1
|
Ragusa R, Caselli C. Focus on cardiac troponin complex: From gene expression to cardiomyopathy. Genes Dis 2024; 11:101263. [PMID: 39211905 PMCID: PMC11357864 DOI: 10.1016/j.gendis.2024.101263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 09/04/2024] Open
Abstract
The cardiac troponin complex (cTn) is a regulatory component of sarcomere. cTn consists of three subunits: cardiac troponin C (cTnC), which confers Ca2+ sensitivity to muscle; cTnI, which inhibits the interaction of cross-bridge of myosin with thin filament during diastole; and cTnT, which has multiple roles in sarcomere, such as promoting the link between the cTnI-cTnC complex and tropomyosin within the thin filament and influencing Ca2+ sensitivity of cTn and force development during contraction. Conditions that interfere with interactions within cTn and/or other thin filament proteins can be key factors in the regulation of cardiac contraction. These conditions include alterations in myofilament Ca2+ sensitivity, direct changes in cTn function, and triggering downstream events that lead to adverse cardiac remodeling and impairment of heart function. This review describes gene expression and post-translational modifications of cTn as well as the conditions that can adversely affect the delicate balance among the components of cTn, thereby promoting contractile dysfunction.
Collapse
Affiliation(s)
- Rosetta Ragusa
- Institute of Clinical Physiology, CNR, via Moruzzi 1, Pisa 56124, Italy
| | - Chiara Caselli
- Institute of Clinical Physiology, CNR, via Moruzzi 1, Pisa 56124, Italy
- Fondazione Toscana Gabriele Monasterio, via Moruzzi 1, Pisa 56124, Italy
| |
Collapse
|
2
|
Hantz ER, Tikunova SB, Belevych N, Davis JP, Reiser PJ, Lindert S. Targeting Troponin C with Small Molecules Containing Diphenyl Moieties: Calcium Sensitivity Effects on Striated Muscles and Structure-Activity Relationship. J Chem Inf Model 2023; 63:3462-3473. [PMID: 37204863 PMCID: PMC10496875 DOI: 10.1021/acs.jcim.3c00196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Despite large investments from academia and industry, heart failure, which results from a disruption of the contractile apparatus, remains a leading cause of death. Cardiac muscle contraction is a calcium-dependent mechanism, which is regulated by the troponin protein complex (cTn) and specifically by the N-terminal domain of its calcium-binding subunit (cNTnC). There is an increasing need for the development of small molecules that increase calcium sensitivity without altering the systolic calcium concentration, thereby strengthening the cardiac function. Here, we examined the effect of our previously identified calcium-sensitizing small molecule, ChemBridge compound 7930079, in the context of several homologous muscle systems. The effect of this molecule on force generation in isolated cardiac trabeculae and slow skeletal muscle fibers was measured. Furthermore, we explored the use of Gaussian accelerated molecular dynamics in sampling highly predictive receptor conformations based on NMR-derived starting structures. Additionally, we took a rational computational approach for lead optimization based on lipophilic diphenyl moieties. This integrated structural-biochemical-physiological approach led to the identification of three novel low-affinity binders, which had similar binding affinities to the known positive inotrope trifluoperazine. The most potent identified calcium sensitizer was compound 16 with an apparent affinity of 117 ± 17 μM.
Collapse
Affiliation(s)
- Eric R. Hantz
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210
| | - Svetlana B. Tikunova
- Davis Heart and Lung Research Institute and Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210
| | - Natalya Belevych
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210
| | - Jonathan P. Davis
- Davis Heart and Lung Research Institute and Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210
| | - Peter J. Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210
| |
Collapse
|
3
|
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
|
4
|
Hantz ER, Tikunova SB, Belevych N, Davis JP, Reiser PJ, Lindert S. Targeting Troponin C with Small Molecules Containing Diphenyl Moieties: Calcium Sensitivity Effects on Striated Muscle and Structure Activity Relationship. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527323. [PMID: 36798160 PMCID: PMC9934531 DOI: 10.1101/2023.02.06.527323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Despite large investments from academia and industry, heart failure, which results from a disruption of the contractile apparatus, remains a leading cause of death. Cardiac muscle contraction is a calcium-dependent mechanism, which is regulated by the troponin protein complex (cTn) and specifically by the N-terminal domain of its calcium binding subunit (cNTnC). There is an increasing need for the development of small molecules that increase calcium sensitivity without altering systolic calcium concentration, thereby strengthening cardiac function. Here, we examined the effect of our previously identified calcium sensitizing small molecule, ChemBridge compound 7930079, in the context of several homologous muscle systems. The effect of this molecule on force generation in isolated cardiac trabeculae and slow skeletal muscle fibers was measured. Furthermore, we explored the use of Gaussian accelerated molecular dynamics in sampling highly predictive receptor conformations based on NMR derived starting structures. Additionally, we took a rational computational approach for lead optimization based on lipophilic diphenyl moieties. This led to the identification of three novel low affinity binders, which had similar binding affinities to known positive inotrope trifluoperazine. The most potent identified calcium sensitizer was compound 16 with an apparent affinity of 117 ± 17 μM .
Collapse
Affiliation(s)
- Eric R. Hantz
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210
| | - Svetlana B. Tikunova
- Davis Heart and Lung Research Institute and Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210
| | - Natalya Belevych
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210
| | - Jonathan P. Davis
- Davis Heart and Lung Research Institute and Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210
| | - Peter J. Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210,Correspondence to: Department of Chemistry and Biochemistry, Ohio State University, 2114 Newman & Wolfrom Laboratory, 100 W. 18th Avenue, Columbus, OH 43210, 614-292-8284 (office), 614-292-1685 (fax),
| |
Collapse
|
5
|
Prill K, Jones MR, Steffensen K, Teng GZ, Dawson JF. Increasing the calcium sensitivity of muscle using trifluoperazine-induced manipulations in silico, in vitro and in vivo systems. Arch Biochem Biophys 2023; 735:109521. [PMID: 36657606 DOI: 10.1016/j.abb.2023.109521] [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: 05/04/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/18/2023]
Abstract
Many therapeutics for cardiomyopathy treat the symptoms of the disease rather than the underlying mechanism. The mechanism of cardiomyopathy onset is believed to include two means: calcium sensitivity changes and myosin activity alteration. Trifluoperazine is a compound that binds troponin, and other components of the calcium pathway, which impacts calcium regulation of contraction. Here, the ability of TFP to shift calcium sensitivity was examined in vitro with purified proteins and the impact of TFP on heart function was assessed in vivo using embryonic zebrafish. The binding of TFP to troponin was modeled in silico and a model of zebrafish troponin was generated. TFP increased regulated cardiac actomyosin activity in vitro and elevated embryonic zebrafish heart rates at effective drug concentrations. Troponin structural changes predicted in silico suggest altered protein interactions within thin filaments that would affect the regulation of heart function.
