1
|
Kochurova AM, Beldiia EA, Nefedova VV, Yampolskaya DS, Koubassova NA, Kleymenov SY, Antonets JY, Ryabkova NS, Katrukha IA, Bershitsky SY, Matyushenko AM, Kopylova GV, Shchepkin DV. The D75N and P161S Mutations in the C0-C2 Fragment of cMyBP-C Associated with Hypertrophic Cardiomyopathy Disturb the Thin Filament Activation, Nucleotide Exchange in Myosin, and Actin-Myosin Interaction. Int J Mol Sci 2024; 25:11195. [PMID: 39456977 PMCID: PMC11508426 DOI: 10.3390/ijms252011195] [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: 08/28/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
About half of the mutations that lead to hypertrophic cardiomyopathy (HCM) occur in the MYBPC3 gene. However, the molecular mechanisms of pathogenicity of point mutations in cardiac myosin-binding protein C (cMyBP-C) remain poorly understood. In this study, we examined the effects of the D75N and P161S substitutions in the C0 and C1 domains of cMyBP-C on the structural and functional properties of the C0-C1-m-C2 fragment (C0-C2). Differential scanning calorimetry revealed that these mutations disorder the tertiary structure of the C0-C2 molecule. Functionally, the D75N mutation reduced the maximum sliding velocity of regulated thin filaments in an in vitro motility assay, while the P161S mutation increased it. Both mutations significantly reduced the calcium sensitivity of the actin-myosin interaction and impaired thin filament activation by cross-bridges. D75N and P161S C0-C2 fragments substantially decreased the sliding velocity of the F-actin-tropomyosin filament. ADP dose-dependently reduced filament sliding velocity in the presence of WT and P161S fragments, but the velocity remained unchanged with the D75N fragment. We suppose that the D75N mutation alters nucleotide exchange kinetics by decreasing ADP affinity to the ATPase pocket and slowing the myosin cycle. Our molecular dynamics simulations mean that the D75N mutation affects myosin S1 function. Both mutations impair cardiac contractility by disrupting thin filament activation. The results offer new insights into the HCM pathogenesis caused by missense mutations in N-terminal domains of cMyBP-C, highlighting the distinct effects of D75N and P161S mutations on cardiac contractile function.
Collapse
Affiliation(s)
- Anastasia M. Kochurova
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | - Evgenia A. Beldiia
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | - Victoria V. Nefedova
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | - Daria S. Yampolskaya
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
| | | | - Sergey Y. Kleymenov
- Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Julia Y. Antonets
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | - Natalia S. Ryabkova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- HyTest Ltd., 20520 Turku, Finland
| | - Ivan A. Katrukha
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- HyTest Ltd., 20520 Turku, Finland
| | - Sergey Y. Bershitsky
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | | | - Galina V. Kopylova
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| | - Daniil V. Shchepkin
- Institute of Immunology and Physiology of the Russian Academy of Sciences, 620049 Yekaterinburg, Russia (D.V.S.)
| |
Collapse
|
2
|
Spudich JA, Nandwani N, Robert-Paganin J, Houdusse A, Ruppel KM. Reassessing the unifying hypothesis for hypercontractility caused by myosin mutations in hypertrophic cardiomyopathy. EMBO J 2024; 43:4139-4155. [PMID: 39192034 PMCID: PMC11445530 DOI: 10.1038/s44318-024-00199-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024] Open
Affiliation(s)
- James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Neha Nandwani
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France
| | - Kathleen M Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
3
|
Iyer A, Lauerova B, Mariano J, Haberlová J, Lassuthova P, Zidkova J, Wright NT, Kontrogianni-Konstantopoulos A. Compound heterozygous variants in MYBPC1 lead to severe distal arthrogryposis type-1 manifestations. Gene 2024; 910:148339. [PMID: 38438057 PMCID: PMC10981553 DOI: 10.1016/j.gene.2024.148339] [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: 01/10/2024] [Revised: 02/17/2024] [Accepted: 03/01/2024] [Indexed: 03/06/2024]
Abstract
Dominant missense variants in MYBPC1 encoding slow Myosin Binding Protein-C (sMyBP-C) have been increasingly linked to arthrogryposis syndromes and congenital myopathy with tremor. Herein, we describe novel compound heterozygous variants - NM_002465.4:[c.2486_2492del];[c.2663A > G] - present in fibronectin-III (Fn-III) C7 and immunoglobulin (Ig) C8 domains, respectively, manifesting as severe, early-onset distal arthrogryposis type-1, with the carrier requiring intensive care and several surgical interventions at an early age. Computational modeling predicts that the c.2486_2492del p.(Lys829IlefsTer7) variant destabilizes the structure of the Fn-III C7 domain, while the c.2663A > G p.(Asp888Gly) variant causes minimal structural alterations in the Ig C8 domain. Although the parents of the proband are heterozygous carriers for a single variant, they exhibit no musculoskeletal defects, suggesting a complex interplay between the two mutant alleles underlying this disorder. As emerging novel variants in MYBPC1 are shown to be causatively associated with musculoskeletal disease, it becomes clear that MYBPC1 should be included in relevant genetic screenings.
Collapse
Affiliation(s)
- Aishwarya Iyer
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Barbora Lauerova
- Department of Paediatric Neurology, Second Faculty of Medicine Charles University and University Hospital Motol, Prague, Czech Republic; Full Member of the ERN Euro-NMD, Prague, Czech Republic
| | - Jennifer Mariano
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jana Haberlová
- Department of Paediatric Neurology, Second Faculty of Medicine Charles University and University Hospital Motol, Prague, Czech Republic; Full Member of the ERN Euro-NMD, Prague, Czech Republic
| | - Petra Lassuthova
- Department of Paediatric Neurology, Second Faculty of Medicine Charles University and University Hospital Motol, Prague, Czech Republic; Full Member of the ERN Euro-NMD, Prague, Czech Republic
| | - Jana Zidkova
- Centre of Molecular Biology and Genetics, University Hospital Brno, Czech Republic
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA, USA
| | | |
Collapse
|
4
|
Desai DA, Baby A, Ananthamohan K, Green LC, Arif M, Duncan BC, Kumar M, Singh RR, Koch SE, Natesan S, Rubinstein J, Jegga AG, Sadayappan S. Roles of cMyBP-C phosphorylation on cardiac contractile dysfunction in db/db mice. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 8:100075. [PMID: 38957358 PMCID: PMC11218625 DOI: 10.1016/j.jmccpl.2024.100075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disease and comorbidity associated with several conditions, including cardiac dysfunction leading to heart failure with preserved ejection fraction (HFpEF), in turn resulting in T2DM-induced cardiomyopathy (T2DM-CM). However, the molecular mechanisms underlying the development of T2DM-CM are poorly understood. It is hypothesized that molecular alterations in myopathic genes induced by diabetes promote the development of HFpEF, whereas cardiac myosin inhibitors can rescue the resultant T2DM-mediated cardiomyopathy. To test this hypothesis, a Leptin receptor-deficient db/db homozygous (Lepr db/db) mouse model was used to define the pathogenesis of T2DM-CM. Echocardiographic studies at 4 and 6 months revealed that Lepr db/db hearts started developing cardiac dysfunction by four months, and left ventricular hypertrophy with diastolic dysfunction was evident at 6 months. RNA-seq data analysis, followed by functional enrichment, revealed the differential regulation of genes related to cardiac dysfunction in Lepr db/db heart tissues. Strikingly, the level of cardiac myosin binding protein-C phosphorylation was significantly increased in Lepr db/db mouse hearts. Finally, using isolated skinned papillary muscles and freshly isolated cardiomyocytes, CAMZYOS ® (mavacamten, MYK-461), a prescription heart medicine used for symptomatic obstructive hypertrophic cardiomyopathy treatment, was tested for its ability to rescue T2DM-CM. Compared with controls, MYK-461 significantly reduced force generation in papillary muscle fibers and cardiomyocyte contractility in the db/db group. This line of evidence shows that 1) T2DM-CM is associated with hyperphosphorylation of cardiac myosin binding protein-C and 2) MYK-461 significantly lessened disease progression in vitro, suggesting its promise as a treatment for HFpEF.
Collapse
Affiliation(s)
- Darshini A. Desai
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Akhil Baby
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Kalyani Ananthamohan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Lisa C. Green
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mohammed Arif
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Brittany C. Duncan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mohit Kumar
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rohit R. Singh
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sheryl E. Koch
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sankar Natesan
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Jack Rubinstein
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Sakthivel Sadayappan
- Center for Cardiovascular Research, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
5
|
Doh CY, Schmidt AV, Chinthalapudi K, Stelzer JE. Bringing into focus the central domains C3-C6 of myosin binding protein C. Front Physiol 2024; 15:1370539. [PMID: 38487262 PMCID: PMC10937550 DOI: 10.3389/fphys.2024.1370539] [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: 01/14/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
Myosin binding protein C (MyBPC) is a multi-domain protein with each region having a distinct functional role in muscle contraction. The central domains of MyBPC have often been overlooked due to their unclear roles. However, recent research shows promise in understanding their potential structural and regulatory functions. Understanding the central region of MyBPC is important because it may have specialized function that can be used as drug targets or for disease-specific therapies. In this review, we provide a brief overview of the evolution of our understanding of the central domains of MyBPC in regard to its domain structures, arrangement and dynamics, interaction partners, hypothesized functions, disease-causing mutations, and post-translational modifications. We highlight key research studies that have helped advance our understanding of the central region. Lastly, we discuss gaps in our current understanding and potential avenues to further research and discovery.
Collapse
Affiliation(s)
- Chang Yoon Doh
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Alexandra V. Schmidt
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
6
|
Leduc-Pessah H, Smith IC, Kernohan KD, Sampaio M, Melkus G, Strasser L, Chisholm C, Huang L, Hanes I, Tran MA, Venkateswaran S, Muir K, Charlesworth L, Warman-Chardon J. Congenital tremor and myopathy secondary to novel MYBPC1 variant. J Neurol Sci 2024; 457:122864. [PMID: 38185014 DOI: 10.1016/j.jns.2023.122864] [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: 10/24/2023] [Revised: 12/03/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
Congenital myopathy with tremor (MYOTREM) is a recently described disorder characterized by mild myopathy and a postural and intention tremor present since early infancy. MYOTREM is associated with pathogenic variants in MYBPC1 which encodes slow myosin-binding protein C, a sarcomere protein with regulatory and structural roles. Here, we describe a family with three generations of variably affected members exhibiting a novel variant in MYBPC1 (c.656 T > C, p.Leu219Pro). Among the unique features of affected family members is the persistence of tremor in sleep. We also present the first muscle magnetic resonance images for this disorder, and report muscle atrophy and fatty infiltration.
Collapse
Affiliation(s)
- Heather Leduc-Pessah
- Department of Pediatrics, Neurology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada.
| | - Ian C Smith
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Kristin D Kernohan
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada; Newborn Screening, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Marcos Sampaio
- Department of Radiology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Gerd Melkus
- Department of Radiology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Lauren Strasser
- Department of Pediatrics, Neurology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Caitlin Chisholm
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Lijia Huang
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Ilana Hanes
- Department of Pediatrics, Neurology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - My-An Tran
- Department of Pediatrics, Neurology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Sunita Venkateswaran
- Department of Pediatrics, Neurology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Katherine Muir
- Department of Pediatrics, Neurology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | | | - Jodi Warman-Chardon
- Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada; Department of Medicine, Neurology, The Ottawa Hospital, Ottawa, ON, Canada
| |
Collapse
|
7
|
Hoh JFY. Mechanism of post-tetanic depression of slow muscle fibres. J Comp Physiol B 2024; 194:41-45. [PMID: 38347296 DOI: 10.1007/s00360-024-01536-6] [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: 10/16/2023] [Revised: 12/04/2023] [Accepted: 01/08/2024] [Indexed: 03/16/2024]
Abstract
A brief tetanic stimulation has a very different effect on the subsequent isometric twitch force of fast and slow skeletal muscles. Fast muscle responds with an enhanced twitch force which doubles that of the pre-tetanic value, whereas slow muscle depresses the post-tetanic twitch by about 20%. Twitch potentiation of fast muscle has long been known to be due to myosin light chain 2 phosphorylation. It is proposed that post-tetanic twitch depression in slow muscle is due to the dephosphorylation of the slow isoform of the thick filament protein, myosin-binding protein-C, by Ca2+/calmodulin-activated phosphatase calcineurin, whilst its phosphorylation underlies the force enhancement due to β-adrenergic stimulation in slow and fast muscle.
