1
|
Wood PT, Seffrood MM, Colson BA, Stelzer JE. cMyBP-C in hypertrophic cardiomyopathy: gene therapy and small-molecule innovations. Front Cardiovasc Med 2025; 12:1550649. [PMID: 40134985 PMCID: PMC11935118 DOI: 10.3389/fcvm.2025.1550649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/13/2025] [Indexed: 03/27/2025] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic disorder in the heart caused by variants in sarcomeric proteins that disrupt myocardial function, leading to hypercontractility, hypertrophy, and fibrosis. Optimal cardiac function relies on the precise coordination of thin and thick filament proteins that control the timing, magnitude of cellular force generation and relaxation, and in vivo systolic and diastolic function. Sarcomeric proteins, such as cardiac myosin binding protein C (cMyBP-C) play a crucial role in myocardial contractile function by modulating actomyosin interactions. Genetic variants in cMyBP-C are a frequent cause of HCM, highlighting its importance in cardiac health. This review explores the molecular mechanisms underpinning HCM and the rapidly advancing field of HCM translational research, including gene therapy and small-molecule interventions targeting sarcomere function. We will highlight novel approaches, including gene therapy using recombinant AAV vectors and small-molecule drugs targeting sarcomere function.
Collapse
Affiliation(s)
- Patrick T. Wood
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Morgan M. Seffrood
- Department of Cellular & Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Brett A. Colson
- Department of Cellular & Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
2
|
Fitts RH, Wang X, Kwok WM, Camara AKS. Cardiomyocyte Adaptation to Exercise: K+ Channels, Contractility and Ischemic Injury. Int J Sports Med 2024; 45:791-803. [PMID: 38648799 DOI: 10.1055/a-2296-7604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality, and exercise-training (TRN) is known to reduce risk factors and protect the heart from ischemia and reperfusion injury. Though the cardioprotective effects of exercise are well-documented, underlying mechanisms are not well understood. This review highlights recent findings and focuses on cardiac factors with emphasis on K+ channel control of the action potential duration (APD), β-adrenergic and adenosine regulation of cardiomyocyte function, and mitochondrial Ca2+ regulation. TRN-induced prolongation and shortening of the APD at low and high activation rates, respectively, is discussed in the context of a reduced response of the sarcolemma delayed rectifier potassium channel (IK) and increased content and activation of the sarcolemma KATP channel. A proposed mechanism underlying the latter is presented, including the phosphatidylinositol-3kinase/protein kinase B pathway. TRN induced increases in cardiomyocyte contractility and the response to adrenergic agonists are discussed. The TRN-induced protection from reperfusion injury is highlighted by the increased content and activation of the sarcolemma KATP channel and the increased phosphorylated glycogen synthase kinase-3β, which aid in preventing mitochondrial Ca2+ overload and mitochondria-triggered apoptosis. Finally, a brief section is presented on the increased incidences of atrial fibrillation associated with age and in life-long exercisers.
Collapse
Affiliation(s)
- Robert H Fitts
- Biological Sciences, Marquette University, Milwaukee, United States
| | - Xinrui Wang
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
| | - Wai-Meng Kwok
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
- Anesthesiology, Medical College of Wisconsin, Milwaukee, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, United States
| | - Amadou K S Camara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States
- Anesthesiology, Medical College of Wisconsin, Milwaukee, United States
- Cancer Center, Medical College of Wisconsin, Milwaukee, United States
- Physiology, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
3
|
Han YS, Pakkam M, Fogarty MJ, Sieck GC, Brozovich FV. Alterations in cardiac contractile and regulatory proteins contribute to age-related cardiac dysfunction in male rats. Physiol Rep 2024; 12:e70012. [PMID: 39169429 PMCID: PMC11338742 DOI: 10.14814/phy2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
Aging is associated with cardiac contractile abnormalities, but the etiology of these contractile deficits is unclear. We hypothesized that cardiac contractile and regulatory protein expression is altered during aging. To investigate this possibility, left ventricular (LV) lysates were prepared from young (6 months) and old (24 months) Fischer344 rats. There are no age-related changes in SERCA2 expression or phospholamban phosphorylation. Additionally, neither titin isoform expression nor phosphorylation differed. However, there is a significant increase in β-isoform of the myosin heavy chain (MyHC) expression and phosphorylation of TnI and MyBP-C during aging. In permeabilized strips of papillary muscle, force and Ca2+ sensitivity are reduced during aging, consistent with the increase in β-MyHC expression and TnI phosphorylation. However, the increase in MyBP-C phosphorylation during aging may represent a mechanism to compensate for age-related contractile deficits. In isolated cardiomyocytes loaded with Fura-2, the peak of the Ca2+ transient is reduced, but the kinetics of the Ca2+ transient are not altered. Furthermore, the extent of shortening and the rates of both sarcomere shortening and re-lengthening are reduced. These results demonstrate that aging is associated with changes in contractile and regulatory protein expression and phosphorylation, which affect the mechanical properties of cardiac muscle.
Collapse
Affiliation(s)
- Young Soo Han
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Madona Pakkam
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Matthew J. Fogarty
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Gary C. Sieck
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Frank V. Brozovich
- Department of Physiology & Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Department of Cardiovascular DiseasesMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
4
|
Steczina S, Mohran S, Bailey LRJ, McMillen TS, Kooiker KB, Wood NB, Davis J, Previs MJ, Olivotto I, Pioner JM, Geeves MA, Poggesi C, Regnier M. MYBPC3-c.772G>A mutation results in haploinsufficiency and altered myosin cycling kinetics in a patient induced stem cell derived cardiomyocyte model of hypertrophic cardiomyopathy. J Mol Cell Cardiol 2024; 191:27-39. [PMID: 38648963 PMCID: PMC11116032 DOI: 10.1016/j.yjmcc.2024.04.010] [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: 10/13/2023] [Revised: 03/13/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Approximately 40% of hypertrophic cardiomyopathy (HCM) mutations are linked to the sarcomere protein cardiac myosin binding protein-C (cMyBP-C). These mutations are either classified as missense mutations or truncation mutations. One mutation whose nature has been inconsistently reported in the literature is the MYBPC3-c.772G > A mutation. Using patient-derived human induced pluripotent stem cells differentiated to cardiomyocytes (hiPSC-CMs), we have performed a mechanistic study of the structure-function relationship for this MYBPC3-c.772G > A mutation versus a mutation corrected, isogenic cell line. Our results confirm that this mutation leads to exon skipping and mRNA truncation that ultimately suggests ∼20% less cMyBP-C protein (i.e., haploinsufficiency). This, in turn, results in increased myosin recruitment and accelerated myofibril cycling kinetics. Our mechanistic studies suggest that faster ADP release from myosin is a primary cause of accelerated myofibril cross-bridge cycling due to this mutation. Additionally, the reduction in force generating heads expected from faster ADP release during isometric contractions is outweighed by a cMyBP-C phosphorylation mediated increase in myosin recruitment that leads to a net increase of myofibril force, primarily at submaximal calcium activations. These results match well with our previous report on contractile properties from myectomy samples of the patients from whom the hiPSC-CMs were generated, demonstrating that these cell lines are a good model to study this pathological mutation and extends our understanding of the mechanisms of altered contractile properties of this HCM MYBPC3-c.772G > A mutation.
Collapse
Affiliation(s)
- Sonette Steczina
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Saffie Mohran
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Logan R J Bailey
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Molecular and Cellular Biology, University of Washington, Seattle, WA 98109, USA; Department of Lab Medicine and Pathology, University of Washington, Seattle, WA 98109, USA
| | - Timothy S McMillen
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA
| | - Kristina B Kooiker
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Neil B Wood
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05404, USA
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Lab Medicine and Pathology, University of Washington, Seattle, WA 98109, USA
| | - Michael J Previs
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05404, USA
| | - Iacopo Olivotto
- Department of Experimental and Clinical Medicine, Division of Physiology, University of Florence, Italy
| | | | | | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, Division of Physiology, University of Florence, Italy
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Center for Translational Muscle Research, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
5
|
Lee S, Vander Roest AS, Blair CA, Kao K, Bremner SB, Childers MC, Pathak D, Heinrich P, Lee D, Chirikian O, Mohran SE, Roberts B, Smith JE, Jahng JW, Paik DT, Wu JC, Gunawardane RN, Ruppel KM, Mack DL, Pruitt BL, Regnier M, Wu SM, Spudich JA, Bernstein D. Incomplete-penetrant hypertrophic cardiomyopathy MYH7 G256E mutation causes hypercontractility and elevated mitochondrial respiration. Proc Natl Acad Sci U S A 2024; 121:e2318413121. [PMID: 38683993 PMCID: PMC11087781 DOI: 10.1073/pnas.2318413121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/05/2024] [Indexed: 05/02/2024] Open
Abstract
Determining the pathogenicity of hypertrophic cardiomyopathy-associated mutations in the β-myosin heavy chain (MYH7) can be challenging due to its variable penetrance and clinical severity. This study investigates the early pathogenic effects of the incomplete-penetrant MYH7 G256E mutation on myosin function that may trigger pathogenic adaptations and hypertrophy. We hypothesized that the G256E mutation would alter myosin biomechanical function, leading to changes in cellular functions. We developed a collaborative pipeline to characterize myosin function across protein, myofibril, cell, and tissue levels to determine the multiscale effects on structure-function of the contractile apparatus and its implications for gene regulation and metabolic state. The G256E mutation disrupts the transducer region of the S1 head and reduces the fraction of myosin in the folded-back state by 33%, resulting in more myosin heads available for contraction. Myofibrils from gene-edited MYH7WT/G256E human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) exhibited greater and faster tension development. This hypercontractile phenotype persisted in single-cell hiPSC-CMs and engineered heart tissues. We demonstrated consistent hypercontractile myosin function as a primary consequence of the MYH7 G256E mutation across scales, highlighting the pathogenicity of this gene variant. Single-cell transcriptomic and metabolic profiling demonstrated upregulated mitochondrial genes and increased mitochondrial respiration, indicating early bioenergetic alterations. This work highlights the benefit of our multiscale platform to systematically evaluate the pathogenicity of gene variants at the protein and contractile organelle level and their early consequences on cellular and tissue function. We believe this platform can help elucidate the genotype-phenotype relationships underlying other genetic cardiovascular diseases.
