,Photonic Pharma LLC, Minneapolis, Minnesota, USA
| |
Collapse
|
36
|
Holmes JB, Stelzer JE. Prof. Cristobal dos Remedios and the Sydney Heart Bank: enabling translatable heart failure research. Biophys Rev 2020; 12:783-784. [PMID: 32572679 DOI: 10.1007/s12551-020-00711-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/17/2020] [Indexed: 01/03/2023] Open
Affiliation(s)
- Joshua B Holmes
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, 2109 Adelbert Rd, Robbins E522, Cleveland, OH, 44106, USA
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, 2109 Adelbert Rd, Robbins E522, Cleveland, OH, 44106, USA.
| |
Collapse
|
37
|
Ge Z, Li A, McNamara J, Dos Remedios C, Lal S. Pathogenesis and pathophysiology of heart failure with reduced ejection fraction: translation to human studies. Heart Fail Rev 2020; 24:743-758. [PMID: 31209771 DOI: 10.1007/s10741-019-09806-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Heart failure represents the end result of different pathophysiologic processes, which culminate in functional impairment. Regardless of its aetiology, the presentation of heart failure usually involves symptoms of pump failure and congestion, which forms the basis for clinical diagnosis. Pathophysiologic descriptions of heart failure with reduced ejection fraction (HFrEF) are being established. Most commonly, HFrEF is centred on a reactive model where a significant initial insult leads to reduced cardiac output, further triggering a cascade of maladaptive processes. Predisposing factors include myocardial injury of any cause, chronically abnormal loading due to hypertension, valvular disease, or tachyarrhythmias. The pathophysiologic processes behind remodelling in heart failure are complex and reflect systemic neurohormonal activation, peripheral vascular effects and localised changes affecting the cardiac substrate. These abnormalities have been the subject of intense research. Much of the translational successes in HFrEF have come from targeting neurohormonal responses to reduced cardiac output, with blockade of the renin-angiotensin-aldosterone system (RAAS) and beta-adrenergic blockade being particularly fruitful. However, mortality and morbidity associated with heart failure remains high. Although systemic neurohormonal blockade slows disease progression, localised ventricular remodelling still adversely affects contractile function. Novel therapy targeted at improving cardiac contractile mechanics in HFrEF hold the promise of alleviating heart failure at its source, yet so far none has found success. Nevertheless, there are increasing calls for a proximal, 'cardiocentric' approach to therapy. In this review, we examine HFrEF therapy aimed at improving cardiac function with a focus on recent trials and emerging targets.
Collapse
Affiliation(s)
- Zijun Ge
- Sydney Medical School, University of Sydney, Camperdown, Australia
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Amy Li
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
- Department of Pharmacy and Biomedical Science, La Trobe University, Melbourne, Australia
| | - James McNamara
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Cris Dos Remedios
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Sean Lal
- Sydney Medical School, University of Sydney, Camperdown, Australia.
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.
- Cardiac Research Laboratory, Discipline of Anatomy and Histology, University of Sydney, Anderson Stuart Building (F13), Camperdown, NSW, 2006, Australia.
| |
Collapse
|
38
|
Holmes JB, Doh CY, Mamidi R, Li J, Stelzer JE. Strategies for targeting the cardiac sarcomere: avenues for novel drug discovery. Expert Opin Drug Discov 2020; 15:457-469. [PMID: 32067508 PMCID: PMC7065952 DOI: 10.1080/17460441.2020.1722637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/24/2020] [Indexed: 01/10/2023]
Abstract
Introduction: Heart failure remains one of the largest clinical challenges in the United States. Researchers have continually searched for more effective heart failure treatments that target the cardiac sarcomere but have found few successes despite numerous expensive cardiovascular clinical trials. Among many reasons, the high failure rate of cardiovascular clinical trials may be partly due to incomplete characterization of a drug candidate's complex interaction with cardiac physiology.Areas covered: In this review, the authors address the issue of preclinical cardiovascular studies of sarcomere-targeting heart failure therapies. The authors consider inherent tradeoffs made between mechanistic transparency and physiological fidelity for several relevant preclinical techniques at the atomic, molecular, heart muscle fiber, whole heart, and whole-organism levels. Thus, the authors suggest a comprehensive, bottom-up approach to preclinical cardiovascular studies that fosters scientific rigor and hypothesis-driven drug discovery.Expert opinion: In the authors' opinion, the implementation of hypothesis-driven drug discovery practices, such as the bottom-up approach to preclinical cardiovascular studies, will be imperative for the successful development of novel heart failure treatments. However, additional changes to clinical definitions of heart failure and current drug discovery culture must accompany the bottom-up approach to maximize the effectiveness of hypothesis-driven drug discovery.