Collapse
Affiliation(s)
- Joseph Foon Yoong Hoh
- Discipline of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
- , A2 Garnet St, Killara, NSW, 2071, Australia.
| |
Collapse
|
8
|
Wong FL, Bunch TA, Lepak VC, Steedman AL, Colson BA. Cardiac myosin-binding protein C N-terminal interactions with myosin and actin filaments: Opposite effects of phosphorylation and M-domain mutations. J Mol Cell Cardiol 2024; 186:125-137. [PMID: 38008210 PMCID: PMC10872421 DOI: 10.1016/j.yjmcc.2023.11.010] [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: 06/21/2023] [Revised: 10/26/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023]
Abstract
N-terminal cardiac myosin-binding protein C (cMyBP-C) domains (C0-C2) bind to thick (myosin) and thin (actin) filaments to coordinate contraction and relaxation of the heart. These interactions are regulated by phosphorylation of the M-domain situated between domains C1 and C2. In cardiomyopathies and heart failure, phosphorylation of cMyBP-C is significantly altered. We aimed to investigate how cMyBP-C interacts with myosin and actin. We developed complementary, high-throughput, C0-C2 FRET-based binding assays for myosin and actin to characterize the effects due to 5 HCM-linked variants or functional mutations in unphosphorylated and phosphorylated C0-C2. The assays indicated that phosphorylation decreases binding to both myosin and actin, whereas the HCM mutations in M-domain generally increase binding. The effects of mutations were greatest in phosphorylated C0-C2, and some mutations had a larger effect on actin than myosin binding. Phosphorylation also altered the spatial relationship of the probes on C0-C2 and actin. The magnitude of these structural changes was dependent on C0-C2 probe location (C0, C1, or M-domain). We conclude that binding can differ between myosin and actin due to phosphorylation or mutations. Additionally, these variables can change the mode of binding, affecting which of the interactions in cMyBP-C N-terminal domains with myosin or actin take place. The opposite effects of phosphorylation and M-domain mutations is consistent with the idea that cMyBP-C phosphorylation is critical for normal cardiac function. The precision of these assays is indicative of their usefulness in high-throughput screening of drug libraries for targeting cMyBP-C as therapy.
Collapse
Affiliation(s)
- Fiona L Wong
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Thomas A Bunch
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Victoria C Lepak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Allison L Steedman
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Brett A Colson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States.
| |
Collapse
|
9
|
Kochurova AM, Beldiia EA, Nefedova VV, Ryabkova NS, Yampolskaya DS, Matyushenko AM, Bershitsky SY, Kopylova GV, Shchepkin DV. N-Terminal Fragment of Cardiac Myosin Binding Protein C Modulates Cooperative Mechanisms of Thin Filament Activation in Atria and Ventricles. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:116-129. [PMID: 38467549 DOI: 10.1134/s0006297924010073] [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: 11/16/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 03/13/2024]
Abstract
Cardiac myosin binding protein C (cMyBP-C) is one of the essential control components of the myosin cross-bridge cycle. The C-terminal part of cMyBP-C is located on the surface of the thick filament, and its N-terminal part interacts with actin, myosin, and tropomyosin, affecting both kinetics of the ATP hydrolysis cycle and lifetime of the cross-bridge, as well as calcium regulation of the actin-myosin interaction, thereby modulating contractile function of myocardium. The role of cMyBP-C in atrial contraction has not been practically studied. We examined effect of the N-terminal C0-C1-m-C2 (C0-C2) fragment of cMyBP-C on actin-myosin interaction using ventricular and atrial myosin in an in vitro motility assay. The C0-C2 fragment of cMyBP-C significantly reduced the maximum sliding velocity of thin filaments on both myosin isoforms and increased the calcium sensitivity of the actin-myosin interaction. The C0-C2 fragment had different effects on the kinetics of ATP and ADP exchange, increasing the affinity of ventricular myosin for ADP and decreasing the affinity of atrial myosin. The effect of the C0-C2 fragment on the activation of the thin filament depended on the myosin isoforms. Atrial myosin activates the thin filament less than ventricular myosin, and the C0-C2 fragment makes these differences in the myosin isoforms more pronounced.
Collapse
Affiliation(s)
- Anastasia M Kochurova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Evgenia A Beldiia
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
- Ural Federal University, Ekaterinburg, 620002, Russia
| | - Victoria V Nefedova
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Natalia S Ryabkova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- HyTest Ltd., Turku, 20520, Finland
| | - Daria S Yampolskaya
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Alexander M Matyushenko
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Sergey Y Bershitsky
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Galina V Kopylova
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia
| | - Daniil V Shchepkin
- Institute of Immunology and Physiology, Russian Academy of Sciences, Yekaterinburg, 620049, Russia.
| |
Collapse
|
10
|
Quedan D, Singh R, Akel A, Bernardino AL, Thang C, Bhaskaruni M, Haldankar A, Tanner BCW, Root DD. Cooperative & competitive binding of anti-myosin tail antibodies revealed by super-resolution microscopy. Arch Biochem Biophys 2023; 747:109753. [PMID: 37714251 DOI: 10.1016/j.abb.2023.109753] [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: 05/12/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
The MF30 monoclonal antibody, which binds to the myosin subfragment-2 (S2), was found to increase the extent of myofibril shortening. Yet, previous observations found no effect of this antibody on actin sliding over myosin during in vitro motility assays with purified proteins in which myosin binding protein C (MyBPC) was absent. MF30 is hypothesized to enhance the availability of myosin heads (subfragment-1 or S1) to bind actin by destabilizing the myosin S2 coiled-coil and sterically blocking S2 from binding S1. The mechanism of action likely includes MF30's substantial size, thereby inhibiting S1 heads and MyBPC from binding S2. Hypothetically, MF30 should enhance the ON state of myosin, thereby increasing muscle contraction. Our findings indicate that MF30 binds preferentially to the unfolded heavy chains of S2, displaying positive cooperativity. However, the dose-response curve of MF30's enhancement of myofibril shortening did not suggest complex interactions with S2. Single, double, and triple-stained myofibrils with increasing amounts of antibodies against myosin rods indicate a possible competition with MyBPC. Additional assays revealed decreased fluorescence intensity at the C-zone (central zone in the sarcomere, where MyBPC is located), where MyBPC may inhibit MF30 binding. Another monoclonal antibody named MF20, which binds to the light meromyosin (LMM) without affecting myofibril contraction, showed less reduction in fluorescence intensity at the C-zone in expansion microscopy than MF30. Expansion microscopy images of myofibrils labeled with MF20 revealed labeling of the A-band (anisotropic band) and a slight reduction in the labeling at the C-zone. The staining pattern obtained from the expansion microscopy image was consistent with images from photolocalization microscopy which required the synthesis of unique photoactivatable quantum dots, and Zeiss Airyscan imaging as well as alternative expansion microscopy digestion methods. Consistent with the hypothesis that MF30 competes with MyBPC binding to S2, cardiac tissue from MyBPC knockout mice was stained more intensely, especially in the C-zone, by MF30 compared to the wild type.
Collapse
Affiliation(s)
- Dua'a Quedan
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX, 76203, USA
| | - Rohit Singh
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX, 76203, USA
| | - Amal Akel
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX, 76203, USA
| | - Andrea L Bernardino
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX, 76203, USA
| | - Christopher Thang
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX, 76203, USA
| | - Mithilesh Bhaskaruni
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX, 76203, USA
| | - Anushka Haldankar
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX, 76203, USA
| | - Bertrand C W Tanner
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, 99164, USA
| | - Douglas D Root
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX, 76203, USA.
| |
Collapse
|
11
|
Huang X, Torre I, Chiappi M, Yin Z, Vydyanath A, Cao S, Raschdorf O, Beeby M, Quigley B, de Tombe PP, Liu J, Morris EP, Luther PK. Cryo-electron tomography of intact cardiac muscle reveals myosin binding protein-C linking myosin and actin filaments. J Muscle Res Cell Motil 2023; 44:165-178. [PMID: 37115473 PMCID: PMC10542292 DOI: 10.1007/s10974-023-09647-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/29/2023] [Indexed: 04/29/2023]
Abstract
Myosin binding protein C (MyBP-C) is an accessory protein of the thick filament in vertebrate cardiac muscle arranged over 9 stripes of intervals of 430 Å in each half of the A-band in the region called the C-zone. Mutations in cardiac MyBP-C are a leading cause of hypertrophic cardiomyopathy the mechanism of which is unknown. It is a rod-shaped protein composed of 10 or 11 immunoglobulin- or fibronectin-like domains labelled C0 to C10 which binds to the thick filament via its C-terminal region. MyBP-C regulates contraction in a phosphorylation dependent fashion that may be through binding of its N-terminal domains with myosin or actin. Understanding the 3D organisation of MyBP-C in the sarcomere environment may provide new light on its function. We report here the fine structure of MyBP-C in relaxed rat cardiac muscle by cryo-electron tomography and subtomogram averaging of refrozen Tokuyasu cryosections. We find that on average MyBP-C connects via its distal end to actin across a disc perpendicular to the thick filament. The path of MyBP-C suggests that the central domains may interact with myosin heads. Surprisingly MyBP-C at Stripe 4 is different; it has weaker density than the other stripes which could result from a mainly axial or wavy path. Given that the same feature at Stripe 4 can also be found in several mammalian cardiac muscles and in some skeletal muscles, our finding may have broader implication and significance. In the D-zone, we show the first demonstration of myosin crowns arranged on a uniform 143 Å repeat.
Collapse
Affiliation(s)
- Xinrui Huang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT, 06516, USA
| | - Iratxe Torre
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Michele Chiappi
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Zhan Yin
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Anupama Vydyanath
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Shuangyi Cao
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | | | - Morgan Beeby
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Bonnie Quigley
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Pieter P de Tombe
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott Ave, Chicago, IL, 60612, USA
- Phymedexp, Université de Montpellier, Inserm, CNRS, Montpellier, France
| | - Jun Liu
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT, 06516, USA
| | - Edward P Morris
- Division of Structural Biology, Institute of Cancer Research, London, SW3 6JB, UK
- School of Molecular Biosciences, University of Glasgow, Garscube Campus, Jarrett Building, 351, Bearsden Road, Glasgow, G61 1QH, UK
| | - Pradeep K Luther
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK.
- Cardiac Function Section, National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
12
|
Song T, Landim-Vieira M, Ozdemir M, Gott C, Kanisicak O, Pinto JR, Sadayappan S. Etiology of genetic muscle disorders induced by mutations in fast and slow skeletal MyBP-C paralogs. Exp Mol Med 2023; 55:502-509. [PMID: 36854776 PMCID: PMC10073172 DOI: 10.1038/s12276-023-00953-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 03/02/2023] Open
Abstract
Skeletal muscle, a highly complex muscle type in the eukaryotic system, is characterized by different muscle subtypes and functions associated with specific myosin isoforms. As a result, skeletal muscle is the target of numerous diseases, including distal arthrogryposes (DAs). Clinically, DAs are a distinct disorder characterized by variation in the presence of contractures in two or more distal limb joints without neurological issues. DAs are inherited, and up to 40% of patients with this condition have mutations in genes that encode sarcomeric protein, including myosin heavy chains, troponins, and tropomyosin, as well as myosin binding protein-C (MYBPC). Our research group and others are actively studying the specific role of MYBPC in skeletal muscles. The MYBPC family of proteins plays a critical role in the contraction of striated muscles. More specifically, three paralogs of the MYBPC gene exist, and these are named after their predominant expression in slow-skeletal, fast-skeletal, and cardiac muscle as sMyBP-C, fMyBP-C, and cMyBP-C, respectively, and encoded by the MYBPC1, MYBPC2, and MYBPC3 genes, respectively. Although the physiology of various types of skeletal muscle diseases is well defined, the molecular mechanism underlying the pathological regulation of DAs remains to be elucidated. In this review article, we aim to highlight recent discoveries involving the role of skeletal muscle-specific sMyBP-C and fMyBP-C as well as their expression profile, localization in the sarcomere, and potential role(s) in regulating muscle contractility. Thus, this review provides an overall summary of MYBPC skeletal paralogs, their potential roles in skeletal muscle function, and future research directions.
Collapse
Affiliation(s)
- Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA.