Collapse
Affiliation(s)
- Kendal Prill
- From the Department of Molecular & Cellular Biology, University of Guelph; Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | - Michael R Jones
- From the Department of Molecular & Cellular Biology, University of Guelph; Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | - Karl Steffensen
- From the Department of Molecular & Cellular Biology, University of Guelph; Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | - Grace Zi Teng
- From the Department of Molecular & Cellular Biology, University of Guelph; Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - John F Dawson
- From the Department of Molecular & Cellular Biology, University of Guelph; Centre for Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada
| |
Collapse
|
6
|
Genchev GZ, Kobayashi M, Kobayashi T, Lu H. Molecular dynamics provides new insights into the mechanism of calcium signal transduction and interdomain interactions in cardiac troponin. FEBS Open Bio 2021; 11:1841-1853. [PMID: 33085832 PMCID: PMC8255835 DOI: 10.1002/2211-5463.13009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/05/2020] [Accepted: 10/17/2020] [Indexed: 12/16/2022] Open
Abstract
Understanding the regulation of cardiac muscle contraction at a molecular level is crucial for the development of therapeutics for heart conditions. Despite the availability of atomic structures of the protein components of cardiac muscle thin filaments, detailed insights into their dynamics and response to calcium are yet to be fully depicted. In this study, we used molecular dynamics simulations of the core domains of the cardiac muscle protein troponin to characterize the equilibrium dynamics of its calcium-bound and calcium-free forms, with a focus on elements of cardiac muscle contraction activation and deactivation, that is, calcium binding to the cardiac troponin Ca2+ -binding subunit (TnC) and the release of the switch region of the troponin inhibitory subunit (TnI) from TnC. The process of calcium binding to the TnC binding site is described as a three-step process commencing with calcium capture by the binding site residues, followed by cooperative residue interplay bringing the calcium ion to the binding site, and finally, calcium-water exchange. Furthermore, we uncovered a set of TnC-TnI interdomain interactions that are critical for TnC N-lobe hydrophobic pocket dynamics. Absence of these interactions allows the closure of the TnC N-lobe hydrophobic pocket while the TnI switch region remains expelled, whereas if the interactions are maintained, the hydrophobic pocket remains open. Modification of these interactions may fine-tune the ability of the TnC N-lobe hydrophobic pocket to close or remain open, modulate cardiac contractility and present potential therapy-relevant targets.
Collapse
Affiliation(s)
- Georgi Z Genchev
- Center for Biomedical Informatics, Shanghai Children's Hospital, Shanghai, China.,SJTU-Yale Joint Center for Biostatistics, Shanghai Jiao Tong University, Shanghai, China.,Bulgarian Institute for Genomics and Precision Medicine, Sofia, Bulgaria.,Bioinformatics Program, Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Minae Kobayashi
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Tomoyoshi Kobayashi
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Hui Lu
- Center for Biomedical Informatics, Shanghai Children's Hospital, Shanghai, China.,SJTU-Yale Joint Center for Biostatistics, Shanghai Jiao Tong University, Shanghai, China.,Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Alsulami K, Marston S. Small Molecules acting on Myofilaments as Treatments for Heart and Skeletal Muscle Diseases. Int J Mol Sci 2020; 21:E9599. [PMID: 33339418 PMCID: PMC7767104 DOI: 10.3390/ijms21249599] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are the most prevalent forms of the chronic and progressive pathological condition known as cardiomyopathy. These diseases have different aetiologies; however, they share the feature of haemodynamic abnormalities, which is mainly due to dysfunction in the contractile proteins that make up the contractile unit known as the sarcomere. To date, pharmacological treatment options are not disease-specific and rather focus on managing the symptoms, without addressing the disease mechanism. Earliest attempts at improving cardiac contractility by modulating the sarcomere indirectly (inotropes) resulted in unwanted effects. In contrast, targeting the sarcomere directly, aided by high-throughput screening systems, could identify small molecules with a superior therapeutic value in cardiac muscle disorders. Herein, an extensive literature review of 21 small molecules directed to five different targets was conducted. A simple scoring system was created to assess the suitability of small molecules for therapy by evaluating them in eight different criteria. Most of the compounds failed due to lack of target specificity or poor physicochemical properties. Six compounds stood out, showing a potential therapeutic value in HCM, DCM or heart failure (HF). Omecamtiv Mecarbil and Danicamtiv (myosin activators), Mavacamten, CK-274 and MYK-581 (myosin inhibitors) and AMG 594 (Ca2+-sensitiser) are all small molecules that allosterically modulate troponin or myosin. Omecamtiv Mecarbil showed limited efficacy in phase III GALACTIC-HF trial, while, results from phase III EXPLORER-HCM trial were recently published, indicating that Mavacamten reduced left ventricular outflow tract (LVOT) obstruction and diastolic dysfunction and improved the health status of patients with HCM. A novel category of small molecules known as "recouplers" was reported to target a phenomenon termed uncoupling commonly found in familial cardiomyopathies but has not progressed beyond preclinical work. In conclusion, the contractile apparatus is a promising target for new drug development.
Collapse
Affiliation(s)
- Khulud Alsulami
- Imperial Centre for Translational and Experimental Medicine, Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology, Riyadh 11461, Saudi Arabia
| | - Steven Marston
- Imperial Centre for Translational and Experimental Medicine, Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
10
|
Coldren WH, Tikunova SB, Davis JP, Lindert S. Discovery of Novel Small-Molecule Calcium Sensitizers for Cardiac Troponin C: A Combined Virtual and Experimental Screening Approach. J Chem Inf Model 2020; 60:3648-3661. [PMID: 32633957 DOI: 10.1021/acs.jcim.0c00452] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Heart failure is a leading cause of death throughout the world and is triggered by a disruption of the cardiac contractile machinery. This machinery is regulated in a calcium-dependent manner by the protein complex troponin. Calcium binds to the N-terminal domain of cardiac troponin C (cNTnC) setting into motion the cascade of events leading to muscle contraction. Because of the severity and prevalence of heart failure, there is a strong need to develop small-molecule therapeutics designed to increase the calcium sensitivity of cardiac troponin in order to treat this devastating condition. Molecules that are able to stabilize an open configuration of cNTnC and additionally facilitate the binding of the cardiac troponin I (cTnI) switch peptide have the potential to enable increased calcium sensitization and strengthened cardiac function. Here, we employed a high throughput virtual screening methodology built upon the ability of computational docking to reproduce known experimental results and to accurately recognize cNTnC conformations conducive to small molecule binding using a receiver operator characteristic curve analysis. This approach combined with concurrent stopped-flow kinetic experimental verification led to the identification of a number of sensitizers, which slowed the calcium off-rate. An initial hit, compound 4, was identified with medium affinity (84 ± 30 μM). Through refinement, a calcium sensitizing agent, compound 5, with an apparent affinity of 1.45 ± 0.09 μM was discovered. This molecule is one of the highest affinity calcium sensitizers known to date.
Collapse
Affiliation(s)
- William H Coldren
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, Ohio 43210, United States
| | - Svetlana B Tikunova
- Davis Heart and Lung Research Institute and Department of Physiology and Cell Biology, Ohio State University, Columbus, Ohio 43210, United States
| | - Jonathan P Davis
- Davis Heart and Lung Research Institute and Department of Physiology and Cell Biology, Ohio State University, Columbus, Ohio 43210, United States
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
11
|
Studying Calcium Ion-Dependent Effect on the Inter-subunit Interaction Between the cTnC N-terminal Domain and cTnI C-terminal Switch Peptide of Human Cardiac Troponin via Chou’s 5-Steps Rule. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-09875-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Smith AS, Choi E, Gray K, Macadangdang J, Ahn EH, Clark EC, Laflamme MA, Wu JC, Murry CE, Tung L, Kim DH. NanoMEA: A Tool for High-Throughput, Electrophysiological Phenotyping of Patterned Excitable Cells. NANO LETTERS 2020; 20:1561-1570. [PMID: 31845810 PMCID: PMC7547911 DOI: 10.1021/acs.nanolett.9b04152] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Matrix nanotopographical cues are known to regulate the structure and function of somatic cells derived from human pluripotent stem cell (hPSC) sources. High-throughput electrophysiological analysis of excitable cells derived from hPSCs is possible via multielectrode arrays (MEAs) but conventional MEA platforms use flat substrates and do not reproduce physiologically relevant tissue-specific architecture. To address this issue, we developed a high-throughput nanotopographically patterned multielectrode array (nanoMEA) by integrating conductive, ion-permeable, nanotopographic patterns with 48-well MEA plates, and investigated the effect of substrate-mediated cytoskeletal organization on hPSC-derived cardiomyocyte and neuronal function at scale. Using our nanoMEA platform, we found patterned hPSC-derived cardiac monolayers exhibit both enhanced structural organization and greater sensitivity to treatment with calcium blocking or conduction inhibiting compounds when subjected to high-throughput dose-response studies. Similarly, hPSC-derived neurons grown on nanoMEA substrates exhibit faster migration and neurite outgrowth speeds, greater colocalization of pre- and postsynaptic markers, and enhanced cell-cell communication only revealed through examination of data sets derived from multiple technical replicates. The presented data highlight the nanoMEA as a new tool to facilitate high-throughput, electrophysiological analysis of ordered cardiac and neuronal monolayers, which can have important implications for preclinical analysis of excitable cell function.