Collapse
Affiliation(s)
- Soah Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Biopharmaceutical Convergence, Sungkyunkwan University School of Pharmacy, Suwon, Gyeonggi-do16419South Korea
- School of Pharmacy, Sungkyunkwan University School of Pharmacy, Suwon, Gyeonggi-do16419, South Korea
| | - Alison S. Vander Roest
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA94305
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Cheavar A. Blair
- Biological Engineering, University of California, Santa Barbara, CA93106
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY40536
| | - Kerry Kao
- Department of Bioengineering, University of Washington School of Medicine and College of Engineering, Seattle, WA98195
| | - Samantha B. Bremner
- Department of Bioengineering, University of Washington School of Medicine and College of Engineering, Seattle, WA98195
| | - Matthew C. Childers
- Department of Bioengineering, University of Washington School of Medicine and College of Engineering, Seattle, WA98195
| | - Divya Pathak
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
| | - Paul Heinrich
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Daniel Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Orlando Chirikian
- Biological Engineering, University of California, Santa Barbara, CA93106
| | - Saffie E. Mohran
- Department of Bioengineering, University of Washington School of Medicine and College of Engineering, Seattle, WA98195
| | | | | | - James W. Jahng
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
| | - David T. Paik
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | | | - Kathleen M. Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
| | - David L. Mack
- Department of Bioengineering, University of Washington School of Medicine and College of Engineering, Seattle, WA98195
| | - Beth L. Pruitt
- Biological Engineering, University of California, Santa Barbara, CA93106
| | - Michael Regnier
- Department of Bioengineering, University of Washington School of Medicine and College of Engineering, Seattle, WA98195
| | - Sean M. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - James A. Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
| | - Daniel Bernstein
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
6
|
Solaro RJ, Goldspink PH, Wolska BM. Emerging Concepts of Mechanisms Controlling Cardiac Tension: Focus on Familial Dilated Cardiomyopathy (DCM) and Sarcomere-Directed Therapies. Biomedicines 2024; 12:999. [PMID: 38790961 PMCID: PMC11117855 DOI: 10.3390/biomedicines12050999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Novel therapies for the treatment of familial dilated cardiomyopathy (DCM) are lacking. Shaping research directions to clinical needs is critical. Triggers for the progression of the disorder commonly occur due to specific gene variants that affect the production of sarcomeric/cytoskeletal proteins. Generally, these variants cause a decrease in tension by the myofilaments, resulting in signaling abnormalities within the micro-environment, which over time result in structural and functional maladaptations, leading to heart failure (HF). Current concepts support the hypothesis that the mutant sarcomere proteins induce a causal depression in the tension-time integral (TTI) of linear preparations of cardiac muscle. However, molecular mechanisms underlying tension generation particularly concerning mutant proteins and their impact on sarcomere molecular signaling are currently controversial. Thus, there is a need for clarification as to how mutant proteins affect sarcomere molecular signaling in the etiology and progression of DCM. A main topic in this controversy is the control of the number of tension-generating myosin heads reacting with the thin filament. One line of investigation proposes that this number is determined by changes in the ratio of myosin heads in a sequestered super-relaxed state (SRX) or in a disordered relaxed state (DRX) poised for force generation upon the Ca2+ activation of the thin filament. Contrasting evidence from nanometer-micrometer-scale X-ray diffraction in intact trabeculae indicates that the SRX/DRX states may have a lesser role. Instead, the proposal is that myosin heads are in a basal OFF state in relaxation then transfer to an ON state through a mechano-sensing mechanism induced during early thin filament activation and increasing thick filament strain. Recent evidence about the modulation of these mechanisms by protein phosphorylation has also introduced a need for reconsidering the control of tension. We discuss these mechanisms that lead to different ideas related to how tension is disturbed by levels of mutant sarcomere proteins linked to the expression of gene variants in the complex landscape of DCM. Resolving the various mechanisms and incorporating them into a unified concept is crucial for gaining a comprehensive understanding of DCM. This deeper understanding is not only important for diagnosis and treatment strategies with small molecules, but also for understanding the reciprocal signaling processes that occur between cardiac myocytes and their micro-environment. By unraveling these complexities, we can pave the way for improved therapeutic interventions for managing DCM.
Collapse
Affiliation(s)
- R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
| | - Paul H. Goldspink
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
| | - Beata M. Wolska
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA; (P.H.G.); (B.M.W.)
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
7
|
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
|
8
|
Prodanovic M, Wang Y, Mijailovich SM, Irving T. Using Multiscale Simulations as a Tool to Interpret Equatorial X-ray Fiber Diffraction Patterns from Skeletal Muscle. Int J Mol Sci 2023; 24:8474. [PMID: 37239821 PMCID: PMC10218096 DOI: 10.3390/ijms24108474] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Synchrotron small-angle X-ray diffraction is the method of choice for nm-scale structural studies of striated muscle under physiological conditions and on millisecond time scales. The lack of generally applicable computational tools for modeling X-ray diffraction patterns from intact muscles has been a significant barrier to exploiting the full potential of this technique. Here, we report a novel "forward problem" approach using the spatially explicit computational simulation platform MUSICO to predict equatorial small-angle X-ray diffraction patterns and the force output simultaneously from resting and isometrically contracting rat skeletal muscle that can be compared to experimental data. The simulation generates families of thick-thin filament repeating units, each with their individually predicted occupancies of different populations of active and inactive myosin heads that can be used to generate 2D-projected electron density models based on known Protein Data Bank structures. We show how, by adjusting only a few selected parameters, we can achieve a good correspondence between experimental and predicted X-ray intensities. The developments presented here demonstrate the feasibility of combining X-ray diffraction and spatially explicit modeling to form a powerful hypothesis-generating tool that can be used to motivate experiments that can reveal emergent properties of muscle.
Collapse
Affiliation(s)
- Momcilo Prodanovic
- Institute for Information Technologies, University of Kragujevac, 34000 Kragujevac, Serbia;
- FilamenTech, Inc., Newton, MA 02458, USA;
| | - Yiwei Wang
- Department of Applied Mathematics, Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
- Department of Mathematics, University of California, Riverside, CA 92521, USA
| | | | - Thomas Irving
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
9
|
Pilagov M, Heling LW, Walklate J, Geeves MA, Kad NM. Single-molecule imaging reveals how mavacamten and PKA modulate ATP turnover in skeletal muscle myofibrils. J Gen Physiol 2022; 155:213694. [PMID: 36394553 PMCID: PMC9674027 DOI: 10.1085/jgp.202213087] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Muscle contraction is controlled at two levels: the thin and the thick filaments. The latter level of control involves three states of myosin heads: active, disordered relaxed (DRX), and super-relaxed (SRX), the distribution of which controls the number of myosins available to interact with actin. How these are controlled is still uncertain. Using fluorescently labeled ATP, we were able to spatially assign the activity of individual myosins within the sarcomere. We observed that SRX comprises 53% of all heads in the C-zone compared with 35% and 44% in the P- and D-zones, respectively. The recently FDA-approved hypertrophic cardiomyopathy drug, mavacamten (mava), significantly decreased DRX, favoring SRX in both the C- and D-zones at 60% and 63%, respectively. Since thick filament regulation is in part regulated by the myosin-binding protein-C (MyBP-C), we also studied PKA phosphorylation. This had the opposite effect as mava, specifically in the C-zone where it decreased SRX to 34%, favoring DRX. These results directly show that excess concentrations of mava do increase SRX, but the effect is limited across the sarcomere, suggesting mava is less effective on skeletal muscle. In addition, we show that PKA directly affects the contractile machinery of skeletal muscle leading to the liberation of repressed heads. Since the effect is focused on the C-zone, this suggests it is likely through MyBP-C phosphorylation, although our data suggest that a further reserve of myosins remain that are not accessible to PKA treatment.
Collapse
Affiliation(s)
- Matvey Pilagov
- School of Biological Sciences, University of Kent, Canterbury, UK
| | | | | | | | - Neil M. Kad
- School of Biological Sciences, University of Kent, Canterbury, UK,Correspondence to Neil M. Kad:
| |
Collapse
|
10
|
Nakanishi T, Oyama K, Tanaka H, Kobirumaki-Shimozawa F, Ishii S, Terui T, Ishiwata S, Fukuda N. Effects of omecamtiv mecarbil on the contractile properties of skinned porcine left atrial and ventricular muscles. Front Physiol 2022; 13:947206. [PMID: 36082222 PMCID: PMC9445838 DOI: 10.3389/fphys.2022.947206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Omecamtiv mecarbil (OM) is a novel inotropic agent for heart failure with systolic dysfunction. OM prolongs the actomyosin attachment duration, which enhances thin filament cooperative activation and accordingly promotes the binding of neighboring myosin to actin. In the present study, we investigated the effects of OM on the steady-state contractile properties in skinned porcine left ventricular (PLV) and atrial (PLA) muscles. OM increased Ca2+ sensitivity in a concentration-dependent manner in PLV, by left shifting the mid-point (pCa50) of the force-pCa curve (ΔpCa50) by ∼0.16 and ∼0.33 pCa units at 0.5 and 1.0 μM, respectively. The Ca2+-sensitizing effect was likewise observed in PLA, but less pronounced with ΔpCa50 values of ∼0.08 and ∼0.22 pCa units at 0.5 and 1.0 μM, respectively. The Ca2+-sensitizing effect of OM (1.0 μM) was attenuated under enhanced thin filament cooperative activation in both PLV and PLA; this attenuation occurred directly via treatment with fast skeletal troponin (ΔpCa50: ∼0.16 and ∼0.10 pCa units in PLV and PLA, respectively) and indirectly by increasing the number of strongly bound cross-bridges in the presence of 3 mM MgADP (ΔpCa50: ∼0.21 and ∼0.08 pCa units in PLV and PLA, respectively). It is likely that this attenuation of the Ca2+-sensitizing effect of OM is due to a decrease in the number of “recruitable” cross-bridges that can potentially produce active force. When cross-bridge detachment was accelerated in the presence of 20 mM inorganic phosphate, the Ca2+-sensitizing effect of OM (1.0 μM) was markedly decreased in both types of preparations (ΔpCa50: ∼0.09 and ∼0.03 pCa units in PLV and PLA, respectively). The present findings suggest that the positive inotropy of OM is more markedly exerted in the ventricle than in the atrium, which results from the strongly bound cross-bridge-dependent allosteric activation of thin filaments.
Collapse
Affiliation(s)
- Tomohiro Nakanishi
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Anesthesiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kotaro Oyama
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Quantum Beam Science Research Directorate, National Institutes for Quantum Science and Technology, Gunma, Japan
| | - Hiroyuki Tanaka
- Laboratory of Marine Biotechnology and Microbiology, Hokkaido University, Hakodate, Japan
| | | | - Shuya Ishii
- Quantum Beam Science Research Directorate, National Institutes for Quantum Science and Technology, Gunma, Japan
| | - Takako Terui
- Department of Anesthesiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shin’ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, Japan
| | - Norio Fukuda
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- *Correspondence: Norio Fukuda,
| |
Collapse
|
11
|
Doh C, Dominic KL, Swanberg CE, Bharambe N, Willard BB, Li L, Ramachandran R, Stelzer JE. Identification of Phosphorylation and Other Post-Translational Modifications in the Central C4C5 Domains of Murine Cardiac Myosin Binding Protein C. ACS OMEGA 2022; 7:14189-14202. [PMID: 35573219 PMCID: PMC9089392 DOI: 10.1021/acsomega.2c00799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/05/2022] [Indexed: 05/06/2023]
Abstract
Cardiac myosin binding protein C (cMyBPC) is a critical multidomain protein that modulates myosin cross bridge behavior and cardiac contractility. cMyBPC is principally regulated by phosphorylation of the residues within the M-domain of its N-terminus. However, not much is known about the phosphorylation or other post-translational modification (PTM) landscape of the central C4C5 domains. In this study, the presence of phosphorylation outside the M-domain was confirmed in vivo using mouse models expressing cMyBPC with nonphosphorylatable serine (S) to alanine substitutions. Purified recombinant mouse C4C5 domain constructs were incubated with 13 different kinases, and samples from the 6 strongest kinases were chosen for mass spectrometry analysis. A total of 26 unique phosphorylated peptides were found, representing 13 different phosphorylation sites including 10 novel sites. Parallel reaction monitoring and subsequent mutagenesis experiments revealed that the S690 site (UniProtKB O70468) was the predominant target of PKA and PKG1. We also report 6 acetylation and 7 ubiquitination sites not previously described in the literature. These PTMs demonstrate the possibility of additional layers of regulation and potential importance of the central domains of cMyBPC in cardiac health and disease. Data are available via ProteomeXchange with identifier PXD031262.