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, 32306, USA
| | - Mustafa Ozdemir
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Caroline Gott
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, 32306, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA.
| |
Collapse
|
13
|
Risi CM, Villanueva E, Belknap B, Sadler RL, Harris SP, White HD, Galkin VE. Cryo-Electron Microscopy Reveals Cardiac Myosin Binding Protein-C M-Domain Interactions with the Thin Filament. J Mol Biol 2022; 434:167879. [PMID: 36370805 PMCID: PMC9771592 DOI: 10.1016/j.jmb.2022.167879] [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: 08/23/2022] [Revised: 10/18/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Cardiac myosin binding protein C (cMyBP-C) modulates cardiac contraction via direct interactions with cardiac thick (myosin) and thin (actin) filaments (cTFs). While its C-terminal domains (e.g. C8-C10) anchor cMyBP-C to the backbone of the thick filament, its N-terminal domains (NTDs) (e.g. C0, C1, M, and C2) bind to both myosin and actin to accomplish its dual roles of inhibiting thick filaments and activating cTFs. While the positions of C0, C1 and C2 on cTF have been reported, the binding site of the M-domain on the surface of the cTF is unknown. Here, we used cryo-EM to reveal that the M-domain interacts with actin via helix 3 of its ordered tri-helix bundle region, while the unstructured part of the M-domain does not maintain extensive interactions with actin. We combined the recently obtained structure of the cTF with the positions of all the four NTDs on its surface to propose a complete model of the NTD binding to the cTF. The model predicts that the interactions of the NTDs with the cTF depend on the activation state of the cTF. At the peak of systole, when bound to the extensively activated cTF, NTDs would inhibit actomyosin interactions. In contrast, at falling Ca2+ levels, NTDs would not compete with the myosin heads for binding to the cTF, but would rather promote formation of active cross-bridges at the adjacent regulatory units located at the opposite cTF strand. Our structural data provides a testable model of the cTF regulation by the cMyBP-C.
Collapse
Affiliation(s)
- Cristina M Risi
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Edwin Villanueva
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Betty Belknap
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Rachel L Sadler
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Samantha P Harris
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Howard D White
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| |
Collapse
|
14
|
Kucher AN, Valiakhmetov NR, Salakhov RR, Golubenko MV, Pavlyukova EN, Nazarenko MS. Phenotype variation of hypertrophic cardiomyopathy in carriers of the p.Arg870His pathogenic variant in the MYH7 gene. BULLETIN OF SIBERIAN MEDICINE 2022. [DOI: 10.20538/1682-0363-2022-3-205-216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The review analyzes variability of clinical manifestations of p.Arg870His in the MYH7 gene, which is repeatedly registered in patients with hypertrophic cardiomyopathy (HCM). The analysis involves the data from scientific publications obtained as a search result in the PubMed, СlinVar, and eLibrary.ru databases, as well as authors’ own results. A wide range of phenotypic manifestations have been revealed in carriers of p.Arg870His, from the asymptomatic to severe course, rapid progression, and early death. The review considers possible factors that modify the effect of the pathogenic variant (i.e. dosage of the pathogenic variant, the presence of other unfavorable genetic variants, etc.). The importance of accumulating information on the clinical features of HCM in the carriers of specific gene variants is emphasized in order to clarify their pathogenicity and to identify factors modifying the clinical outcome, which is important for the choice of the treatment strategy for HCM.
Collapse
Affiliation(s)
- A. N. Kucher
- Research Institute of Medical Genetics, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | - N. R. Valiakhmetov
- Research Institute of Medical Genetics, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | - R. R. Salakhov
- Research Institute of Medical Genetics, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences; Siberian State Medical University
| | - M. V. Golubenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | - E. N. Pavlyukova
- Cardiology Research Institute, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences
| | - M. S. Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center (NRMC), Russian Academy of Sciences; Siberian State Medical University
| |
Collapse
|
15
|
Kawana M, Spudich JA, Ruppel KM. Hypertrophic cardiomyopathy: Mutations to mechanisms to therapies. Front Physiol 2022; 13:975076. [PMID: 36225299 PMCID: PMC9548533 DOI: 10.3389/fphys.2022.975076] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/22/2022] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) affects more than 1 in 500 people in the general population with an extensive burden of morbidity in the form of arrhythmia, heart failure, and sudden death. More than 25 years since the discovery of the genetic underpinnings of HCM, the field has unveiled significant insights into the primary effects of these genetic mutations, especially for the myosin heavy chain gene, which is one of the most commonly mutated genes. Our group has studied the molecular effects of HCM mutations on human β-cardiac myosin heavy chain using state-of-the-art biochemical and biophysical tools for the past 10 years, combining insights from clinical genetics and structural analyses of cardiac myosin. The overarching hypothesis is that HCM-causing mutations in sarcomere proteins cause hypercontractility at the sarcomere level, and we have shown that an increase in the number of myosin molecules available for interaction with actin is a primary driver. Recently, two pharmaceutical companies have developed small molecule inhibitors of human cardiac myosin to counteract the molecular consequences of HCM pathogenesis. One of these inhibitors (mavacamten) has recently been approved by the FDA after completing a successful phase III trial in HCM patients, and the other (aficamten) is currently being evaluated in a phase III trial. Myosin inhibitors will be the first class of medication used to treat HCM that has both robust clinical trial evidence of efficacy and that targets the fundamental mechanism of HCM pathogenesis. The success of myosin inhibitors in HCM opens the door to finding other new drugs that target the sarcomere directly, as we learn more about the genetics and fundamental mechanisms of this disease.
Collapse
Affiliation(s)
- Masataka Kawana
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States
| | - Kathleen M. Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, United States,*Correspondence: Kathleen M. Ruppel,
| |
Collapse
|
16
|
Touma AM, Tang W, Rasicci DV, Vang D, Rai A, Previs SB, Warshaw DM, Yengo CM, Sivaramakrishnan S. Nanosurfer assay dissects β-cardiac myosin and cardiac myosin-binding protein C interactions. Biophys J 2022; 121:2449-2460. [PMID: 35591788 PMCID: PMC9279167 DOI: 10.1016/j.bpj.2022.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 04/13/2022] [Accepted: 05/13/2022] [Indexed: 11/02/2022] Open
Abstract
Cardiac myosin-binding protein C (cMyBP-C) modulates cardiac contractility through putative interactions with the myosin S2 tail and/or the thin filament. The relative contribution of these binding-partner interactions to cMyBP-C modulatory function remains unclear. Hence, we developed a "nanosurfer" assay as a model system to interrogate these cMyBP-C binding-partner interactions. Synthetic thick filaments were generated using recombinant human β-cardiac myosin subfragments (HMM or S1) attached to DNA nanotubes, with 14- or 28-nm spacing, corresponding to the 14.3-nm myosin spacing in native thick filaments. The nanosurfer assay consists of DNA nanotubes added to the in vitro motility assay so that myosins on the motility surface effectively deliver thin filaments to the DNA nanotubes, enhancing thin filament gliding probability on the DNA nanotubes. Thin filament velocities on nanotubes with either 14- or 28-nm myosin spacing were no different. We then characterized the effects of cMyBP-C on thin filament motility by alternating HMM and cMyBP-C N-terminal fragments (C0-C2 or C1-C2) on nanotubes every 14 nm. Both C0-C2 and C1-C2 reduced thin filament velocity four- to sixfold relative to HMM alone. Similar inhibition occurred using the myosin S1 construct, which lacks the myosin S2 region proposed to interact with cMyBP-C, suggesting that the cMyBP-C N terminus must interact with other myosin head domains and/or actin to slow thin filament velocity. Thin filament velocity was unaffected by the C0-C1f fragment, which lacks the majority of the M-domain, supporting the importance of this domain for inhibitory interaction(s). A C0-C2 fragment with phospho-mimetic replacement in the M-domain showed markedly less inhibition of thin filament velocity compared with its phospho-null counterpart, highlighting the modulatory role of M-domain phosphorylation on cMyBP-C function. Therefore, the nanosurfer assay provides a platform to precisely manipulate spatially dependent cMyBP-C binding-partner interactions, shedding light on the molecular regulation of β-cardiac myosin contractility.
Collapse
Affiliation(s)
- Anja M Touma
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | - Wanjian Tang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - David V Rasicci
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Duha Vang
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | - Ashim Rai
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | - Samantha B Previs
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, Vermont
| | - David M Warshaw
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, Vermont
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Sivaraj Sivaramakrishnan
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
17
|
Suay-Corredera C, Alegre-Cebollada J. The mechanics of the heart: zooming in on hypertrophic cardiomyopathy and cMyBP-C. FEBS Lett 2022; 596:703-746. [PMID: 35224729 DOI: 10.1002/1873-3468.14301] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Hypertrophic cardiomyopathy (HCM), a disease characterized by cardiac muscle hypertrophy and hypercontractility, is the most frequently inherited disorder of the heart. HCM is mainly caused by variants in genes encoding proteins of the sarcomere, the basic contractile unit of cardiomyocytes. The most frequently mutated among them is MYBPC3, which encodes cardiac myosin-binding protein C (cMyBP-C), a key regulator of sarcomere contraction. In this review, we summarize clinical and genetic aspects of HCM and provide updated information on the function of the healthy and HCM sarcomere, as well as on emerging therapeutic options targeting sarcomere mechanical activity. Building on what is known about cMyBP-C activity, we examine different pathogenicity drivers by which MYBPC3 variants can cause disease, focussing on protein haploinsufficiency as a common pathomechanism also in nontruncating variants. Finally, we discuss recent evidence correlating altered cMyBP-C mechanical properties with HCM development.
Collapse
|
18
|
Shah AS, Sadayappan S, Urbina EM. Lipids: a Potential Molecular Pathway Towards Diastolic Dysfunction in Youth-Onset Type 2 Diabetes. Curr Atheroscler Rep 2022; 24:109-117. [PMID: 35080716 PMCID: PMC8930525 DOI: 10.1007/s11883-022-00989-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 02/03/2023]
Abstract
PURPOSE OF THE REVIEW Obesity and type 2 diabetes (T2D) with onset in youth are emerging public health concerns. Youth with obesity and T2D are at risk for the development of heart failure with preserved ejection fraction (HFpEF) due to diabetes-related cardiomyopathy with evidence of precursor stages, namely diastolic dysfunction, present in youth. We review the literature regarding diastolic dysfunction in youth with obesity and T2D; discuss the potential mechanisms including the role of lipids, contractile proteins and their post-translational modifications, and conclude with studies to guide future treatments. RECENT FINDINGS The diabetes milieu namely hyperglycemia, hyperinsulinemia, and lipotoxicity favor development of diastolic dysfunction and HFpEF. Recent studies show HFpEF is associated with slow left ventricular relaxation and sarcomere stiffness induced by reduced calcium (Ca2+) and β-adrenergic responses. There are currently no effective therapies available for treating HFpEF. Targeting the sarcomere is an area of ongoing research.
Collapse
Affiliation(s)
- Amy S Shah
- Department of Pediatrics, Division of Endocrinology, Cincinnati Children's Hospital Medical Center and The University of Cincinnati, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Elaine M Urbina
- Department of Pediatrics, The Heart Institute, Cincinnati Children's Hospital Medical Center and The University of Cincinnati, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| |
Collapse
|
19
|
Kaviarasan V, Mohammed V, Veerabathiran R. Genetic predisposition study of heart failure and its association with cardiomyopathy. Egypt Heart J 2022; 74:5. [PMID: 35061126 PMCID: PMC8782994 DOI: 10.1186/s43044-022-00240-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) is a clinical condition distinguished by structural and functional defects in the myocardium, which genetic and environmental factors can induce. HF is caused by various genetic factors that are both heterogeneous and complex. The incidence of HF varies depending on the definition and area, but it is calculated to be between 1 and 2% in developed countries. There are several factors associated with the progression of HF, ranging from coronary artery disease to hypertension, of which observed the most common genetic cause to be cardiomyopathy. The main objective of this study is to investigate heart failure and its association with cardiomyopathy with their genetic variants. The selected novel genes that have been linked to human inherited cardiomyopathy play a critical role in the pathogenesis and progression of HF. Research sources collected from the human gene mutation and several databases revealed that numerous genes are linked to cardiomyopathy and thus explained the hereditary influence of such a condition. Our findings support the understanding of the genetics aspect of HF and will provide more accurate evidence of the role of changing disease accuracy. Furthermore, a better knowledge of the molecular pathophysiology of genetically caused HF could contribute to the emergence of personalized therapeutics in future.