Collapse
Affiliation(s)
- Alec S.T. Smith
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Eunpyo Choi
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, South Korea
| | - Kevin Gray
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- NanoSurface Biomedical, Inc. Seattle, WA 98195, USA
| | - Jesse Macadangdang
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- NanoSurface Biomedical, Inc. Seattle, WA 98195, USA
| | - Eun Hyun Ahn
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Elisa C. Clark
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Michael A. Laflamme
- Toronto General Hospital Research Institute, McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| | - Charles E. Murry
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- To whom correspondence should be addressed: Dr. Deok-Ho Kim, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Ross Research Building, 715B, 720 Rutland Avenue, Baltimore, MD 21205,
| |
Collapse
|
13
|
Szatkowski L, Lynn ML, Holeman T, Williams MR, Baldo AP, Tardiff JC, Schwartz SD. Proof of Principle that Molecular Modeling Followed by a Biophysical Experiment Can Develop Small Molecules that Restore Function to the Cardiac Thin Filament in the Presence of Cardiomyopathic Mutations. ACS OMEGA 2019; 4:6492-6501. [PMID: 31342001 PMCID: PMC6649307 DOI: 10.1021/acsomega.8b03340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/28/2019] [Indexed: 06/10/2023]
Abstract
This article reports a coupled computational experimental approach to design small molecules aimed at targeting genetic cardiomyopathies. We begin with a fully atomistic model of the cardiac thin filament. To this we dock molecules using accepted computational drug binding methodologies. The candidates are screened for their ability to repair alterations in biophysical properties caused by mutation. Hypertrophic and dilated cardiomyopathies caused by mutation are initially biophysical in nature, and the approach we take is to correct the biophysical insult prior to irreversible cardiac damage. Candidate molecules are then tested experimentally for both binding and biophysical properties. This is a proof of concept study-eventually candidate molecules will be tested in transgenic animal models of genetic (sarcomeric) cardiomyopathies.
Collapse
Affiliation(s)
- Lukasz Szatkowski
- Department
of Chemistry and Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Melissa L. Lynn
- Department of Physiological
Sciences and Department of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Teryn Holeman
- Department of Physiological
Sciences and Department of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Michael R. Williams
- Department
of Chemistry and Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Anthony P. Baldo
- Department
of Chemistry and Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Jil C. Tardiff
- Department of Physiological
Sciences and Department of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Steven D. Schwartz
- Department
of Chemistry and Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
14
|
Potluri PR, Cordina NM, Kachooei E, Brown LJ. Characterization of the L29Q Hypertrophic Cardiomyopathy Mutation in Cardiac Troponin C by Paramagnetic Relaxation Enhancement Nuclear Magnetic Resonance. Biochemistry 2019; 58:908-917. [PMID: 30620548 DOI: 10.1021/acs.biochem.8b01140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The key events in regulating muscle contraction involve the troponin (Tn) heterotrimeric protein complex in which the binding to and release of Ca2+ from the highly conserved troponin C (TnC) subunit trigger a series of structural changes within Tn, and the other thin filament proteins, to result in contraction. In the heart, the control of contraction and relaxation events can be altered by many single-point mutations that may result in cardiomyopathy and sometimes sudden cardiac death. Here we have examined the structural effects of one hypertrophic cardiomyopathy mutation, L29Q, on Ca2+-induced structural transitions within whole TnC. This mutation is of particular interest as several physiological and structural studies have indicated that the response of TnC to Ca2+ binding is altered in the presence of the L29Q mutation, but the structural nature of these changes continues to be debated. In addition, little is known about the effect of this mutation in the Ca2+ free state. Here we have used paramagnetic relaxation enhancement nuclear magnetic resonance (PRE-NMR) to assess the structural effects arising from the L29Q mutation. PRE-NMR distances obtained from a nitroxide spin-label at Cys84 showed that the L29Q mutation perturbs the structure of the TnC N-domain in the presence and absence of Ca2+, with a more "open" TnC N-domain observed in the apo form. In addition, binding of Ca2+ to the TnC-L29Q construct triggers a change in the orientation between the two domains of TnC. Together, these structural perturbations, revealed by PRE-NMR, provide insight into the pathogenesis of this mutation.
Collapse
Affiliation(s)
- Phani R Potluri
- Department of Molecular Sciences , Macquarie University , Sydney , NSW 2109 , Australia
| | - Nicole M Cordina
- Department of Molecular Sciences , Macquarie University , Sydney , NSW 2109 , Australia
| | - Ehsan Kachooei
- Department of Molecular Sciences , Macquarie University , Sydney , NSW 2109 , Australia
| | - Louise J Brown
- Department of Molecular Sciences , Macquarie University , Sydney , NSW 2109 , Australia
| |
Collapse
|
15
|
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
|
16
|
Li MX, Gelozia S, Danmaliki GI, Wen Y, Liu PB, Lemieux MJ, West FG, Sykes BD, Hwang PM. The calcium sensitizer drug MCI-154 binds the structural C-terminal domain of cardiac troponin C. Biochem Biophys Rep 2018; 16:145-151. [PMID: 30417133 PMCID: PMC6218639 DOI: 10.1016/j.bbrep.2018.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 10/12/2018] [Accepted: 10/19/2018] [Indexed: 11/27/2022] Open
Abstract
The compound MCI-154 was previously shown to increase the calcium sensitivity of cardiac muscle contraction. Using solution NMR spectroscopy, we demonstrate that MCI-154 interacts with the calcium-sensing subunit of the cardiac troponin complex, cardiac troponin C (cTnC). Surprisingly, however, it binds only to the structural C-terminal domain of cTnC (cCTnC), and not to the regulatory N-terminal domain (cNTnC) that determines the calcium sensitivity of cardiac muscle. Physiologically, cTnC is always bound to cardiac troponin I (cTnI), so we examined its interaction with MCI-154 in the presence of two soluble constructs, cTnI1–77 and cTnI135–209, which contain all of the segments of cTnI known to interact with cTnC. Neither the cTnC-cTnI1–77 complex nor the cTnC-cTnI135–209 complex binds to MCI-154. Since residues 39–60 of cTnI are known to bind tightly to the cCTnC domain to form a structured core that is invariant throughout the cardiac cycle, we conclude that MCI-154 does not bind to cTnC when it is part of the intact cardiac troponin complex. Thus, MCI-154 likely exerts its calcium sensitizing effect by interacting with a target other than cardiac troponin. MCI-154 is a small molecule calcium sensitizer in cardiac muscle. The N-domain of cardiac troponin C controls calcium sensitivity in cardiac muscle. MCI-154 binds weakly to the promiscuous C-terminal domain of troponin C. Cardiac troponin C does not bind MCI-154 in the presence of troponin I. MCI-154 does not exert its calcium sensitizing effect directly through troponin C.