Collapse
Affiliation(s)
- Chang
Yoon Doh
- Department
of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Katherine L. Dominic
- Department
of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Caitlin E. Swanberg
- Department
of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Nikhil Bharambe
- Department
of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Belinda B. Willard
- Proteomics
and Metabolomics Laboratory, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, United States
| | - Ling Li
- Proteomics
and Metabolomics Laboratory, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, United States
| | - Rajesh Ramachandran
- Department
of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Julian E. Stelzer
- Department
of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
12
|
Hou L, Kumar M, Anand P, Chen Y, El-Bizri N, Pickens CJ, Seganish WM, Sadayappan S, Swaminath G. Modulation of myosin by cardiac myosin binding protein-C peptides improves cardiac contractility in ex-vivo experimental heart failure models. Sci Rep 2022; 12:4337. [PMID: 35288601 PMCID: PMC8921245 DOI: 10.1038/s41598-022-08169-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/03/2022] [Indexed: 01/23/2023] Open
Abstract
Cardiac myosin binding protein-C (cMyBP-C) is an important regulator of sarcomeric function. Reduced phosphorylation of cMyBP-C has been linked to compromised contractility in heart failure patients. Here, we used previously published cMyBP-C peptides 302A and 302S, surrogates of the regulatory phosphorylation site serine 302, as a tool to determine the effects of modulating the dephosphorylation state of cMyBP-C on cardiac contraction and relaxation in experimental heart failure (HF) models in vitro. Both peptides increased the contractility of papillary muscle fibers isolated from a mouse model expressing cMyBP-C phospho-ablation (cMyBP-CAAA) constitutively. Peptide 302A, in particular, could also improve the force redevelopment rate (ktr) in papillary muscle fibers from cMyBP-CAAA (nonphosphorylated alanines) mice. Consistent with the above findings, both peptides increased ATPase rates in myofibrils isolated from rats with myocardial infarction (MI), but not from sham rats. Furthermore, in the cMyBP-CAAA mouse model, both peptides improved ATPase hydrolysis rates. These changes were not observed in non-transgenic (NTG) mice or sham rats, indicating the specific effects of these peptides in regulating the dephosphorylation state of cMyBP-C under the pathological conditions of HF. Taken together, these studies demonstrate that modulation of cMyBP-C dephosphorylation state can be a therapeutic approach to improve myosin function, sarcomere contractility and relaxation after an adverse cardiac event. Therefore, targeting cMyBP-C could potentially improve overall cardiac performance as a complement to standard-care drugs in HF patients.
Collapse
Affiliation(s)
- Luqia Hou
- Cardiometabolic Department, Merck & Co., Inc., 213 East Grand Ave., South San Francisco, CA, 94080, USA
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Priti Anand
- Cardiometabolic Department, Merck & Co., Inc., 213 East Grand Ave., South San Francisco, CA, 94080, USA
| | - Yinhong Chen
- Cardiometabolic Department, Merck & Co., Inc., 213 East Grand Ave., South San Francisco, CA, 94080, USA
| | - Nesrine El-Bizri
- Cardiometabolic Department, Merck & Co., Inc., 213 East Grand Ave., South San Francisco, CA, 94080, USA
| | - Chad J Pickens
- Analytical R&D, Merck & Co., Inc., South San Francisco, CA, 94080, USA
| | - W Michael Seganish
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, CA, 94080, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Gayathri Swaminath
- Cardiometabolic Department, Merck & Co., Inc., 213 East Grand Ave., South San Francisco, CA, 94080, USA.
| |
Collapse
|
13
|
Ma W, Irving TC. Small Angle X-ray Diffraction as a Tool for Structural Characterization of Muscle Disease. Int J Mol Sci 2022; 23:3052. [PMID: 35328477 PMCID: PMC8949570 DOI: 10.3390/ijms23063052] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023] Open
Abstract
Small angle X-ray fiber diffraction is the method of choice for obtaining molecular level structural information from striated muscle fibers under hydrated physiological conditions. For many decades this technique had been used primarily for investigating basic biophysical questions regarding muscle contraction and regulation and its use confined to a relatively small group of expert practitioners. Over the last 20 years, however, X-ray diffraction has emerged as an important tool for investigating the structural consequences of cardiac and skeletal myopathies. In this review we show how simple and straightforward measurements, accessible to non-experts, can be used to extract biophysical parameters that can help explain and characterize the physiology and pathology of a given experimental system. We provide a comprehensive guide to the range of the kinds of measurements that can be made and illustrate how they have been used to provide insights into the structural basis of pathology in a comprehensive review of the literature. We also show how these kinds of measurements can inform current controversies and indicate some future directions.
Collapse
Affiliation(s)
- Weikang Ma
- The Biophysics Collaborative Access Team (BioCAT), Center for Synchrotron Radiation Research and Instrumentation (CSSRI), Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Thomas C. Irving
- The Biophysics Collaborative Access Team (BioCAT), Center for Synchrotron Radiation Research and Instrumentation (CSSRI), Illinois Institute of Technology, Chicago, IL 60616, USA;
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA
| |
Collapse
|
14
|
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: 0.3] [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
|
15
|
Waddingham MT, Tsuchimochi H, Sonobe T, Asano R, Jin H, Ow CPC, Schwenke DO, Katare R, Aoyama K, Umetani K, Hoshino M, Uesugi K, Shirai M, Ogo T, Pearson JT. Using Synchrotron Radiation Imaging Techniques to Elucidate the Actions of Hexarelin in the Heart of Small Animal Models. Front Physiol 2022; 12:766818. [PMID: 35126171 PMCID: PMC8814524 DOI: 10.3389/fphys.2021.766818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
The majority of the conventional techniques that are utilized for investigating the pathogenesis of cardiovascular disease in preclinical animal models do not permit microlevel assessment of in situ cardiomyocyte and microvascular functions. Therefore, it has been difficult to establish whether cardiac dysfunction in complex multiorgan disease states, such as heart failure with preserved ejection fraction and pulmonary hypertension, have their origins in microvascular dysfunction or rather in the cardiomyocyte. Herein, we describe our approach of utilizing synchrotron radiation microangiography to, first, ascertain whether the growth hormone secretagogue (GHS) hexarelin is a vasodilator in the coronary circulation of normal and anesthetized Sprague-Dawley rats, and next investigate if hexarelin is able to prevent the pathogenesis of right ventricle (RV) dysfunction in pulmonary hypertension in the sugen chronic hypoxia model rat. We show that acute hexarelin administration evokes coronary microvascular dilation through GHS-receptor 1a and nitric oxide, and through endothelium-derived hyperpolarization. Previous work indicated that chronic exogenous administration of ghrelin largely prevented the pathogenesis of pulmonary hypertension in chronic hypoxia and in monocrotaline models. Unexpectedly, chronic hexarelin administration prior to sugen chronic hypoxia did not prevent RV hypertrophy or RV cardiomyocyte relaxation impairment. Small-angle X-ray scattering revealed that super relaxed myosin filaments contributed to diastolic dysfunction, and that length-dependent activation might contribute to sustained contractility of the RV. Thus, synchrotron-based imaging approaches can reveal novel insights into cardiac and coronary functions in vivo.
Collapse
Affiliation(s)
- Mark T. Waddingham
- Department of Advanced Medical Research for Pulmonary Hypertension, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Takashi Sonobe
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Ryotaro Asano
- Department of Advanced Medical Research for Pulmonary Hypertension, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Huiling Jin
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Connie P. C. Ow
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Daryl O. Schwenke
- Department of Physiology, School of Biomedical Sciences, Heart Otago, University of Otago, Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology, School of Biomedical Sciences, Heart Otago, University of Otago, Dunedin, New Zealand
| | - Kohki Aoyama
- Japan Synchrotron Radiation Research Institute, Harima, Japan
| | - Keiji Umetani
- Japan Synchrotron Radiation Research Institute, Harima, Japan
| | - Masato Hoshino
- Japan Synchrotron Radiation Research Institute, Harima, Japan
| | - Kentaro Uesugi
- Japan Synchrotron Radiation Research Institute, Harima, Japan
| | - Mikiyasu Shirai
- Department of Advanced Medical Research for Pulmonary Hypertension, National Cerebral and Cardiovascular Center, Suita, Japan
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Takeshi Ogo
- Department of Advanced Medical Research for Pulmonary Hypertension, National Cerebral and Cardiovascular Center, Suita, Japan
| | - James T. Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- *Correspondence: James T. Pearson
| |
Collapse
|
16
|
Microscale thermophoresis suggests a new model of regulation of cardiac myosin function via interaction with cardiac myosin-binding protein C. J Biol Chem 2021; 298:101485. [PMID: 34915024 PMCID: PMC8733265 DOI: 10.1016/j.jbc.2021.101485] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/24/2021] [Accepted: 12/03/2021] [Indexed: 12/02/2022] Open
Abstract
The cardiac isoform of myosin-binding protein C (cMyBP-C) is a key regulatory protein found in cardiac myofilaments that can control the activation state of both the actin-containing thin and myosin-containing thick filaments. However, in contrast to thin filament–based mechanisms of regulation, the mechanism of myosin-based regulation by cMyBP-C has yet to be defined in detail. To clarify its function in this process, we used microscale thermophoresis to build an extensive interaction map between cMyBP-C and isolated fragments of β-cardiac myosin. We show here that the regulatory N-terminal domains (C0C2) of cMyBP-C interact with both the myosin head (myosin S1) and tail domains (myosin S2) with micromolar affinity via phosphorylation-independent and phosphorylation-dependent interactions of domain C1 and the cardiac-specific m-motif, respectively. Moreover, we show that the interaction sites with the highest affinity between cMyBP-C and myosin S1 are localized to its central domains, which bind myosin with submicromolar affinity. We identified two separate interaction regions in the central C2C4 and C5C7 segments that compete for the same binding site on myosin S1, suggesting that cMyBP-C can crosslink the two myosin heads of a single myosin molecule and thereby stabilize it in the folded OFF state. Phosphorylation of the cardiac-specific m-motif by protein kinase A had no effect on the binding of either the N-terminal or the central segments to the myosin head domain, suggesting this might therefore represent a constitutively bound state of myosin associated with cMyBP-C. Based on our results, we propose a new model of regulation of cardiac myosin function by cMyBP-C.