Collapse
Affiliation(s)
- Vaishak Kaviarasan
- Human Cytogenetics and Genomics Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, 603103, India
| | - Vajagathali Mohammed
- Human Cytogenetics and Genomics Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, 603103, India
| | - Ramakrishnan Veerabathiran
- Human Cytogenetics and Genomics Laboratory, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, 603103, India.
| |
Collapse
|
20
|
Schwäbe FV, Peter EK, Taft MH, Manstein DJ. Assessment of the Contribution of a Thermodynamic and Mechanical Destabilization of Myosin-Binding Protein C Domain C2 to the Pathomechanism of Hypertrophic Cardiomyopathy-Causing Double Mutation MYBPC3Δ25bp/D389V. Int J Mol Sci 2021; 22:ijms222111949. [PMID: 34769381 PMCID: PMC8584774 DOI: 10.3390/ijms222111949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 01/01/2023] Open
Abstract
Mutations in the gene encoding cardiac myosin-binding protein-C (MyBPC), a thick filament assembly protein that stabilizes sarcomeric structure and regulates cardiac function, are a common cause for the development of hypertrophic cardiomyopathy. About 10% of carriers of the Δ25bp variant of MYBPC3, which is common in individuals from South Asia, are also carriers of the D389V variant on the same allele. Compared with noncarriers and those with MYBPC3Δ25bp alone, indicators for the development of hypertrophic cardiomyopathy occur with increased frequency in MYBPC3Δ25bp/D389V carriers. Residue D389 lies in the IgI-like C2 domain that is part of the N-terminal region of MyBPC. To probe the effects of mutation D389V on structure, thermostability, and protein–protein interactions, we produced and characterized wild-type and mutant constructs corresponding to the isolated 10 kDa C2 domain and a 52 kDa N-terminal fragment that includes subdomains C0 to C2. Our results show marked reductions in the melting temperatures of D389V mutant constructs. Interactions of construct C0–C2 D389V with the cardiac isoforms of myosin-2 and actin remain unchanged. Molecular dynamics simulations reveal changes in the stiffness and conformer dynamics of domain C2 caused by mutation D389V. Our results suggest a pathomechanism for the development of HCM based on the toxic buildup of misfolded protein in young MYBPC3Δ25bp/D389V carriers that is supplanted and enhanced by C-zone haploinsufficiency at older ages.
Collapse
Affiliation(s)
- Frederic V. Schwäbe
- Fritz Hartmann Centre for Medical Research, Institute for Biophysical Chemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany; (F.V.S.); (E.K.P.); (M.H.T.)
| | - Emanuel K. Peter
- Fritz Hartmann Centre for Medical Research, Institute for Biophysical Chemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany; (F.V.S.); (E.K.P.); (M.H.T.)
| | - Manuel H. Taft
- Fritz Hartmann Centre for Medical Research, Institute for Biophysical Chemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany; (F.V.S.); (E.K.P.); (M.H.T.)
| | - Dietmar J. Manstein
- Fritz Hartmann Centre for Medical Research, Institute for Biophysical Chemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany; (F.V.S.); (E.K.P.); (M.H.T.)
- Division for Structural Biochemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany
- Correspondence:
| |
Collapse
|
21
|
Suay-Corredera C, Pricolo MR, Velázquez-Carreras D, Pathak D, Nandwani N, Pimenta-Lopes C, Sánchez-Ortiz D, Urrutia-Irazabal I, Vilches S, Dominguez F, Frisso G, Monserrat L, García-Pavía P, de Sancho D, Spudich JA, Ruppel KM, Herrero-Galán E, Alegre-Cebollada J. Nanomechanical Phenotypes in Cardiac Myosin-Binding Protein C Mutants That Cause Hypertrophic Cardiomyopathy. ACS NANO 2021; 15:10203-10216. [PMID: 34060810 PMCID: PMC8514129 DOI: 10.1021/acsnano.1c02242] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a disease of the myocardium caused by mutations in sarcomeric proteins with mechanical roles, such as the molecular motor myosin. Around half of the HCM-causing genetic variants target contraction modulator cardiac myosin-binding protein C (cMyBP-C), although the underlying pathogenic mechanisms remain unclear since many of these mutations cause no alterations in protein structure and stability. As an alternative pathomechanism, here we have examined whether pathogenic mutations perturb the nanomechanics of cMyBP-C, which would compromise its modulatory mechanical tethers across sliding actomyosin filaments. Using single-molecule atomic force spectroscopy, we have quantified mechanical folding and unfolding transitions in cMyBP-C domains targeted by HCM mutations that do not induce RNA splicing alterations or protein thermodynamic destabilization. Our results show that domains containing mutation R495W are mechanically weaker than wild-type at forces below 40 pN and that R502Q mutant domains fold faster than wild-type. None of these alterations are found in control, nonpathogenic variants, suggesting that nanomechanical phenotypes induced by pathogenic cMyBP-C mutations contribute to HCM development. We propose that mutation-induced nanomechanical alterations may be common in mechanical proteins involved in human pathologies.
Collapse
Affiliation(s)
| | - Maria Rosaria Pricolo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131, Naples, Italy
| | | | - Divya Pathak
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Neha Nandwani
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | | | - David Sánchez-Ortiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | | | - Silvia Vilches
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, 28222, Madrid, Spain
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART, http://guardheart.ern-net.eu/), 28222, Madrid, Spain
| | - Fernando Dominguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, 28222, Madrid, Spain
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART, http://guardheart.ern-net.eu/), 28222, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain
| | - Giulia Frisso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate, scarl, 80145, Naples, Italy
| | | | - Pablo García-Pavía
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, 28222, Madrid, Spain
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART, http://guardheart.ern-net.eu/), 28222, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain
- Universidad Francisco de Vitoria (UFV), 28223, Pozuelo de Alarcón, Madrid, Spain
| | - David de Sancho
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, 20018, Donostia-San Sebastián, Spain
- Donostia International Physics Center (DIPC), 20018, Donostia-San Sebastián, Spain
| | - James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Kathleen M Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Elías Herrero-Galán
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | | |
Collapse
|
22
|
Bunch TA, Guhathakurta P, Lepak VC, Thompson AR, Kanassatega RS, Wilson A, Thomas DD, Colson BA. Cardiac myosin-binding protein C interaction with actin is inhibited by compounds identified in a high-throughput fluorescence lifetime screen. J Biol Chem 2021; 297:100840. [PMID: 34052227 PMCID: PMC8233204 DOI: 10.1016/j.jbc.2021.100840] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 11/26/2022] Open
Abstract
Cardiac myosin-binding protein C (cMyBP-C) interacts with actin and myosin to modulate cardiac muscle contractility. These interactions are disfavored by cMyBP-C phosphorylation. Heart failure patients often display decreased cMyBP-C phosphorylation, and phosphorylation in model systems has been shown to be cardioprotective against heart failure. Therefore, cMyBP-C is a potential target for heart failure drugs that mimic phosphorylation or perturb its interactions with actin/myosin. Here we have used a novel fluorescence lifetime-based assay to identify small-molecule inhibitors of actin-cMyBP-C binding. Actin was labeled with a fluorescent dye (Alexa Fluor 568, AF568) near its cMyBP-C binding sites; when combined with the cMyBP-C N-terminal fragment, C0-C2, the fluorescence lifetime of AF568-actin decreases. Using this reduction in lifetime as a readout of actin binding, a high-throughput screen of a 1280-compound library identified three reproducible hit compounds (suramin, NF023, and aurintricarboxylic acid) that reduced C0-C2 binding to actin in the micromolar range. Binding of phosphorylated C0-C2 was also blocked by these compounds. That they specifically block binding was confirmed by an actin-C0-C2 time-resolved FRET (TR-FRET) binding assay. Isothermal titration calorimetry (ITC) and transient phosphorescence anisotropy (TPA) confirmed that these compounds bind to cMyBP-C, but not to actin. TPA results were also consistent with these compounds inhibiting C0-C2 binding to actin. We conclude that the actin-cMyBP-C fluorescence lifetime assay permits detection of pharmacologically active compounds that affect cMyBP-C-actin binding. We now have, for the first time, a validated high-throughput screen focused on cMyBP-C, a regulator of cardiac muscle contractility and known key factor in heart failure.
Collapse
Affiliation(s)
- Thomas A Bunch
- Department of Cellular & Molecular Medicine, University of Arizona, Tucson Arizona, USA
| | - Piyali Guhathakurta
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Victoria C Lepak
- Department of Cellular & Molecular Medicine, University of Arizona, Tucson Arizona, USA
| | - Andrew R Thompson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Anna Wilson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brett A Colson
- Department of Cellular & Molecular Medicine, University of Arizona, Tucson Arizona, USA.
| |
Collapse
|
23
|
Singh RR, McNamara JW, Sadayappan S. Mutations in myosin S2 alter cardiac myosin-binding protein-C interaction in hypertrophic cardiomyopathy in a phosphorylation-dependent manner. J Biol Chem 2021; 297:100836. [PMID: 34051236 PMCID: PMC8239744 DOI: 10.1016/j.jbc.2021.100836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 12/31/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited cardiovascular disorder primarily caused by mutations in the β-myosin heavy-chain gene. The proximal subfragment 2 region (S2), 126 amino acids of myosin, binds with the C0-C2 region of cardiac myosin-binding protein-C to regulate cardiac muscle contractility in a manner dependent on PKA-mediated phosphorylation. However, it is unknown if HCM-associated mutations within S2 dysregulate actomyosin dynamics by disrupting its interaction with C0-C2, ultimately leading to HCM. Herein, we study three S2 mutations known to cause HCM: R870H, E924K, and E930Δ. First, experiments using recombinant proteins, solid-phase binding, and isothermal titrating calorimetry assays independently revealed that mutant S2 proteins displayed significantly reduced binding with C0-C2. In addition, CD revealed greater instability of the coiled-coil structure in mutant S2 proteins compared with S2Wt proteins. Second, mutant S2 exhibited 5-fold greater affinity for PKA-treated C0-C2 proteins. Third, skinned papillary muscle fibers treated with mutant S2 proteins showed no change in the rate of force redevelopment as a measure of actin–myosin cross-bridge kinetics, whereas S2Wt showed increased the rate of force redevelopment. In summary, S2 and C0-C2 interaction mediated by phosphorylation is altered by mutations in S2, which augment the speed and force of contraction observed in HCM. Modulating this interaction could be a potential strategy to treat HCM in the future.
Collapse
Affiliation(s)
- Rohit R Singh
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, Ohio, USA
| | - James W McNamara
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, Ohio, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, Ohio, USA.
| |
Collapse
|
24
|
Powers JD, Malingen SA, Regnier M, Daniel TL. The Sliding Filament Theory Since Andrew Huxley: Multiscale and Multidisciplinary Muscle Research. Annu Rev Biophys 2021; 50:373-400. [PMID: 33637009 DOI: 10.1146/annurev-biophys-110320-062613] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Two groundbreaking papers published in 1954 laid out the theory of the mechanism of muscle contraction based on force-generating interactions between myofilaments in the sarcomere that cause filaments to slide past one another during muscle contraction. The succeeding decades of research in muscle physiology have revealed a unifying interest: to understand the multiscale processes-from atom to organ-that govern muscle function. Such an understanding would have profound consequences for a vast array of applications, from developing new biomimetic technologies to treating heart disease. However, connecting structural and functional properties that are relevant at one spatiotemporal scale to those that are relevant at other scales remains a great challenge. Through a lens of multiscale dynamics, we review in this article current and historical research in muscle physiology sparked by the sliding filament theory.