Collapse
Affiliation(s)
- Monica X Li
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2R3
| | - Shorena Gelozia
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Gaddafi I Danmaliki
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Yurong Wen
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7.,School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Philip B Liu
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2R3
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Frederick G West
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Brian D Sykes
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Peter M Hwang
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2R3.,Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| |
Collapse
|
17
|
Schlecht W, Dong WJ. Dynamic Equilibrium of Cardiac Troponin C's Hydrophobic Cleft and Its Modulation by Ca 2+ Sensitizers and a Ca 2+ Sensitivity Blunting Phosphomimic, cTnT(T204E). Bioconjug Chem 2017; 28:2581-2590. [PMID: 28876897 DOI: 10.1021/acs.bioconjchem.7b00418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Several studies have suggested that conformational dynamics are important in the regulation of thin filament activation in cardiac troponin C (cTnC); however, little direct evidence has been offered to support these claims. In this study, a dye homodimerization approach is developed and implemented that allows the determination of the dynamic equilibrium between open and closed conformations in cTnC's hydrophobic cleft. Modulation of this equilibrium by Ca2+, cardiac troponin I (cTnI), cardiac troponin T (cTnT), Ca2+-sensitizers, and a Ca2+-desensitizing phosphomimic of cTnT (cTnT(T204E) is characterized. Isolated cTnC contained a small open conformation population in the absence of Ca2+ that increased significantly upon the addition of saturating levels of Ca2+. This suggests that the Ca2+-induced activation of thin filament arises from an increase in the probability of hydrophobic cleft opening. The inclusion of cTnI increased the population of open cTnC, and the inclusion of cTnT had the opposite effect. Samples containing Ca2+-desensitizing cTnT(T204E) showed a slight but insignificant decrease in open conformation probability compared to samples with cardiac troponin T, wild type [cTnT(wt)], while Ca2+ sensitizer treated samples generally increased open conformation probability. These findings show that an equilibrium between the open and closed conformations of cTnC's hydrophobic cleft play a significant role in tuning the Ca2+ sensitivity of the heart.
Collapse
Affiliation(s)
- William Schlecht
- The Voiland School of Chemical Engineering and Bioengineering and ‡The Department of Integrated Neuroscience and Physiology, Washington State University , Pullman, Washington 99164, United States
| | - Wen-Ji Dong
- The Voiland School of Chemical Engineering and Bioengineering and ‡The Department of Integrated Neuroscience and Physiology, Washington State University , Pullman, Washington 99164, United States
| |
Collapse
|
18
|
Robertson IM, Pineda-Sanabria SE, Yan Z, Kampourakis T, Sun YB, Sykes BD, Irving M. Reversible Covalent Binding to Cardiac Troponin C by the Ca2+-Sensitizer Levosimendan. Biochemistry 2016; 55:6032-6045. [DOI: 10.1021/acs.biochem.6b00758] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ian M. Robertson
- Randall
Division of Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s College London, New Hunt’s
House, Guy’s Campus, London, SE1 1UL, U.K
- Department
of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Sandra E. Pineda-Sanabria
- Department
of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Ziqian Yan
- Randall
Division of Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s College London, New Hunt’s
House, Guy’s Campus, London, SE1 1UL, U.K
| | - Thomas Kampourakis
- Randall
Division of Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s College London, New Hunt’s
House, Guy’s Campus, London, SE1 1UL, U.K
| | - Yin-Biao Sun
- Randall
Division of Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s College London, New Hunt’s
House, Guy’s Campus, London, SE1 1UL, U.K
| | - Brian D. Sykes
- Department
of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Malcolm Irving
- Randall
Division of Cell and Molecular Biophysics and British Heart Foundation
Centre of Research Excellence, King’s College London, New Hunt’s
House, Guy’s Campus, London, SE1 1UL, U.K
| |
Collapse
|
19
|
Thompson BR, Martindale J, Metzger JM. Sarcomere neutralization in inherited cardiomyopathy: small-molecule proof-of-concept to correct hyper-Ca2+-sensitive myofilaments. Am J Physiol Heart Circ Physiol 2016; 311:H36-43. [PMID: 27199134 DOI: 10.1152/ajpheart.00981.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 05/05/2016] [Indexed: 11/22/2022]
Abstract
The sarcomere is the functional unit of the heart. Alterations in sarcomere activation lead to disease states such as hypertrophic and restrictive cardiomyopathy (HCM/RCM). Mutations in many of the sarcomeric genes are causal for HCM/RCM. In most cases, these mutations result in increased Ca(2+) sensitivity of the sarcomere, giving rise to altered systolic and diastolic function. There is emerging evidence that small-molecule sarcomere neutralization is a potential therapeutic strategy for HCM/RCM. To pursue proof-of-concept, W7 was used here because of its well-known Ca(2+) desensitizer biochemical effects at the level of cardiac troponin C. Acute treatment of adult cardiac myocytes with W7 caused a dose-dependent (1-10 μM) decrease in contractility in a Ca(2+)-independent manner. Alkalosis was used as an in vitro experimental model of acquired heightened Ca(2+) sensitivity, resulting in increased live cell contractility and decreased baseline sarcomere length, which were rapidly corrected with W7. As an inherited cardiomyopathy model, R193H cardiac troponin I (cTnI) transgenic myocytes showed significant decreased baseline sarcomere length and slowed relaxation that were rapidly and dose-dependently corrected by W7. Langendorff whole heart pacing stress showed that R193H cTnI transgenic hearts had elevated end-diastolic pressures at all pacing frequencies compared with hearts from nontransgenic mice. Acute treatment with W7 rapidly restored end-diastolic pressures to normal values in R193H cTnI hearts, supporting a sarcomere intrinsic mechanism of dysfunction. The known off-target effects of W7 notwithstanding, these results provide further proof-of-concept that small-molecule-based sarcomere neutralization is a potential approach to remediate hyper-Ca(2+)-sensitive sarcomere function.
Collapse
Affiliation(s)
- Brian R Thompson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Joshua Martindale
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
20
|
Pineda-Sanabria SE, Robertson IM, Sun YB, Irving M, Sykes BD. Probing the mechanism of cardiovascular drugs using a covalent levosimendan analog. J Mol Cell Cardiol 2016; 92:174-84. [PMID: 26853943 PMCID: PMC4831045 DOI: 10.1016/j.yjmcc.2016.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/24/2016] [Accepted: 02/02/2016] [Indexed: 01/16/2023]
Abstract
One approach to improve contraction in the failing heart is the administration of calcium (Ca2 +) sensitizers. Although it is known that levosimendan and other sensitizers bind to troponin C (cTnC), their in vivo mechanism is not fully understood. Based on levosimendan, we designed a covalent Ca2 + sensitizer (i9) that targets C84 of cTnC and exchanged this complex into cardiac muscle. The NMR structure of the covalent complex showed that i9 binds deep in the hydrophobic pocket of cTnC. Despite slightly reducing troponin I affinity, i9 enhanced the Ca2 + sensitivity of cardiac muscle. We conclude that i9 enhances Ca2 + sensitivity by stabilizing the open conformation of cTnC. These findings provide new insights into the in vivo mechanism of Ca2 + sensitization and demonstrate that directly targeting cTnC has significant potential in cardiovascular therapy. A Ca2 + sensitizer, i9 was designed that forms a covalent bond with C84 of cTnC. i9 stabilized the open state of the N-domain of cTnC. The structure of the covalent cTnC-cTnI-i9 complex was solved by NMR. The structure showed that i9 binds deep in the hydrophobic pocket of cTnC. Despite slightly reducing cTnI affinity, i9 enhanced the Ca2 + sensitivity of cardiac muscle.