Collapse
Affiliation(s)
- Joshua B Holmes
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Chang Yoon Doh
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
39
|
Robertson IM, Klein BA, Sykes BD. Optimizing fluorine labelling for 19F solid-state NMR in oriented biological systems. JOURNAL OF BIOMOLECULAR NMR 2020; 74:1-7. [PMID: 31912345 DOI: 10.1007/s10858-019-00296-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/31/2019] [Indexed: 06/10/2023]
Abstract
When planning a fluorine labeling strategy for 19F solid state NMR (ssNMR) studies of the structure and/or mobility of fluorine labeled compounds in situ in an oriented biological system, it is important to characterize the NMR properties of the label. This manuscript focuses on the characterization of a selection of aromatic fluorine compounds in dimyristoylphosphatidylcholine bilayers using 19F ssNMR from the standpoint of determining the optimum arrangement of fluorine nuclei on a pendant aromatic ring before incorporation into more complex biological systems.
Collapse
Affiliation(s)
- Ian M Robertson
- Ministry of Health, Government of Alberta, Edmonton, AB, T5J 1S6, Canada
| | - Brittney A Klein
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G2H7, Canada
| | - Brian D Sykes
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G2H7, Canada.
| |
Collapse
|
40
|
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: 1.5] [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
|
41
|
Gene expression profiling of skeletal myogenesis in human embryonic stem cells reveals a potential cascade of transcription factors regulating stages of myogenesis, including quiescent/activated satellite cell-like gene expression. PLoS One 2019; 14:e0222946. [PMID: 31560727 PMCID: PMC6764674 DOI: 10.1371/journal.pone.0222946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/10/2019] [Indexed: 01/05/2023] Open
Abstract
Human embryonic stem cell (hESC)-derived skeletal muscle progenitors (SMP)—defined as PAX7-expressing cells with myogenic potential—can provide an abundant source of donor material for muscle stem cell therapy. As in vitro myogenesis is decoupled from in vivo timing and 3D-embryo structure, it is important to characterize what stage or type of muscle is modeled in culture. Here, gene expression profiling is analyzed in hESCs over a 50 day skeletal myogenesis protocol and compared to datasets of other hESC-derived skeletal muscle and adult murine satellite cells. Furthermore, day 2 cultures differentiated with high or lower concentrations of CHIR99021, a GSK3A/GSK3B inhibitor, were contrasted. Expression profiling of the 50 day time course identified successively expressed gene subsets involved in mesoderm/paraxial mesoderm induction, somitogenesis, and skeletal muscle commitment/formation which could be regulated by a putative cascade of transcription factors. Initiating differentiation with higher CHIR99021 concentrations significantly increased expression of MSGN1 and TGFB-superfamily genes, notably NODAL, resulting in enhanced paraxial mesoderm and reduced ectoderm/neuronal gene expression. Comparison to adult satellite cells revealed that genes expressed in 50-day cultures correlated better with those expressed by quiescent or early activated satellite cells, which have the greatest therapeutic potential. Day 50 cultures were similar to other hESC-derived skeletal muscle and both expressed known and novel SMP surface proteins. Overall, a putative cascade of transcription factors has been identified which regulates four stages of myogenesis. Subsets of these factors were upregulated by high CHIR99021 or their binding sites were significantly over-represented during SMP activation, ranging from quiescent to late-activated stages. This analysis serves as a resource to further study the progression of in vitro skeletal myogenesis and could be mined to identify novel markers of pluripotent-derived SMPs or regulatory transcription/growth factors. Finally, 50-day hESC-derived SMPs appear similar to quiescent/early activated satellite cells, suggesting they possess therapeutic potential.