Collapse
Affiliation(s)
- Joseph D Powers
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, USA
| | - Sage A Malingen
- Department of Biology, University of Washington, Seattle, Washington 98195, USA;
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington 98185, USA
- Center for Translational Muscle Research, University of Washington, Seattle, Washington 98185, USA
| | - Thomas L Daniel
- Department of Biology, University of Washington, Seattle, Washington 98195, USA;
- Department of Bioengineering, University of Washington, Seattle, Washington 98185, USA
- Center for Translational Muscle Research, University of Washington, Seattle, Washington 98185, USA
| |
Collapse
|
25
|
Tanner BCW, Previs MJ, Wang Y, Robbins J, Palmer BM. Cardiac myosin binding protein-C phosphorylation accelerates β-cardiac myosin detachment rate in mouse myocardium. Am J Physiol Heart Circ Physiol 2021; 320:H1822-H1835. [PMID: 33666504 PMCID: PMC8163640 DOI: 10.1152/ajpheart.00673.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 02/08/2021] [Accepted: 03/01/2021] [Indexed: 11/22/2022]
Abstract
Cardiac myosin binding protein-C (cMyBP-C) is a thick filament protein that influences sarcomere stiffness and modulates cardiac contraction-relaxation through its phosphorylation. Phosphorylation of cMyBP-C and ablation of cMyBP-C have been shown to increase the rate of MgADP release in the acto-myosin cross-bridge cycle in the intact sarcomere. The influence of cMyBP-C on Pi-dependent myosin kinetics has not yet been examined. We investigated the effect of cMyBP-C, and its phosphorylation, on myosin kinetics in demembranated papillary muscle strips bearing the β-cardiac myosin isoform from nontransgenic and homozygous transgenic mice lacking cMyBP-C. We used quick stretch and stochastic length-perturbation analysis to characterize rates of myosin detachment and force development over 0-12 mM Pi and at maximal (pCa 4.8) and near-half maximal (pCa 5.75) Ca2+ activation. Protein kinase A (PKA) treatment was applied to half the strips to probe the effect of cMyBP-C phosphorylation on Pi sensitivity of myosin kinetics. Increasing Pi increased myosin cross-bridge detachment rate similarly for muscles with and without cMyBP-C, although these rates were higher in muscle without cMyBP-C. Treating myocardial strips with PKA accelerated detachment rate when cMyBP-C was present over all Pi, but not when cMyBP-C was absent. The rate of force development increased with Pi in all muscles. However, Pi sensitivity of the rate force development was reduced when cMyBP-C was present versus absent, suggesting that cMyBP-C inhibits Pi-dependent reversal of the power stroke or stabilizes cross-bridge attachment to enhance the probability of completing the power stroke. These results support a functional role for cMyBP-C in slowing myosin detachment rate, possibly through a direct interaction with myosin or by altering strain-dependent myosin detachment via cMyBP-C-dependent stiffness of the thick filament and myofilament lattice. PKA treatment reduces the role for cMyBP-C to slow myosin detachment and thus effectively accelerates β-myosin detachment in the intact myofilament lattice.NEW & NOTEWORTHY Length perturbation analysis was used to demonstrate that β-cardiac myosin characteristic rates of detachment and recruitment in the intact myofilament lattice are accelerated by Pi, phosphorylation of cMyBP-C, and the absence of cMyBP-C. The results suggest that cMyBP-C normally slows myosin detachment, including Pi-dependent detachment, and that this inhibition is released with phosphorylation or absence of cMyBP-C.
Collapse
Affiliation(s)
- Bertrand C W Tanner
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont
| | - Michael J Previs
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont
| | - Yuan Wang
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont
| | - Jeffrey Robbins
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Bradley M Palmer
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, Vermont
| |
Collapse
|
26
|
Sodhi SS, Sharma N, Ghosh M, Sethi RS, Jeong DK, Lee SJ. Differential expression patterns of myogenic regulatory factors in the postnatal longissimus dorsi muscle of Jeju Native Pig and Berkshire breeds along with their co-expression with Pax7. ELECTRON J BIOTECHN 2021. [DOI: 10.1016/j.ejbt.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
27
|
Mamidi R, Holmes JB, Doh CY, Dominic KL, Madugula N, Stelzer JE. cMyBPC phosphorylation modulates the effect of omecamtiv mecarbil on myocardial force generation. J Gen Physiol 2021; 153:211867. [PMID: 33688929 PMCID: PMC7953254 DOI: 10.1085/jgp.202012816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/27/2021] [Indexed: 01/15/2023] Open
Abstract
Omecamtiv mecarbil (OM), a direct myosin motor activator, is currently being tested as a therapeutic replacement for conventional inotropes in heart failure (HF) patients. It is known that HF patients exhibit dysregulated β-adrenergic signaling and decreased cardiac myosin-binding protein C (cMyBPC) phosphorylation, a critical modulator of myocardial force generation. However, the functional effects of OM in conditions of altered cMyBPC phosphorylation have not been established. Here, we tested the effects of OM on force generation and cross-bridge (XB) kinetics using murine myocardial preparations isolated from wild-type (WT) hearts and from hearts expressing S273A, S282A, and S302A substitutions (SA) in the M domain, between the C1 and C2 domains of cMyBPC, which cannot be phosphorylated. At submaximal Ca2+ activations, OM-mediated force enhancements were less pronounced in SA than in WT myocardial preparations. Additionally, SA myocardial preparations lacked the dose-dependent increases in force that were observed in WT myocardial preparations. Following OM incubation, the basal differences in the rate of XB detachment (krel) between WT and SA myocardial preparations were abolished, suggesting that OM differentially affects the XB behavior when cMyBPC phosphorylation is reduced. Similarly, in myocardial preparations pretreated with protein kinase A to phosphorylate cMyBPC, incubation with OM significantly slowed krel in both the WT and SA myocardial preparations. Collectively, our data suggest there is a strong interplay between the effects of OM and XB behavior, such that it effectively uncouples the sarcomere from cMyBPC phosphorylation levels. Our findings imply that OM may significantly alter the in vivo cardiac response to β-adrenergic stimulation.
Collapse
Affiliation(s)
- Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Joshua B Holmes
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Chang Yoon Doh
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Katherine L Dominic
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Nikhil Madugula
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
28
|
Harris SP. Making waves: A proposed new role for myosin-binding protein C in regulating oscillatory contractions in vertebrate striated muscle. J Gen Physiol 2021; 153:e202012729. [PMID: 33275758 PMCID: PMC7721898 DOI: 10.1085/jgp.202012729] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Myosin-binding protein C (MyBP-C) is a critical regulator of muscle performance that was first identified through its strong binding interactions with myosin, the force-generating protein of muscle. Almost simultaneously with its discovery, MyBP-C was soon found to bind to actin, the physiological catalyst for myosin's activity. However, the two observations posed an apparent paradox, in part because interactions of MyBP-C with myosin were on the thick filament, whereas MyBP-C interactions with actin were on the thin filament. Despite the intervening decades since these initial discoveries, it is only recently that the dual binding modes of MyBP-C are becoming reconciled in models that place MyBP-C at a central position between actin and myosin, where MyBP-C alternately stabilizes a newly discovered super-relaxed state (SRX) of myosin on thick filaments in resting muscle and then prolongs the "on" state of actin on thin filaments in active muscle. Recognition of these dual, alternating functions of MyBP-C reveals how it is central to the regulation of both muscle contraction and relaxation. The purpose of this Viewpoint is to briefly summarize the roles of MyBP-C in binding to myosin and actin and then to highlight a possible new role for MyBP-C in inducing and damping oscillatory waves of contraction and relaxation. Because the contractile waves bear similarity to cycles of contraction and relaxation in insect flight muscles, which evolved for fast, energetically efficient contraction, the ability of MyBP-C to damp so-called spontaneous oscillatory contractions (SPOCs) has broad implications for previously unrecognized regulatory mechanisms in vertebrate striated muscle. While the molecular mechanisms by which MyBP-C can function as a wave maker or a wave breaker are just beginning to be explored, it is likely that MyBP-C dual interactions with both myosin and actin will continue to be important for understanding the new functions of this enigmatic protein.
Collapse
|
29
|
Schmid M, Toepfer CN. Cardiac myosin super relaxation (SRX): a perspective on fundamental biology, human disease and therapeutics. Biol Open 2021; 10:bio057646. [PMID: 33589442 PMCID: PMC7904003 DOI: 10.1242/bio.057646] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The fundamental basis of muscle contraction 'the sliding filament model' (Huxley and Niedergerke, 1954; Huxley and Hanson, 1954) and the 'swinging, tilting crossbridge-sliding filament mechanism' (Huxley, 1969; Huxley and Brown, 1967) nucleated a field of research that has unearthed the complex and fascinating role of myosin structure in the regulation of contraction. A recently discovered energy conserving state of myosin termed the super relaxed state (SRX) has been observed in filamentous myosins and is central to modulating force production and energy use within the sarcomere. Modulation of myosin function through SRX is a rapidly developing theme in therapeutic development for both cardiovascular disease and infectious disease. Some 70 years after the first discoveries concerning muscular function, modulation of myosin SRX may bring the first myosin targeted small molecule to the clinic, for treating hypertrophic cardiomyopathy (Olivotto et al., 2020). An often monogenic disease HCM afflicts 1 in 500 individuals, and can cause heart failure and sudden cardiac death. Even as we near therapeutic translation, there remain many questions about the governance of muscle function in human health and disease. With this review, we provide a broad overview of contemporary understanding of myosin SRX, and explore the complexities of targeting this myosin state in human disease.This article has an associated Future Leaders to Watch interview with the authors of the paper.
Collapse
Affiliation(s)
- Manuel Schmid
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Christopher N Toepfer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
30
|
Abstract
Since the discovery of muscle in the 19th century, myosins as molecular motors have been extensively studied. However, in the last decade, a new functional super-relaxed (SRX) state of myosin has been discovered, which has a 10-fold slower ATP turnover rate than the already-known non-actin-bound, disordered relaxed (DRX) state. These two states are in dynamic equilibrium under resting muscle conditions and are thought to be significant contributors to adaptive thermogenesis in skeletal muscle and can act as a reserve pool that may be recruited when there is a sustained demand for increased cardiac muscle power. This report provides an evolutionary perspective of how striated muscle contraction is regulated by modulating this myosin DRX↔SRX state equilibrium. We further discuss this equilibrium with respect to different physiological and pathophysiological perturbations, including insults causing hypertrophic cardiomyopathy, and small-molecule effectors that modulate muscle contractility in diseased pathology.
Collapse
Affiliation(s)
- Suman Nag
- Department of Biology, MyoKardia IncBrisbaneUnited States
| | - Darshan V Trivedi
- Department of Biochemistry, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
31
|
Bobyleva LG, Shumeyko SA, Yakupova EI, Surin AK, Galzitskaya OV, Kihara H, Timchenko AA, Timchenko MA, Penkov NV, Nikulin AD, Suvorina MY, Molochkov NV, Lobanov MY, Fadeev RS, Vikhlyantsev IM, Bobylev AG. Myosin Binding Protein-C Forms Amyloid-Like Aggregates In Vitro. Int J Mol Sci 2021; 22:ijms22020731. [PMID: 33450960 PMCID: PMC7828380 DOI: 10.3390/ijms22020731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 11/17/2022] Open
Abstract
This work investigated in vitro aggregation and amyloid properties of skeletal myosin binding protein-C (sMyBP-C) interacting in vivo with proteins of thick and thin filaments in the sarcomeric A-disc. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) found a rapid (5–10 min) formation of large (>2 μm) aggregates. sMyBP-C oligomers formed both at the initial 5–10 min and after 16 h of aggregation. Small angle X-ray scattering (SAXS) and DLS revealed sMyBP-C oligomers to consist of 7–10 monomers. TEM and atomic force microscopy (AFM) showed sMyBP-C to form amorphous aggregates (and, to a lesser degree, fibrillar structures) exhibiting no toxicity on cell culture. X-ray diffraction of sMyBP-C aggregates registered reflections attributed to a cross-β quaternary structure. Circular dichroism (CD) showed the formation of the amyloid-like structure to occur without changes in the sMyBP-C secondary structure. The obtained results indicating a high in vitro aggregability of sMyBP-C are, apparently, a consequence of structural features of the domain organization of proteins of this family. Formation of pathological amyloid or amyloid-like sMyBP-C aggregates in vivo is little probable due to amino-acid sequence low identity (<26%), alternating ordered/disordered regions in the protein molecule, and S–S bonds providing for general stability.
Collapse
Affiliation(s)
- Liya G. Bobyleva
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Sergey A. Shumeyko
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Elmira I. Yakupova
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Alexey K. Surin
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
- Biological Testing Laboratory, Branch of the Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
- Laboratory of the Biochemistry of Pathogenic Microorganisms, State Research Centre for Applied Microbiology and Biotechnology, Obolensk, 142279 Serpukhov District, Russia
| | - Oxana V. Galzitskaya
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Hiroshi Kihara
- Department of Early Childhood Education, Himeji-Hinomoto College, 890 Koro, Kodera-cho, Himeji 679-2151, Japan;
| | - Alexander A. Timchenko
- Group of Experimental Research and Engineering of Oligomeric Structures, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Maria A. Timchenko
- Laboratory of Applied Enzymology, FRC PSCBR, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Nikita V. Penkov
- Laboratory of the Methods of Optical Spectral Analysis, Institute of Cell Biophysics, Russian Academy of Sciences, FRC PSCBR RAS, 142290 Pushchino, Russia;
| | - Alexey D. Nikulin
- Laboratory for Structural Studies of the Translational Apparatus, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Mariya Yu. Suvorina
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Nikolay V. Molochkov
- Laboratory of NMR Investigations of Biosystems, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Mikhail Yu. Lobanov
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Roman S. Fadeev
- Laboratory of Pharmacological Regulation of Cell Resistance, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Ivan M. Vikhlyantsev
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Correspondence: (I.M.V.); (A.G.B.)
| | - Alexander G. Bobylev
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Correspondence: (I.M.V.); (A.G.B.)
| |
Collapse
|
32
|
Kim K, Kim D, Min Y, Jeong D, Son YO, Do K. Myogenic regulatory factors are key players in determining muscle mass and meat quality in Jeju native and Berkshire pigs. Vet Med Sci 2020; 7:735-745. [PMID: 33372732 PMCID: PMC8136949 DOI: 10.1002/vms3.418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/30/2020] [Accepted: 12/09/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Meat from Jeju native pigs (JNPs) is highly popular among Korean consumers; however, the production efficiency is limited due to the low adult body weight. In contrast, the Berkshire breed, which has a genetic background closely related to Asian native pigs, gains weight more efficiently. OBJECTIVES This study focused on the differential expression of genes related to muscle growth in postnatal myogenesis between Berkshire and JNPs, specifically the myogenic regulatory factor (MRF) genes (MyoD, Pax7, Myf5, Myf6 and MyBPH). The MRF family is primarily involved in the proliferation and development of muscle. METHODS Qualitative reverse transcription-polymerase chain reaction and western blot analyses revealed that expression of MyoD and Pax7 was significantly higher in Berkshire pigs than in JNPs. In addition, co-expression of MyoD and Pax7 was observed in myotubes formed in cultured C2C12 cells. ToppCluster was used to elucidate the relationship between biological processes of the MRFs and muscle-related signalling pathways. RESULTS MyoD and Pax7 are factors essential for the activation of satellite cell during myogenesis. However, the mRNA and protein levels of MyBPH (which is responsible for meat quality, e.g. water content, colour and tenderness) are significantly higher in both 1-day-old piglets and adult JNPs than in Berkshire pigs. CONCLUSIONS This study provides a genetic understanding of myogenesis in the postnatal and adult stages of Berkshire pigs and JNPs. Moreover, these results will help identify marker genes related to muscle mass, growth performance and meat quality in indigenous Korean pig breeds.