Collapse
Affiliation(s)
- Sandra E Pineda-Sanabria
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ian M Robertson
- Randall Division of Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Yin-Biao Sun
- Randall Division of Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Malcolm Irving
- Randall Division of Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Brian D Sykes
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
21
|
Schlecht W, Li KL, Hu D, Dong W. Fluorescence Based Characterization of Calcium Sensitizer Action on the Troponin Complex. Chem Biol Drug Des 2015; 87:171-81. [PMID: 26375298 DOI: 10.1111/cbdd.12651] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/15/2015] [Accepted: 08/05/2015] [Indexed: 11/29/2022]
Abstract
Calcium sensitizers enhance the transduction of the Ca(2+) signal into force within the heart and have found use in treating heart failure. However the mechanisms of action for most Ca(2+) sensitizers remain unclear. To address this issue an efficient fluorescence based approach to Ca(2+) sensitizer screening was developed which monitors cardiac troponin C's (cTnC's) hydrophobic cleft. This approach was tested on four common Ca(2+) -sensitizers, EMD 57033, levosimendan, bepridil and pimobendan with the aim of elucidating the mechanisms of action for each as well as proving the efficacy of the new screening method. Ca(2+) -titration experiments were employed to determine the effect on Ca(2+) sensitivity and cooperativity of cTnC opening, while stopped flow experiments were used to investigate the impact on cTnC relaxation kinetics. Bepridil was shown to increase the sensitivity of cTnC for Ca(2+) under all reconstitution conditions, sensitization by the other drugs was context dependent. Levosimendan and pimobendan reduced the rate of cTnC closing consistent with a stabilization of cTnC's open conformation while bepridil increased the rate of relaxation. Experiments were also run on samples containing cTnT(T204E), a known Ca(2+) -desensitizing phosphorylation mimic. Levosimendan, bepridil, and pimobendan were found to elevate the Ca(2+) -sensitivity of cTnT(T204E) containing samples in this context.
Collapse
Affiliation(s)
- William Schlecht
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, PO Box 646515, Washington State University, Pullman, WA 99164-6515, USA
| | - King-Lun Li
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, PO Box 646515, Washington State University, Pullman, WA 99164-6515, USA
| | - Dehong Hu
- The Environmental and Molecular Science Laboratory, Pacific Northwest National Laboratory, 3335 Innovation Boulevard Richland, WA 99354, USA
| | - Wenji Dong
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, PO Box 646515, Washington State University, Pullman, WA 99164-6515, USA
| |
Collapse
|
22
|
The structural and functional effects of the familial hypertrophic cardiomyopathy-linked cardiac troponin C mutation, L29Q. J Mol Cell Cardiol 2015; 87:257-69. [PMID: 26341255 PMCID: PMC4640586 DOI: 10.1016/j.yjmcc.2015.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/09/2015] [Accepted: 08/23/2015] [Indexed: 01/02/2023]
Abstract
Familial hypertrophic cardiomyopathy (FHC) is characterized by severe abnormal cardiac muscle growth. The traditional view of disease progression in FHC is that an increase in the Ca2 +-sensitivity of cardiac muscle contraction ultimately leads to pathogenic myocardial remodeling, though recent studies suggest this may be an oversimplification. For example, FHC may be developed through altered signaling that prevents downstream regulation of contraction. The mutation L29Q, found in the Ca2 +-binding regulatory protein in heart muscle, cardiac troponin C (cTnC), has been linked to cardiac hypertrophy. However, reports on the functional effects of this mutation are conflicting, and our goal was to combine in vitro and in situ structural and functional data to elucidate its mechanism of action. We used nuclear magnetic resonance and circular dichroism to solve the structure and characterize the backbone dynamics and stability of the regulatory domain of cTnC with the L29Q mutation. The overall structure and dynamics of cTnC were unperturbed, although a slight rearrangement of site 1, an increase in backbone flexibility, and a small decrease in protein stability were observed. The structure and function of cTnC was also assessed in demembranated ventricular trabeculae using fluorescence for in situ structure. L29Q reduced the cooperativity of the Ca2 +-dependent structural change in cTnC in trabeculae under basal conditions and abolished the effect of force-generating myosin cross-bridges on this structural change. These effects could contribute to the pathogenesis of this mutation. The cTnC L29Q mutation causes a small change in the NMR structure of site 1 in cTnC. L29Q reduces the cooperativity of Ca2 +-dependent structural changes in cTnC in situ. L29Q removes the impact of force-generating myosin heads on cTnC structural changes.
Collapse
|
23
|
Papadaki M, Vikhorev PG, Marston SB, Messer AE. Uncoupling of myofilament Ca2+ sensitivity from troponin I phosphorylation by mutations can be reversed by epigallocatechin-3-gallate. Cardiovasc Res 2015; 108:99-110. [PMID: 26109583 DOI: 10.1093/cvr/cvv181] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/11/2015] [Indexed: 02/02/2023] Open
Abstract
AIMS Heart muscle contraction is regulated via the β-adrenergic response that leads to phosphorylation of Troponin I (TnI) at Ser22/23, which changes the Ca(2+) sensitivity of the cardiac myofilament. Mutations in thin filament proteins that cause dilated cardiomyopathy (DCM) and some mutations that cause hypertrophic cardiomyopathy (HCM) abolish the relationship between TnI phosphorylation and Ca(2+) sensitivity (uncoupling). Small molecule Ca(2+) sensitizers and Ca(2+) desensitizers that act upon troponin alter the Ca(2+) sensitivity of the thin filament, but their relationship with TnI phosphorylation has never been studied before. METHODS AND RESULTS Quantitative in vitro motility assay showed that 30 µM EMD57033 and 100 µM Bepridil increase Ca(2+) sensitivity of phosphorylated cardiac thin filaments by 3.1- and 2.8-fold, respectively. Additionally they uncoupled Ca(2+) sensitivity from TnI phosphorylation, mimicking the effect of HCM mutations. Epigallocatechin-3-gallate (EGCG) decreased Ca(2+) sensitivity of phosphorylated and unphosphorylated wild-type thin filaments equally (by 2.15 ± 0.45- and 2.80 ± 0.48-fold, respectively), retaining the coupling. Moreover, EGCG also reduced Ca(2+) sensitivity of phosphorylated but not unphosphorylated thin filaments containing DCM and HCM-causing mutations; thus, the dependence of Ca(2+) sensitivity upon TnI phosphorylation of uncoupled mutant thin filaments was restored in every case. In single mouse heart myofibrils, EGCG reduced Ca(2+) sensitivity of force and kACT and also preserved coupling. Myofibrils from the ACTC E361G (DCM) mouse were uncoupled; EGCG reduced Ca(2+) sensitivity more for phosphorylated than for unphosphorylated myofibrils, thus restoring coupling. CONCLUSION We conclude that it is possible to both mimic and reverse the pathological defects in troponin caused by cardiomyopathy mutations pharmacologically. Re-coupling by EGCG may be of potential therapeutic significance for treating cardiomyopathies.
