1
|
Bogomolova AP, Katrukha IA. Troponins and Skeletal Muscle Pathologies. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2083-2106. [PMID: 39865025 DOI: 10.1134/s0006297924120010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/19/2024] [Accepted: 12/01/2024] [Indexed: 01/28/2025]
Abstract
Skeletal muscles account for ~30-40% of the total weight of human body and are responsible for its most important functions, including movement, respiration, thermogenesis, and glucose and protein metabolism. Skeletal muscle damage negatively impacts the whole-body functioning, leading to deterioration of the quality of life and, in severe cases, death. Therefore, timely diagnosis and therapy for skeletal muscle dysfunction are important goals of modern medicine. In this review, we focused on the skeletal troponins that are proteins in the thin filaments of muscle fibers. Skeletal troponins play a key role in regulation of muscle contraction. Biochemical properties of these proteins and their use as biomarkers of skeletal muscle damage are described in this review. One of the most convenient and sensitive methods of protein biomarker measurement in biological liquids is immunochemical analysis; hence, we examined the factors that influence immunochemical detection of skeletal troponins and should be taken into account when developing diagnostic test systems. Also, we reviewed the available data on the skeletal troponin mutations that are considered to be associated with pathologies leading to the development of diseases and discussed utilization of troponins as drug targets for treatment of the skeletal muscle disorders.
Collapse
Affiliation(s)
- Agnessa P Bogomolova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Hytest Ltd., Turku, Finland
| | - Ivan A Katrukha
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Hytest Ltd., Turku, Finland
| |
Collapse
|
2
|
Sevrieva IR, Kampourakis T, Irving M. Structural changes in troponin during activation of skeletal and heart muscle determined in situ by polarised fluorescence. Biophys Rev 2024; 16:753-772. [PMID: 39830118 PMCID: PMC11735716 DOI: 10.1007/s12551-024-01245-y] [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: 08/04/2024] [Accepted: 09/30/2024] [Indexed: 01/22/2025] Open
Abstract
Calcium binding to troponin triggers the contraction of skeletal and heart muscle through structural changes in the thin filaments that allow myosin motors from the thick filaments to bind to actin and drive filament sliding. Here, we review studies in which those changes were determined in demembranated fibres of skeletal and heart muscle using fluorescence for in situ structure (FISS), which determines domain orientations using polarised fluorescence from bifunctional rhodamine attached to cysteine pairs in the target domain. We describe the changes in the orientations of the N-terminal lobe of troponin C (TnCN) and the troponin IT arm in skeletal and cardiac muscle cells associated with contraction and compare the orientations with those determined in isolated cardiac thin filaments by cryo-electron microscopy. We show that the orientations of the IT arm determined by the two approaches are essentially the same and that this region acts as an almost rigid scaffold for regulatory changes in the more mobile regions of troponin. However, the TnCN orientations determined by the two methods are clearly distinct in both low- and high-calcium conditions. We discuss the implications of these results for the role of TnCN in mediating the multiple signalling pathways acting through troponin in heart muscle cells and the general advantages and limitations of FISS and cryo-EM for determining protein domain orientations in cells and multiprotein complexes.
Collapse
Affiliation(s)
- Ivanka R Sevrieva
- Randall Centre for Cell & Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, UK
| | - Thomas Kampourakis
- Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Malcolm Irving
- Randall Centre for Cell & Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, UK
| |
Collapse
|
3
|
Spahiu E, Uta P, Kraft T, Nayak A, Amrute-Nayak M. Influence of native thin filament type on the regulation of atrial and ventricular myosin motor activity. J Biol Chem 2024; 300:107854. [PMID: 39369990 PMCID: PMC11570844 DOI: 10.1016/j.jbc.2024.107854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
Ca2+-mediated activation of thin filaments is a crucial step in initiating striated muscle contraction. To gain mechanistic insight into this regulatory process, thin filament (TF) components and myosin motors from diverse species and tissue sources are often combined in minimal in vitro systems. The contribution of tissue-specific TF composition with native myosin motors in generating contraction speed remains unclear. To examine TF-mediated regulation, we established a procedure to purify native TFs (nTF) and myosin motors (M-II) from the same cardiac tissue samples as low as 10 mg and investigated their influence on gliding speeds and Ca2+ sensitivity. The rabbit atrial and ventricular nTFs and M-II were assessed in in vitro nTF motility experiments under varying Ca2+ concentrations. The speed-pCa relationship yielded a maximum TF speed of 2.58 μm/s for atrial (aM-II) and 1.51 μm/s for ventricular myosin (vM-II), both higher than the respective unregulated actin filament gliding speeds. The Ca2+ sensitivity was different for both protein sources. After swapping the nTFs, the ventricular TFs increased their gliding speed on atrial myosin, while the atrial nTFs reduced their gliding speed on ventricular myosin. Swapping of the nTFs decreased the calcium sensitivity for both vM-II and aM-II, indicating a strong influence of the thin filament source. These studies suggest that the nTF-myosin combination is critical to understanding the Ca2+ sensitivity of the shortening speed. Our approach is highly relevant to studying precious human cardiac samples, that is, small myectomy samples, to address the alteration of contraction speed and Ca2+ sensitivity in cardiomyopathies.
Collapse
Affiliation(s)
- Emrulla Spahiu
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Petra Uta
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Arnab Nayak
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany.
| | - Mamta Amrute-Nayak
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
4
|
Lungu CN, Mangalagiu II, Gurau G, Mehedinti MC. Variations of VEGFR2 Chemical Space: Stimulator and Inhibitory Peptides. Int J Mol Sci 2024; 25:7787. [PMID: 39063029 PMCID: PMC11276785 DOI: 10.3390/ijms25147787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The kinase pathway plays a crucial role in blood vessel function. Particular attention is paid to VEGFR type 2 angiogenesis and vascular morphogenesis as the tyrosine kinase pathway is preferentially activated. In silico studies were performed on several peptides that affect VEGFR2 in both stimulating and inhibitory ways. This investigation aims to examine the molecular properties of VEGFR2, a molecule primarily involved in the processes of vasculogenesis and angiogenesis. These relationships were defined by the interactions between Vascular Endothelial Growth Factor receptor 2 (VEGFR2) and the structural features of the systems. The chemical space of the inhibitory peptides and stimulators was described using topological and energetic properties. Furthermore, chimeric models of stimulating and inhibitory proteins (for VEGFR2) were computed using the protein system structures. The interaction between the chimeric proteins and VEGFR was computed. The chemical space was further characterized using complex manifolds and high-dimensional data visualization. The results show that a slightly similar chemical area is shared by VEGFR2 and stimulating and inhibitory proteins. On the other hand, the stimulator peptides and the inhibitors have distinct chemical spaces.
Collapse
Affiliation(s)
- Claudiu N. Lungu
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania; (G.G.); (M.C.M.)
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
| | - Ionel I. Mangalagiu
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
| | - Gabriela Gurau
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania; (G.G.); (M.C.M.)
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
| | - Mihaela Cezarina Mehedinti
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania; (G.G.); (M.C.M.)
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 11 Carol 1st Bvd, 700506 Iasi, Romania
| |
Collapse
|
5
|
Donkervoort S, van de Locht M, Ronchi D, Reunert J, McLean CA, Zaki M, Orbach R, de Winter JM, Conijn S, Hoomoedt D, Neto OLA, Magri F, Viaene AN, Foley AR, Gorokhova S, Bolduc V, Hu Y, Acquaye N, Napoli L, Park JH, Immadisetty K, Miles LB, Essawi M, McModie S, Ferreira LF, Zanotti S, Neuhaus SB, Medne L, ElBagoury N, Johnson KR, Zhang Y, Laing NG, Davis MR, Bryson-Richardson RJ, Hwee DT, Hartman JJ, Malik FI, Kekenes-Huskey PM, Comi GP, Sharaf-Eldin W, Marquardt T, Ravenscroft G, Bönnemann CG, Ottenheijm CAC. Pathogenic TNNI1 variants disrupt sarcomere contractility resulting in hypo- and hypercontractile muscle disease. Sci Transl Med 2024; 16:eadg2841. [PMID: 38569017 DOI: 10.1126/scitranslmed.adg2841] [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: 05/23/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
Troponin I (TnI) regulates thin filament activation and muscle contraction. Two isoforms, TnI-fast (TNNI2) and TnI-slow (TNNI1), are predominantly expressed in fast- and slow-twitch myofibers, respectively. TNNI2 variants are a rare cause of arthrogryposis, whereas TNNI1 variants have not been conclusively established to cause skeletal myopathy. We identified recessive loss-of-function TNNI1 variants as well as dominant gain-of-function TNNI1 variants as a cause of muscle disease, each with distinct physiological consequences and disease mechanisms. We identified three families with biallelic TNNI1 variants (F1: p.R14H/c.190-9G>A, F2 and F3: homozygous p.R14C), resulting in loss of function, manifesting with early-onset progressive muscle weakness and rod formation on histology. We also identified two families with a dominantly acting heterozygous TNNI1 variant (F4: p.R174Q and F5: p.K176del), resulting in gain of function, manifesting with muscle cramping, myalgias, and rod formation in F5. In zebrafish, TnI proteins with either of the missense variants (p.R14H; p.R174Q) incorporated into thin filaments. Molecular dynamics simulations suggested that the loss-of-function p.R14H variant decouples TnI from TnC, which was supported by functional studies showing a reduced force response of sarcomeres to submaximal [Ca2+] in patient myofibers. This contractile deficit could be reversed by a slow skeletal muscle troponin activator. In contrast, patient myofibers with the gain-of-function p.R174Q variant showed an increased force to submaximal [Ca2+], which was reversed by the small-molecule drug mavacamten. Our findings demonstrated that TNNI1 variants can cause muscle disease with variant-specific pathomechanisms, manifesting as either a hypo- or a hypercontractile phenotype, suggesting rational therapeutic strategies for each mechanism.