Collapse
|
17
|
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
|
18
|
Lertwanakarn T, Suntravat M, Sánchez EE, Wolska BM, Solaro RJ, de Tombe PP, Tachampa K. Negative inotropic mechanisms of β-cardiotoxin in cardiomyocytes by depression of myofilament ATPase activity without activation of the classical β-adrenergic pathway. Sci Rep 2021; 11:21154. [PMID: 34707114 PMCID: PMC8551325 DOI: 10.1038/s41598-021-00282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/06/2021] [Indexed: 11/09/2022] Open
Abstract
Beta-cardiotoxin (β-CTX) from the king cobra venom (Ophiophagus hannah) was previously proposed as a novel β-adrenergic blocker. However, the involvement of β-adrenergic signaling by this compound has never been elucidated. The objectives of this study were to investigate the underlying mechanisms of β-CTX as a β-blocker and its association with the β-adrenergic pathway. The effects of β-CTX on isolated cardiac myocyte functions, calcium homeostasis, the phosphorylation level of targeted proteins, and the myofibrillar ATPase activity were studied. Healthy Sprague Dawley rats were used for cardiomyocytes isolation. Like propranolol, β-CTX attenuated the cardiomyocyte inotropy and calcium transient alterations as induced by isoproterenol stimulation. In contrast, these effects were not observed in forskolin-treated cells. Interestingly, cardiomyocytes treated with β-CTX showed no changes in phosphorylation level at any PKA-targeted sites in the myofilaments as demonstrated in Western blot analysis. The skinned fibers study revealed no change in myofilament kinetics by β-CTX. However, this protein exhibited the direct inhibition of myofibrillar ATPase activity with calcium de-sensitization of the enzyme. In summary, the negative inotropic mechanism of β-CTX was discovered. β-CTX exhibits an atypical β-blocker mechanism. These properties of β-CTX may benefit in developing a novel agent aid to treat hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Tuchakorn Lertwanakarn
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Montamas Suntravat
- National Natural Toxins Research Center, Texas-A&M University-Kingsville, Kingsville, TX, USA.,Department of Chemistry, Texas A&M University-Kingsville, Kingsville, TX, USA
| | - Elda E Sánchez
- National Natural Toxins Research Center, Texas-A&M University-Kingsville, Kingsville, TX, USA.,Department of Chemistry, Texas A&M University-Kingsville, Kingsville, TX, USA
| | - Beata M Wolska
- Department of Physiology and Biophysics, the University of Illinois at Chicago, Chicago, IL, USA.,Department of Medicine, the University of Illinois at Chicago, Chicago, IL, USA
| | - R John Solaro
- Department of Physiology and Biophysics, the University of Illinois at Chicago, Chicago, IL, USA
| | - Pieter P de Tombe
- Department of Physiology and Biophysics, the University of Illinois at Chicago, Chicago, IL, USA.,Phymedexp, Université de Montpellier, Inserm, CNRS, Montpellier, France
| | - Kittipong Tachampa
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
19
|
Lynch TL, Kumar M, McNamara JW, Kuster DWD, Sivaguru M, Singh RR, Previs MJ, Lee KH, Kuffel G, Zilliox MJ, Lin BL, Ma W, Gibson AM, Blaxall BC, Nieman ML, Lorenz JN, Leichter DM, Leary OP, Janssen PML, de Tombe PP, Gilbert RJ, Craig R, Irving T, Warshaw DM, Sadayappan S. Amino terminus of cardiac myosin binding protein-C regulates cardiac contractility. J Mol Cell Cardiol 2021; 156:33-44. [PMID: 33781820 PMCID: PMC8217138 DOI: 10.1016/j.yjmcc.2021.03.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022]
Abstract
Phosphorylation of cardiac myosin binding protein-C (cMyBP-C) regulates cardiac contraction through modulation of actomyosin interactions mediated by the protein's amino terminal (N')-region (C0-C2 domains, 358 amino acids). On the other hand, dephosphorylation of cMyBP-C during myocardial injury results in cleavage of the 271 amino acid C0-C1f region and subsequent contractile dysfunction. Yet, our current understanding of amino terminus region of cMyBP-C in the context of regulating thin and thick filament interactions is limited. A novel cardiac-specific transgenic mouse model expressing cMyBP-C, but lacking its C0-C1f region (cMyBP-C∆C0-C1f), displayed dilated cardiomyopathy, underscoring the importance of the N'-region in cMyBP-C. Further exploring the molecular basis for this cardiomyopathy, in vitro studies revealed increased interfilament lattice spacing and rate of tension redevelopment, as well as faster actin-filament sliding velocity within the C-zone of the transgenic sarcomere. Moreover, phosphorylation of the unablated phosphoregulatory sites was increased, likely contributing to normal sarcomere morphology and myoarchitecture. These results led us to hypothesize that restoration of the N'-region of cMyBP-C would return actomyosin interaction to its steady state. Accordingly, we administered recombinant C0-C2 (rC0-C2) to permeabilized cardiomyocytes from transgenic, cMyBP-C null, and human heart failure biopsies, and we found that normal regulation of actomyosin interaction and contractility was restored. Overall, these data provide a unique picture of selective perturbations of the cardiac sarcomere that either lead to injury or adaptation to injury in the myocardium.
Collapse
Affiliation(s)
- Thomas L Lynch
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Mohit Kumar
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL 60153, USA; Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - James W McNamara
- Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Diederik W D Kuster
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL 60153, USA; Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Mayandi Sivaguru
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rohit R Singh
- Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Michael J Previs
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT 05405, USA
| | - Kyoung Hwan Lee
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Gina Kuffel
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL 60153, USA
| | - Michael J Zilliox
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL 60153, USA
| | - Brian Leei Lin
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Weikang Ma
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Aaron M Gibson
- Department of Pediatrics, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Burns C Blaxall
- Department of Pediatrics, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Michelle L Nieman
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - John N Lorenz
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Dana M Leichter
- Research Service, Providence VA Medical Center, Providence, RI 02908, USA
| | - Owen P Leary
- Research Service, Providence VA Medical Center, Providence, RI 02908, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Pieter P de Tombe
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL 60153, USA; Department of Physiology, University of Illinois at Chicago, Chicago 60612, USA; Phymedexp, Université de Montpellier, Inserm, CNRS, Montpellier, France
| | - Richard J Gilbert
- Research Service, Providence VA Medical Center, Providence, RI 02908, USA
| | - Roger Craig
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Thomas Irving
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - David M Warshaw
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, University of Vermont, Burlington, VT 05405, USA
| | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL 60153, USA; Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
20
|
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: 0.5] [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
|
21
|
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: 1.0] [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
|
22
|
Giles J, Fitzsimons DP, Patel JR, Knudtsen C, Neuville Z, Moss RL. cMyBP-C phosphorylation modulates the time-dependent slowing of unloaded shortening in murine skinned myocardium. J Gen Physiol 2021; 153:e202012782. [PMID: 33566084 PMCID: PMC7879488 DOI: 10.1085/jgp.202012782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/14/2021] [Indexed: 11/20/2022] Open
Abstract
In myocardium, phosphorylation of cardiac myosin-binding protein-C (cMyBP-C) is thought to modulate the cooperative activation of the thin filament by binding to myosin and/or actin, thereby regulating the probability of cross-bridge binding to actin. At low levels of Ca2+ activation, unloaded shortening velocity (Vo) in permeabilized cardiac muscle is comprised of an initial high-velocity phase and a subsequent low-velocity phase. The velocities in these phases scale with the level of activation, culminating in a single high-velocity phase (Vmax) at saturating Ca2+. To test the idea that cMyBP-C phosphorylation contributes to the activation dependence of Vo, we measured Vo before and following treatment with protein kinase A (PKA) in skinned trabecula isolated from mice expressing either wild-type cMyBP-C (tWT), nonphosphorylatable cMyBP-C (t3SA), or phosphomimetic cMyBP-C (t3SD). During maximal Ca2+ activation, Vmax was monophasic and not significantly different between the three groups. Although biphasic shortening was observed in all three groups at half-maximal activation under control conditions, the high- and low-velocity phases were faster in the t3SD myocardium compared with values obtained in either tWT or t3SA myocardium. Treatment with PKA significantly accelerated both the high- and low-velocity phases in tWT myocardium but had no effect on Vo in either the t3SD or t3SA myocardium. These results can be explained in terms of a model in which the level of cMyBP-C phosphorylation modulates the extent and rate of cooperative spread of myosin binding to actin.
Collapse
Affiliation(s)
- Jasmine Giles
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, and the University of Wisconsin Cardiovascular Research Center, Madison, WI
| | - Daniel P. Fitzsimons
- Department of Animal, Veterinary and Food Sciences, College of Agricultural and Life Sciences, University of Idaho, Moscow, ID
| | - Jitandrakumar R. Patel
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, and the University of Wisconsin Cardiovascular Research Center, Madison, WI
| | - Chloe Knudtsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, and the University of Wisconsin Cardiovascular Research Center, Madison, WI
| | - Zander Neuville
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, and the University of Wisconsin Cardiovascular Research Center, Madison, WI
| | - Richard L. Moss
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, and the University of Wisconsin Cardiovascular Research Center, Madison, WI
| |
Collapse
|
23
|
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: 29] [Impact Index Per Article: 7.3] [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
|
24
|
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: 56] [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
|
25
|
Abstract
Heart failure (HF) is a common consequence of several cardiovascular diseases and is understood as a vicious cycle of cardiac and hemodynamic decline. The current inventory of treatments either alleviates the pathophysiological features (eg, cardiac dysfunction, neurohumoral activation, and ventricular remodeling) and/or targets any underlying pathologies (eg, hypertension and myocardial infarction). Yet, since these do not provide a cure, the morbidity and mortality associated with HF remains high. Therefore, the disease constitutes an unmet medical need, and novel therapies are desperately needed. Cyclic guanosine-3',5'-monophosphate (cGMP), synthesized by nitric oxide (NO)- and natriuretic peptide (NP)-responsive guanylyl cyclase (GC) enzymes, exerts numerous protective effects on cardiac contractility, hypertrophy, fibrosis, and apoptosis. Impaired cGMP signaling, which can occur after GC deactivation and the upregulation of cyclic nucleotide-hydrolyzing phosphodiesterases (PDEs), promotes cardiac dysfunction. In this study, we review the role that NO/cGMP and NP/cGMP signaling plays in HF. After considering disease etiology, the physiological effects of cGMP in the heart are discussed. We then assess the evidence from preclinical models and patients that compromised cGMP signaling contributes to the HF phenotype. Finally, the potential of pharmacologically harnessing cardioprotective cGMP to rectify the present paucity of effective HF treatments is examined.
Collapse
|
26
|
Mullins PD, Bondarenko VE. Mathematical model for β1-adrenergic regulation of the mouse ventricular myocyte contraction. Am J Physiol Heart Circ Physiol 2020; 318:H264-H282. [DOI: 10.1152/ajpheart.00492.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The β1-adrenergic regulation of cardiac myocyte contraction plays an important role in regulating heart function. Activation of this system leads to an increased heart rate and stronger myocyte contraction. However, chronic stimulation of the β1-adrenergic signaling system can lead to cardiac hypertrophy and heart failure. To understand the mechanisms of action of β1-adrenoceptors, a mathematical model of cardiac myocyte contraction that includes the β1-adrenergic system was developed and studied. The model was able to simulate major experimental protocols for measurements of steady-state force-calcium relationships, cross-bridge release rate and force development rate, force-velocity relationship, and force redevelopment rate. It also reproduced quite well frequency and isoproterenol dependencies for intracellular Ca2+ concentration ([Ca2+]i) transients, total contraction force, and sarcomere shortening. The mathematical model suggested the mechanisms of increased contraction force and myocyte shortening on stimulation of β1-adrenergic receptors is due to phosphorylation of troponin I and myosin-binding protein C and increased [Ca2+]i transient resulting from activation of the β1-adrenergic signaling system. The model was used to simulate work-loop contractions and estimate the power during the cardiac cycle as well as the effects of 4-aminopyridine and tedisamil on the myocyte contraction. The developed mathematical model can be used further for simulations of contraction of ventricular myocytes from genetically modified mice and myocytes from mice with chronic cardiac diseases. NEW & NOTEWORTHY A new mathematical model of mouse ventricular myocyte contraction that includes the β1-adrenergic system was developed. The model simulated major experimental protocols for myocyte contraction and predicted the effects of 4-aminopyridine and tedisamil on the myocyte contraction. The model also allowed for simulations of work-loop contractions and estimation of the power during the cardiac cycle.