Collapse
Affiliation(s)
- Maria Papadaki
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Petr G Vikhorev
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Steven B Marston
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Andrew E Messer
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| |
Collapse
|
24
|
Messer AE, Marston SB. Investigating the role of uncoupling of troponin I phosphorylation from changes in myofibrillar Ca(2+)-sensitivity in the pathogenesis of cardiomyopathy. Front Physiol 2014; 5:315. [PMID: 25202278 PMCID: PMC4142463 DOI: 10.3389/fphys.2014.00315] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/02/2014] [Indexed: 12/12/2022] Open
Abstract
Contraction in the mammalian heart is controlled by the intracellular Ca(2+) concentration as it is in all striated muscle, but the heart has an additional signaling system that comes into play to increase heart rate and cardiac output during exercise or stress. β-adrenergic stimulation of heart muscle cells leads to release of cyclic-AMP and the activation of protein kinase A which phosphorylates key proteins in the sarcolemma, sarcoplasmic reticulum and contractile apparatus. Troponin I (TnI) and Myosin Binding Protein C (MyBP-C) are the prime targets in the myofilaments. TnI phosphorylation lowers myofibrillar Ca(2+)-sensitivity and increases the speed of Ca(2+)-dissociation and relaxation (lusitropic effect). Recent studies have shown that this relationship between Ca(2+)-sensitivity and TnI phosphorylation may be unstable. In familial cardiomyopathies, both dilated and hypertrophic (DCM and HCM), a mutation in one of the proteins of the thin filament often results in the loss of the relationship (uncoupling) and blunting of the lusitropic response. For familial dilated cardiomyopathy in thin filament proteins it has been proposed that this uncoupling is causative of the phenotype. Uncoupling has also been found in human heart tissue from patients with hypertrophic obstructive cardiomyopathy as a secondary effect. Recently, it has been found that Ca(2+)-sensitizing drugs can promote uncoupling, whilst one Ca(2+)-desensitizing drug Epigallocatechin 3-Gallate (EGCG) can reverse uncoupling. We will discuss recent findings about the role of uncoupling in the development of cardiomyopathies and the molecular mechanism of the process.
Collapse
Affiliation(s)
- Andrew E. Messer
- National Heart & Lung Institute, Imperial College LondonLondon, UK
| | | |
Collapse
|
25
|
Katrukha IA. Human cardiac troponin complex. Structure and functions. BIOCHEMISTRY (MOSCOW) 2014; 78:1447-65. [DOI: 10.1134/s0006297913130063] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Cordina NM, Liew CK, Gell DA, Fajer PG, Mackay JP, Brown LJ. Effects of calcium binding and the hypertrophic cardiomyopathy A8V mutation on the dynamic equilibrium between closed and open conformations of the regulatory N-domain of isolated cardiac troponin C. Biochemistry 2013; 52:1950-62. [PMID: 23425245 DOI: 10.1021/bi4000172] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Troponin C (TnC) is the calcium-binding subunit of the troponin complex responsible for initiating striated muscle contraction in response to calcium influx. In the skeletal TnC isoform, calcium binding induces a structural change in the regulatory N-domain of TnC that involves a transition from a closed to open structural state and accompanying exposure of a large hydrophobic patch for troponin I (TnI) to subsequently bind. However, little is understood about how calcium primes the N-domain of the cardiac isoform (cTnC) for interaction with the TnI subunit as the open conformation of the regulatory domain of cTnC has been observed only in the presence of bound TnI. Here we use paramagnetic relaxation enhancement (PRE) to characterize the closed to open transition of isolated cTnC in solution, a process that cannot be observed by traditional nuclear magnetic resonance methods. Our PRE data from four spin-labeled monocysteine constructs of isolated cTnC reveal that calcium binding triggers movement of the N-domain helices toward an open state. Fitting of the PRE data to a closed to open transition model reveals the presence of a small population of cTnC molecules in the absence of calcium that possess an open conformation, the level of which increases substantially upon Ca(2+) binding. These data support a model in which calcium binding creates a dynamic equilibrium between the closed and open structural states to prime cTnC for interaction with its target peptide. We also used PRE data to assess the structural effects of a familial hypertrophic cardiomyopathy point mutation located within the N-domain of cTnC (A8V). The PRE data show that the Ca(2+) switch mechanism is perturbed by the A8V mutation, resulting in a more open N-domain conformation in both the apo and holo states.
Collapse
Affiliation(s)
- Nicole M Cordina
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | | | | | | | | | | |
Collapse
|
27
|
The green tea polyphenol (-)-epigallocatechin-3-gallate inhibits magnesium binding to the C-domain of cardiac troponin C. J Muscle Res Cell Motil 2013; 34:107-13. [PMID: 23417789 DOI: 10.1007/s10974-013-9338-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/11/2013] [Indexed: 01/26/2023]
Abstract
Cardiac muscle contraction is activated via the single Ca(2+)-binding site (site II) in the N-domain of troponin C (cTnC). The two Ca(2+)/Mg(2+) binding sites in the C-domain of cTnC (sites III and IV) have been considered to play a purely structural role in anchoring cTnC to the thin filament. However, several recent discoveries suggest a possible role of this domain in contractile regulation. The green tea polyphenol (-)-epigallocatechin 3-gallate (EGCg), which binds specifically to the C-domain of cTnC, reduces cardiac myofilament Ca(2+) sensitivity along with maximum force and acto-myosin ATPase activity. We have determined the effect of EGCg on Ca(2+) and Mg(2+) binding to the C-domain of cTnC. In the absence of Mg(2+) there was no significant effect of EGCg on the Ca(2+)-cTnC affinity. Surprisingly, in the presence of Mg(2+) EGCg caused an increase in Ca(2+) affinity for sites III and IV of cTnC. However, in the absence of Ca(2+) the addition of EGCg caused a significant reduction in Mg(2+)-cTnC affinity. This reduction is presumably responsible for the increase in Ca(2+)-cTnC affinity produced by EGCg in the presence of Mg(2+). We propose that the inhibitory effect of EGCg on myofilament Ca(2+) activation may be related to an enhanced Ca(2+)-Mg(2+)exchange at sites III and IV of cTnC, which might reduce the myosin crossbridge dependent component of thin filament activation.
Collapse
|
28
|
Cordina NM, Liew CK, Gell DA, Fajer PG, Mackay JP, Brown LJ. Interdomain orientation of cardiac troponin C characterized by paramagnetic relaxation enhancement NMR reveals a compact state. Protein Sci 2013; 21:1376-87. [PMID: 22811351 DOI: 10.1002/pro.2124] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiac troponin C (cTnC) is the calcium binding subunit of the troponin complex that triggers the thin filament response to calcium influx into the sarcomere. cTnC consists of two globular EF-hand domains (termed the N- and C-domains) connected by a flexible linker. While the conformation of each domain of cTnC has been thoroughly characterized through NMR studies involving either the isolated N-domain (N-cTnC) or C-domain (C-cTnC), little attention has been paid to the range of interdomain orientations possible in full-length cTnC that arises as a consequence of the flexibility of the domain linker. Flexibility in the domain linker of cTnC is essential for effective regulatory function of troponin. We have therefore utilized paramagnetic relaxation enhancement (PRE) NMR to assess the interdomain orientation of cTnC. Ensemble fitting of our interdomain PRE measurements reveals that isolated cTnC has considerable interdomain flexibility and preferentially adopts a bent conformation in solution, with a defined range of relative domain orientations.
Collapse
Affiliation(s)
- Nicole M Cordina
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | | | | | | | | | | |
Collapse
|
29
|
Colson BA, Gruber SJ, Thomas DD. Structural dynamics of muscle protein phosphorylation. J Muscle Res Cell Motil 2012; 33:419-29. [PMID: 22930331 DOI: 10.1007/s10974-012-9317-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 08/07/2012] [Indexed: 02/03/2023]
Abstract
We have used site-directed spectroscopic probes to detect structural changes, motions, and interactions due to phosphorylation of proteins involved in the regulation of muscle contraction and relaxation. Protein crystal structures provide static snapshots that provide clues to the conformations that are sampled dynamically by proteins in the cellular environment. Our site-directed spectroscopic experiments, combined with computational simulations, extend these studies into functional assemblies in solution, and reveal details of protein regions that are too dynamic or disordered for crystallographic approaches. Here, we discuss phosphorylation-mediated structural transitions in the smooth muscle myosin regulatory light chain, the striated muscle accessory protein myosin binding protein-C, and the cardiac membrane Ca(2+) pump modulator phospholamban. In each of these systems, phosphorylation near the N terminus of the regulatory protein relieves an inhibitory interaction between the phosphoprotein and its regulatory target. Several additional unifying themes emerge from our studies: (a) The effect of phosphorylation is not to change the affinity of the phosphoprotein for its regulated binding partner, but to change the structure of the bound complex without dissociation. (b) Phosphorylation induces transitions between order and dynamic disorder. (c) Structural states are only loosely coupled to phosphorylation; i.e., complete phosphorylation induces dramatic functional effects with only a partial shift in the equilibrium between ordered and disordered structural states. These studies, which offer atomic-resolution insight into the structural and functional dynamics of these phosphoproteins, were inspired in part by the ground-breaking work in this field by Michael and Kate Barany.