Collapse
Affiliation(s)
- Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Martijn van de Locht
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Dario Ronchi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, 20135, Italy
| | - Janine Reunert
- Department of General Pediatrics, University of Münster, Münster, 48149, Germany
| | - Catriona A McLean
- Department of Anatomical Pathology, Alfred Hospital, Melbourne, Victoria, 3004, Australia
- Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Victoria, 3168, Australia
| | - Maha Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Rotem Orbach
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Josine M de Winter
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Stefan Conijn
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Daan Hoomoedt
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| | - Osorio Lopes Abath Neto
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Francesca Magri
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, 20122, Italy
| | - Angela N Viaene
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104 PA, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Svetlana Gorokhova
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Medical Genetics, Timone Children's Hospital, APHM, Marseille, 13005, France
- INSERM, U1251-MMG, Aix-Marseille Université, Marseille, 13009, France
| | - Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Acquaye
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura Napoli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Julien H Park
- Department of General Pediatrics, University Hospital Münster, Münster, 48149 Germany
| | - Kalyan Immadisetty
- Department of Cell and Molecular Physiology, Loyola University, Chicago, IL 60153, USA
| | - Lee B Miles
- School of Biological Sciences, Monash University, Melbourne, Victoria, 3800, Australia
| | - Mona Essawi
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Salar McModie
- Department of Neurology, Alfred Health, Melbourne, Victoria, 3004, Australia
| | - Leonardo F Ferreira
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Simona Zanotti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Sarah B Neuhaus
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Livija Medne
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nagham ElBagoury
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Kory R Johnson
- Bioinformatics Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong Zhang
- Bioinformatics Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nigel G Laing
- Neurogenetics Unit, Department of Diagnostic Genomics, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
- Centre for Medical Research University of Western Australia, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Mark R Davis
- Neurogenetics Unit, Department of Diagnostic Genomics, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | | | - Darren T Hwee
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - James J Hartman
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Fady I Malik
- Research and Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | | | - Giacomo Pietro Comi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, 20135, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Disease Unit, Milan, 20122, Italy
| | - Wessam Sharaf-Eldin
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12622, Egypt
| | - Thorsten Marquardt
- Department of General Pediatrics, University of Münster, Münster, 48149, Germany
| | - Gianina Ravenscroft
- Centre for Medical Research University of Western Australia, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, 1081 HV Netherlands
| |
Collapse
|
6
|
Ronca F, Raggi A. Role of the interaction between troponin T and AMP deaminase by zinc bridge in modulating muscle contraction and ammonia production. Mol Cell Biochem 2024; 479:793-809. [PMID: 37184757 PMCID: PMC11016001 DOI: 10.1007/s11010-023-04763-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/06/2023] [Indexed: 05/16/2023]
Abstract
The N-terminal region of troponin T (TnT) does not bind any protein of the contractile machinery and the role of its hypervariability remains uncertain. In this review we report the evidence of the interaction between TnT and AMP deaminase (AMPD), a regulated zinc enzyme localized on the myofibril. In periods of intense muscular activity, a decrease in the ATP/ADP ratio, together with a decrease in the tissue pH, is the stimulus for the activation of the enzyme that deaminating AMP to IMP and NH3 displaces the myokinase reaction towards the formation of ATP. In skeletal muscle subjected to strong tetanic contractions, a calpain-like proteolytic activity produces the removal in vivo of a 97-residue N-terminal fragment from the enzyme that becomes desensitized towards the inhibition by ATP, leading to an unrestrained production of NH3. When a 95-residue N-terminal fragment is removed from AMPD by trypsin, simulating in vitro the calpain action, rabbit fast TnT or its phosphorylated 50-residue N-terminal peptide binds AMPD restoring the inhibition by ATP. Taking in consideration that the N-terminus of TnT expressed in human as well as rabbit white muscle contains a zinc-binding motif, we suggest that TnT might mimic the regulatory action of the inhibitory N-terminal domain of AMPD due to the presence of a zinc ion connecting the N-terminal and C-terminal regions of the enzyme, indicating that the two proteins might physiologically associate to modulate muscle contraction and ammonia production in fast-twitching muscle under strenuous conditions.
Collapse
Affiliation(s)
- Francesca Ronca
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Antonio Raggi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| |
Collapse
|
7
|
Ashrafi E, Radisic M, Elliott JAW. Systematic cryopreservation study of cardiac myoblasts in suspension. PLoS One 2024; 19:e0295131. [PMID: 38446773 PMCID: PMC10917286 DOI: 10.1371/journal.pone.0295131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/15/2023] [Indexed: 03/08/2024] Open
Abstract
H9c2 myoblasts are a cell line derived from embryonic rat heart tissue and demonstrate the ability to differentiate to cardiac myotubes upon reduction of the serum concentration (from 10% to 1%) and addition of all-trans retinoic acid in the growth medium. H9c2 cells are increasingly being used as an easy-to-culture proxy for some functions of cardiomyocytes. The cryobiology of cardiac cells including H9c2 myoblasts has not been studied as extensively as that of some cell types. Consequently, it is important to characterize the cryobiological response and systematically develop well-optimized cryopreservation protocols for H9c2 cells to have optimal and consistent viability and functionality after thaw for high quality studies with this cell type. In this work, an interrupted slow cooling protocol (graded freezing) was applied to characterize H9c2 response throughout the cooling profile. Important factors that affect the cell response were examined, and final protocols that provided the highest post-thaw viability are reported. One protocol uses the common cryoprotectant dimethyl sulfoxide combined with hydroxyethyl starch, which will be suitable for applications in which the presence of dimethyl sulfoxide is not an issue; and the other protocol uses glycerol as a substitute when there is a desire to avoid dimethyl sulfoxide. Both protocols achieved comparable post-thaw viabilities (higher than 80%) based on SYTO 13/GelRed flow cytometry results. H9c2 cells cryopreserved by either protocol showed ability to differentiate to cardiac myotubes comparable to fresh (unfrozen) H9c2 cells, and their differentiation to cardiac myotubes was confirmed with i) change in cell morphology, ii) expression of cardiac marker troponin I, and iii) increase in mitochondrial mass.
Collapse
Affiliation(s)
- Elham Ashrafi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Janet A. W. Elliott
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
8
|
Barry ME, Rynkiewicz MJ, Pavadai E, Viana A, Lehman W, Moore JR. Glutamate 139 of tropomyosin is critical for cardiac thin filament blocked-state stabilization. J Mol Cell Cardiol 2024; 188:30-37. [PMID: 38266978 PMCID: PMC11654406 DOI: 10.1016/j.yjmcc.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/14/2023] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
The cardiac thin filament proteins troponin and tropomyosin control actomyosin formation and thus cardiac contractility. Calcium binding to troponin changes tropomyosin position along the thin filament, allowing myosin head binding to actin required for heart muscle contraction. The thin filament regulatory proteins are hot spots for genetic mutations causing heart muscle dysfunction. While much of the thin filament structure has been characterized, critical regions of troponin and tropomyosin involved in triggering conformational changes remain unresolved. A poorly resolved region, helix-4 (H4) of troponin I, is thought to stabilize tropomyosin in a position on actin that blocks actomyosin interactions at low calcium concentrations during muscle relaxation. We have proposed that contact between glutamate 139 on tropomyosin and positively charged residues on H4 leads to blocking-state stabilization. In this study, we attempted to disrupt these interactions by replacing E139 with lysine (E139K) to define the importance of this residue in thin filament regulation. Comparison of mutant and wild-type tropomyosin was carried out using in-vitro motility assays, actin co-sedimentation, and molecular dynamics simulations to determine perturbations in troponin-tropomyosin function caused by the tropomyosin mutation. Motility assays revealed that mutant thin filaments moved at higher velocity at low calcium with increased calcium sensitivity demonstrating that tropomyosin residue 139 is vital for proper tropomyosin-mediated inhibition during relaxation. Similarly, molecular dynamic simulations revealed a mutation-induced decrease in interaction energy between tropomyosin-E139K and troponin I (R170 and K174). These results suggest that salt-bridge stabilization of tropomyosin position by troponin IH4 is essential to prevent actomyosin interactions during cardiac muscle relaxation.
Collapse
Affiliation(s)
- Meaghan E Barry
- Department of Biological Sciences, University of Massachusetts Lowell, One University Ave, Lowell, MA 01854, United States of America
| | - Michael J Rynkiewicz
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisan School of Medicine, 700 Albany Street, W-408E, Boston, MA 02118, United States of America
| | - Elumalai Pavadai
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisan School of Medicine, 700 Albany Street, W-408E, Boston, MA 02118, United States of America
| | - Alex Viana
- Department of Biological Sciences, University of Massachusetts Lowell, One University Ave, Lowell, MA 01854, United States of America
| | - William Lehman
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisan School of Medicine, 700 Albany Street, W-408E, Boston, MA 02118, United States of America
| | - Jeffrey R Moore
- Department of Biological Sciences, University of Massachusetts Lowell, One University Ave, Lowell, MA 01854, United States of America.
| |
Collapse
|
9
|
Bai J, Wei X. Identification of teleost tnnc1a enhancers for specific pan-cardiac transcription. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582099. [PMID: 38464177 PMCID: PMC10925198 DOI: 10.1101/2024.02.26.582099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Troponin C regulates muscle contraction by forming the troponin complex with troponin I and troponin T. Different muscle types express different troponin C genes. The mechanisms of such differential transcription are not fully understood. The Zebrafish tnnc1a gene is restrictively expressed in cardiac muscles. We here identify the enhancers and promoters of the zebrafish and medaka tnnc1a genes, including intronic enhancers in zebrafish and medaka and an upstream enhancer in the medaka. The intronic and upstream enhancers are likely functionally redundant. The GFP transgenic reporter driven by these enhancers is expressed more strongly in the ventricle than in the atrium, recapitulating the expression pattern of the endogenous zebrafish tnnc1a gene. Our study identifies a new set of enhancers for cardiac-specific transgenic expression in zebrafish. These enhancers can serve as tools for future identification of transcription factor networks that drive cardiac-specific gene transcription.
Collapse
|
10
|
Chapman EA, Roberts DS, Tiambeng TN, Andrews J, Wang MD, Reasoner EA, Melby JA, Li BH, Kim D, Alpert AJ, Jin S, Ge Y. Structure and dynamics of endogenous cardiac troponin complex in human heart tissue captured by native nanoproteomics. Nat Commun 2023; 14:8400. [PMID: 38110393 PMCID: PMC10728164 DOI: 10.1038/s41467-023-43321-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/07/2023] [Indexed: 12/20/2023] Open
Abstract
Protein complexes are highly dynamic entities that display substantial diversity in their assembly, post-translational modifications, and non-covalent interactions, allowing them to play critical roles in various biological processes. The heterogeneity, dynamic nature, and low abundance of protein complexes in their native states present challenges to study using conventional structural biology techniques. Here we develop a native nanoproteomics strategy for the enrichment and subsequent native top-down mass spectrometry (nTDMS) analysis of endogenous cardiac troponin (cTn) complex directly from human heart tissue. The cTn complex is enriched and purified using peptide-functionalized superparamagnetic nanoparticles under non-denaturing conditions to enable the isotopic resolution of cTn complex, revealing their complex structure and assembly. Moreover, nTDMS elucidates the stoichiometry and composition of the cTn complex, localizes Ca2+ binding domains, defines cTn-Ca2+ binding dynamics, and provides high-resolution mapping of the proteoform landscape. This native nanoproteomics strategy opens a paradigm for structural characterization of endogenous native protein complexes.
Collapse
Affiliation(s)
- Emily A Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - David S Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Timothy N Tiambeng
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jãán Andrews
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Man-Di Wang
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Emily A Reasoner
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jake A Melby
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Brad H Li
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Donguk Kim
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Song Jin
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
11
|
Yang Z, Marston SB, Gould IR. Modulation of Structure and Dynamics of Cardiac Troponin by Phosphorylation and Mutations Revealed by Molecular Dynamics Simulations. J Phys Chem B 2023; 127:8736-8748. [PMID: 37791815 PMCID: PMC10591477 DOI: 10.1021/acs.jpcb.3c02337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/08/2023] [Indexed: 10/05/2023]
Abstract
Adrenaline acts on β1 receptors in the heart muscle to enhance contractility, increase the heart rate, and increase the rate of relaxation (lusitropy) via activation of the cyclic AMP-dependent protein kinase, PKA. Phosphorylation of serines 22 and 23 in the N-terminal peptide of cardiac troponin I is responsible for lusitropy. Mutations associated with cardiomyopathy suppress the phosphorylation-dependent change. Key parts of troponin responsible for this modulatory system are disordered and cannot be resolved by conventional structural approaches. We performed all-atom molecular dynamics simulations (5 × 1.5 μs runs) of the troponin core (419 amino acids) in the presence of Ca2+ in the bisphosphorylated and unphosphorylated states for both wild-type troponin and the troponin C (cTnC) G159D mutant. PKA phosphorylation affects troponin dynamics. There is significant rigidification of the structure involving rearrangement of the cTnI(1-33)-cTnC interaction and changes in the distribution of the cTnC helix A/B angle, troponin I (cTnI) switch peptide (149-164) docking, and the angle between the regulatory head and ITC arm domains. The familial dilated cardiomyopathy cTnC G159D mutation whose Ca2+ sensitivity is not modulated by cTnI phosphorylation exhibits a structure inherently more rigid than the wild type, with phosphorylation reversing the direction of all metrics relative to the wild type.
Collapse
Affiliation(s)
- Zeyu Yang
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, Shepherd’s Bush, London W12 0BZ, U.K.