Collapse
Affiliation(s)
- Kyoungho Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea.,Livestock Division, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea
| | - Dahye Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea.,Practical Translational Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea
| | - Yunhui Min
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea
| | - DongKee Jeong
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea.,Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea.,Bio-Health Materials Core-Facility Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea.,Practical Translational Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea
| | - Kyoungtag Do
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Republic of Korea
| |
Collapse
|
33
|
Main A, Fuller W, Baillie GS. Post-translational regulation of cardiac myosin binding protein-C: A graphical review. Cell Signal 2020; 76:109788. [DOI: 10.1016/j.cellsig.2020.109788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 01/01/2023]
|
34
|
Diering S, Stathopoulou K, Goetz M, Rathjens L, Harder S, Piasecki A, Raabe J, Schulz S, Brandt M, Pflaumenbaum J, Fuchs U, Donzelli S, Sadayappan S, Nikolaev VO, Flenner F, Ehler E, Cuello F. Receptor-independent modulation of cAMP-dependent protein kinase and protein phosphatase signaling in cardiac myocytes by oxidizing agents. J Biol Chem 2020; 295:15342-15365. [PMID: 32868295 PMCID: PMC7650233 DOI: 10.1074/jbc.ra120.014467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/28/2020] [Indexed: 02/04/2023] Open
Abstract
The contraction and relaxation of the heart is controlled by stimulation of the β1-adrenoreceptor (AR) signaling cascade, which leads to activation of cAMP-dependent protein kinase (PKA) and subsequent cardiac protein phosphorylation. Phosphorylation is counteracted by the main cardiac protein phosphatases, PP2A and PP1. Both kinase and phosphatases are sensitive to intramolecular disulfide formation in their catalytic subunits that inhibits their activity. Additionally, intermolecular disulfide formation between PKA type I regulatory subunits (PKA-RI) has been described to enhance PKA's affinity for protein kinase A anchoring proteins, which alters its subcellular distribution. Nitroxyl donors have been shown to affect contractility and relaxation, but the mechanistic basis for this effect is unclear. The present study investigates the impact of several nitroxyl donors and the thiol-oxidizing agent diamide on cardiac myocyte protein phosphorylation and oxidation. Although all tested compounds equally induced intermolecular disulfide formation in PKA-RI, only 1-nitrosocyclohexalycetate (NCA) and diamide induced reproducible protein phosphorylation. Phosphorylation occurred independently of β1-AR activation, but was abolished after pharmacological PKA inhibition and thus potentially attributable to increased PKA activity. NCA treatment of cardiac myocytes induced translocation of PKA and phosphatases to the myofilament compartment as shown by fractionation, immunofluorescence, and proximity ligation assays. Assessment of kinase and phosphatase activity within the myofilament fraction of cardiac myocytes after exposure to NCA revealed activation of PKA and inhibition of phosphatase activity thus explaining the increase in phosphorylation. The data suggest that the NCA-mediated effect on cardiac myocyte protein phosphorylation orchestrates alterations in the kinase/phosphatase balance.
Collapse
Affiliation(s)
- Simon Diering
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konstantina Stathopoulou
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mara Goetz
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Rathjens
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sönke Harder
- Institut für Klinische Chemie und Laboratoriumsmedizin, Massenspektrometrische Proteomanalytik, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Angelika Piasecki
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janice Raabe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Steven Schulz
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mona Brandt
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Pflaumenbaum
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrike Fuchs
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonia Donzelli
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sakthivel Sadayappan
- University of Cincinnati Heart, Lung and Vascular Institute, Cardiovascular Center, Cincinnati, Ohio USA
| | - Viacheslav O Nikolaev
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Experimental Cardiovascular Research, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederik Flenner
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics (School of Basic and Medical Biosciences) and School of Cardiovascular Medicine and Sciences, British Heart Foundation Research Excellence Center, King's College London, London, United Kingdom
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
35
|
Arif M, Nabavizadeh P, Song T, Desai D, Singh R, Bazrafshan S, Kumar M, Wang Y, Gilbert RJ, Dhandapany PS, Becker RC, Kranias EG, Sadayappan S. Genetic, clinical, molecular, and pathogenic aspects of the South Asian-specific polymorphic MYBPC3 Δ25bp variant. Biophys Rev 2020; 12:1065-1084. [PMID: 32656747 PMCID: PMC7429610 DOI: 10.1007/s12551-020-00725-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a cardiac genetic disease characterized by ventricular enlargement, diastolic dysfunction, and increased risk for sudden cardiac death. Sarcomeric genetic defects are the predominant known cause of HCM. In particular, mutations in the myosin-binding protein C gene (MYBPC3) are associated with ~ 40% of all HCM cases in which a genetic basis has been established. A decade ago, our group reported a 25-base pair deletion in intron 32 of MYBPC3 (MYBPC3Δ25bp) that is uniquely prevalent in South Asians and is associated with autosomal dominant cardiomyopathy. Although our studies suggest that this deletion results in left ventricular dysfunction, cardiomyopathies, and heart failure, the precise mechanism by which this variant predisposes to heart disease remains unclear. Increasingly appreciated, however, is the contribution of secondary risk factors, additional mutations, and lifestyle choices in augmenting or modifying the HCM phenotype in MYBPC3Δ25bp carriers. Therefore, the goal of this review article is to summarize the current research dedicated to understanding the molecular pathophysiology of HCM in South Asians with the MYBPC3Δ25bp variant. An emphasis is to review the latest techniques currently applied to explore the MYBPC3Δ25bp pathogenesis and to provide a foundation for developing new diagnostic strategies and advances in therapeutics.
Collapse
Affiliation(s)
- Mohammed Arif
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA.
| | - Pooneh Nabavizadeh
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Darshini Desai
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Rohit Singh
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Sholeh Bazrafshan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Richard J Gilbert
- Research Service, Providence VA Medical Center, Providence, RI, 02908, USA
| | - Perundurai S Dhandapany
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Medicine, Oregon Health and Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Richard C Becker
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| |
Collapse
|
36
|
Sarkar SS, Trivedi DV, Morck MM, Adhikari AS, Pasha SN, Ruppel KM, Spudich JA. The hypertrophic cardiomyopathy mutations R403Q and R663H increase the number of myosin heads available to interact with actin. SCIENCE ADVANCES 2020; 6:eaax0069. [PMID: 32284968 PMCID: PMC7124958 DOI: 10.1126/sciadv.aax0069] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 01/09/2020] [Indexed: 05/20/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) mutations in β-cardiac myosin and myosin binding protein-C (MyBP-C) lead to hypercontractility of the heart, an early hallmark of HCM. We show that hypercontractility caused by the HCM-causing mutation R663H cannot be explained by changes in fundamental myosin contractile parameters, much like the HCM-causing mutation R403Q. Using enzymatic assays with purified human β-cardiac myosin, we provide evidence that both mutations cause hypercontractility by increasing the number of functionally accessible myosin heads. We also demonstrate that the myosin mutation R403Q, but not R663H, ablates the binding of myosin with the C0-C7 fragment of MyBP-C. Furthermore, addition of C0-C7 decreases the wild-type myosin basal ATPase single turnover rate, while the mutants do not show a similar reduction. These data suggest that a primary mechanism of action for these mutations is to increase the number of myosin heads functionally available for interaction with actin, which could contribute to hypercontractility.
Collapse
Affiliation(s)
- Saswata S. Sarkar
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Darshan V. Trivedi
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Makenna M. Morck
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arjun S. Adhikari
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shaik N. Pasha
- National Centre for Biological Sciences (TIFR), GKVK Campus, Bellary Road, Bangalore, India
| | - Kathleen M. Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
37
|
Bunch TA, Kanassatega RS, Lepak VC, Colson BA. Human cardiac myosin-binding protein C restricts actin structural dynamics in a cooperative and phosphorylation-sensitive manner. J Biol Chem 2019; 294:16228-16240. [PMID: 31519753 PMCID: PMC6827302 DOI: 10.1074/jbc.ra119.009543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/11/2019] [Indexed: 11/20/2022] Open
Abstract
Cardiac myosin-binding protein C (cMyBP-C) is a thick filament-associated protein that influences actin-myosin interactions. cMyBP-C alters myofilament structure and contractile properties in a protein kinase A (PKA) phosphorylation-dependent manner. To determine the effects of cMyBP-C and its phosphorylation on the microsecond rotational dynamics of actin filaments, we attached a phosphorescent probe to F-actin at Cys-374 and performed transient phosphorescence anisotropy (TPA) experiments. Binding of cMyBP-C N-terminal domains (C0-C2) to labeled F-actin reduced rotational flexibility by 20-25°, indicated by increased final anisotropy of the TPA decay. The effects of C0-C2 on actin TPA were highly cooperative (n = ∼8), suggesting that the cMyBP-C N terminus impacts the rotational dynamics of actin spanning seven monomers (i.e. the length of tropomyosin). PKA-mediated phosphorylation of C0-C2 eliminated the cooperative effects on actin flexibility and modestly increased actin rotational rates. Effects of Ser to Asp phosphomimetic substitutions in the M-domain of C0-C2 on actin dynamics only partially recapitulated the phosphorylation effects. C0-C1 (lacking M-domain/C2) similarly exhibited reduced cooperativity, but not as reduced as by phosphorylated C0-C2. These results suggest an important regulatory role of the M-domain in cMyBP-C effects on actin structural dynamics. In contrast, phosphomimetic substitution of the glycogen synthase kinase (GSK3β) site in the Pro/Ala-rich linker of C0-C2 did not significantly affect the TPA results. We conclude that cMyBP-C binding and PKA-mediated phosphorylation can modulate actin dynamics. We propose that these N-terminal cMyBP-C-induced changes in actin dynamics help explain the functional effects of cMyBP-C phosphorylation on actin-myosin interactions.
Collapse
Affiliation(s)
- Thomas A Bunch
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona 85724
| | | | - Victoria C Lepak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona 85724
| | - Brett A Colson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona 85724
| |
Collapse
|
38
|
Skeletal MyBP-C isoforms tune the molecular contractility of divergent skeletal muscle systems. Proc Natl Acad Sci U S A 2019; 116:21882-21892. [PMID: 31591218 DOI: 10.1073/pnas.1910549116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle myosin-binding protein C (MyBP-C) is a myosin thick filament-associated protein, localized through its C terminus to distinct regions (C-zones) of the sarcomere. MyBP-C modulates muscle contractility, presumably through its N terminus extending from the thick filament and interacting with either the myosin head region and/or the actin thin filament. Two isoforms of MyBP-C (fast- and slow-type) are expressed in a muscle type-specific manner. Are the expression, localization, and Ca2+-dependent modulatory capacities of these isoforms different in fast-twitch extensor digitorum longus (EDL) and slow-twitch soleus (SOL) muscles derived from Sprague-Dawley rats? By mass spectrometry, 4 MyBP-C isoforms (1 fast-type MyBP-C and 3 N-terminally spliced slow-type MyBP-C) were expressed in EDL, but only the 3 slow-type MyBP-C isoforms in SOL. Using EDL and SOL native thick filaments in which the MyBP-C stoichiometry and localization are preserved, native thin filament sliding over these thick filaments showed that, only in the C-zone, MyBP-C Ca2+ sensitizes the thin filament and slows thin filament velocity. These modulatory properties depended on MyBP-C's N terminus as N-terminal proteolysis attenuated MyBP-C's functional capacities. To determine each MyBP-C isoform's contribution to thin filament Ca2+ sensitization and slowing in the C-zone, we used a combination of in vitro motility assays using expressed recombinant N-terminal fragments and in silico mechanistic modeling. Our results suggest that each skeletal MyBP-C isoform's N terminus is functionally distinct and has modulatory capacities that depend on the muscle type in which they are expressed, providing the potential for molecular tuning of skeletal muscle performance through differential MyBP-C expression.