Collapse
Affiliation(s)
- Brett A Colson
- Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | |
Collapse
|
30
|
Li AY, Lee J, Borek D, Otwinowski Z, Tibbits GF, Paetzel M. Crystal structure of cardiac troponin C regulatory domain in complex with cadmium and deoxycholic acid reveals novel conformation. J Mol Biol 2011; 413:699-711. [PMID: 21920370 PMCID: PMC4068330 DOI: 10.1016/j.jmb.2011.08.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 08/23/2011] [Accepted: 08/24/2011] [Indexed: 01/07/2023]
Abstract
The amino-terminal regulatory domain of cardiac troponin C (cNTnC) plays an important role as the calcium sensor for the troponin complex. Calcium binding to cNTnC results in conformational changes that trigger a cascade of events that lead to cardiac muscle contraction. The cardiac N-terminal domain of TnC consists of two EF-hand calcium binding motifs, one of which is dysfunctional in binding calcium. Nevertheless, the defunct EF-hand still maintains a role in cNTnC function. For its structural analysis by X-ray crystallography, human cNTnC with the wild-type primary sequence was crystallized under a novel crystallization condition. The crystal structure was solved by the single-wavelength anomalous dispersion method and refined to 2.2 Å resolution. The structure displays several novel features. Firstly, both EF-hand motifs coordinate cadmium ions derived from the crystallization milieu. Secondly, the ion coordination in the defunct EF-hand motif accompanies unusual changes in the protein conformation. Thirdly, deoxycholic acid, also derived from the crystallization milieu, is bound in the central hydrophobic cavity. This is reminiscent of the interactions observed for cardiac calcium sensitizer drugs that bind to the same core region and maintain the "open" conformational state of calcium-bound cNTnC. The cadmium ion coordination in the defunct EF-hand indicates that this vestigial calcium binding site retains the structural and functional elements that allow it to coordinate a cadmium ion. However, it is a result of, or concomitant with, large and unusual structural changes in cNTnC.
Collapse
Affiliation(s)
- Alison Yueh Li
- Department of Molecular Biology and Biochemistry, Simon Fraser University, South Science Building, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
- Department of Biomedical Physiology and Kinesiology, Molecular Cardiac Physiology Group, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
| | - Jaeyong Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, South Science Building, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
| | - Dominika Borek
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Zbyszek Otwinowski
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Glen F. Tibbits
- Department of Molecular Biology and Biochemistry, Simon Fraser University, South Science Building, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
- Department of Biomedical Physiology and Kinesiology, Molecular Cardiac Physiology Group, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
- Cardiovascular Sciences, Child and Family Research Institute, 950 West 28 Ave, Vancouver, BC, Canada V5Z 4H4
| | - Mark Paetzel
- Department of Molecular Biology and Biochemistry, Simon Fraser University, South Science Building, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
- Department of Biomedical Physiology and Kinesiology, Molecular Cardiac Physiology Group, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, Canada, V5A 1S6
- Address correspondence to: Dr. Mark Paetzel, Simon Fraser University, Department of Molecular Biology and Biochemistry, South Science Building, 8888 University Drive, Burnaby, British Columbia, Canada V5A 1S6, Tel.: 778-782-4230, Fax.: 778-782-5583,
| |
Collapse
|
31
|
Huang RYC, Rempel DL, Gross ML. HD exchange and PLIMSTEX determine the affinities and order of binding of Ca2+ with troponin C. Biochemistry 2011; 50:5426-35. [PMID: 21574565 PMCID: PMC3115450 DOI: 10.1021/bi200377c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Troponin C (TnC), present in all striated muscle, is the Ca(2+)-activated trigger that initiates myocyte contraction. The binding of Ca(2+) to TnC initiates a cascade of conformational changes involving the constituent proteins of the thin filament. The functional properties of TnC and its ability to bind Ca(2+) have significant regulatory influence on the contractile reaction of muscle. Changes in TnC may also correlate with cardiac and various other muscle-related diseases. We report here the implementation of the PLIMSTEX strategy (protein ligand interaction by mass spectrometry, titration, and H/D exchange) to elucidate the binding affinity of TnC with Ca(2+) and, more importantly, to determine the order of Ca(2+) binding of the four EF hands of the protein. The four equilibrium constants, K(1) = (5 ± 5) × 10(7) M(-1), K(2) = (1.8 ± 0.8) × 10(7) M(-1), K(3) = (4.2 ± 0.9) × 10(6) M(-1), and K(4) = (1.6 ± 0.6) × 10(6) M(-1), agree well with determinations by other methods and serve to increase our confidence in the PLIMSTEX approach. We determined the order of binding to the four EF hands to be III, IV, II, and I by extracting from the H/DX results the deuterium patterns for each EF hand for each state of the protein (apo through fully Ca(2+) bound). This approach, demonstrated for the first time, may be general for determining binding orders of metal ions and other ligands to proteins.
Collapse
Affiliation(s)
- Richard Y-C. Huang
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130
| | - Don L. Rempel
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130
| | - Michael L. Gross
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
32
|
Pineda-Sanabria SE, Robertson IM, Sykes BD. Structure of trans-resveratrol in complex with the cardiac regulatory protein troponin C. Biochemistry 2011; 50:1309-20. [PMID: 21226534 DOI: 10.1021/bi101985j] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiac troponin, a heterotrimeric protein complex that regulates heart contraction, represents an attractive target for the development of drugs for treating heart disease. Cardiovascular diseases are one of the chief causes of morbidity and mortality worldwide. In France, however, the death rate from heart disease is remarkably low relative to fat consumption. This so-called "French paradox" has been attributed to the high level of consumption of wine in France, and the antioxidant trans-resveratrol is thought to be the primary basis for wine's cardioprotective nature. It has been demonstrated that trans-resveratrol increases the myofilament Ca(2+) sensitivity of guinea pig myocytes [Liew, R., Stagg, M. A., MacLeod, K. T., and Collins, P. (2005) Eur. J. Pharmacol. 519, 1-8]; however, the specific mode of its action is unknown. In this study, the structure of trans-resveratrol free and bound to the calcium-binding protein, troponin C, was determined by nuclear magnetic resonance spectroscopy. The results indicate that trans-resveratrol undergoes a minor conformational change upon binding to the hydrophobic pocket of the C-domain of troponin C. The location occupied by trans-resveratrol coincides with the binding site of troponin I, troponin C's natural binding partner. This has been seen for other troponin C-targeting inotropes and implicates the modulation of the troponin C-troponin I interaction as a possible mechanism of action for trans-resveratrol.