- Institute
of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, Shepherd’s Bush, London W12 0BZ, U.K.
| | - Steven B. Marston
- National
Heart & Lung Institute, Imperial College
London, London W12 0NN, U.K.
| | - Ian R. Gould
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, Shepherd’s Bush, London W12 0BZ, U.K.
- Institute
of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, Shepherd’s Bush, London W12 0BZ, U.K.
| |
Collapse
|
12
|
Bradshaw M, Squire JM, Morris E, Atkinson G, Richardson R, Lees J, Caputo M, Bigotti GM, Paul DM. Zebrafish as a model for cardiac disease; Cryo-EM structure of native cardiac thin filaments from Danio Rerio. J Muscle Res Cell Motil 2023; 44:179-192. [PMID: 37480427 PMCID: PMC10542308 DOI: 10.1007/s10974-023-09653-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 07/04/2023] [Indexed: 07/24/2023]
Abstract
Actin, tropomyosin and troponin, the proteins that comprise the contractile apparatus of the cardiac thin filament, are highly conserved across species. We have used cryo-EM to study the three-dimensional structure of the zebrafish cardiac thin and actin filaments. With 70% of human genes having an obvious zebrafish orthologue, and conservation of 85% of disease-causing genes, zebrafish are a good animal model for the study of human disease. Our structure of the zebrafish thin filament reveals the molecular interactions between the constituent proteins, showing that the fundamental organisation of the complex is the same as that reported in the human reconstituted thin filament. A reconstruction of zebrafish cardiac F-actin demonstrates no deviations from human cardiac actin over an extended length of 14 actin subunits. Modelling zebrafish homology models into our maps enabled us to compare, in detail, the similarity with human models. The structural similarities of troponin-T in particular, a region known to contain a hypertrophic cardiomyopathy 'hotspot', confirm the suitability of zebrafish to study these disease-causing mutations.
Collapse
Affiliation(s)
- Marston Bradshaw
- Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - John M Squire
- Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Edward Morris
- University of Glasgow, Glasgow, UK
- Institute of Cancer Research, London, UK
| | - Georgia Atkinson
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Rebecca Richardson
- Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Jon Lees
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Massimo Caputo
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Giulia M Bigotti
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Danielle M Paul
- Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK.
| |
Collapse
|
13
|
Marston S. Recent studies of the molecular mechanism of lusitropy due to phosphorylation of cardiac troponin I by protein kinase A. J Muscle Res Cell Motil 2023; 44:201-208. [PMID: 36131171 PMCID: PMC10541847 DOI: 10.1007/s10974-022-09630-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/28/2022] [Indexed: 10/14/2022]
Abstract
Ca2+ acts on troponin and tropomyosin to switch the thin filament on and off, however in cardiac muscle a more graded form of regulation is essential to tailor cardiac output to the body's needs. This is achieved by the action of adrenaline on β1 receptors of heart muscle cells leading to enhanced contractility, faster heart rate and faster relaxation (lusitropy) via activation of the cyclic AMP-dependent protein kinase, PKA. PKA phosphorylates serines 22 and 23 in the N-terminal peptide of cardiac troponin I. As a consequence the rate of Ca2+release from troponin is increased. This is the key determinant of lusitropy. The molecular mechanism of this process has remained unknown long after the mechanism of the troponin Ca2+ switch itself was defined. Investigation of this subtle process at the atomic level poses a challenge, since the change in Ca2+-sensitivity is only about twofold and key parts of the troponin modulation and regulation system are disordered and cannot be fully resolved by conventional structural approaches. We will review recent studies using molecular dynamics simulations together with functional, cryo-em and NMR techniques that have started to give us a precise picture of how phosphorylation of troponin I modulates the dynamics of troponin to produce the lusitropic effect.
Collapse
|
14
|
Chapman EA, Roberts DS, Tiambeng TN, Andrews J, Wang MD, Reasoner EA, Melby JA, Li BH, Kim D, Alpert AJ, Jin S, Ge Y. Structure and dynamics of endogenous protein complexes in human heart tissue captured by native nanoproteomics. RESEARCH SQUARE 2023:rs.3.rs-3108087. [PMID: 37461709 PMCID: PMC10350235 DOI: 10.21203/rs.3.rs-3108087/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Protein complexes are highly dynamic entities that display substantial diversity in their assembly, post-translational modifications, and non-covalent interactions, allowing them to play critical roles in various biological processes. The heterogeneity, dynamic nature, and low abundance of protein complexes in their native states present tremendous challenges to study using conventional structural biology techniques. Here we develop a "native nanoproteomics" strategy for the native enrichment and subsequent native top-down mass spectrometry (nTDMS) of low-abundance protein complexes. Specifically, we demonstrate the first comprehensive characterization of the structure and dynamics of cardiac troponin (cTn) complexes directly from human heart tissue. The endogenous cTn complex is effectively enriched and purified using peptide-functionalized superparamagnetic nanoparticles under non-denaturing conditions to enable the isotopic resolution of cTn complexes, revealing their complex structure and assembly. Moreover, nTDMS elucidates the stoichiometry and composition of the heterotrimeric cTn complex, localizes Ca2+ binding domains (II-IV), defines cTn-Ca2+ binding dynamics, and provides high-resolution mapping of the proteoform landscape. This native nanoproteomics strategy opens a new paradigm for structural characterization of low-abundance native protein complexes.
Collapse
Affiliation(s)
- Emily A. Chapman
- 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
| | - Timothy N. Tiambeng
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Jãán Andrews
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Man-Di Wang
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Emily A. Reasoner
- 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
| | - Brad H. Li
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Donguk Kim
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | - Song Jin
- 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
|
15
|
Chapman EA, Roberts DS, Tiambeng TN, Andrews J, Wang MD, Reasoner EA, Melby JA, Li BH, Kim D, Alpert AJ, Jin S, Ge Y. Structure and dynamics of endogenous protein complexes in human heart tissue captured by native nanoproteomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.544817. [PMID: 37398031 PMCID: PMC10312745 DOI: 10.1101/2023.06.13.544817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Protein complexes are highly dynamic entities that display substantial diversity in their assembly, post-translational modifications, and non-covalent interactions, allowing them to play critical roles in various biological processes. The heterogeneity, dynamic nature, and low abundance of protein complexes in their native states present tremendous challenges to study using conventional structural biology techniques. Here we develop a "native nanoproteomics" strategy for the native enrichment and subsequent native top-down mass spectrometry (nTDMS) of low-abundance protein complexes. Specifically, we demonstrate the first comprehensive characterization of the structure and dynamics of cardiac troponin (cTn) complexes directly from human heart tissue. The endogenous cTn complex is effectively enriched and purified using peptide-functionalized superparamagnetic nanoparticles under non-denaturing conditions to enable the isotopic resolution of cTn complexes, revealing their complex structure and assembly. Moreover, nTDMS elucidates the stoichiometry and composition of the heterotrimeric cTn complex, localizes Ca2+ binding domains (II-IV), defines cTn-Ca2+ binding dynamics, and provides high-resolution mapping of the proteoform landscape. This native nanoproteomics strategy opens a new paradigm for structural characterization of low-abundance native protein complexes.
Collapse
|
16
|
Brunello E, Marcucci L, Irving M, Fusi L. Activation of skeletal muscle is controlled by a dual-filament mechano-sensing mechanism. Proc Natl Acad Sci U S A 2023; 120:e2302837120. [PMID: 37216507 PMCID: PMC10235942 DOI: 10.1073/pnas.2302837120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Contraction of skeletal muscle is triggered by a transient rise in intracellular calcium concentration leading to a structural change in the actin-containing thin filaments that allows binding of myosin motors from the thick filaments. Most myosin motors are unavailable for actin binding in resting muscle because they are folded back against the thick filament backbone. Release of the folded motors is triggered by thick filament stress, implying a positive feedback loop in the thick filaments. However, it was unclear how thin and thick filament activation mechanisms are coordinated, partly because most previous studies of the thin filament regulation were conducted at low temperatures where the thick filament mechanisms are inhibited. Here, we use probes on both troponin in the thin filaments and myosin in the thick filaments to monitor the activation states of both filaments in near-physiological conditions. We characterize those activation states both in the steady state, using conventional titrations with calcium buffers, and during activation on the physiological timescale, using calcium jumps produced by photolysis of caged calcium. The results reveal three activation states of the thin filament in the intact filament lattice of a muscle cell that are analogous to those proposed previously from studies on isolated proteins. We characterize the rates of the transitions between these states in relation to thick filament mechano-sensing and show how thin- and thick-filament-based mechanisms are coupled by two positive feedback loops that switch on both filaments to achieve rapid cooperative activation of skeletal muscle.
Collapse
Affiliation(s)
- Elisabetta Brunello
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and British Heart Foundation Centre of Research Excellence, King’s College London, LondonSE1 1UL, United Kingdom
| | - Lorenzo Marcucci
- Department of Biomedical Sciences, University of Padova, Padova35131, Italy
- RIKEN Centre for Biosystems Dynamics Research, Suita565-0874, Japan
| | - Malcolm Irving
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and British Heart Foundation Centre of Research Excellence, King’s College London, LondonSE1 1UL, United Kingdom
| | - Luca Fusi
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences and British Heart Foundation Centre of Research Excellence, King’s College London, LondonSE1 1UL, United Kingdom
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, King’s College London, LondonSE1 1UL, United Kingdom
| |
Collapse
|
17
|
Huang SC, Huang CC, Ko CY, Huang CY, Liu CH, Lee YK, Chen TY, Hsueh CW, Tzou SJ, Tai MH, Hu TH, Tsai MC, Lee WC, Ho YC, Wu CC, Chang YC, Chang JJ, Liu KH, Li CC, Wen ZH, Chang CL, Chu TH. Slow skeletal muscle troponin T acts as a potential prognostic biomarker and therapeutic target for hepatocellular carcinoma. Gene 2023; 865:147331. [PMID: 36871674 DOI: 10.1016/j.gene.2023.147331] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Slow skeletal muscle troponin T (TNNT1) as a poor prognostic indicator is upregulated in colon and breast cancers. However, the role of TNNT1 in the disease prognosis and biological functions of hepatocellular carcinoma (HCC) is still unclear. The Cancer Genome Atlas (TCGA), real-time quantitative RT-PCR (qRT-PCR), immunoblot, and immunohistochemical analyses were applied to evaluate the TNNT1 expression of human HCC. The impact of TNNT1 levels on disease progression and survival outcome was studied using TCGA analysis. Moreover, the bioinformatics analysis and HCC cell culture were used to investigate the biological functions of TNNT1. Besides, the immunoblot analysis and enzyme-linked immunosorbent assay (ELISA) were used to detect the extracellular TNNT1 of HCC cells and circulating TNNT1 of HCC patients, respectively. The effect of TNNT1 neutralization on oncogenic behaviors and signaling was further validated in the cultured hepatoma cells. In this study, tumoral and blood TNNT1 was upregulated in HCC patients based on the analyses using bioinformatics, fresh tissues, paraffin sections, and serum. From the multiple bioinformatics tools, the TNNT1 overexpression was associated with advanced stage, high grade, metastasis, vascular invasion, recurrence, and poor survival outcome in HCC patients. By the cell culture and TCGA analyses, TNNT1 expression and release were positively correlated with epithelial-mesenchymal transition (EMT) processes in HCC tissues and cells. Moreover, TNNT1 neutralization suppressed oncogenic behaviors and EMT in hepatoma cells. In conclusion, TNNT1 may serve as a non-invasive biomarker and drug target for HCC management. This research finding may provide a new insight for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Shih-Chung Huang
- Department of Internal Medicine, Division of Cardiology, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan; Department of Internal Medicine, Division of Cardiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chao-Cheng Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chou-Yuan Ko
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Cheng-Yi Huang
- Department of Pathology, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Ching-Han Liu
- Department of Internal Medicine, Division of Cardiology, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Yung-Kuo Lee
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Tung-Yuan Chen
- Department of Surgery, Division of Colorectal Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Chao-Wen Hsueh
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Shiow-Jyu Tzou
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Nursing, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, Taiwan; Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Center for Neuroscience, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Tsung-Hui Hu
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ming-Chao Tsai
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Chin Lee
- Department of Internal Medicine, Division of Nephrology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Cheng Ho
- School of Medicine, Medical College, I-Shou University, Kaohsiung, Taiwan
| | - Cheng-Chun Wu
- School of Medicine, Medical College, I-Shou University, Kaohsiung, Taiwan
| | - Yi-Chen Chang
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, Taiwan
| | - Jung-Jui Chang
- Division of Orthopedics, Department of Surgery, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Kai-Hsi Liu
- Department of Internal Medicine, Division of Cardiology, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Chiao-Ching Li
- Division of Urology, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, Asia-Pacific Ocean Research Center, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chen-Lin Chang
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Psychiatry, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan.