Collapse
|
39
|
Viswanathan SK, Puckelwartz MJ, Mehta A, Ramachandra CJA, Jagadeesan A, Fritsche-Danielson R, Bhat RV, Wong P, Kandoi S, Schwanekamp JA, Kuffel G, Pesce LL, Zilliox MJ, Durai UNB, Verma RS, Molokie RE, Suresh DP, Khoury PR, Thomas A, Sanagala T, Tang HC, Becker RC, Knöll R, Shim W, McNally EM, Sadayappan S. Association of Cardiomyopathy With MYBPC3 D389V and MYBPC3Δ25bpIntronic Deletion in South Asian Descendants. JAMA Cardiol 2019; 3:481-488. [PMID: 29641836 DOI: 10.1001/jamacardio.2018.0618] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Importance The genetic variant MYBPC3Δ25bp occurs in 4% of South Asian descendants, with an estimated 100 million carriers worldwide. MYBPC3 Δ25bp has been linked to cardiomyopathy and heart failure. However, the high prevalence of MYBPC3Δ25bp suggests that other stressors act in concert with MYBPC3Δ25bp. Objective To determine whether there are additional genetic factors that contribute to the cardiomyopathic expression of MYBPC3Δ25bp. Design, Setting, andParticipants South Asian individuals living in the United States were screened for MYBPC3Δ25bp, and a subgroup was clinically evaluated using electrocardiograms and echocardiograms at Loyola University, Chicago, Illinois, between January 2015 and July 2016. Main Outcomes and Measures Next-generation sequencing of 174 cardiovascular disease genes was applied to identify additional modifying gene mutations and correlate genotype-phenotype parameters. Cardiomyocytes derived from human-induced pluripotent stem cells were established and examined to assess the role of MYBPC3Δ25bp. Results In this genotype-phenotype study, individuals of South Asian descent living in the United States from both sexes (36.23% female) with a mean population age of 48.92 years (range, 18-84 years) were recruited. Genetic screening of 2401 US South Asian individuals found an MYBPC3Δ25bpcarrier frequency of 6%. A higher frequency of missense TTN variation was found in MYBPC3Δ25bp carriers compared with noncarriers, identifying distinct genetic backgrounds within the MYBPC3Δ25bp carrier group. Strikingly, 9.6% of MYBPC3Δ25bp carriers also had a novel MYBPC3 variant, D389V. Family studies documented D389V was in tandem on the same allele as MYBPC3Δ25bp, and D389V was only seen in the presence of MYBPC3Δ25bp. In contrast to MYBPC3Δ25bp, MYBPC3Δ25bp/D389V was associated with hyperdynamic left ventricular performance (mean [SEM] left ventricular ejection fraction, 66.7 [0.7%]; left ventricular fractional shortening, 36.6 [0.6%]; P < .03) and stem cell-derived cardiomyocytes exhibited cellular hypertrophy with abnormal Ca2+ transients. Conclusions and Relevance MYBPC3Δ25bp/D389V is associated with hyperdynamic features, which are an early finding in hypertrophic cardiomyopathy and thought to reflect an unfavorable energetic state. These findings support that a subset of MYBPC3Δ25bp carriers, those with D389V, account for the increased risk attributed to MYBPC3Δ25bp.
Collapse
Affiliation(s)
- Shiv Kumar Viswanathan
- Heart, Lung and Vascular Institute, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio.,Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois
| | | | - Ashish Mehta
- National Heart Research Institute Singapore.,Cardiovascular Academic Clinical Program, DUKE-NUS Medical School, Singapore.,PSC and Phenotyping Laboratory, Victor Chang Cardiac Research Institute, Sydney, Australia
| | | | | | - Regina Fritsche-Danielson
- Cardiovascular and Metabolic Disease Innovative Medicines and Early Development Unit, AstraZeneca Research and Development, Gothenburg, Sweden
| | - Ratan V Bhat
- Cardiovascular and Metabolic Disease Innovative Medicines and Early Development Unit, AstraZeneca Research and Development, Gothenburg, Sweden
| | - Philip Wong
- National Heart Research Institute Singapore.,Cardiovascular and Metabolic Disorders Program, DUKE-NUS Medical School, Singapore.,Department of Cardiology, National Heart Centre Singapore, Singapore
| | - Sangeetha Kandoi
- Heart, Lung and Vascular Institute, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio.,Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois.,Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Jennifer A Schwanekamp
- Heart, Lung and Vascular Institute, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Gina Kuffel
- Department of Public Health Sciences, Loyola University Chicago, Maywood, Illinois
| | - Lorenzo L Pesce
- Computation Institute, The University of Chicago, Chicago, Illinois
| | - Michael J Zilliox
- Department of Public Health Sciences, Loyola University Chicago, Maywood, Illinois
| | - U Nalla B Durai
- Divison of Hematology and Oncology, University of Illinois at Chicago
| | - Rama Shanker Verma
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Robert E Molokie
- Divison of Hematology and Oncology, University of Illinois at Chicago
| | | | - Philip R Khoury
- Heart Institute, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Annie Thomas
- Marcella Niehoff School of Nursing, Loyola University Chicago, Maywood, Illinois
| | - Thriveni Sanagala
- Department of Cardiology and Echocardiography and Cardiographics, Loyola University Chicago, Maywood, Illinois
| | - Hak Chiaw Tang
- Department of Cardiology, National Heart Centre Singapore, Singapore
| | - Richard C Becker
- Heart, Lung and Vascular Institute, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Ralph Knöll
- Cardiovascular and Metabolic Disease Innovative Medicines and Early Development Unit, AstraZeneca Research and Development, Gothenburg, Sweden.,Integrated Cardio-Metabolic Centre, Myocardial Genetics, Karolinska Institutet, University Hospital, Heart and Vascular Theme, Stockholm, Sweden
| | - Winston Shim
- National Heart Research Institute Singapore.,Cardiovascular and Metabolic Disorders Program, DUKE-NUS Medical School, Singapore
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois.,Associate Editor for Translational Science
| | - Sakthivel Sadayappan
- Heart, Lung and Vascular Institute, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio.,Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois
| |
Collapse
|
40
|
Nabiev SR, Kopylova GV, Shchepkin DV. The Effect of Cardiac Myosin-Binding Protein C on Calcium Regulation of the Actin–Myosin Interaction Depends on Myosin Light Chain Isoforms. Biophysics (Nagoya-shi) 2019. [DOI: 10.1134/s000635091905018x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
41
|
Cardiac myosin binding protein-C phosphorylation regulates the super-relaxed state of myosin. Proc Natl Acad Sci U S A 2019; 116:11731-11736. [PMID: 31142654 DOI: 10.1073/pnas.1821660116] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Phosphorylation of cardiac myosin binding protein-C (cMyBP-C) accelerates cardiac contractility. However, the mechanisms by which cMyBP-C phosphorylation increases contractile kinetics have not been fully elucidated. In this study, we tested the hypothesis that phosphorylation of cMyBP-C releases myosin heads from the inhibited super-relaxed state (SRX), thereby determining the fraction of myosin available for contraction. Mice with various alanine (A) or aspartic acid (D) substitutions of the three main phosphorylatable serines of cMyBP-C (serines 273, 282, and 302) were used to address the association between cMyBP-C phosphorylation and SRX. Single-nucleotide turnover in skinned ventricular preparations demonstrated that phosphomimetic cMyBP-C destabilized SRX, whereas phospho-ablated cMyBP-C had a stabilizing effect on SRX. Strikingly, phosphorylation at serine 282 site was found to play a critical role in regulating the SRX. Treatment of WT preparations with protein kinase A (PKA) reduced the SRX, whereas, in nonphosphorylatable cMyBP-C preparations, PKA had no detectable effect. Mice with stable SRX exhibited reduced force production. Phosphomimetic cMyBP-C with reduced SRX exhibited increased rates of tension redevelopment and reduced binding to myosin. We also used recombinant myosin subfragment-2 to disrupt the endogenous interaction between cMyBP-C and myosin and observed a significant reduction in the population of SRX myosin. This peptide also increased force generation and rate of tension redevelopment in skinned fibers. Taken together, this study demonstrates that the phosphorylation-dependent interaction between cMyBP-C and myosin is a determinant of the fraction of myosin available for contraction. Furthermore, the binding between cMyBP-C and myosin may be targeted to improve contractile function.
Collapse
|
42
|
Spudich JA. Three perspectives on the molecular basis of hypercontractility caused by hypertrophic cardiomyopathy mutations. Pflugers Arch 2019; 471:701-717. [PMID: 30767072 PMCID: PMC6475635 DOI: 10.1007/s00424-019-02259-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/11/2019] [Accepted: 01/20/2019] [Indexed: 01/10/2023]
Abstract
Several lines of evidence suggest that the primary effect of hypertrophic cardiomyopathy mutations in human β-cardiac myosin is hypercontractility of the heart, which leads to subsequent hypertrophy, fibrosis, and myofilament disarray. Here, I describe three perspectives on the molecular basis of this hypercontractility. The first is that hypercontractility results from changes in the fundamental parameters of the actin-activated β-cardiac myosin chemo-mechanical ATPase cycle. The second considers that hypercontractility results from an increase in the number of functionally accessible heads in the sarcomere for interaction with actin. The final and third perspective is that load dependence of contractility is affected by cardiomyopathy mutations and small-molecule effectors in a manner that changes the power output of cardiac contraction. Experimental approaches associated with each perspective are described along with concepts of therapeutic approaches that could prove valuable in treating hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
43
|
Role of intrinsic disorder in muscle sarcomeres. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 166:311-340. [PMID: 31521234 DOI: 10.1016/bs.pmbts.2019.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The role and utility of intrinsically disordered regions (IDRs) is reviewed for two groups of sarcomeric proteins, such as members of tropomodulin/leiomodin (Tmod/Lmod) protein homology group and myosin binding protein C (MyBP-C). These two types of sarcomeric proteins represent very different but strongly interdependent functions, being responsible for maintaining structure and operation of the muscle sarcomere. The role of IDRs in the formation of complexes between thin filaments and Tmods/Lmods is discussed within the framework of current understanding of the thin filament length regulation. For MyBP-C, the function of IDRs is discussed in the context of MYBP-C-dependent sarcomere contraction and actomyosin activation.
Collapse
|
44
|
Revealing the mechanism of how cardiac myosin-binding protein C N-terminal fragments sensitize thin filaments for myosin binding. Proc Natl Acad Sci U S A 2019; 116:6828-6835. [PMID: 30877248 PMCID: PMC6452674 DOI: 10.1073/pnas.1816480116] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Diverse demands on cardiac muscle require the fine-tuning of contraction. Cardiac myosin binding protein-C (cMyBP-C) is involved in this regulation; however, its precise molecular mechanism of action remains uncertain. By imaging the interactions of single myosin and cMyBP-C molecules interacting with suspended thin filaments in vitro we observe cMyBP-C N-terminal fragments assist activation and modulate contraction velocity by affecting myosin binding to the thin filament. Fluorescent imaging of Cy3-labeled cMyBP-C revealed that it diffusively scans the thin filament and then strongly binds to displace tropomyosin and activate at low calcium. At high calcium, cMyBP-C decorates the filament more extensively, reducing myosin binding through competing with binding sites. Understanding the mechanism of MyBP-C action has important implications for heart disease. Cardiac muscle contraction is triggered by calcium binding to troponin. The consequent movement of tropomyosin permits myosin binding to actin, generating force. Cardiac myosin-binding protein C (cMyBP-C) plays a modulatory role in this activation process. One potential mechanism for the N-terminal domains of cMyBP-C to achieve this is by binding directly to the actin-thin filament at low calcium levels to enhance the movement of tropomyosin. To determine the molecular mechanisms by which cMyBP-C enhances myosin recruitment to the actin-thin filament, we directly visualized fluorescently labeled cMyBP-C N-terminal fragments and GFP-labeled myosin molecules binding to suspended actin-thin filaments in a fluorescence-based single-molecule microscopy assay. Binding of the C0C3 N-terminal cMyBP-C fragment to the thin filament enhanced myosin association at low calcium levels. However, at high calcium levels, C0C3 bound in clusters, blocking myosin binding. Dynamic imaging of thin filament-bound Cy3-C0C3 molecules demonstrated that these fragments diffuse along the thin filament before statically binding, suggesting a mechanism that involves a weak-binding mode to search for access to the thin filament and a tight-binding mode to sensitize the thin filament to calcium, thus enhancing myosin binding. Although shorter N-terminal fragments (Cy3-C0C1 and Cy3-C0C1f) bound to the thin filaments and displayed modes of motion on the thin filament similar to that of the Cy3-C0C3 fragment, the shorter fragments were unable to sensitize the thin filament. Therefore, the longer N-terminal fragment (C0C3) must possess the requisite domains needed to bind specifically to the thin filament in order for the cMyBP-C N terminus to modulate cardiac contractility.