Collapse
|
42
|
Marston S. Small molecule studies: the fourth wave of muscle research. J Muscle Res Cell Motil 2019; 40:69-76. [PMID: 31228047 PMCID: PMC6726831 DOI: 10.1007/s10974-019-09526-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/13/2019] [Indexed: 12/28/2022]
Abstract
The study of muscle and contractility is an unusual scientific endeavour since it has from the start been focussed on one problem-What makes muscle work?-and yet has needed a vast range of different approaches and techniques to study it. Its uniqueness lies in the fundamental fascination of a large scale molecular machine that converts chemical energy into mechanical energy at ambient temperature and with high efficiency that is also controlled by an exquisitely intricate yet utterly reliable regulatory system and is an essential component of animal life. The investigation of muscle is as innovative as any other field of research. As soon as one approach appears to be played out another comes along. It is instructive to consider this as a series of waves of novel and heightened activity starting in the 1950s. The thesis of this article is that we are approaching the fourth wave with the recent rise of interest in small molecules as research tools and possible therapies for muscle diseases.
Collapse
Affiliation(s)
- Steven Marston
- Cardiovascular Division, National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
43
|
Cai M, Han L, Liu L, He F, Chu W, Zhang J, Tian Z, Du S. Defective sarcomere assembly in smyd1a and smyd1b zebrafish mutants. FASEB J 2019; 33:6209-6225. [PMID: 30817176 PMCID: PMC6463926 DOI: 10.1096/fj.201801578r] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/22/2019] [Indexed: 12/16/2022]
Abstract
Two smyd1 paralogues, smyd1a and smyd1b, have been identified in zebrafish. Although Smyd1b function has been reported in fast muscle, its function in slow muscle and the function of Smyd1a, in general, are uncertain. In this study, we generated 2 smyd1a mutant alleles and analyzed the muscle defects in smyd1a and smyd1b single and double mutants in zebrafish. We demonstrated that knockout of smyd1a alone had no visible effect on muscle development and fish survival. This was in contrast to the smyd1b mutant, which exhibited skeletal and cardiac muscle defects, leading to early embryonic lethality. The smyd1a and smyd1b double mutants, however, showed a stronger muscle defect compared with smyd1a or smyd1b mutation alone, namely, the complete disruption of sarcomere organization in slow and fast muscles. Immunostaining revealed that smyd1a; smyd1b double mutations had no effect on myosin gene expression but resulted in a dramatic reduction of myosin protein levels in muscle cells of zebrafish embryos. This was accompanied by the up-regulation of hsp40 and hsp90-α1 gene expression. Together, our studies indicate that both Smyd1a and Smyd1b partake in slow and fast muscle development although Smyd1b plays a dominant role compared with Smyd1a.-Cai, M., Han, L., Liu, L., He, F., Chu, W., Zhang, J., Tian, Z., Du, S. Defective sarcomere assembly in smyd1a and smyd1b zebrafish mutants.
Collapse
Affiliation(s)
- Mengxin Cai
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi’an, China
| | - Lichen Han
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lusha Liu
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Feng He
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- School of Fisheries, Ocean University of China, Qingdao, China
| | - Wuying Chu
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, China
| | - Jianshe Zhang
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, China
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi’an, China
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
44
|
Campbell KS, Yengo CM, Lee LC, Kotter J, Sorrell VL, Guglin M, Wenk JF. Closing the therapeutic loop. Arch Biochem Biophys 2019; 663:129-131. [PMID: 30639169 PMCID: PMC6377839 DOI: 10.1016/j.abb.2019.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/12/2018] [Accepted: 01/08/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Kenneth S Campbell
- Department of Physiology, University of Kentucky, United States; Division of Cardiovascular Medicine, University of Kentucky, United States.