| | - Tian-Huei Chu
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
18
|
Seyama R, Uchiyama Y, Kaneshi Y, Hamanaka K, Fujita A, Tsuchida N, Koshimizu E, Misawa K, Miyatake S, Mizuguchi T, Makino S, Itakura A, Okamoto N, Matsumoto N. Distal arthrogryposis in a girl arising from a novel TNNI2 variant inherited from paternal somatic mosaicism. J Hum Genet 2023; 68:363-367. [PMID: 36631501 DOI: 10.1038/s10038-022-01117-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/22/2022] [Accepted: 12/25/2022] [Indexed: 01/13/2023]
Abstract
TNNI2 at 11p15.5 encodes troponin I2, fast skeletal type, which is a member of the troponin I gene family and a component of the troponin complex. Distal arthrogryposis (DA) is characterized by congenital limb contractures without primary neurological or muscular effects. DA is inherited in an autosomal dominant fashion and is clinically and genetically heterogeneous. Exome sequencing identified a causative variant in TNNI2 [NM_003282.4:c.532T>C p.(Phe178Leu)] in a Japanese girl with typical DA2b. Interestingly, the familial study using Sanger sequencing suggested a mosaic variant in her healthy father. Subsequent targeted amplicon-based deep sequencing detected the TNNI2 variant with variant allele frequencies of 9.4-17.7% in genomic DNA derived from peripheral blood leukocytes, saliva, hair, and nails in the father. We confirmed a disease-causing variant in TNNI2 in the proband inherited from her asymptomatic father with its somatic variant. Our case demonstrates that careful clinical and genetic evaluation is required in DA.
Collapse
Affiliation(s)
- Rie Seyama
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Obstetrics and Gynecology, Juntendo University, Tokyo, Japan
| | - Yuri Uchiyama
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Yosuke Kaneshi
- Department of Pediatrics, Kushiro Red Cross Hospital, Kushiro, Japan
| | - Kohei Hamanaka
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naomi Tsuchida
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Eriko Koshimizu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazuharu Misawa
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shintaro Makino
- Department of Obstetrics and Gynecology, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Atsuo Itakura
- Department of Obstetrics and Gynecology, Juntendo University, Tokyo, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
19
|
Cai F, Kampourakis T, Parijat P, Cockburn KT, Sykes BD. Conversion of a Cardiac Muscle Modulator from an Inhibitor to an Activator. ACS Med Chem Lett 2023; 14:530-533. [PMID: 37077384 PMCID: PMC10108394 DOI: 10.1021/acsmedchemlett.3c00033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
The binding of calcium to cardiac troponin C (cTnC) enhances the binding of troponin I (cTnI) switch region to the regulatory domain of cTnC (cNTnC) and triggers muscle contraction. Several molecules alter the response of the sarcomere by targeting this interface; virtually all have an aromatic core that binds to the hydrophobic pocket of cNTnC and an aliphatic tail that interacts with the switch region of cTnI. W7 has been extensively studied, and the positively charged tail has been shown to be important for its inhibitory action. Herein we investigate the importance of the aromatic core of W7 by synthesizing compounds that have the core region of calcium activator dfbp-o with various lengths of the same tail (D-series). These compounds all bind more tightly to cNTnC-cTnI chimera (cChimera) than the analogous W-series compounds and show increased calcium sensitivity of force generation and ATPase activity, demonstrating that the cardiovascular system is tightly balanced.
Collapse
Affiliation(s)
- Fangze Cai
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Thomas Kampourakis
- Randall
Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, United Kingdom
| | - Priyanka Parijat
- Randall
Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, United Kingdom
| | - Kieran T. Cockburn
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Brian D. Sykes
- Department
of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
20
|
Issahaku AR, Ibrahim MAA, Mukelabai N, Soliman MES. Intermolecular And Dynamic Investigation of The Mechanism of Action of Reldesemtiv on Fast Skeletal Muscle Troponin Complex Toward the Treatment of Impaired Muscle Function. Protein J 2023:10.1007/s10930-023-10091-y. [PMID: 36959428 DOI: 10.1007/s10930-023-10091-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2023] [Indexed: 03/25/2023]
Abstract
Muscle weakness as a secondary feature of attenuated neuronal input often leads to disability and sometimes death in patients with neurogenic neuromuscular diseases. These impaired muscle function has been observed in several diseases including amyotrophic lateral sclerosis, Charcot-Marie-Tooth, spinal muscular atrophy and Myasthenia gravis. This has spurred the search for small molecules which could activate fast skeletal muscle troponin complex as a means to increase muscle strength. Discovered small molecules have however been punctuated by off-target and side effects leading to the development of the second-generation small molecule, Reldesemtiv. In this study, we investigated the impact of Reldesemtiv binding to the fast skeletal troponin complex and the molecular determinants that condition the therapeutic prowess of Redesemtiv through computational techniques. It was revealed that Reldesemtiv binding possibly potentiates troponin C compacting characterized by reduced exposure to solvent molecules which could favor the slow release of calcium ions and the resultant sensitization of the subunit to calcium. These conformational changes were underscored by conventional and carbon hydrogen bonds, pi-alkyl, pi-sulfur and halogen interactions between Reldesemtiv the binding site residues. Arg113 (-3.96 kcal/mol), Met116 (-2.23 kcal/mol), Val114 (-1.28 kcal/mol) and Met121 (-0.63 kcal/mol) of the switch region of the inhibitory subunit were among the residues that contributed the most to the total free binding energy of Reldesemtiv highlighting their importance. These findings present useful insights which could lay the foundation for the development of fast skeletal muscle small molecule activators with high specificity and potency.
Collapse
Affiliation(s)
- Abdul Rashid Issahaku
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
- West African Centre for Computational Research and Innovation, Accra, Ghana
| | - Mahmoud A A Ibrahim
- CompChem Research Group, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Namutula Mukelabai
- Department of Physiotherapy, School of Health Sciences, University of KwaZulu- Natal, Westville Campus, Durban, 4001, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa.
| |
Collapse
|
21
|
Molecular Dynamics Assessment of Mechanical Properties of the Thin Filaments in Cardiac Muscle. Int J Mol Sci 2023; 24:ijms24054792. [PMID: 36902223 PMCID: PMC10003134 DOI: 10.3390/ijms24054792] [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: 01/24/2023] [Revised: 02/19/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Contraction of cardiac muscle is regulated by Ca2+ ions via regulatory proteins, troponin (Tn), and tropomyosin (Tpm) associated with the thin (actin) filaments in myocardial sarcomeres. The binding of Ca2+ to a Tn subunit causes mechanical and structural changes in the multiprotein regulatory complex. Recent cryo-electron microscopy (cryo-EM) models of the complex allow one to study the dynamic and mechanical properties of the complex using molecular dynamics (MD). Here we describe two refined models of the thin filament in the calcium-free state that include protein fragments unresolved by cryo-EM and reconstructed using structure prediction software. The parameters of the actin helix and the bending, longitudinal, and torsional stiffness of the filaments estimated from the MD simulations performed with these models were close to those found experimentally. However, problems revealed from the MD simulation suggest that the models require further refinement by improving the protein-protein interaction in some regions of the complex. The use of relatively long refined models of the regulatory complex of the thin filament allows one to perform MD simulation of the molecular mechanism of Ca2+ regulation of contraction without additional constraints and study the effects of cardiomyopathy-associated mutation of the thin filament proteins of cardiac muscle.
Collapse
|
22
|
Sun S, Xu H, Xie Y, Sanchez JE, Guo W, Liu D, Li L. HIT-2: Implementing machine learning algorithms to treat bound ions in biomolecules. Comput Struct Biotechnol J 2023; 21:1383-1389. [PMID: 36817955 PMCID: PMC9929202 DOI: 10.1016/j.csbj.2023.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Electrostatic features are fundamental to protein functions and protein-protein interactions. Studying highly charged biomolecules is challenging given the heterogeneous distribution of the ionic cloud around such biomolecules. Here we report a new computational method, Hybridizing Ions Treatment-2 (HIT-2), which is used to model biomolecule-bound ions using the implicit solvation model. By modeling ions, HIT-2 allows the user to calculate important electrostatic features of the biomolecules. HIT-2 applies an efficient algorithm to calculate the position of bound ions from molecular dynamics simulations. Modeling parameters were optimized by machine learning methods from thousands of datasets. The optimized parameters produced results with errors lower than 0.2 Å. The testing results on bound Ca2+ and Zn2+ in NAMD simulations also proved that HIT-2 can effectively identify bound ion types, numbers, and positions. Also, multiple tests performed on HIT-2 suggest the method can handle biomolecules that undergo remarkable conformational changes. HIT-2 can significantly improve electrostatic calculations for many problems in computational biophysics.
Collapse
Affiliation(s)
- Shengjie Sun
- Computational Science Program, The University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Honglun Xu
- Computational Science Program, The University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Yixin Xie
- Department of Information Technology, College of Computing and Software Engineering, Kennesaw State University, 1000 Chastain Rd NW, Kennesaw, GA 30144, USA
| | - Jason E. Sanchez
- Computational Science Program, The University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Wenhan Guo
- Computational Science Program, The University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Dongfang Liu
- Department of Computer Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA
| | - Lin Li
- Computational Science Program, The University of Texas at El Paso, 500 W University Ave, TX 79968, USA
- Department of Physics, the University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| |
Collapse
|
23
|
Lin L, Liu S, Chen Z, Xia Y, Xie J, Fu M, Lu D, Wu Y, Shen H, Yang P, Qian J. Anatomically resolved transcriptome and proteome landscapes reveal disease‐relevant molecular signatures and systematic changes in heart function of end‐stage dilated cardiomyopathy. VIEW 2022. [DOI: 10.1002/viw.20220040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Ling Lin
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Shanshan Liu
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
| | - Zhangwei Chen
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Yan Xia
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Juanjuan Xie
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
| | - Mingqiang Fu
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Danbo Lu
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Yuan Wu
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| | - Huali Shen
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
| | - Pengyuan Yang
- Institutes of Biomedical Sciences of Shanghai Medical School & Minhang Hospital Fudan University Shanghai China
- Department of chemistry Fudan University Shanghai China
| | - Juying Qian
- Department of Cardiology Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital Fudan University Shanghai China
| |
Collapse
|
24
|
Early Divergence of the C-Terminal Variable Region of Troponin T Via a Pair of Mutually Exclusive Alternatively Spliced Exons Followed by a Selective Fixation in Vertebrate Heart. J Mol Evol 2022; 90:452-467. [PMID: 36171395 PMCID: PMC10080876 DOI: 10.1007/s00239-022-10075-z] [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: 01/25/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
Abstract
Troponin T (TnT) is the thin filament anchoring subunit of troponin complex and plays an organizer role in the Ca2+-regulation of striated muscle contraction. From an ancestral gene emerged ~ 700 million years ago in Bilateria, three homologous genes have evolved in vertebrates to encode muscle type-specific isoforms of TnT. Alternative splicing variants of TnT are present in vertebrate and invertebrate muscles to add functional diversity. While the C-terminal region of TnT is largely conserved, it contains an alternatively spliced segment emerged early in C. elegans, which has evolved into a pair of mutually exclusive exons in arthropods (10A and 10B of Drosophila TpnT gene) and vertebrates (16 and 17 of fast skeletal muscle Tnnt3 gene). The C-terminal alternatively spliced segment of TnT interfaces with the other two subunits of troponin with functional significance. The vertebrate cardiac TnT gene that emerged from duplication of the fast TnT gene has eliminated this alternative splicing by the fixation of an exon 17-like constitutive exon, indicating a functional value in slower and rhythmic contractions. The vertebrate slow skeletal muscle TnT gene that emerged from duplication of the cardiac TnT gene has the exon 17-like structure conserved, indicating its further function in sustained and fatigue resistant contractions. This functionality-based evolution is consistent with the finding that exon 10B-encoded segment of Drosophila TnT homologous to the exon 17-encoded segment of vertebrate fast TnT is selectively expressed in insect heart and leg muscles. The evolution of the C-terminal variable region of TnT demonstrates a submolecular mechanism in modifying striated muscle contractility and for the treatment of muscle and heart diseases.