Collapse
Affiliation(s)
- Sandra E Pineda-Sanabria
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | |
Collapse
|
33
|
Robertson IM, Sun YB, Li MX, Sykes BD. A structural and functional perspective into the mechanism of Ca2+-sensitizers that target the cardiac troponin complex. J Mol Cell Cardiol 2010; 49:1031-41. [PMID: 20801130 DOI: 10.1016/j.yjmcc.2010.08.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 08/17/2010] [Accepted: 08/19/2010] [Indexed: 11/25/2022]
Abstract
The Ca(2+) dependent interaction between troponin I (cTnI) and troponin C (cTnC) triggers contraction in heart muscle. Heart failure is characterized by a decrease in cardiac output, and compounds that increase the sensitivity of cardiac muscle to Ca(2+) have therapeutic potential. The Ca(2+)-sensitizer, levosimendan, targets cTnC; however, detailed understanding of its mechanism has been obscured by its instability. In order to understand how this class of positive inotropes function, we investigated the mode of action of two fluorine containing novel analogs of levosimendan; 2',4'-difluoro(1,1'-biphenyl)-4-yloxy acetic acid (dfbp-o) and 2',4'-difluoro(1,1'-biphenyl)-4-yl acetic acid (dfbp). The affinities of dfbp and dfbp-o for the regulatory domain of cTnC were measured in the absence and presence of cTnI by NMR spectroscopy, and dfbp-o was found to bind more strongly than dfbp. Dfbp-o also increased the affinity of cTnI for cTnC. Dfbp-o increased the Ca(2+)-sensitivity of demembranated cardiac trabeculae in a manner similar to levosimendan. The high resolution NMR solution structure of the cTnC-cTnI-dfbp-o ternary complex showed that dfbp-o bound at the hydrophobic interface formed by cTnC and cTnI making critical interactions with residues such as Arg147 of cTnI. In the absence of cTnI, docking localized dfbp-o to the same position in the hydrophobic groove of cTnC. The structural and functional data reveal that the levosimendan class of Ca(2+)-sensitizers work by binding to the regulatory domain of cTnC and stabilizing the pivotal cTnC-cTnI regulatory unit via a network of hydrophobic and electrostatic interactions, in contrast to the destabilizing effects of antagonists such as W7 at the same interface.
Collapse
Affiliation(s)
- Ian M Robertson
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
34
|
Hoffman RMB, Sykes BD. Structure of the inhibitor W7 bound to the regulatory domain of cardiac troponin C. Biochemistry 2009; 48:5541-52. [PMID: 19419198 PMCID: PMC2697600 DOI: 10.1021/bi9001826] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The calmodulin antagonist W7 binds to troponin C in the presence of Ca(2+) and inhibits striated muscle contraction. This study integrates multiple data into the structure of the regulatory domain of human cardiac troponin C (cNTnC) bound to Ca(2+) and W7. The protein-W7 interface is defined through a three-dimensional {(1)H,(13)C}-edited-{(1)H,(12)C}-detected NOESY NMR experiment, and other aspects of the structure are modeled as perturbations to previously known coordinates and restraints. The structure determination protocol optimizes the protein-W7 contacts prior to the introduction of protein-W7 steric interactions or conformational changes in the protein. The structure determination protocol gives families of conformers that all have an optimal docking as assessed by satisfaction of the target function. The structure supports the previously proposed troponin I blocking mechanism for the activity of W7 in striated muscle and suggests a role for the flexible tail of W7 in stabilization of the bound state. This clarifies the structure-activity relationships of W7 and implicates an electrostatically mediated component of activity in common analogues of W7, including the antipsychotic trifluoroperazine and the cardiotonic levosimendan.
Collapse
Affiliation(s)
- Ryan M B Hoffman
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | | |
Collapse
|
35
|
Robertson IM, Li MX, Sykes BD. Solution structure of human cardiac troponin C in complex with the green tea polyphenol, (-)-epigallocatechin 3-gallate. J Biol Chem 2009; 284:23012-23. [PMID: 19542563 DOI: 10.1074/jbc.m109.021352] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Heart muscle contraction is regulated by Ca(2+) binding to the thin filament protein troponin C. In cardiovascular disease, the myofilament response to Ca(2+) is often altered. Compounds that rectify this perturbation are of considerable interest as therapeutics. Plant flavonoids have been found to provide protection against a variety of human illnesses such as cancer, infection, and heart disease. (-)-Epigallocatechin gallate (EGCg), the prevalent flavonoid in green tea, modulates force generation in isolated guinea pig hearts (Hotta, Y., Huang, L., Muto, T., Yajima, M., Miyazeki, K., Ishikawa, N., Fukuzawa, Y., Wakida, Y., Tushima, H., Ando, H., and Nonogaki, T. (2006) Eur. J. Pharmacol. 552, 123-130) and in skinned cardiac muscle fibers (Liou, Y. M., Kuo, S. C., and Hsieh, S. R. (2008) Pflugers Arch. 456, 787-800; and Tadano, N., Yumoto, F., Tanokura, M., Ohtsuki, I., and Morimoto, S. (2005) Biophys. J. 88, 314a). In this study we describe the solution structure of the Ca(2+)-saturated C-terminal domain of troponin C in complex with EGCg. Moreover, we show that EGCg forms a ternary complex with the C-terminal domain of troponin C and the anchoring region of troponin I. The structural evidence indicates that the binding site of EGCg on the C-terminal domain of troponin C is in the hydrophobic pocket in the absence of troponin I, akin to EMD 57033. Based on chemical shift mapping, the binding of EGCg to the C-terminal domain of troponin C in the presence of troponin I may be to a new site formed by the troponin C.troponin I complex. This interaction of EGCg with the C-terminal domain of troponin C.troponin I complex has not been shown with other cardiotonic molecules and illustrates the potential mechanism by which EGCg modulates heart contraction.
Collapse
Affiliation(s)
- Ian M Robertson
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | | | | |
Collapse
|
36
|
Robertson IM, Baryshnikova OK, Li MX, Sykes BD. Defining the binding site of levosimendan and its analogues in a regulatory cardiac troponin C-troponin I complex. Biochemistry 2008; 47:7485-95. [PMID: 18570382 PMCID: PMC2652250 DOI: 10.1021/bi800438k] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The interaction of Cardiac Troponin C (cTnC) and Cardiac Troponin I (cTnI) plays a critical role in transmitting the Ca (2+) signal to the other myofilament proteins in the activation of cardiac muscle contraction. As such, the cTnC-cTnI interface is a logical target for cardiotonic agents such as levosimendan that can modulate the Ca (2+) sensitivity of the myofilaments. Evidence indicates that drug candidates may exert their effects by targeting a site formed by binding of the switch region of cTnI to the regulatory N domain of cTnC (cNTnC). In this study, we utilized two-dimensional (1)H- (15)N HSQC NMR spectroscopy to monitor the binding of levosimendan and its analogues, CMDP, AMDP, CI-930, imazodan, and MPDP, to cNTnC.Ca (2+) in complex with two versions of the switch region of cTnI (cTnI 147-163 and cTnI 144-163). Levosimendan, CMDP, AMDP, and CI-930 were found to bind to both cNTnC.Ca (2+).cTnI 147-163 and cNTnC.Ca (2+).cTnI 144-163 complexes. These compounds contain a methyl group that is absent in MPDP or imazodan. Thus, the methyl group is one of the pharmacophores responsible for the action of these pyridazinone drugs on cTnC. Furthermore, the results showed that the cNTnC.Ca (2+).cTnI 144-163 complex presents a higher-affinity binding site for these compounds than the cNTnC.Ca (2+).cTnI 147-163 complex. This is consistent with our observation that the affinity of cTnI 144-163 for cNTnC.Ca (2+) is approximately 10-fold stronger than that of cTnI 147-163, likely a result of electrostatic forces between the N-terminal RRV extension in cTnI 144-163 and the acidic residues in the C and D helices of cNTnC. These results will help in the delineation of the mode of action of levosimendan on the important functional unit of cardiac troponin that constitutes the regulatory domain of cTnC and the switch region of cTnI.
Collapse
Affiliation(s)
- Ian M Robertson
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | | | | | | |
Collapse
|