Collapse
Affiliation(s)
- Paula D. Mullins
- Department of Mathematics, University of North Georgia, Blue Ridge, Georgia
- Department of Mathematics and Statistics and Neuroscience Institute, Georgia State University, Atlanta, Georgia
| | - Vladimir E. Bondarenko
- Department of Mathematics and Statistics and Neuroscience Institute, Georgia State University, Atlanta, Georgia
| |
Collapse
|
27
|
Waddingham MT, Sonobe T, Tsuchimochi H, Edgley AJ, Sukumaran V, Chen YC, Hansra SS, Schwenke DO, Umetani K, Aoyama K, Yagi N, Kelly DJ, Gaderi S, Herwig M, Kolijn D, Mügge A, Paulus WJ, Ogo T, Shirai M, Hamdani N, Pearson JT. Diastolic dysfunction is initiated by cardiomyocyte impairment ahead of endothelial dysfunction due to increased oxidative stress and inflammation in an experimental prediabetes model. J Mol Cell Cardiol 2019; 137:119-131. [DOI: 10.1016/j.yjmcc.2019.10.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/10/2019] [Accepted: 10/18/2019] [Indexed: 12/19/2022]
|
28
|
Site-specific phosphorylation of myosin binding protein-C coordinates thin and thick filament activation in cardiac muscle. Proc Natl Acad Sci U S A 2019; 116:15485-15494. [PMID: 31308242 PMCID: PMC6681757 DOI: 10.1073/pnas.1903033116] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Phosphorylation of cardiac myosin binding protein-C (cMyBP-C) is a key regulator of myocardial contractility, and dephosphorylation of cMyBP-C is associated with heart failure. However, the molecular mechanisms underlying contractile regulation by cMyBP-C phosphorylation are poorly understood. We describe the kinase specificity of the multiple phosphorylation sites on cMyBP-C and show that they are interdependent and have distinct effects on the structure of the thin and thick filaments. The results lead to a model of regulation by cMyBP-C phosphorylation through altered affinity of cMyBP-C’s N terminus for thin and thick filaments, as well as their structures and associated regulatory states. Impairment of these mechanisms is likely to underlie the functional effects of mutations in filament proteins associated with cardiomyopathy. The heart’s response to varying demands of the body is regulated by signaling pathways that activate protein kinases which phosphorylate sarcomeric proteins. Although phosphorylation of cardiac myosin binding protein-C (cMyBP-C) has been recognized as a key regulator of myocardial contractility, little is known about its mechanism of action. Here, we used protein kinase A (PKA) and Cε (PKCε), as well as ribosomal S6 kinase II (RSK2), which have different specificities for cMyBP-C’s multiple phosphorylation sites, to show that individual sites are not independent, and that phosphorylation of cMyBP-C is controlled by positive and negative regulatory coupling between those sites. PKA phosphorylation of cMyBP-C’s N terminus on 3 conserved serine residues is hierarchical and antagonizes phosphorylation by PKCε, and vice versa. In contrast, RSK2 phosphorylation of cMyBP-C accelerates PKA phosphorylation. We used cMyBP-C’s regulatory N-terminal domains in defined phosphorylation states for protein–protein interaction studies with isolated cardiac native thin filaments and the S2 domain of cardiac myosin to show that site-specific phosphorylation of this region of cMyBP-C controls its interaction with both the actin-containing thin and myosin-containing thick filaments. We also used fluorescence probes on the myosin-associated regulatory light chain in the thick filaments and on troponin C in the thin filaments to monitor structural changes in the myofilaments of intact heart muscle cells associated with activation of myocardial contraction by the N-terminal region of cMyBP-C in its different phosphorylation states. Our results suggest that cMyBP-C acts as a sarcomeric integrator of multiple signaling pathways that determines downstream physiological function.
Collapse
|
29
|
Ding Y, Lyons SA, Scott GR, Gillis TE. Characterizing the influence of chronic hypobaric hypoxia on diaphragmatic myofilament contractile function and phosphorylation in high-altitude deer mice and low-altitude white-footed mice. J Comp Physiol B 2019; 189:489-499. [PMID: 31278612 DOI: 10.1007/s00360-019-01224-w] [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: 02/09/2019] [Revised: 06/06/2019] [Accepted: 06/24/2019] [Indexed: 11/27/2022]
Abstract
Deer mice, Peromyscusmaniculatus, live at high altitudes where limited O2 represents a challenge to maintaining oxygen delivery to tissues. Previous work has demonstrated that hypoxia acclimation of deer mice and low altitude white-footed mice (P. leucopus) increases the force generating capacity of the diaphragm. The mechanism behind this improved contractile function is not known. Within myocytes, the myofilament plays a critical role in setting the rate and level of force production, and its ability to generate force can change in response to changes in physiological conditions. In the current study, we examined how chronic hypobaric hypoxia exposure of deer mice and white-footed mice influences the Ca2+ activation of force generation by skinned diaphragmatic myofilaments, and the phosphorylation of myofilament proteins. Results demonstrate that myofilament force production, and the Ca2+ sensitivity of force generation, were not impacted by acclimation to hypobaric hypoxia, and did not differ between preparations from the two species. The cooperativity of the force-pCa relationship, and the maximal rate of force generation were also the same in the preparations from both species, and not impacted by acclimation. Finally, the relative phosphorylation of TnT, and MLC was lower in deer mice than white-footed mice, but was not affected by acclimation. These results indicate that species differences in diaphragm function, and the increase in force production with hypoxia acclimation, are not due to differences, or changes, in myofilament function. However, it appears that diaphragmatic myofilament function in these species is not affected by chronic hypobaric hypoxia exposure.
Collapse
Affiliation(s)
- Y Ding
- Department of Integrative Biology, University of Guelph, Guelph, ON, N1G-2W1, Canada
| | - S A Lyons
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - G R Scott
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Todd E Gillis
- Department of Integrative Biology, University of Guelph, Guelph, ON, N1G-2W1, Canada.
| |
Collapse
|
30
|
Powers JD, Yuan CC, McCabe KJ, Murray JD, Childers MC, Flint GV, Moussavi-Harami F, Mohran S, Castillo R, Zuzek C, Ma W, Daggett V, McCulloch AD, Irving TC, Regnier M. Cardiac myosin activation with 2-deoxy-ATP via increased electrostatic interactions with actin. Proc Natl Acad Sci U S A 2019; 116:11502-11507. [PMID: 31110001 PMCID: PMC6561254 DOI: 10.1073/pnas.1905028116] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The naturally occurring nucleotide 2-deoxy-adenosine 5'-triphosphate (dATP) can be used by cardiac muscle as an alternative energy substrate for myosin chemomechanical activity. We and others have previously shown that dATP increases contractile force in normal hearts and models of depressed systolic function, but the structural basis of these effects has remained unresolved. In this work, we combine multiple techniques to provide structural and functional information at the angstrom-nanometer and millisecond time scales, demonstrating the ability to make both structural measurements and quantitative kinetic estimates of weak actin-myosin interactions that underpin sarcomere dynamics. Exploiting dATP as a molecular probe, we assess how small changes in myosin structure translate to electrostatic-based changes in sarcomere function to augment contractility in cardiac muscle. Through Brownian dynamics simulation and computational structural analysis, we found that deoxy-hydrolysis products [2-deoxy-adenosine 5'-diphosphate (dADP) and inorganic phosphate (Pi)] bound to prepowerstroke myosin induce an allosteric restructuring of the actin-binding surface on myosin to increase the rate of cross-bridge formation. We then show experimentally that this predicted effect translates into increased electrostatic interactions between actin and cardiac myosin in vitro. Finally, using small-angle X-ray diffraction analysis of sarcomere structure, we demonstrate that the proposed increased electrostatic affinity of myosin for actin causes a disruption of the resting conformation of myosin motors, resulting in their repositioning toward the thin filament before activation. The dATP-mediated structural alterations in myosin reported here may provide insight into an improved criterion for the design or selection of small molecules to be developed as therapeutic agents to treat systolic dysfunction.
Collapse
Affiliation(s)
- Joseph D Powers
- Department of Bioengineering, University of Washington, Seattle, WA 98109;
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Chen-Ching Yuan
- Department of Bioengineering, University of Washington, Seattle, WA 98109
| | - Kimberly J McCabe
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Jason D Murray
- Department of Bioengineering, University of Washington, Seattle, WA 98109
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98109
| | | | - Galina V Flint
- Department of Bioengineering, University of Washington, Seattle, WA 98109
| | - Farid Moussavi-Harami
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Saffie Mohran
- Department of Bioengineering, University of Washington, Seattle, WA 98109
| | - Romi Castillo
- Department of Bioengineering, University of Washington, Seattle, WA 98109
| | - Carla Zuzek
- Department of Bioengineering, University of Washington, Seattle, WA 98109
| | - Weikang Ma
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616
| | - Valerie Daggett
- Department of Bioengineering, University of Washington, Seattle, WA 98109
| | - Andrew D McCulloch
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Thomas C Irving
- Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98109;
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98109
| |
Collapse
|
31
|
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: 65] [Impact Index Per Article: 10.8] [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
|
32
|
Fenwick AJ, Awinda PO, Yarbrough-Jones JA, Eldridge JA, Rodgers BD, Tanner BCW. Demembranated skeletal and cardiac fibers produce less force with altered cross-bridge kinetics in a mouse model for limb-girdle muscular dystrophy 2i. Am J Physiol Cell Physiol 2019; 317:C226-C234. [PMID: 31091146 DOI: 10.1152/ajpcell.00524.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Limb-girdle muscular dystrophy 2i (LGMD2i) is a dystroglycanopathy that compromises myofiber integrity and primarily reduces power output in limb muscles but can influence cardiac muscle as well. Previous studies of LGMD2i made use of a transgenic mouse model in which a proline-to-leucine (P448L) mutation in fukutin-related protein severely reduces glycosylation of α-dystroglycan. Muscle function is compromised in P448L mice in a manner similar to human patients with LGMD2i. In situ studies reported lower maximal twitch force and depressed force-velocity curves in medial gastrocnemius (MG) muscles from male P448L mice. Here, we measured Ca2+-activated force generation and cross-bridge kinetics in both demembranated MG fibers and papillary muscle strips from P448L mice. Maximal activated tension was 37% lower in MG fibers and 18% lower in papillary strips from P448L mice than controls. We also found slightly faster rates of cross-bridge recruitment and detachment in MG fibers from P448L than control mice. These increases in skeletal cross-bridge cycling could reduce the unitary force output from individual cross bridges by lowering the ratio of time spent in a force-bearing state to total cycle time. This suggests that the decreased force production in LGMD2i may be due (at least in part) to altered cross-bridge kinetics. This finding is notable, as the majority of studies germane to muscular dystrophies have focused on sarcolemma or whole muscle properties, whereas our findings suggest that the disease pathology is also influenced by potential downstream effects on cross-bridge behavior.