Collapse
|
25
|
Pavadai E, Rynkiewicz MJ, Yang Z, Gould IR, Marston SB, Lehman W. Modulation of cardiac thin filament structure by phosphorylated troponin-I analyzed by protein-protein docking and molecular dynamics simulation. Arch Biochem Biophys 2022; 725:109282. [PMID: 35577070 PMCID: PMC10680062 DOI: 10.1016/j.abb.2022.109282] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/14/2022] [Accepted: 05/04/2022] [Indexed: 12/16/2022]
Abstract
Tropomyosin, controlled by troponin-linked Ca2+-binding, regulates muscle contraction by a macromolecular scale steric-mechanism that governs myosin-crossbridge-actin interactions. At low-Ca2+, C-terminal domains of troponin-I (TnI) trap tropomyosin in a position on thin filaments that interferes with myosin-binding, thus causing muscle relaxation. Steric inhibition is reversed at high-Ca2+ when TnI releases from F-actin-tropomyosin as Ca2+ and the TnI switch-peptide bind to the N-lobe of troponin-C (TnC). The opposite end of cardiac TnI contains a phosphorylation-sensitive ∼30 residue-long N-terminal peptide that is absent in skeletal muscle, and likely modifies these interactions in hearts. Here, PKA-dependent phosphorylation of serine 23 and 24 modulates Ca2+ and possibly switch-peptide binding to TnC, causing faster relaxation during the cardiac-cycle (lusitropy). The cardiac-specific N-terminal TnI domain is not captured in crystal structures of troponin or in cryo-EM reconstructions of thin filaments; thus, its global impact on thin filament structure and function is uncertain. Here, we used protein-protein docking and molecular dynamics simulation-based protocols to build a troponin model that was guided by and hence consistent with the recent seminal Yamada structure of Ca2+-activated thin filaments. We find that when present on thin filaments, phosphorylated Ser23/24 along with adjacent polar TnI residues interact closely with both tropomyosin and the N-lobe of TnC during our simulations. These interactions would likely bias tropomyosin to an off-state positioning on actin. In situ, such enhanced relaxation kinetics would promote cardiac lusitropy.
Collapse
Affiliation(s)
- Elumalai Pavadai
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, MA, 02118, USA
| | - Michael J Rynkiewicz
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, MA, 02118, USA
| | - Zeyu Yang
- Department of Chemistry and Institute of Chemical Biology, Imperial College London, Shepard's Bush, London, W12 0BZ, UK
| | - Ian R Gould
- Department of Chemistry and Institute of Chemical Biology, Imperial College London, Shepard's Bush, London, W12 0BZ, UK
| | - Steven B Marston
- National Heart & Lung Institute, Imperial College London, Dovehouse Street, W12 0NN, UK
| | - William Lehman
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, MA, 02118, USA.
| |
Collapse
|
26
|
Szikora S, Görög P, Mihály J. The Mechanisms of Thin Filament Assembly and Length Regulation in Muscles. Int J Mol Sci 2022; 23:5306. [PMID: 35628117 PMCID: PMC9140763 DOI: 10.3390/ijms23105306] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
The actin containing tropomyosin and troponin decorated thin filaments form one of the crucial components of the contractile apparatus in muscles. The thin filaments are organized into densely packed lattices interdigitated with myosin-based thick filaments. The crossbridge interactions between these myofilaments drive muscle contraction, and the degree of myofilament overlap is a key factor of contractile force determination. As such, the optimal length of the thin filaments is critical for efficient activity, therefore, this parameter is precisely controlled according to the workload of a given muscle. Thin filament length is thought to be regulated by two major, but only partially understood mechanisms: it is set by (i) factors that mediate the assembly of filaments from monomers and catalyze their elongation, and (ii) by factors that specify their length and uniformity. Mutations affecting these factors can alter the length of thin filaments, and in human cases, many of them are linked to debilitating diseases such as nemaline myopathy and dilated cardiomyopathy.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, H-6726 Szeged, Hungary;
| | - Péter Görög
- Institute of Genetics, Biological Research Centre, H-6726 Szeged, Hungary;
- Doctoral School of Multidisciplinary Medical Science, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary
| | - József Mihály
- Institute of Genetics, Biological Research Centre, H-6726 Szeged, Hungary;
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary
| |
Collapse
|
27
|
Rasmussen M, Jin JP. Monoclonal Antibodies as Probes to Study Ligand-Induced Conformations of Troponin Subunits. Front Physiol 2022; 13:828144. [PMID: 35399275 PMCID: PMC8990283 DOI: 10.3389/fphys.2022.828144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/25/2022] [Indexed: 11/20/2022] Open
Abstract
Striated muscle contraction and relaxation is regulated by Ca2+ at the myofilament level via conformational modulations of the troponin complex. To understand the structure-function relationship of troponin in normal muscle and in myopathies, it is necessary to study the functional effects of troponin isoforms and mutations at the level of allosteric conformations of troponin subunits. Traditional methodologies assessing such conformational studies are laborious and require significant amounts of purified protein, while many current methodologies require non-physiological conditions or labeling of the protein, which may affect their physiological conformation and function. To address these issues, we developed a novel approach using site-specific monoclonal antibodies (mAb) as molecular probes to detect and monitor conformational changes of proteins. Here, we present examples for its application in studies of two subunits of troponin: the Ca2+-binding subunit, TnC, and the tropomyosin-binding/thin filament-anchoring subunit, TnT. Studies using a high-throughput microplate assay are compared with that using localized surface plasmon resonance (LSPR) to demonstrate the effectiveness of using mAb probes to assess ligand-induced conformations of troponin subunits in physiological conditions. The assays utilize relatively small amounts of protein and are free of protein modification, which may bias results. Detailed methodologies using various monoclonal antibodies (mAbs) are discussed with considerations for the optimization of assay conditions and the broader application in studies of other proteins as well as in screening of therapeutic reagents that bind a specific target site with conformational and functional effects.
Collapse
Affiliation(s)
- Monica Rasmussen
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
28
|
Ma W, Irving TC. Small Angle X-ray Diffraction as a Tool for Structural Characterization of Muscle Disease. Int J Mol Sci 2022; 23:3052. [PMID: 35328477 PMCID: PMC8949570 DOI: 10.3390/ijms23063052] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023] Open
Abstract
Small angle X-ray fiber diffraction is the method of choice for obtaining molecular level structural information from striated muscle fibers under hydrated physiological conditions. For many decades this technique had been used primarily for investigating basic biophysical questions regarding muscle contraction and regulation and its use confined to a relatively small group of expert practitioners. Over the last 20 years, however, X-ray diffraction has emerged as an important tool for investigating the structural consequences of cardiac and skeletal myopathies. In this review we show how simple and straightforward measurements, accessible to non-experts, can be used to extract biophysical parameters that can help explain and characterize the physiology and pathology of a given experimental system. We provide a comprehensive guide to the range of the kinds of measurements that can be made and illustrate how they have been used to provide insights into the structural basis of pathology in a comprehensive review of the literature. We also show how these kinds of measurements can inform current controversies and indicate some future directions.
Collapse
Affiliation(s)
- Weikang Ma
- The Biophysics Collaborative Access Team (BioCAT), Center for Synchrotron Radiation Research and Instrumentation (CSSRI), Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Thomas C. Irving
- The Biophysics Collaborative Access Team (BioCAT), Center for Synchrotron Radiation Research and Instrumentation (CSSRI), Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
29
|
Zonneveld MH, Abbel D, le Cessie S, Jukema JW, Noordam R, Trompet S. Cardiac Troponin, Cognitive Function, and Dementia: A Systematic Review. Aging Dis 2022; 14:386-397. [PMID: 37008066 PMCID: PMC10017151 DOI: 10.14336/ad.2022.0818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022] Open
Abstract
Elevated cardiac troponin, a biomarker of myocardial injury, has been found in individuals with brain damage and lower cognitive function. We conducted a systematic review to examine the association of troponin with cognitive function, incidence of dementia and dementia-related outcomes. PubMed, Web of Science and EMBASE were searched from inception to August 2022. Inclusion criteria were: (i) population-based cohort studies; (ii) troponin measured as determinant; and (iii) cognitive function in any metric or diagnosis of any type of dementia or dementia-related measures as outcomes. Fourteen studies were identified and included, with a combined total of 38,286 participants. Of these studies, four examined dementia-related outcomes, eight studies examined cognitive function, and two studies examined both dementia-related outcomes and cognitive function. Studies report higher troponin to be associated with higher prevalence of cognitive impairment (n=1), incident dementia (n=1), increased risk of dementia hospitalization (specifically due to vascular dementia) (n=1), but not with incident Alzheimer's Disease (n=2). Majority of studies on cognitive function found elevated troponin also associated with worse global cognitive function (n=3), attention (n=2), reaction time (n=1) and visuomotor speed (n=1), both cross-sectionally and prospectively. Evidence regarding the association between higher troponin and memory, executive function, processing speed, language and visuospatial function was mixed. This was the first systematic review on the association between troponin, cognitive function, and dementia. Higher troponin is associated with subclinical cerebrovascular damage and might act as a risk-marker of cognitive vulnerability.
Collapse
Affiliation(s)
- Michelle H Zonneveld
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
- Department of Cardiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
- Correspondence should be addressed to: Michelle Zonneveld, M.S., Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.
| | - Denise Abbel
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Saskia le Cessie
- Department of Clinical Epidemiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
- Department of Biomedical Data Sciences, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
30
|
Syomin F, Osepyan A, Tsaturyan A. Computationally efficient model of myocardial electromechanics for multiscale simulations. PLoS One 2021; 16:e0255027. [PMID: 34293046 PMCID: PMC8297763 DOI: 10.1371/journal.pone.0255027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/08/2021] [Indexed: 11/19/2022] Open
Abstract
A model of myocardial electromechanics is suggested. It combines modified and simplified versions of previously published models of cardiac electrophysiology, excitation-contraction coupling, and mechanics. The mechano-calcium and mechano-electrical feedbacks, including the strain-dependence of the propagation velocity of the action potential, are also accounted for. The model reproduces changes in the twitch amplitude and Ca2+-transients upon changes in muscle strain including the slow response. The model also reproduces the Bowditch effect and changes in the twitch amplitude and duration upon changes in the interstimulus interval, including accelerated relaxation at high stimulation frequency. Special efforts were taken to reduce the stiffness of the differential equations of the model. As a result, the equations can be integrated numerically with a relatively high time step making the model suitable for multiscale simulation of the human heart and allowing one to study the impact of myocardial mechanics on arrhythmias.