Collapse
|
45
|
van der Velden J, Stienen GJM. Cardiac Disorders and Pathophysiology of Sarcomeric Proteins. Physiol Rev 2019; 99:381-426. [PMID: 30379622 DOI: 10.1152/physrev.00040.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The sarcomeric proteins represent the structural building blocks of heart muscle, which are essential for contraction and relaxation. During recent years, it has become evident that posttranslational modifications of sarcomeric proteins, in particular phosphorylation, tune cardiac pump function at rest and during exercise. This delicate, orchestrated interaction is also influenced by mutations, predominantly in sarcomeric proteins, which cause hypertrophic or dilated cardiomyopathy. In this review, we follow a bottom-up approach starting from a description of the basic components of cardiac muscle at the molecular level up to the various forms of cardiac disorders at the organ level. An overview is given of sarcomere changes in acquired and inherited forms of cardiac disease and the underlying disease mechanisms with particular reference to human tissue. A distinction will be made between the primary defect and maladaptive/adaptive secondary changes. Techniques used to unravel functional consequences of disease-induced protein changes are described, and an overview of current and future treatments targeted at sarcomeric proteins is given. The current evidence presented suggests that sarcomeres not only form the basis of cardiac muscle function but also represent a therapeutic target to combat cardiac disease.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Ger J M Stienen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| |
Collapse
|
46
|
Giles J, Patel JR, Miller A, Iverson E, Fitzsimons D, Moss RL. Recovery of left ventricular function following in vivo reexpression of cardiac myosin binding protein C. J Gen Physiol 2019; 151:77-89. [PMID: 30573635 PMCID: PMC6314388 DOI: 10.1085/jgp.201812238] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/27/2018] [Indexed: 01/26/2023] Open
Abstract
The loss of cardiac myosin binding protein C (cMyBP-C) results in left ventricular dilation, cardiac hypertrophy, and impaired ventricular function in both constitutive and conditional cMyBP-C knockout (MYBPC3 null) mice. It remains unclear whether the structural and functional phenotypes expressed in the MYBPC3 null mouse are reversible, which is an important question, since reduced expression of cMyBP-C is an important cause of hypertrophic cardiomyopathy in humans. To investigate this question, we generated a cardiac-specific transgenic mouse model using a Tet-Off inducible system to permit the controlled expression of WT cMyBP-C on the MYBPC3 null background. Functional Tet-Off mice expressing WT cMyBP-C (FT-WT) were generated by crossing tetracycline transactivator mice with responder mice carrying the WT cMyBP-C transgene. Prior to dietary doxycycline administration, cMyBP-C was expressed at normal levels in FT-WT myocardium, which exhibited similar levels of steady-state force and in vivo left ventricular function as WT mice. Introduction of dietary doxycycline for four weeks resulted in a partial knockdown of cMyBP-C expression and commensurate impairment of systolic and diastolic function to levels approaching those observed in MYBPC 3 null mice. Subsequent withdrawal of doxycycline from the diet resulted in the reexpression of cMyBP-C to levels comparable to those observed in WT mice, along with near-complete recovery of in vivo ventricular function. These results show that the cardiac phenotypes associated with MYBPC3 null mice are reversible. Our work also validates the use of the Tet-Off inducible system as a means to study the mechanisms underlying hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Jasmine Giles
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Jitandrakumar R Patel
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Adam Miller
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Elizabeth Iverson
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Daniel Fitzsimons
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Richard L Moss
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
47
|
Doh CY, Li J, Mamidi R, Stelzer JE. The HCM-causing Y235S cMyBPC mutation accelerates contractile function by altering C1 domain structure. Biochim Biophys Acta Mol Basis Dis 2019; 1865:661-677. [PMID: 30611859 DOI: 10.1016/j.bbadis.2019.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/18/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022]
Abstract
Mutations in cardiac myosin binding protein C (cMyBPC) are a major cause of hypertrophic cardiomyopathy (HCM). In particular, a single amino acid substitution of tyrosine to serine at residue 237 in humans (residue 235 in mice) has been linked to HCM with strong disease association. Although cMyBPC truncations, deletions and insertions, and frame shift mutations have been studied, relatively little is known about the functional consequences of missense mutations in cMyBPC. In this study, we characterized the functional and structural effects of the HCM-causing Y235S mutation by performing mechanical experiments and molecular dynamics simulations (MDS). cMyBPC null mouse myocardium was virally transfected with wild-type (WT) or Y235S cMyBPC (KOY235S). We found that Y235S cMyBPC was properly expressed and incorporated into the cardiac sarcomere, suggesting that the mechanism of disease of the Y235S mutation is not haploinsufficiency or poison peptides. Mechanical experiments in detergent-skinned myocardium isolated from KOY235S hearts revealed hypercontractile behavior compared to KOWT hearts, evidenced by accelerated cross-bridge kinetics and increased Ca2+ sensitivity of force generation. In addition, MDS revealed that the Y235S mutation causes alterations in important intramolecular interactions, surface conformations, and electrostatic potential of the C1 domain of cMyBPC. Our combined in vitro and in silico data suggest that the Y235S mutation directly disrupts internal and surface properties of the C1 domain of cMyBPC, which potentially alters its ligand-binding interactions. These molecular changes may underlie the mechanism for hypercontractile cross-bridge behavior, which ultimately results in the development of cardiac hypertrophy and in vivo cardiac dysfunction.
Collapse
Affiliation(s)
- Chang Yoon Doh
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
48
|
Risi C, Belknap B, Forgacs-Lonart E, Harris SP, Schröder GF, White HD, Galkin VE. N-Terminal Domains of Cardiac Myosin Binding Protein C Cooperatively Activate the Thin Filament. Structure 2018; 26:1604-1611.e4. [PMID: 30270174 PMCID: PMC6281772 DOI: 10.1016/j.str.2018.08.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/25/2018] [Accepted: 08/09/2018] [Indexed: 10/28/2022]
Abstract
Muscle contraction relies on interaction between myosin-based thick filaments and actin-based thin filaments. Myosin binding protein C (MyBP-C) is a key regulator of actomyosin interactions. Recent studies established that the N'-terminal domains (NTDs) of MyBP-C can either activate or inhibit thin filaments, but the mechanism of their collective action is poorly understood. Cardiac MyBP-C (cMyBP-C) harbors an extra NTD, which is absent in skeletal isoforms of MyBP-C, and its role in regulation of cardiac contraction is unknown. Here we show that the first two domains of human cMyPB-C (i.e., C0 and C1) cooperate to activate the thin filament. We demonstrate that C1 interacts with tropomyosin via a positively charged loop and that this interaction, stabilized by the C0 domain, is required for thin filament activation by cMyBP-C. Our data reveal a mechanism by which cMyBP-C can modulate cardiac contraction and demonstrate a function of the C0 domain.
Collapse
Affiliation(s)
- Cristina Risi
- Department of Physiological Sciences, Eastern Virginia Medical School, 700 West Olney Road, Lewis Hall, Room 3126, Norfolk, VA 23507, USA
| | - Betty Belknap
- Department of Physiological Sciences, Eastern Virginia Medical School, 700 West Olney Road, Lewis Hall, Room 3126, Norfolk, VA 23507, USA
| | - Eva Forgacs-Lonart
- Department of Physiological Sciences, Eastern Virginia Medical School, 700 West Olney Road, Lewis Hall, Room 3126, Norfolk, VA 23507, USA
| | - Samantha P Harris
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Gunnar F Schröder
- Institute of Complex Systems ICS-6, Forschungszentrum Jülich, 52425 Jülich, Germany; Physics Department, Heinrich-Heine Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Howard D White
- Department of Physiological Sciences, Eastern Virginia Medical School, 700 West Olney Road, Lewis Hall, Room 3126, Norfolk, VA 23507, USA
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, 700 West Olney Road, Lewis Hall, Room 3126, Norfolk, VA 23507, USA.
| |
Collapse
|
49
|
Bunch TA, Lepak VC, Kanassatega RS, Colson BA. N-terminal extension in cardiac myosin-binding protein C regulates myofilament binding. J Mol Cell Cardiol 2018; 125:140-148. [PMID: 30359561 DOI: 10.1016/j.yjmcc.2018.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/22/2018] [Accepted: 10/10/2018] [Indexed: 10/28/2022]
Abstract
RATIONALE Mutations in the gene encoding the sarcomeric protein cardiac myosin-binding protein C (cMyBP-C) are a leading cause of hypertrophic cardiomyopathy (HCM). Mouse models targeting cMyBP-C and use of recombinant proteins have been effective in studying its roles in contractile function and disease. Surprisingly, while the N-terminus of cMyBP-C is important to regulate myofilament binding and contains many HCM mutations, an incorrect sequence, lacking the N-terminal 8 amino acids has been used in many studies. OBJECTIVES To determine the N-terminal cMyBP-C sequences in ventricles and investigate the roles of species-specific differences in cMyBP-C on myofilament binding. METHODS AND RESULTS We determined cMyBP-C sequences in mouse and human by inspecting available sequence databases. N-terminal differences were confirmed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Cosedimentation assays with actin or myosin were used to examine binding in mouse, human and chimeric fusion proteins of cMyBP-C. Time-resolved FRET (TR-FRET) with site-directed probes on cMyBP-C was employed to measure structural dynamics. LC-MS/MS supported the sequencing data that mouse cMyBP-C contains an eight-residue N-terminal extension (NTE) not found in human. Cosedimentation assays revealed that cardiac myosin binding was strongly influenced by the presence of the NTE, which reduced binding by 60%. 75% more human C0-C2 than mouse bound to myosin. Actin binding of mouse C0-C2 was not affected by the NTE. 50% more human C0-C2 than mouse bound to actin. TR-FRET indicates that the NTE did not significantly affect structural dynamics across domains C0 and C1. CONCLUSIONS Our functional results are consistent with the idea that cardiac myosin binding of N-terminal cMyBP-C is reduced in the mouse protein due to the presence of the NTE, which is proposed to interfere with myosin regulatory light chain (RLC) binding. The NTE is a critical component of mouse cMyBP-C, and should be considered in extrapolation of studies to cMyBP-C and HCM mechanisms in human.
Collapse
Affiliation(s)
- Thomas A Bunch
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Victoria C Lepak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Rhye-Samuel Kanassatega
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States
| | - Brett A Colson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, United States.
| |
Collapse
|
50
|
McNamara JW, Sadayappan S. Skeletal myosin binding protein-C: An increasingly important regulator of striated muscle physiology. Arch Biochem Biophys 2018; 660:121-128. [PMID: 30339776 DOI: 10.1016/j.abb.2018.10.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/07/2018] [Accepted: 10/11/2018] [Indexed: 12/22/2022]
Abstract
The Myosin Binding Protein-C (MyBP-C) family is a group of sarcomeric proteins important for striated muscle structure and function. Comprising approximately 2% of the myofilament mass, MyBP-C has important roles in both contraction and relaxation. Three paralogs of MyBP-C are encoded by separate genes with distinct expression profiles in striated muscle. In mammals, cardiac MyBP-C is limited to the heart, and it is the most extensively studied owing to its involvement in cardiomyopathies. However, the roles of two skeletal paralogs, slow and fast, in muscle biology remain poorly characterized. Nonetheless, both have been recently implicated in the development of skeletal myopathies. This calls for a better understanding of their function in the pathophysiology of distal arthrogryposis. This review characterizes MyBP-C as a whole and points out knowledge gaps that still remain with respect to skeletal MyBP-C.
Collapse
Affiliation(s)
- James W McNamara
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45236, USA
| | - Sakthivel Sadayappan
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45236, USA.
| |
Collapse
|