Collapse
Affiliation(s)
- Axel J Fenwick
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington.,Washington Center for Muscle Biology, Washington State University , Pullman, Washington
| | - Peter O Awinda
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington.,Washington Center for Muscle Biology, Washington State University , Pullman, Washington
| | - Jacob A Yarbrough-Jones
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington.,Washington Center for Muscle Biology, Washington State University , Pullman, Washington
| | - Jennifer A Eldridge
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington.,Washington Center for Muscle Biology, Washington State University , Pullman, Washington
| | - Buel D Rodgers
- Washington Center for Muscle Biology, Washington State University , Pullman, Washington.,AAVogen, Inc. , Rockville, Maryland
| | - Bertrand C W Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington.,Washington Center for Muscle Biology, Washington State University , Pullman, Washington
| |
Collapse
|
33
|
Abstract
Colson discusses a recent investigation of the functional effect of slow myosin binding protein-C in slow-twitch skeletal muscle fibers.
Collapse
Affiliation(s)
- Brett A Colson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
34
|
Robinett JC, Hanft LM, Geist J, Kontrogianni-Konstantopoulos A, McDonald KS. Regulation of myofilament force and loaded shortening by skeletal myosin binding protein C. J Gen Physiol 2019; 151:645-659. [PMID: 30705121 PMCID: PMC6504288 DOI: 10.1085/jgp.201812200] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 01/11/2019] [Indexed: 12/28/2022] Open
Abstract
Myosin binding protein C (MyBP-C) is thought to regulate the contraction of skeletal muscle. Robinett et al. show that phosphorylation of slow skeletal MyBP-C modulates contraction by recruiting cross-bridges, modifying cross-bridge kinetics, and altering internal drag forces in the C-zone. Myosin binding protein C (MyBP-C) is a 125–140-kD protein located in the C-zone of each half-thick filament. It is thought to be an important regulator of contraction, but its precise role is unclear. Here we investigate mechanisms by which skeletal MyBP-C regulates myofilament function using rat permeabilized skeletal muscle fibers. We mount either slow-twitch or fast-twitch skeletal muscle fibers between a force transducer and motor, use Ca2+ to activate a range of forces, and measure contractile properties including transient force overshoot, rate of force development, and loaded sarcomere shortening. The transient force overshoot is greater in slow-twitch than fast-twitch fibers at all Ca2+ activation levels. In slow-twitch fibers, protein kinase A (PKA) treatment (a) augments phosphorylation of slow skeletal MyBP-C (sMyBP-C), (b) doubles the magnitude of the relative transient force overshoot at low Ca2+ activation levels, and (c) increases force development rates at all Ca2+ activation levels. We also investigate the role that phosphorylated and dephosphorylated sMyBP-C plays in loaded sarcomere shortening. We test the hypothesis that MyBP-C acts as a brake to filament sliding within the myofilament lattice by measuring sarcomere shortening as thin filaments traverse into the C-zone during lightly loaded slow-twitch fiber contractions. Before PKA treatment, shortening velocity decelerates as sarcomeres traverse from ∼3.10 to ∼3.00 µm. After PKA treatment, sarcomeres shorten a greater distance and exhibit less deceleration during similar force clamps. After sMyBP-C dephosphorylation, sarcomere length traces display a brief recoil (i.e., “bump”) that initiates at ∼3.06 µm during loaded shortening. Interestingly, the timing of the bump shifts with changes in load but manifests at the same sarcomere length. Our results suggest that sMyBP-C and its phosphorylation state regulate sarcomere contraction by a combination of cross-bridge recruitment, modification of cross-bridge cycling kinetics, and alteration of drag forces that originate in the C-zone.
Collapse
Affiliation(s)
- Joel C Robinett
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO
| | - Laurin M Hanft
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD
| | | | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO
| |
Collapse
|
35
|
Li J, Gresham KS, Mamidi R, Doh CY, Wan X, Deschenes I, Stelzer JE. Sarcomere-based genetic enhancement of systolic cardiac function in a murine model of dilated cardiomyopathy. Int J Cardiol 2018; 273:168-176. [PMID: 30279005 DOI: 10.1016/j.ijcard.2018.09.073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/27/2018] [Accepted: 09/20/2018] [Indexed: 01/28/2023]
Abstract
Diminished cardiac contractile function is a characteristic feature of dilated cardiomyopathy (DCM) and many other heart failure (HF) causing etiologies. We tested the hypothesis that targeting the sarcomere to increase cardiac contractility can effectively prevent the DCM phenotype in muscle-LIM protein knockout (MLP-/-) mice. The ablation of cardiac myosin binding protein C (MYBPC3-/-) protected the MLP-/- mice from developing the DCM phenotype. We examined the in vivo cardiac function and morphology of the resultant mouse model lacking both MLP and MYBPC3 (DKO) by echocardiography and pressure-volume catheterization and found a significant reduction in hypertrophy, as evidenced by normalized wall thickness and chamber dimensions, and improved systolic function, as evidenced by enhanced ejection fraction (~26% increase compared MLP-/- mice) and rate of pressure development (DKO 7851.0 ± 504.8 vs. MLP-/- 4496.4 ± 196.8 mmHg/s). To investigate the molecular basis for the improved DKO phenotype we performed mechanical experiments in skinned myocardium isolated from WT and the individual KO mice. Skinned myocardium isolated from DKO mice displayed increased Ca2+ sensitivity of force generation, and significantly accelerated rate of cross-bridge detachment (+63% compared to MLP-/-) and rate of XB recruitment (+58% compared to MLP-/-) at submaximal Ca2+ activations. The in vivo and in vitro functional enhancement of DKO mice demonstrates that enhancing the sarcomeric contractility can be cardioprotective in HF characterized by reduced cardiac output, such as in cases of DCM.
Collapse
Affiliation(s)
- Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Kenneth S Gresham
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States of America
| | - Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Chang Yoon Doh
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Xiaoping Wan
- The Heart and Vascular Research Center, Metro Health, Cleveland, OH, United States of America
| | - Isabelle Deschenes
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America; The Heart and Vascular Research Center, Metro Health, Cleveland, OH, United States of America
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States of America.
| |
Collapse
|
36
|
Cai W, Hite ZL, Lyu B, Wu Z, Lin Z, Gregorich ZR, Messer AE, McIlwain SJ, Marston SB, Kohmoto T, Ge Y. Temperature-sensitive sarcomeric protein post-translational modifications revealed by top-down proteomics. J Mol Cell Cardiol 2018; 122:11-22. [PMID: 30048711 DOI: 10.1016/j.yjmcc.2018.07.247] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/11/2018] [Accepted: 07/21/2018] [Indexed: 10/28/2022]
Abstract
Despite advancements in symptom management for heart failure (HF), this devastating clinical syndrome remains the leading cause of death in the developed world. Studies using animal models have greatly advanced our understanding of the molecular mechanisms underlying HF; however, differences in cardiac physiology and the manifestation of HF between animals, particularly rodents, and humans necessitates the direct interrogation of human heart tissue samples. Nevertheless, an ever-present concern when examining human heart tissue samples is the potential for artefactual changes related to temperature changes during tissue shipment or sample processing. Herein, we examined the effects of temperature on the post-translational modifications (PTMs) of sarcomeric proteins, the proteins responsible for muscle contraction, under conditions mimicking those that might occur during tissue shipment or sample processing. Using a powerful top-down proteomics method, we found that sarcomeric protein PTMs were differentially affected by temperature. Specifically, cardiac troponin I and enigma homolog isoform 2 showed robust increases in phosphorylation when tissue was incubated at either 4 °C or 22 °C. The observed increase is likely due to increased cyclic AMP levels and activation of protein kinase A in the tissue. On the contrary, cardiac troponin T and myosin regulatory light chain phosphorylation decreased when tissue was incubated at 4 °C or 22 °C. Furthermore, significant protein degradation was also observed after incubation at 4 °C or 22 °C. Overall, these results indicate that temperature exerts various effects on sarcomeric protein PTMs and careful tissue handling is critical for studies involving human heart samples. Moreover, these findings highlight the power of top-down proteomics for examining the integrity of cardiac tissue samples.
Collapse
Affiliation(s)
- Wenxuan Cai
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary L Hite
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Beini Lyu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zhijie Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ziqing Lin
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachery R Gregorich
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Andrew E Messer
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sean J McIlwain
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Steve B Marston
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Takushi Kohmoto
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ying Ge
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
37
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
38
|
Ait Mou Y, Lacampagne A, Irving T, Scheuermann V, Blot S, Ghaleh B, de Tombe PP, Cazorla O. Altered myofilament structure and function in dogs with Duchenne muscular dystrophy cardiomyopathy. J Mol Cell Cardiol 2017; 114:345-353. [PMID: 29275006 DOI: 10.1016/j.yjmcc.2017.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 11/16/2022]
Abstract
AIM Duchenne Muscular Dystrophy (DMD) is associated with progressive depressed left ventricular (LV) function. However, DMD effects on myofilament structure and function are poorly understood. Golden Retriever Muscular Dystrophy (GRMD) is a dog model of DMD recapitulating the human form of DMD. OBJECTIVE The objective of this study is to evaluate myofilament structure and function alterations in GRMD model with spontaneous cardiac failure. METHODS AND RESULTS We have employed synchrotron X-rays diffraction to evaluate myofilament lattice spacing at various sarcomere lengths (SL) on permeabilized LV myocardium. We found a negative correlation between SL and lattice spacing in both sub-epicardium (EPI) and sub-endocardium (ENDO) LV layers in control dog hearts. In the ENDO of GRMD hearts this correlation is steeper due to higher lattice spacing at short SL (1.9μm). Furthermore, cross-bridge cycling indexed by the kinetics of tension redevelopment (ktr) was faster in ENDO GRMD myofilaments at short SL. We measured post-translational modifications of key regulatory contractile proteins. S-glutathionylation of cardiac Myosin Binding Protein-C (cMyBP-C) was unchanged and PKA dependent phosphorylation of the cMyBP-C was significantly reduced in GRMD ENDO tissue and more modestly in EPI tissue. CONCLUSIONS We found a gradient of contractility in control dogs' myocardium that spreads across the LV wall, negatively correlated with myofilament lattice spacing. Chronic stress induced by dystrophin deficiency leads to heart failure that is tightly associated with regional structural changes indexed by increased myofilament lattice spacing, reduced phosphorylation of regulatory proteins and altered myofilament contractile properties in GRMD dogs.