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, United States
| | - Lik-Chuan Lee
- Department of Mechanical Engineering, Michigan State University, United States
| | - John Kotter
- Division of Cardiovascular Medicine, University of Kentucky, United States
| | - Vincent L Sorrell
- Division of Cardiovascular Medicine, University of Kentucky, United States
| | - Maya Guglin
- Division of Cardiovascular Medicine, University of Kentucky, United States
| | - Jonathan F Wenk
- Department of Mechanical Engineering and Department of Surgery, University of Kentucky, United States
| |
Collapse
|
45
|
Amin M, Kushida Y, Wakao S, Kitada M, Tatsumi K, Dezawa M. Cardiotrophic Growth Factor-Driven Induction of Human Muse Cells Into Cardiomyocyte-Like Phenotype. Cell Transplant 2019; 27:285-298. [PMID: 29637816 PMCID: PMC5898685 DOI: 10.1177/0963689717721514] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are endogenous nontumorigenic stem cells collectable as stage-specific embryonic antigen 3 (SSEA-3) + from various organs including the bone marrow and are pluripotent-like. The potential of human bone marrow-derived Muse cells to commit to cardiac lineage cells was evaluated. We found that (1) initial treatment of Muse cells with 5'-azacytidine in suspension culture successfully accelerated demethylation of cardiac marker Nkx2.5 promoter; (2) then transferring the cells onto adherent culture and treatment with early cardiac differentiation factors including wingless-int (Wnt)-3a, bone morphogenetic proteins (BMP)-2/4, and transforming growth factor (TGF) β1; and (3) further treatment with late cardiac differentiation cytokines including cardiotrophin-1 converted Muse cells into cardiomyocyte-like cells that expressed α-actinin and troponin-I with a striation-like pattern. MLC2a expression in the final step suggested differentiation of the cells into an atrial subtype. MLC2v, a marker for a mature ventricular subtype, was expressed when cells were treated with Dickkopf-related protein 1 (DKK-1) and Noggin, inhibitors of Wnt3a and BMP-4, respectively, between steps (2) and (3). None of the steps included exogenous gene transfection, making induced cells feasible for future clinical application.
Collapse
Affiliation(s)
- Mohamed Amin
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,2 Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia, Egypt
| | - Yoshihiro Kushida
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shohei Wakao
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masaaki Kitada
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuki Tatsumi
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,3 Life Science Institute Inc., Regenerative Medicine Division, Nagoya, Japan
| | - Mari Dezawa
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
46
|
Mamidi R, Li J, Doh CY, Holmes JB, Stelzer JE. Lost in translation: Interpreting cardiac muscle mechanics data in clinical practice. Arch Biochem Biophys 2019; 662:213-218. [PMID: 30576628 PMCID: PMC6345594 DOI: 10.1016/j.abb.2018.12.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/25/2018] [Accepted: 12/17/2018] [Indexed: 01/10/2023]
Abstract
Current inotropic therapies improve systolic function in heart failure patients but also elicit undesirable side effects such as arrhythmias and increased intracellular Ca2+ transients. In order to maintain myocyte Ca2+ homeostasis, the increased cytosolic Ca2+ needs to be actively transported back to sarcoplasmic reticulum leading to depleted ATP reserves. Thus, an emerging approach is to design sarcomere-based treatments to correct impaired contractility via a direct and allosteric modulation of myosin's intrinsic force-generating behavior -a concept that potentially avoids the "off-target" effects. To achieve this goal, various biophysical approaches are utilized to investigate the mechanistic impact of sarcomeric modulators but information derived from diverse approaches is not fully integrated into therapeutic applications. This is in part due to the lack of information that provides a coherent connecting link between biophysical data to in vivo function. Hence, our ability to clearly discern the drug-mediated impact on whole-heart function is diminished. Reducing this translational barrier can significantly accelerate clinical progress related to sarcomere-based therapies by optimizing drug-dosing and treatment duration protocols based on information obtained from biophysical studies. Therefore, we attempt to link biophysical mechanical measurements obtained in isolated cardiac muscle and in vivo contractile function.