Collapse
Affiliation(s)
- Fyodor Syomin
- Institute of Mechanics, Lomonosov Moscow State University, Moscow, Russia
- * E-mail:
| | - Anna Osepyan
- Institute of Mechanics, Lomonosov Moscow State University, Moscow, Russia
| | - Andrey Tsaturyan
- Institute of Mechanics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
31
|
Shakur R, Ochoa JP, Robinson AJ, Niroula A, Chandran A, Rahman T, Vihinen M, Monserrat L. Prognostic implications of troponin T variations in inherited cardiomyopathies using systems biology. NPJ Genom Med 2021; 6:47. [PMID: 34127679 PMCID: PMC8203786 DOI: 10.1038/s41525-021-00204-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/11/2021] [Indexed: 11/24/2022] Open
Abstract
The cardiac troponin T variations have often been used as an example of the application of clinical genotyping for prognostication and risk stratification measures for the management of patients with a family history of sudden cardiac death or familial cardiomyopathy. Given the disparity in patient outcomes and therapy options, we investigated the impact of variations on the intermolecular interactions across the thin filament complex as an example of an unbiased systems biology method to better define clinical prognosis to aid future management options. We present a novel unbiased dynamic model to define and analyse the functional, structural and physico-chemical consequences of genetic variations among the troponins. This was subsequently integrated with clinical data from accessible global multi-centre systematic reviews of familial cardiomyopathy cases from 106 articles of the literature: 136 disease-causing variations pertaining to 981 global clinical cases. Troponin T variations showed distinct pathogenic hotspots for dilated and hypertrophic cardiomyopathies; considering the causes of cardiovascular death separately, there was a worse survival in terms of sudden cardiac death for patients with a variation at regions 90–129 and 130–179 when compared to amino acids 1–89 and 200–288. Our data support variations among 90–130 as being a hotspot for sudden cardiac death and the region 131–179 for heart failure death/transplantation outcomes wherein the most common phenotype was dilated cardiomyopathy. Survival analysis into regions of high risk (regions 90–129 and 130–180) and low risk (regions 1–89 and 200–288) was significant for sudden cardiac death (p = 0.011) and for heart failure death/transplant (p = 0.028). Our integrative genomic, structural, model from genotype to clinical data integration has implications for enhancing clinical genomics methodologies to improve risk stratification.
Collapse
Affiliation(s)
- Rameen Shakur
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Boston, Massachusetts, 02459, United States. .,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1RQ, UK.
| | - Juan Pablo Ochoa
- Institute of Biomedical Investigation of A Coruña (INIBIC), University of A Coruña, Hospital Marítimo de Oza (15006), A Coruña, Spain.,Cardiology department, Health In Code. As Xubias s/n, Edificio El Fortín, 15006, A Coruña, Spain
| | - Alan J Robinson
- Medical Research Council Mitochondrial Biology Unit, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - Abhishek Niroula
- Protein Structure and Bioinformatics, Department of Experimental Medical Science, Lund University, SE-22 184, Lund, Sweden
| | - Aneesh Chandran
- Department of Biotechnology & Microbiology, Kannur University, Kannur, 670 661, Kerala, India.,Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Mauno Vihinen
- Protein Structure and Bioinformatics, Department of Experimental Medical Science, Lund University, SE-22 184, Lund, Sweden
| | - Lorenzo Monserrat
- Cardiology department, Health In Code. As Xubias s/n, Edificio El Fortín, 15006, A Coruña, Spain.
| |
Collapse
|
32
|
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
|
33
|
Wang Z, Grange M, Wagner T, Kho AL, Gautel M, Raunser S. The molecular basis for sarcomere organization in vertebrate skeletal muscle. Cell 2021; 184:2135-2150.e13. [PMID: 33765442 PMCID: PMC8054911 DOI: 10.1016/j.cell.2021.02.047] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/27/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022]
Abstract
Sarcomeres are force-generating and load-bearing devices of muscles. A precise molecular picture of how sarcomeres are built underpins understanding their role in health and disease. Here, we determine the molecular architecture of native vertebrate skeletal sarcomeres by electron cryo-tomography. Our reconstruction reveals molecular details of the three-dimensional organization and interaction of actin and myosin in the A-band, I-band, and Z-disc and demonstrates that α-actinin cross-links antiparallel actin filaments by forming doublets with 6-nm spacing. Structures of myosin, tropomyosin, and actin at ~10 Å further reveal two conformations of the "double-head" myosin, where the flexible orientation of the lever arm and light chains enable myosin not only to interact with the same actin filament, but also to split between two actin filaments. Our results provide unexpected insights into the fundamental organization of vertebrate skeletal muscle and serve as a strong foundation for future investigations of muscle diseases.
Collapse
Affiliation(s)
- Zhexin Wang
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Michael Grange
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Thorsten Wagner
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Ay Lin Kho
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Kings College London BHF Excellence Centre, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Mathias Gautel
- The Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, Kings College London BHF Excellence Centre, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany.
| |
Collapse
|
34
|
Mutations Q93H and E97K in TPM2 Disrupt Ca-Dependent Regulation of Actin Filaments. Int J Mol Sci 2021; 22:ijms22084036. [PMID: 33919826 PMCID: PMC8070786 DOI: 10.3390/ijms22084036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Tropomyosin is a two-chain coiled coil protein, which together with the troponin complex controls interactions of actin with myosin in a Ca2+-dependent manner. In fast skeletal muscle, the contractile actin filaments are regulated by tropomyosin isoforms Tpm1.1 and Tpm2.2, which form homo- and heterodimers. Mutations in the TPM2 gene encoding isoform Tpm2.2 are linked to distal arthrogryposis and congenital myopathy-skeletal muscle diseases characterized by hyper- and hypocontractile phenotypes, respectively. In this work, in vitro functional assays were used to elucidate the molecular mechanisms of mutations Q93H and E97K in TPM2. Both mutations tended to decrease actin affinity of homo-and heterodimers in the absence and presence of troponin and Ca2+, although the effect of Q93H was stronger. Changes in susceptibility of tropomyosin to trypsin digestion suggested that the mutations diversified dynamics of tropomyosin homo- and heterodimers on the filament. The presence of Q93H in homo- and heterodimers strongly decreased activation of the actomyosin ATPase and reduced sensitivity of the thin filament to [Ca2+]. In contrast, the presence of E97K caused hyperactivation of the ATPase and increased sensitivity to [Ca2+]. In conclusion, the hypo- and hypercontractile phenotypes associated with mutations Q93H and E97K in Tpm2.2 are caused by defects in Ca2+-dependent regulation of actin-myosin interactions.
Collapse
|
35
|
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
|
36
|
Silva AMM, Heeley DH. Existence in the actin world of a specialized slow skeletal muscle isoform. Comp Biochem Physiol B Biochem Mol Biol 2021; 254:110568. [PMID: 33545366 DOI: 10.1016/j.cbpb.2021.110568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 11/24/2022]
Affiliation(s)
- A Madhushika M Silva
- Department of Biochemistry, Memorial University, St. John's, Newfoundland A1B 3X9, Canada
| | - David H Heeley
- Department of Biochemistry, Memorial University, St. John's, Newfoundland A1B 3X9, Canada.
| |
Collapse
|
37
|
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
|
38
|
Tobacman LS. Troponin Revealed: Uncovering the Structure of the Thin Filament On-Off Switch in Striated Muscle. Biophys J 2021; 120:1-9. [PMID: 33221250 PMCID: PMC7820733 DOI: 10.1016/j.bpj.2020.11.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/25/2022] Open
Abstract
Recently, our understanding of the structural basis of troponin-tropomyosin's Ca2+-triggered regulation of striated muscle contraction has advanced greatly, particularly via cryo-electron microscopy data. Compelling atomic models of troponin-tropomyosin-actin were published for both apo- and Ca2+-saturated states of the cardiac thin filament. Subsequent electron microscopy and computational analyses have supported and further elaborated the findings. Per cryo-electron microscopy, each troponin is highly extended and contacts both tropomyosin strands, which lie on opposite sides of the actin filament. In the apo-state characteristic of relaxed muscle, troponin and tropomyosin hinder strong myosin-actin binding in several different ways, apparently barricading the actin more substantially than does tropomyosin alone. The troponin core domain, the C-terminal third of TnI, and tropomyosin under the influence of a 64-residue helix of TnT located at the overlap of adjacent tropomyosins are all in positions that would hinder strong myosin binding to actin. In the Ca2+-saturated state, the TnI C-terminus dissociates from actin and binds in part to TnC; the core domain pivots significantly; the N-lobe of TnC binds specifically to actin and tropomyosin; and tropomyosin rotates partially away from myosin's binding site on actin. At the overlap domain, Ca2+ causes much less tropomyosin movement, so a more inhibitory orientation persists. In the myosin-saturated state of the thin filament, there is a large additional shift in tropomyosin, with molecular interactions now identified between tropomyosin and both actin and myosin. A new era has arrived for investigation of the thin filament and for functional understandings that increasingly accommodate the recent structural results.
Collapse
Affiliation(s)
- Larry S Tobacman
- Departments of Medicine and of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
39
|
Hornos F, Feng HZ, Rizzuti B, Palomino-Schätzlein M, Wieczorek D, Neira JL, Jin JP. The muscle-relaxing C-terminal peptide from troponin I populates a nascent helix, facilitating binding to tropomyosin with a potent therapeutic effect. J Biol Chem 2021; 296:100228. [PMID: 33814345 PMCID: PMC7948816 DOI: 10.1074/jbc.ra120.016012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 11/06/2022] Open
Abstract
The conserved C-terminal end segment of troponin I (TnI) plays a critical role in regulating muscle relaxation. This function is retained in the isolated C-terminal 27 amino acid peptide (residues 184-210) of human cardiac TnI (HcTnI-C27): When added to skinned muscle fibers, HcTnI-C27 reduces the Ca2+-sensitivity of activated myofibrils and facilitates relaxation without decreasing the maximum force production. However, the underlying mechanism of HcTnI-C27 function is unknown. We studied the conformational preferences of HcTnI-C27 and a myopathic mutant, Arg192His, (HcTnI-C27-H). Both peptides were mainly disordered in aqueous solution with a nascent helix involving residues from Trp191 to Ile195, as shown by NMR analysis and molecular dynamics simulations. The population of nascent helix was smaller in HcTnI-C27-H than in HcTnI-C27, as shown by circular dichroism (CD) titrations. Fluorescence and isothermal titration calorimetry (ITC) showed that both peptides bound tropomyosin (αTm), with a detectably higher affinity (∼10 μM) of HcTnI-C27 than that of HcTnI-C27-H (∼15 μM), consistent with an impaired Ca2+-desensitization effect of the mutant peptide on skinned muscle strips. Upon binding to αTm, HcTnI-C27 acquired a weakly stable helix-like conformation involving residues near Trp191, as shown by transferred nuclear Overhauser effect spectroscopy and hydrogen/deuterium exchange experiments. With the potent Ca2+-desensitization effect of HcTnI-C27 on skinned cardiac muscle from a mouse model of hypertrophic cardiomyopathy, the data support that the C-terminal end domain of TnI can function as an isolated peptide with the intrinsic capacity of binding tropomyosin, providing a promising therapeutic approach to selectively improve diastolic function of the heart.