Collapse
Affiliation(s)
- Younss Ait Mou
- College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar; Department of Cell and Molecular Physiology, Heath Science Division, Loyola University Chicago, Maywood, IL 60153, USA
| | - Alain Lacampagne
- INSERM U1046, CNRS UMR 9214, Université de Montpellier, Physiologie et Médecine Expérimentale du cœur et des muscles - PHYMEDEXP, CHU Arnaud de Villeneuve, 34295 Montpellier cedex 05, France
| | - Thomas Irving
- Department of Cell and Molecular Physiology, Heath Science Division, Loyola University Chicago, Maywood, IL 60153, USA; Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Valérie Scheuermann
- INSERM U1046, CNRS UMR 9214, Université de Montpellier, Physiologie et Médecine Expérimentale du cœur et des muscles - PHYMEDEXP, CHU Arnaud de Villeneuve, 34295 Montpellier cedex 05, France
| | - Stéphane Blot
- Inserm U955-E10, IMRB, Université Paris Est, Ecole nationale vétérinaire d'Alfort, Maisons-Alfort 94700, France
| | | | - Pieter P de Tombe
- Department of Cell and Molecular Physiology, Heath Science Division, Loyola University Chicago, Maywood, IL 60153, USA
| | - Olivier Cazorla
- INSERM U1046, CNRS UMR 9214, Université de Montpellier, Physiologie et Médecine Expérimentale du cœur et des muscles - PHYMEDEXP, CHU Arnaud de Villeneuve, 34295 Montpellier cedex 05, France..
| |
Collapse
|
39
|
Singh RR, Dunn JW, Qadan MM, Hall N, Wang KK, Root DD. Whole length myosin binding protein C stabilizes myosin S2 as measured by gravitational force spectroscopy. Arch Biochem Biophys 2017; 638:41-51. [PMID: 29229286 DOI: 10.1016/j.abb.2017.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 01/21/2023]
Abstract
The mechanical stability of the myosin subfragment-2 (S2) was tested with simulated force spectroscopy (SFS) and gravitational force spectroscopy (GFS). Experiments examined unzipping S2, since it required less force than stretching parallel to the coiled coil. Both GFS and SFS demonstrated that the force required to destabilize the light meromyosin (LMM) was greater than the force required to destabilize the coiled coil at each of three different locations along S2. GFS data also conveyed that the mechanical stability of the S2 region is independent from its association with the myosin thick filament using cofilaments of myosin tail and a single intact myosin. The C-terminal end of myosin binding protein C (MyBPC) binds to LMM and the N-terminal end can bind either S2 or actin. The force required to destabilize the myosin coiled coil molecule was 3 times greater in the presence of MyBPC than in its absence. Furthermore, the in vitro motility assay with full length slow skeletal MyBPC slowed down the actin filament sliding over myosin thick filaments. This study demonstrates that skeletal MyBPC both enhanced the mechanical stability of the S2 coiled coil and reduced the sliding velocity of actin filaments over polymerized myosin filaments.
Collapse
Affiliation(s)
- Rohit R Singh
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX 76203, USA
| | - James W Dunn
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX 76203, USA
| | - Motamed M Qadan
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX 76203, USA
| | - Nakiuda Hall
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX 76203, USA
| | - Kathy K Wang
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX 76203, USA
| | - Douglas D Root
- Department of Biological Sciences, Division of Biochemistry and Molecular Biology, University of North Texas, Denton, TX 76203, USA.
| |
Collapse
|
40
|
MYBPC3 mutations are associated with a reduced super-relaxed state in patients with hypertrophic cardiomyopathy. PLoS One 2017; 12:e0180064. [PMID: 28658286 PMCID: PMC5489194 DOI: 10.1371/journal.pone.0180064] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 06/08/2017] [Indexed: 11/23/2022] Open
Abstract
The “super-relaxed state” (SRX) of myosin represents a ‘reserve’ of motors in the heart. Myosin heads in the SRX are bound to the thick filament and have a very low ATPase rate. Changes in the SRX are likely to modulate cardiac contractility. We previously demonstrated that the SRX is significantly reduced in mouse cardiomyocytes lacking cardiac myosin binding protein–C (cMyBP-C). Here, we report the effect of mutations in the cMyBP-C gene (MYBPC3) using samples from human patients with hypertrophic cardiomyopathy (HCM). Left ventricular (LV) samples from 11 HCM patients were obtained following myectomy surgery to relieve LV outflow tract obstruction. HCM samples were genotyped as either MYBPC3 mutation positive (MYBPC3mut) or negative (HCMsmn) and were compared to eight non-failing donor hearts. Compared to donors, only MYBPC3mut samples display a significantly diminished SRX, characterised by a decrease in both the number of myosin heads in the SRX and the lifetime of ATP turnover. These changes were not observed in HCMsmn samples. There was a positive correlation (p < 0.01) between the expression of cMyBP-C and the proportion of myosin heads in the SRX state, suggesting cMyBP-C modulates and maintains the SRX. Phosphorylation of the myosin regulatory light chain in MYBPC3mut samples was significantly decreased compared to the other groups, suggesting a potential mechanism to compensate for the diminished SRX. We conclude that by altering both contractility and sarcomeric energy requirements, a reduced SRX may be an important disease mechanism in patients with MYBPC3 mutations.
Collapse
|
41
|
Mamidi R, Gresham KS, Li J, Stelzer JE. Cardiac myosin binding protein-C Ser 302 phosphorylation regulates cardiac β-adrenergic reserve. SCIENCE ADVANCES 2017; 3:e1602445. [PMID: 28345052 PMCID: PMC5345928 DOI: 10.1126/sciadv.1602445] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 02/02/2017] [Indexed: 05/22/2023]
Abstract
Phosphorylation of cardiac myosin binding protein-C (MyBP-C) modulates cardiac contractile function; however, the specific roles of individual serines (Ser) within the M-domain that are targets for β-adrenergic signaling are not known. Recently, we demonstrated that significant accelerations in in vivo pressure development following β-agonist infusion can occur in transgenic (TG) mouse hearts expressing phospho-ablated Ser282 (that is, TGS282A) but not in hearts expressing phospho-ablation of all three serines [that is, Ser273, Ser282, and Ser302 (TG3SA)], suggesting an important modulatory role for other Ser residues. In this regard, there is evidence that Ser302 phosphorylation may be a key contributor to the β-agonist-induced positive inotropic responses in the myocardium, but its precise functional role has not been established. Thus, to determine the in vivo and in vitro functional roles of Ser302 phosphorylation, we generated TG mice expressing nonphosphorylatable Ser302 (that is, TGS302A). Left ventricular pressure-volume measurements revealed that TGS302A mice displayed no accelerations in the rate of systolic pressure rise and an inability to maintain systolic pressure following dobutamine infusion similar to TG3SA mice, implicating Ser302 phosphorylation as a critical regulator of enhanced systolic performance during β-adrenergic stress. Dynamic strain-induced cross-bridge (XB) measurements in skinned myocardium isolated from TGS302A hearts showed that the molecular basis for impaired β-adrenergic-mediated enhancements in systolic function is due to the absence of protein kinase A-mediated accelerations in the rate of cooperative XB recruitment. These results demonstrate that Ser302 phosphorylation regulates cardiac contractile reserve by enhancing contractile responses during β-adrenergic stress.
Collapse
Affiliation(s)
- Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Kenneth S. Gresham
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Corresponding author.
| |
Collapse
|
42
|
Kensler RW, Craig R, Moss RL. Phosphorylation of cardiac myosin binding protein C releases myosin heads from the surface of cardiac thick filaments. Proc Natl Acad Sci U S A 2017; 114:E1355-E1364. [PMID: 28167762 PMCID: PMC5338423 DOI: 10.1073/pnas.1614020114] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cardiac myosin binding protein C (cMyBP-C) has a key regulatory role in cardiac contraction, but the mechanism by which changes in phosphorylation of cMyBP-C accelerate cross-bridge kinetics remains unknown. In this study, we isolated thick filaments from the hearts of mice in which the three serine residues (Ser273, Ser282, and Ser302) that are phosphorylated by protein kinase A in the m-domain of cMyBP-C were replaced by either alanine or aspartic acid, mimicking the fully nonphosphorylated and the fully phosphorylated state of cMyBP-C, respectively. We found that thick filaments from the cMyBP-C phospho-deficient hearts had highly ordered cross-bridge arrays, whereas the filaments from the cMyBP-C phospho-mimetic hearts showed a strong tendency toward disorder. Our results support the hypothesis that dephosphorylation of cMyBP-C promotes or stabilizes the relaxed/superrelaxed quasi-helical ordering of the myosin heads on the filament surface, whereas phosphorylation weakens this stabilization and binding of the heads to the backbone. Such structural changes would modulate the probability of myosin binding to actin and could help explain the acceleration of cross-bridge interactions with actin when cMyBP-C is phosphorylated because of, for example, activation of β1-adrenergic receptors in myocardium.
Collapse
Affiliation(s)
- Robert W Kensler
- Department of Anatomy and Neurobiology, University of Puerto Rico Medical School, San Juan, PR 00936;
| | - Roger Craig
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Richard L Moss
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705
| |
Collapse
|
43
|
Gresham KS, Mamidi R, Li J, Kwak H, Stelzer JE. Sarcomeric protein modification during adrenergic stress enhances cross-bridge kinetics and cardiac output. J Appl Physiol (1985) 2016; 122:520-530. [PMID: 27909224 DOI: 10.1152/japplphysiol.00306.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 10/21/2016] [Accepted: 11/23/2016] [Indexed: 12/23/2022] Open
Abstract
Molecular adaptations to chronic neurohormonal stress, including sarcomeric protein cleavage and phosphorylation, provide a mechanism to increase ventricular contractility and enhance cardiac output, yet the link between sarcomeric protein modifications and changes in myocardial function remains unclear. To examine the effects of neurohormonal stress on posttranslational modifications of sarcomeric proteins, mice were administered combined α- and β-adrenergic receptor agonists (isoproterenol and phenylephrine, IPE) for 14 days using implantable osmotic pumps. In addition to significant cardiac hypertrophy and increased maximal ventricular pressure, IPE treatment accelerated pressure development and relaxation (74% increase in dP/dtmax and 14% decrease in τ), resulting in a 52% increase in cardiac output compared with saline (SAL)-treated mice. Accelerated pressure development was maintained when accounting for changes in heart rate and preload, suggesting that myocardial adaptations contribute to enhanced ventricular contractility. Ventricular myocardium isolated from IPE-treated mice displayed a significant reduction in troponin I (TnI) and myosin-binding protein C (MyBP-C) expression and a concomitant increase in the phosphorylation levels of the remaining TnI and MyBP-C protein compared with myocardium isolated from saline-treated control mice. Skinned myocardium isolated from IPE-treated mice displayed a significant acceleration in the rate of cross-bridge (XB) detachment (46% increase) and an enhanced magnitude of XB recruitment (43% increase) at submaximal Ca2+ activation compared with SAL-treated mice but unaltered myofilament Ca2+ sensitivity of force generation. These findings demonstrate that sarcomeric protein modifications during neurohormonal stress are molecular adaptations that enhance in vivo ventricular contractility through accelerated XB kinetics to increase cardiac output.NEW & NOTEWORTHY Posttranslational modifications to sarcomeric regulatory proteins provide a mechanism to modulate cardiac function in response to stress. In this study, we demonstrate that neurohormonal stress produces modifications to myosin-binding protein C and troponin I, including a reduction in protein expression within the sarcomere and increased phosphorylation of the remaining protein, which serve to enhance cross-bridge kinetics and increase cardiac output. These findings highlight the importance of sarcomeric regulatory protein modifications in modulating ventricular function during cardiac stress.