Collapse
Affiliation(s)
- Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Chang Yoon Doh
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Joshua B Holmes
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
47
|
Gregorich ZR, Patel JR, Cai W, Lin Z, Heurer R, Fitzsimons DP, Moss RL, Ge Y. Deletion of Enigma Homologue from the Z-disc slows tension development kinetics in mouse myocardium. J Gen Physiol 2019; 151:670-679. [PMID: 30642915 PMCID: PMC6504290 DOI: 10.1085/jgp.201812214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Enigma Homologue (ENH) is a component of the Z-disc, a structure that anchors actin filaments in the contractile unit of muscle, the sarcomere. Cardiac-specific ablation of ENH protein expression causes contractile dysfunction that ultimately culminates in dilated cardiomyopathy. However, whether ENH is involved in the regulation of myocardial contractility is unknown. To determine if ENH is required for the mechanical activity of cardiac muscle, we analyze muscle mechanics of isolated trabeculae from the hearts of ENH +/+ and ENH -/- mice. We detected no differences in steady-state mechanical properties but show that when muscle fibers are allowed to relax and then are restretched, the rate at which tension redevelops is depressed in ENH -/- mouse myocardium relative to that in ENH +/+ myocardium. SDS-PAGE analysis demonstrated that the expression of β-myosin heavy chain is increased in ENH -/- mouse myocardium, which could partially, but not completely, account for the depression in tension redevelopment kinetics. Using top-down proteomics analysis, we found that the expression of other thin/thick filament regulatory proteins is unaltered, although the phosphorylation of a cardiac troponin T isoform, cardiac troponin I, and myosin regulatory light chain is decreased in ENH -/- mouse myocardium. Nevertheless, these alterations are very small and thus insufficient to explain slowed tension redevelopment kinetics in ENH -/- mouse myocardium. These data suggest that the ENH protein influences tension redevelopment kinetics in mouse myocardium, possibly by affecting cross-bridge cycling kinetics. Previous studies also indicate that ablation of specific Z-disc proteins in myocardium slows contraction kinetics, which could also be a contributing factor in this study.
Collapse
Affiliation(s)
- Zachery R Gregorich
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI.,Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI
| | - Jitandrakumar R Patel
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI.,University of Wisconsin-Madison Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI
| | - Wenxuan Cai
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI.,Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI
| | - Ziqing Lin
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI.,Human Proteomics Program, University of Wisconsin-Madison, Madison, WI
| | - Rachel Heurer
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Daniel P Fitzsimons
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Richard L Moss
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI .,University of Wisconsin-Madison Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI.,Human Proteomics Program, University of Wisconsin-Madison, Madison, WI
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI .,Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI.,University of Wisconsin-Madison Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI.,Human Proteomics Program, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
48
|
Abstract
Regulation of muscle contraction has been viewed as principally involving Ca2+ binding to regulatory proteins on the thin filament, but while this is an important element of regulation, the mechanism does not explain the precise matching of muscle performance to the load it must lift or move. Now, it is increasingly evident that mechanisms instrinsic to the thick filament activate myosin cross-bridges as the force or load on a muscle increases. Both thick and thin filament regulatory mechanisms are featured in this special issue of the Journal of General Physiology .
Collapse
Affiliation(s)
- Richard L Moss
- Cardiovascular Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, Madison, WI
| | - Pieter P de Tombe
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - R John Solaro
- Center for Cardiovascular Research, Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
49
|
Tikunova SB, Cuesta A, Price M, Li MX, Belevych N, Biesiadecki BJ, Reiser PJ, Hwang PM, Davis JP. 3-Chlorodiphenylamine activates cardiac troponin by a mechanism distinct from bepridil or TFP. J Gen Physiol 2018; 151:9-17. [PMID: 30442775 PMCID: PMC6314390 DOI: 10.1085/jgp.201812131] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/02/2018] [Indexed: 01/14/2023] Open
Abstract
Cardiac troponin activators could be beneficial in systolic heart failure. Tikunova et al. demonstrate that, unlike previously known calcium sensitizers, the small molecule 3-chlorodiphenylamine does not activate isolated cardiac troponin C but instead activates the intact troponin complex. Despite extensive efforts spanning multiple decades, the development of highly effective Ca2+ sensitizers for the heart remains an elusive goal. Existing Ca2+ sensitizers have other targets in addition to cardiac troponin (cTn), which can lead to adverse side effects, such as hypotension or arrhythmias. Thus, there is a need to design Ca2+-sensitizing drugs with higher affinity and selectivity for cTn. Previously, we determined that many compounds based on diphenylamine (DPA) were able to bind to a cTnC–cTnI chimera with moderate affinity (Kd ∼10–120 µM). Of these compounds, 3-chlorodiphenylamine (3-Cl-DPA) bound most tightly (Kd of 10 µM). Here, we investigate 3-Cl-DPA further and find that it increases the Ca2+ sensitivity of force development in skinned cardiac muscle. Using NMR, we show that, like the known Ca2+ sensitizers, trifluoperazine (TFP) and bepridil, 3-Cl-DPA is able to bind to the isolated N-terminal domain (N-domain) of cTnC (Kd of 6 µM). However, while the bulky molecules of TFP and bepridil stabilize the open state of the N-domain of cTnC, the small and flexible 3-Cl-DPA molecule is able to bind without stabilizing this open state. Thus, unlike TFP, which drastically slows the rate of Ca2+ dissociation from the N-domain of isolated cTnC in a dose-dependent manner, 3-Cl-DPA has no effect on the rate of Ca2+ dissociation. On the other hand, the affinity of 3-Cl-DPA for a cTnC–TnI chimera is at least an order of magnitude higher than that of TFP or bepridil, likely because 3-Cl-DPA is less disruptive of cTnI binding to cTnC. Therefore, 3-Cl-DPA has a bigger effect on the rate of Ca2+ dissociation from the entire cTn complex than TFP and bepridil. Our data suggest that 3-Cl-DPA activates the cTn complex via a unique mechanism and could be a suitable scaffold for the development of novel treatments for systolic heart failure.