Collapse
MESH Headings
- Amino Acid Sequence
- Amino Acid Substitution
- Animals
- Binding Sites
- Calcium/metabolism
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/metabolism
- Cardiomyopathy, Hypertrophic/pathology
- Cardiomyopathy, Hypertrophic/prevention & control
- Disease Models, Animal
- Gene Expression
- Humans
- Kinetics
- Mice
- Molecular Docking Simulation
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Relaxation
- Mutation
- Myofibrils/drug effects
- Myofibrils/metabolism
- Myofibrils/pathology
- Peptides/chemistry
- Peptides/genetics
- Peptides/metabolism
- Peptides/pharmacology
- Protein Binding
- Protein Conformation, alpha-Helical
- Protein Interaction Domains and Motifs
- Sequence Alignment
- Sequence Homology, Amino Acid
- Substrate Specificity
- Tropomyosin/chemistry
- Tropomyosin/genetics
- Tropomyosin/metabolism
- Troponin I/chemistry
- Troponin I/genetics
- Troponin I/metabolism
Collapse
Affiliation(s)
- Felipe Hornos
- IDIBE, Universidad Miguel Hernández, Alicante, Spain
| | - Han-Zhong Feng
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bruno Rizzuti
- CNR-NANOTEC, Licryl-UOS Cosenza and CEMIF.Cal, Department of Physics, University of Calabria, Cosenza, Italy
| | | | - David Wieczorek
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, Cinncinnnati, Ohio, USA
| | - José L Neira
- IDIBE, Universidad Miguel Hernández, Alicante, Spain; Instituto de Biocomputación y Física de Sistemas Complejos, Zaragoza, Spain.
| | - J-P Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA.
| |
Collapse
|
40
|
McNamara JW, Schuckman M, Becker RC, Sadayappan S. A Novel Homozygous Intronic Variant in TNNT2 Associates With Feline Cardiomyopathy. Front Physiol 2020. [PMID: 33304277 DOI: 10.3389/fphys.2020.608473.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Hypertrophic cardiomyopathy (HCM) is a genetic disease of the heart and the most common cause of sudden cardiac death in the young. HCM is considered a disease of the sarcomere owing to the large number of mutations in genes encoding sarcomeric proteins. The riddle lies in discovering how these mutations lead to disease. As a result, treatments to prevent and/or treat HCM are limited to invasive surgical myectomies or ablations. The A31P variant of cardiac myosin binding protein-C, encoded by MYBPC3, was found to be more prevalent in a cohort of Maine Coon cats with HCM. However, other mutations in MYBPC3 and MYH7 have also been associated with HCM in cats of other breeds. In this study, we expand the spectrum of genes associated with HCM in cats. Results Next Generation Whole Genome sequencing was performed using DNA isolated from peripheral blood of a Maine Coon with cardiomyopathy that tested negative for the MYBPC3 A31P variant. Through risk stratification of variants, we identified a novel, homozygous intronic variant in cardiac troponin T (TNNT2). In silico analysis of the variant suggested that it may affect normal splicing of exon 3 of TNNT2. Both parents tested heterozygous for the mutation, but were unaffected by the disease. Echocardiography analyses revealed that the proband had shown early onset congestive heart failure, which is managed with a treatment regime including ACE and aldosterone inhibitors. Conclusion In summary, we are the first to demonstrate the association between TNNT2 mutations and HCM in felines, suggesting that this gene should be included in the testing panel of genes when performing genetic testing for HCM in cats.
Collapse
Affiliation(s)
- James W McNamara
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, United States
| | - Maggie Schuckman
- Department of Cardiology, MedVet Cincinnati, Fairfax, OH, United States
| | - Richard C Becker
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, United States
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
41
|
McNamara JW, Schuckman M, Becker RC, Sadayappan S. A Novel Homozygous Intronic Variant in TNNT2 Associates With Feline Cardiomyopathy. Front Physiol 2020; 11:608473. [PMID: 33304277 PMCID: PMC7701303 DOI: 10.3389/fphys.2020.608473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 10/26/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a genetic disease of the heart and the most common cause of sudden cardiac death in the young. HCM is considered a disease of the sarcomere owing to the large number of mutations in genes encoding sarcomeric proteins. The riddle lies in discovering how these mutations lead to disease. As a result, treatments to prevent and/or treat HCM are limited to invasive surgical myectomies or ablations. The A31P variant of cardiac myosin binding protein-C, encoded by MYBPC3, was found to be more prevalent in a cohort of Maine Coon cats with HCM. However, other mutations in MYBPC3 and MYH7 have also been associated with HCM in cats of other breeds. In this study, we expand the spectrum of genes associated with HCM in cats. RESULTS Next Generation Whole Genome sequencing was performed using DNA isolated from peripheral blood of a Maine Coon with cardiomyopathy that tested negative for the MYBPC3 A31P variant. Through risk stratification of variants, we identified a novel, homozygous intronic variant in cardiac troponin T (TNNT2). In silico analysis of the variant suggested that it may affect normal splicing of exon 3 of TNNT2. Both parents tested heterozygous for the mutation, but were unaffected by the disease. Echocardiography analyses revealed that the proband had shown early onset congestive heart failure, which is managed with a treatment regime including ACE and aldosterone inhibitors. CONCLUSION In summary, we are the first to demonstrate the association between TNNT2 mutations and HCM in felines, suggesting that this gene should be included in the testing panel of genes when performing genetic testing for HCM in cats.
Collapse
Affiliation(s)
- James W. McNamara
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, United States
| | - Maggie Schuckman
- Department of Cardiology, MedVet Cincinnati, Fairfax, OH, United States
| | - Richard C. Becker
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, United States
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
42
|
A comprehensive guide to genetic variants and post-translational modifications of cardiac troponin C. J Muscle Res Cell Motil 2020; 42:323-342. [PMID: 33179204 DOI: 10.1007/s10974-020-09592-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023]
Abstract
Familial cardiomyopathy is an inherited disease that affects the structure and function of heart muscle and has an extreme range of phenotypes. Among the millions of affected individuals, patients with hypertrophic (HCM), dilated (DCM), or left ventricular non-compaction (LVNC) cardiomyopathy can experience morphologic changes of the heart which lead to sudden death in the most detrimental cases. TNNC1, the gene that codes for cardiac troponin C (cTnC), is a sarcomere gene associated with cardiomyopathies in which probands exhibit young age of presentation and high death, transplant or ventricular fibrillation events relative to TNNT2 and TNNI3 probands. Using GnomAD, ClinVar, UniProt and PhosphoSitePlus databases and published literature, an extensive list to date of identified genetic variants in TNNC1 and post-translational modifications (PTMs) in cTnC was compiled. Additionally, a recent cryo-EM structure of the cardiac thin filament regulatory unit was used to localize each functionally studied amino acid variant and each PTM (acetylation, glycation, s-nitrosylation, phosphorylation) in the structure of cTnC. TNNC1 has a large number of variants (> 100) relative to other genes of the same transcript size. Surprisingly, the mapped variant amino acids and PTMs are distributed throughout the cTnC structure. While many cardiomyopathy-associated variants are localized in α-helical regions of cTnC, this was not statistically significant χ2 (p = 0.72). Exploring the variants in TNNC1 and PTMs of cTnC in the contexts of cardiomyopathy association, physiological modulation and potential non-canonical roles provides insights into the normal function of cTnC along with the many facets of TNNC1 as a cardiomyopathic gene.
Collapse
|
43
|
Solís C, Robinson JM. Cardiac troponin and tropomyosin bind to F-actin cooperatively, as revealed by fluorescence microscopy. FEBS Open Bio 2020; 10:1362-1372. [PMID: 32385956 PMCID: PMC7327902 DOI: 10.1002/2211-5463.12876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 12/22/2022] Open
Abstract
In cardiac muscle, binding of troponin (Tn) and tropomyosin (Tpm) to filamentous (F)‐actin forms thin filaments capable of Ca2+‐dependent regulation of contraction. Tpm binds to F‐actin in a head‐to‐tail fashion, while Tn stabilizes these linkages. Valuable structural and functional information has come from biochemical, X‐ray, and electron microscopy data. However, the use of fluorescence microscopy to study thin filament assembly remains relatively underdeveloped. Here, triple fluorescent labeling of Tn, Tpm, and F‐actin allowed us to track thin filament assembly by fluorescence microscopy. It is shown here that Tn and Tpm molecules self‐organize on actin filaments and give rise to decorated and undecorated regions. Binding curves based on colocalization of Tn and Tpm on F‐actin exhibit cooperative binding with a dissociation constant Kd of ~ 0.5 µm that is independent of the Ca2+ concentration. Binding isotherms based on the intensity profile of fluorescently labeled Tn and Tpm on F‐actin show that binding of Tn is less cooperative relative to Tpm. Computational modeling of Tn‐Tpm binding to F‐actin suggests two equilibrium steps involving the binding of an initial Tn‐Tpm unit (nucleation) and subsequent recruitment of adjacent Tn‐Tpm units (elongation) that stabilize the assembly. The results presented here highlight the utility of employing fluorescence microscopy to study supramolecular protein assemblies.
Collapse
Affiliation(s)
- Christopher Solís
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, USA
| | | |
Collapse
|
44
|
López-Dávila AJ, Chalovich JM, Zittrich S, Piep B, Matinmehr F, Málnási-Csizmadia A, Rauscher AÁ, Kraft T, Brenner B, Stehle R. Cycling Cross-Bridges Contribute to Thin Filament Activation in Human Slow-Twitch Fibers. Front Physiol 2020; 11:144. [PMID: 32265723 PMCID: PMC7105683 DOI: 10.3389/fphys.2020.00144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/11/2020] [Indexed: 11/13/2022] Open
Abstract
It has been shown that not only calcium but also strong binding myosin heads contribute to thin filament activation in isometrically contracting animal fast-twitch and cardiac muscle preparations. This behavior has not been studied in human muscle fibers or animal slow-twitch fibers. Human slow-twitch fibers are interesting since they contain the same myosin heavy chain isoform as the human heart. To explore myosin-induced activation of the thin filament in isometrically contracting human slow-twitch fibers, the endogenous troponin complex was exchanged for a well-characterized fast-twitch skeletal troponin complex labeled with the fluorescent dye N-((2-(Iodoacetoxy)ethyl)-N-methyl)amino-7-nitrobenz-2-oxa-1,3-diazole (fsTn-IANBD). The exchange was ≈70% complete (n = 8). The relative contributions of calcium and strong binding cross-bridges to thin filament activation were dissected by increasing the concentration of calcium from relaxing (pCa 7.5) to saturating levels (pCa 4.5) before and after incubating the exchanged fibers in the myosin inhibitor para-aminoblebbistatin (AmBleb). At pCa 4.5, the relative contributions of calcium and strong binding cross-bridges to thin filament activation were ≈69 and ≈31%, respectively. Additionally, switching from isometric to isotonic contraction at pCa 4.5 revealed that strong binding cross-bridges contributed ≈29% to thin filament activation (i.e., virtually the same magnitude obtained with AmBleb). Thus, we showed through two different approaches that lowering the number of strong binding cross-bridges, at saturating calcium, significantly reduced the activation of the thin filament in human slow-twitch fibers. The contribution of myosin to activation resembled that which was previously reported in rat cardiac and rabbit fast-twitch muscle preparations. This method could be applied to slow-twitch human fibers obtained from the soleus muscle of cardiomyopathy patients. Such studies could lead to a better understanding of the effect of point mutations of the cardiac myosin head on the regulation of muscle contraction and could lead to better management by pharmacological approaches.