Collapse
Affiliation(s)
- Svetlana B Tikunova
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH
| | - Andres Cuesta
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH
| | - Morgan Price
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH
| | - Monica X Li
- Departments of Medicine and Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Natalya Belevych
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH
| | | | - Peter J Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH
| | - Peter M Hwang
- Departments of Medicine and Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH
| |
Collapse
|
50
|
Scheiper S, Ramos-Luis E, Blanco-Verea A, Niess C, Beckmann BM, Schmidt U, Kettner M, Geisen C, Verhoff MA, Brion M, Kauferstein S. Sudden unexpected death in the young - Value of massive parallel sequencing in postmortem genetic analyses. Forensic Sci Int 2018; 293:70-76. [PMID: 30415094 DOI: 10.1016/j.forsciint.2018.09.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/27/2018] [Accepted: 09/29/2018] [Indexed: 01/22/2023]
Abstract
Cases of sudden cardiac death (SCD) in young and apparently healthy individuals represent a devastating event in affected families. Hereditary arrhythmia syndromes, which include primary electrical heart disorders as well as cardiomyopathies, are known to contribute to a significant number of these sudden death cases. We performed postmortem genetic analyses in young sudden death cases (aged <45years) by means of a defined gene panel using massive parallel sequencing (MPS). The data were evaluated bioinformatically and detected sequence variants were assessed using common databases and applying in silico prediction tools. In this study, we identified variants with likely pathogenic effect in 6 of 9 sudden unexpected death (SUD) cases. Due to the detection of numerous unknown and unclassified variants, interpretation of the results proved to be challenging. However, by means of an appropriate evaluation of the findings, MPS represents an important tool to support the forensic investigation and implies great progress for relatives of young SCD victims facilitating adequate risk stratification and genetic counseling.
Collapse
Affiliation(s)
- Stefanie Scheiper
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt, Germany; German Red Cross Blood Center, Institute of Transfusion Medicine and Immunohaematology, University Hospital Frankfurt, Frankfurt, Germany.
| | - Eva Ramos-Luis
- Xenética Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela (A Coruña), Spain; Medicina Xenómica, Universidade de Santiago de Compostela-Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela (A Coruña), Spain
| | - Alejandro Blanco-Verea
- Xenética Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela (A Coruña), Spain; Medicina Xenómica, Universidade de Santiago de Compostela-Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela (A Coruña), Spain
| | - Constanze Niess
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Britt-Maria Beckmann
- Department of Medicine I, University Hospital Munich, Ludwig Maximilians University, Munich, Germany
| | - Ulrike Schmidt
- Institute of Legal Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mattias Kettner
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Christof Geisen
- German Red Cross Blood Center, Institute of Transfusion Medicine and Immunohaematology, University Hospital Frankfurt, Frankfurt, Germany
| | - Marcel A Verhoff
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Maria Brion
- Xenética Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela (A Coruña), Spain; Medicina Xenómica, Universidade de Santiago de Compostela-Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela (A Coruña), Spain
| | - Silke Kauferstein
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| |
Collapse
|