Collapse
Affiliation(s)
| | - Joseph M Chalovich
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Stefan Zittrich
- Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Birgit Piep
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hanover, Germany
| | - Faramarz Matinmehr
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hanover, Germany
| | - Andras Málnási-Csizmadia
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | | | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hanover, Germany
| | - Bernhard Brenner
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hanover, Germany
| | - Robert Stehle
- Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| |
Collapse
|
45
|
Arata T. Myosin and Other Energy-Transducing ATPases: Structural Dynamics Studied by Electron Paramagnetic Resonance. Int J Mol Sci 2020; 21:E672. [PMID: 31968570 PMCID: PMC7014194 DOI: 10.3390/ijms21020672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/05/2020] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
The objective of this article was to document the energy-transducing and regulatory interactions in supramolecular complexes such as motor, pump, and clock ATPases. The dynamics and structural features were characterized by motion and distance measurements using spin-labeling electron paramagnetic resonance (EPR) spectroscopy. In particular, we focused on myosin ATPase with actin-troponin-tropomyosin, neural kinesin ATPase with microtubule, P-type ion-motive ATPase, and cyanobacterial clock ATPase. Finally, we have described the relationships or common principles among the molecular mechanisms of various energy-transducing systems and how the large-scale thermal structural transition of flexible elements from one state to the other precedes the subsequent irreversible chemical reactions.
Collapse
Affiliation(s)
- Toshiaki Arata
- Department of Biology, Graduate School of Science, Osaka City University, Osaka 558-8585, Japan
| |
Collapse
|
46
|
Yamada Y, Namba K, Fujii T. Cardiac muscle thin filament structures reveal calcium regulatory mechanism. Nat Commun 2020; 11:153. [PMID: 31919429 PMCID: PMC6952405 DOI: 10.1038/s41467-019-14008-1] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/12/2019] [Indexed: 01/04/2023] Open
Abstract
Contraction of striated muscles is driven by cyclic interactions of myosin head projecting from the thick filament with actin filament and is regulated by Ca2+ released from sarcoplasmic reticulum. Muscle thin filament consists of actin, tropomyosin and troponin, and Ca2+ binding to troponin triggers conformational changes of troponin and tropomyosin to allow actin-myosin interactions. However, the structural changes involved in this regulatory mechanism remain unknown. Here we report the structures of human cardiac muscle thin filament in the absence and presence of Ca2+ by electron cryomicroscopy. Molecular models in the two states built based on available crystal structures reveal the structures of a C-terminal region of troponin I and an N-terminal region of troponin T in complex with the head-to-tail junction of tropomyosin together with the troponin core on actin filament. Structural changes of the thin filament upon Ca2+ binding now reveal the mechanism of Ca2+ regulation of muscle contraction.
Collapse
Affiliation(s)
- Yurika Yamada
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Keiichi Namba
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- RIKEN Center for Biosystems Dynamics Research and SPring-8 Center, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- JEOL YOKOGUSHI Research Alliance Laboratories, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takashi Fujii
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- JEOL YOKOGUSHI Research Alliance Laboratories, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
47
|
Pavadai E, Rynkiewicz MJ, Ghosh A, Lehman W. Docking Troponin T onto the Tropomyosin Overlapping Domain of Thin Filaments. Biophys J 2019; 118:325-336. [PMID: 31864661 DOI: 10.1016/j.bpj.2019.11.3393] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/14/2019] [Accepted: 11/26/2019] [Indexed: 01/02/2023] Open
Abstract
Complete description of thin filament conformational transitions accompanying muscle regulation requires ready access to atomic structures of actin-bound tropomyosin-troponin. To date, several molecular-docking protocols have been employed to identify troponin interactions on actin-tropomyosin because high-resolution experimentally determined structures of filament-associated troponin are not available. However, previously published all-atom models of the thin filament show chain separation and corruption of components during our molecular dynamics simulations of the models, implying artifactual subunit organization, possibly due to incorporation of unorthodox tropomyosin-TnT crystal structures and complex FRET measurements during model construction. For example, the recent Williams et al. (2016) atomistic model of the thin filament displays a paucity of salt bridges and hydrophobic complementarity between the TnT tail (TnT1) and tropomyosin, which is difficult to reconcile with the high, 20 nM Kd binding of TnT onto tropomyosin. Indeed, our molecular dynamics simulations show the TnT1 component in their model partially dissociates from tropomyosin in under 100 ns, whereas actin-tropomyosin and TnT1 models themselves remain intact. We therefore revisited computational work aiming to improve TnT1-thin filament models by employing unbiased docking methodologies, which test billions of trial rotations and translations of TnT1 over three-dimensional grids covering end-to-end bonded tropomyosin alone or tropomyosin on F-actin. We limited conformational searches to the association of well-characterized TnT1 helical domains and either isolated tropomyosin or actin-tropomyosin yet avoided docking TnT domains that lack known or predicted structure. The docking programs PIPER and ClusPro were used, followed by interaction energy optimization and extensive molecular dynamics. TnT1 docked to either side of isolated tropomyosin but uniquely onto one location of actin-bound tropomyosin. The antiparallel interaction with tropomyosin contained abundant salt bridges and intimately integrated hydrophobic networks joining TnT1 and the tropomyosin N-/C-terminal overlapping domain. The TnT1-tropomyosin linkage yields well-defined molecular crevices. Interaction energy measurements strongly favor this TnT1-tropomyosin design over previously proposed models.
Collapse
Affiliation(s)
- Elumalai Pavadai
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts
| | - Michael J Rynkiewicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts
| | - Anita Ghosh
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts
| | - William Lehman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
48
|
Kharitonov AV, Shubina MY, Nosov GA, Mamontova AV, Arifulin EA, Lisitsyna OM, Nalobin DS, Musinova YR, Sheval EV. Switching of cardiac troponin I between nuclear and cytoplasmic localization during muscle differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118601. [PMID: 31733262 DOI: 10.1016/j.bbamcr.2019.118601] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 11/02/2019] [Accepted: 11/11/2019] [Indexed: 11/18/2022]
Abstract
The nuclear accumulation of proteins may depend on the presence of short targeting sequences, which are known as nuclear localization signals (NLSs). Here, we found that NLSs are predicted in some cytosolic proteins and examined the hypothesis that these NLSs may be functional under certain conditions. As a model, human cardiac troponin I (hcTnI) was used. After expression in cultured non-muscle or undifferentiated muscle cells, hcTnI accumulated inside nuclei. Several NLSs were predicted and confirmed by site-directed mutagenesis in hcTnI. Nuclear import occurred via the classical karyopherin-α/β nuclear import pathway. However, hcTnI expressed in cultured myoblasts redistributed from the nucleus to the cytoplasm, where it was integrated into forming myofibrils after the induction of muscle differentiation. It appears that the dynamic retention of proteins inside cytoplasmic structures can lead to switching between nuclear and cytoplasmic localization.
Collapse
Affiliation(s)
- Alexey V Kharitonov
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Maria Y Shubina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Georgii A Nosov
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; Institute of Medical Physics and Biophysics, 48149 Muenster, Germany
| | - Anastasia V Mamontova
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Eugene A Arifulin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Olga M Lisitsyna
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Denis S Nalobin
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Yana R Musinova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia; Skobelkin State Scientific Center of Laser Medicine FMBA, 121165 Moscow, Russia
| | - Eugene V Sheval
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; LIA 1066 LFR2O French-Russian Joint Cancer Research Laboratory, 94805 Villejuif, France.
| |
Collapse
|
49
|
Gutiérrez-Mejía FA, Moerland CP, van IJzendoorn LJ, Prins MWJ. Conformation switching of single native proteins revealed by nanomechanical probing without a pulling force. NANOSCALE 2019; 11:19933-19942. [PMID: 31599908 DOI: 10.1039/c9nr01448a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Protein conformational changes are essential to biological function, and the heterogeneous nature of the corresponding protein states provokes an interest to measure conformational changes at the single molecule level. Here we demonstrate that conformational changes in single native proteins can be revealed by non-covalent antibody-targeting of specific domains within the protein, using nanomechanical probing without an applied pulling force. The protein of interest was captured between a particle and a substrate and three properties were quantified: the twist amplitude related to an applied torque, torsional compliance related to rotational Brownian motion, and translational Brownian displacement. Calcium-dependent conformation switching was studied in native human cardiac troponin, a heterotrimer protein complex that regulates the contraction and relaxation of heart muscle cells and is also a key biomarker for diagnosing myocardial infarction. The data reveal a change in mechanical properties upon conformation switching from the non-saturated to the calcium-saturated state, which in cardiomyocytes gives myosin motor proteins access to actin filaments. A clear increase was observed in the molecular stiffness for the calcium-saturated protein conformation. Using libraries of monoclonal antibodies, the nanomechanical probing of conformation by antibody targeting opens avenues for characterizing single native protein complexes for research as well as for diagnostic applications.
Collapse
Affiliation(s)
- Fabiola A Gutiérrez-Mejía
- Department of Applied Physics, Eindhoven University of Technology (TU/e), Eindhoven, The Netherlands. and Institute for Complex Molecular Systems (ICMS), TU/e, Eindhoven, The Netherlands
| | - Christian P Moerland
- Department of Applied Physics, Eindhoven University of Technology (TU/e), Eindhoven, The Netherlands. and Institute for Complex Molecular Systems (ICMS), TU/e, Eindhoven, The Netherlands
| | - Leo J van IJzendoorn
- Department of Applied Physics, Eindhoven University of Technology (TU/e), Eindhoven, The Netherlands. and Institute for Complex Molecular Systems (ICMS), TU/e, Eindhoven, The Netherlands
| | - Menno W J Prins
- Department of Applied Physics, Eindhoven University of Technology (TU/e), Eindhoven, The Netherlands. and Institute for Complex Molecular Systems (ICMS), TU/e, Eindhoven, The Netherlands and Department of Biomedical Engineering, TU/e, Eindhoven, The Netherlands
| |
Collapse
|
50
|
Wong S, Feng HZ, Jin JP. The evolutionarily conserved C-terminal peptide of troponin I is an independently configured regulatory structure to function as a myofilament Ca 2+-desensitizer. J Mol Cell Cardiol 2019; 136:42-52. [PMID: 31505197 DOI: 10.1016/j.yjmcc.2019.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 12/22/2022]
Abstract
The C-terminal end segment of troponin subunit I (TnI) is a structure highly conserved among the three muscle type-specific isoforms and across vertebrate species. Partial deletion or point mutation in this segment impairs cardiac muscle relaxation. In the present study, we characterized the C-terminal 27 amino acid peptide of human cardiac TnI (HcTnI-C27) for its role in modulating muscle contractility. Biologically or chemically synthesized HcTnI-C27 peptide retains an epitope structure in physiological solutions similarly to that in intact TnI as recognized by an anti-TnI C-terminus monoclonal antibody (mAb TnI-1). Protein binding studies found that HcTnI-C27 retains the binding affinity for tropomyosin as previously shown with intact cardiac TnI. A restrictive cardiomyopathy mutation R192H in this segment abolishes the bindings to mAb TnI-1 and tropomyosin, demonstrating a pathogenic loss of function. Contractility studies using skinned muscle preparations demonstrated that addition of HcTnI-C27 peptide reduces the Ca2+-sensitivity of myofibrils without decreasing maximum force production. The results indicate that the C-terminal end segment of TnI is a regulatory element of troponin, which retains the native configuration in the form of free peptide to confer an effect on myofilament Ca2+-desensitization. Without negative inotropic impact, this short peptide may be developed into a novel reagent to selectively facilitate cardiac muscle relaxation at the activated state as a potential treatment for heart failure.
Collapse
Affiliation(s)
- Sienna Wong
- Department of Physiology, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA
| | - Han-Zhong Feng
- Department of Physiology, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA
| | - J-P Jin
- Department of Physiology, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA.
| |
Collapse
|