Collapse
Affiliation(s)
- Kenneth S Gresham
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jiayang Li
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Hyerin Kwak
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
44
|
Cheng Y, Lindert S, Oxenford L, Tu AY, McCulloch AD, Regnier M. Effects of Cardiac Troponin I Mutation P83S on Contractile Properties and the Modulation by PKA-Mediated Phosphorylation. J Phys Chem B 2016; 120:8238-53. [PMID: 27150586 PMCID: PMC5001945 DOI: 10.1021/acs.jpcb.6b01859] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
cTnI(P82S) (cTnI(P83S) in rodents) resides at the I-T arm of cardiac troponin I (cTnI) and was initially identified as a disease-causing mutation of hypertrophic cardiomyopathy (HCM). However, later studies suggested this may not be true. We recently reported that introduction of an HCM-associated mutation in either inhibitory-peptide (cTnI(R146G)) or cardiac-specific N-terminus (cTnI(R21C)) of cTnI blunts the PKA-mediated modulation on myofibril activation/relaxation kinetics by prohibiting formation of intrasubunit contacts between these regions. Here, we tested whether this also occurs for cTnI(P83S). cTnI(P83S) increased both Ca(2+) binding affinity to cTn (KCa) and affinity of cTnC for cTnI (KC-I), and eliminated the reduction of KCa and KC-I observed for phosphorylated-cTnI(WT). In isolated myofibrils, cTnI(P83S) maintained maximal tension (TMAX) and Ca(2+) sensitivity of tension (pCa50). For cTnI(WT) myofibrils, PKA-mediated phosphorylation decreased pCa50 and sped up the slow-phase relaxation (especially for those Ca(2+) conditions that heart performs in vivo). Those effects were blunted for cTnI(P83S) myofibrils. Molecular-dynamics simulations suggested cTnI(P83S) moderately inhibited an intrasubunit interaction formation between inhibitory-peptide and N-terminus, but this "blunting" effect was weaker than that with cTnI(R146G) or cTnI(R21C). In summary, cTnI(P83S) has similar effects as other HCM-associated cTnI mutations on troponin and myofibril function even though it is in the I-T arm of cTnI.
Collapse
Affiliation(s)
- Yuanhua Cheng
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States
- National Biomedical Computation Resource, University of California San Diego, La Jolla, California 92093, United States
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Lucas Oxenford
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States
| | - An-yue Tu
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States
| | - Andrew D. McCulloch
- National Biomedical Computation Resource, University of California San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
45
|
Krishnamoorthy N, Gajendrarao P, Olivotto I, Yacoub M. Impact of disease-causing mutations on inter-domain interactions in cMyBP-C: a steered molecular dynamics study. J Biomol Struct Dyn 2016; 35:1916-1922. [DOI: 10.1080/07391102.2016.1199329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Navaneethakrishnan Krishnamoorthy
- Qatar Cardiovascular Research Centre, Qatar Foundation, Doha, Qatar
- Division of Experimental Genetics, Sidra Medical and Research Centre, Doha, Qatar
- Heart Science Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Poornima Gajendrarao
- Qatar Cardiovascular Research Centre, Qatar Foundation, Doha, Qatar
- Heart Science Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Iacopo Olivotto
- Referral Centre for Myocardial Diseases, Careggi University Hospital, Florence, Italy
| | - Magdi Yacoub
- Qatar Cardiovascular Research Centre, Qatar Foundation, Doha, Qatar
- Heart Science Centre, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
46
|
McNamara JW, Li A, Smith NJ, Lal S, Graham RM, Kooiker KB, van Dijk SJ, Remedios CGD, Harris SP, Cooke R. Ablation of cardiac myosin binding protein-C disrupts the super-relaxed state of myosin in murine cardiomyocytes. J Mol Cell Cardiol 2016; 94:65-71. [PMID: 27021517 DOI: 10.1016/j.yjmcc.2016.03.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/17/2016] [Accepted: 03/19/2016] [Indexed: 11/24/2022]
Abstract
Cardiac myosin binding protein-C (cMyBP-C) is a structural and regulatory component of cardiac thick filaments. It is observed in electron micrographs as seven to nine transverse stripes in the central portion of each half of the A band. Its C-terminus binds tightly to the myosin rod and contributes to thick filament structure, while the N-terminus can bind both myosin S2 and actin, influencing their structure and function. Mutations in the MYBPC3 gene (encoding cMyBP-C) are commonly associated with hypertrophic cardiomyopathy (HCM). In cardiac cells there exists a population of myosin heads in the super-relaxed (SRX) state, which are bound to the thick filament core with a highly inhibited ATPase activity. This report examines the role cMyBP-C plays in regulating the population of the SRX state of cardiac myosin by using an assay that measures single ATP turnover of myosin. We report a significant decrease in the proportion of myosin heads in the SRX state in homozygous cMyBP-C knockout mice, however heterozygous cMyBP-C knockout mice do not significantly differ from the wild type. A smaller, non-significant decrease is observed when thoracic aortic constriction is used to induce cardiac hypertrophy in mutation negative mice. These results support the proposal that cMyBP-C stabilises the thick filament and that the loss of cMyBP-C results in an untethering of myosin heads. This results in an increased myosin ATP turnover, further consolidating the relationship between thick filament structure and the myosin ATPase.
Collapse
Affiliation(s)
- James W McNamara
- Discipline of Anatomy & Histology, Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| | - Amy Li
- Discipline of Anatomy & Histology, Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Nicola J Smith
- Molecular Cardiology Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Sean Lal
- Discipline of Anatomy & Histology, Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Robert M Graham
- Molecular Cardiology Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Kristina Bezold Kooiker
- Department of Pediatrics (Cardiology), Cardiovascular Institute, Stanford University, CA 94304, USA
| | - Sabine J van Dijk
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - Cristobal G Dos Remedios
- Discipline of Anatomy & Histology, Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Samantha P Harris
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - Roger Cooke
- Department of Biochemistry & Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158-2517, USA
| |
Collapse
|
47
|
Site-directed spectroscopy of cardiac myosin-binding protein C reveals effects of phosphorylation on protein structural dynamics. Proc Natl Acad Sci U S A 2016; 113:3233-8. [PMID: 26908877 DOI: 10.1073/pnas.1521281113] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We have used the site-directed spectroscopies of time-resolved fluorescence resonance energy transfer (TR-FRET) and double electron-electron resonance (DEER), combined with complementary molecular dynamics (MD) simulations, to resolve the structure and dynamics of cardiac myosin-binding protein C (cMyBP-C), focusing on the N-terminal region. The results have implications for the role of this protein in myocardial contraction, with particular relevance to β-adrenergic signaling, heart failure, and hypertrophic cardiomyopathy. N-terminal cMyBP-C domains C0-C2 (C0C2) contain binding regions for potential interactions with both thick and thin filaments. Phosphorylation by PKA in the MyBP-C motif regulates these binding interactions. Our spectroscopic assays detect distances between pairs of site-directed probes on cMyBP-C. We engineered intramolecular pairs of labeling sites within cMyBP-C to measure, with high resolution, the distance and disorder in the protein's flexible regions using TR-FRET and DEER. Phosphorylation reduced the level of molecular disorder and the distribution of C0C2 intramolecular distances became more compact, with probes flanking either the motif between C1 and C2 or the Pro/Ala-rich linker (PAL) between C0 and C1. Further insight was obtained from microsecond MD simulations, which revealed a large structural change in the disordered motif region in which phosphorylation unmasks the surface of a series of residues on a stable α-helix within the motif with high potential as a protein-protein interaction site. These experimental and computational findings elucidate structural transitions in the flexible and dynamic portions of cMyBP-C, providing previously unidentified molecular insight into the modulatory role of this protein in cardiac muscle contractility.
Collapse
|
48
|
Mamidi R, Gresham KS, Verma S, Stelzer JE. Cardiac Myosin Binding Protein-C Phosphorylation Modulates Myofilament Length-Dependent Activation. Front Physiol 2016; 7:38. [PMID: 26913007 PMCID: PMC4753332 DOI: 10.3389/fphys.2016.00038] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/28/2016] [Indexed: 11/13/2022] Open
Abstract
Cardiac myosin binding protein-C (cMyBP-C) phosphorylation is an important regulator of contractile function, however, its contributions to length-dependent changes in cross-bridge (XB) kinetics is unknown. Therefore, we performed mechanical experiments to quantify contractile function in detergent-skinned ventricular preparations isolated from wild-type (WT) hearts, and hearts expressing non-phosphorylatable cMyBP-C [Ser to Ala substitutions at residues Ser273, Ser282, and Ser302 (i.e., 3SA)], at sarcomere length (SL) 1.9 μm or 2.1μm, prior and following protein kinase A (PKA) treatment. Steady-state force generation measurements revealed a blunting in the length-dependent increase in myofilament Ca(2+)-sensitivity of force generation (pCa50) following an increase in SL in 3SA skinned myocardium compared to WT skinned myocardium. Dynamic XB behavior was assessed at submaximal Ca(2+)-activations by imposing an acute rapid stretch of 2% of initial muscle length, and measuring both the magnitudes and rates of resultant phases of force decay due to strain-induced XB detachment and delayed force rise due to recruitment of additional XBs with increased SL (i.e., stretch activation). The magnitude (P2) and rate of XB detachment (k rel) following stretch was significantly reduced in 3SA skinned myocardium compared to WT skinned myocardium at short and long SL, and prior to and following PKA treatment. Furthermore, the length-dependent acceleration of k rel due to decreased SL that was observed in WT skinned myocardium was abolished in 3SA skinned myocardium. PKA treatment accelerated the rate of XB recruitment (k df) following stretch at both SL's in WT but not in 3SA skinned myocardium. The amplitude of the enhancement in force generation above initial pre-stretch steady-state levels (P3) was not different between WT and 3SA skinned myocardium at any condition measured. However, the magnitude of the entire delayed force phase which can dip below initial pre-stretch steady-state levels (Pdf) was significantly lower in 3SA skinned myocardium under all conditions, in part due to a reduced magnitude of XB detachment (P2) in 3SA skinned myocardium compared to WT skinned myocardium. These findings demonstrate that cMyBP-C phospho-ablation regulates SL- and PKA-mediated effects on XB kinetics in the myocardium, which would be expected to contribute to the regulation of the Frank-Starling mechanism.
Collapse
Affiliation(s)
- Ranganath Mamidi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University Cleveland, OH, USA
| | - Kenneth S Gresham
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University Cleveland, OH, USA
| | - Sujeet Verma
- Department of Horticultural Science, Institute of Food and Agricultural Sciences Gulf Coast Research and Education Center, University of Florida Wimauma, FL, USA
| | - Julian E Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University Cleveland, OH, USA
| |
Collapse
|
49
|
Hanft LM, Cornell TD, McDonald CA, Rovetto MJ, Emter CA, McDonald KS. Molecule specific effects of PKA-mediated phosphorylation on rat isolated heart and cardiac myofibrillar function. Arch Biochem Biophys 2016; 601:22-31. [PMID: 26854722 DOI: 10.1016/j.abb.2016.01.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/27/2016] [Accepted: 01/29/2016] [Indexed: 01/08/2023]
Abstract
Increased cardiac myocyte contractility by the β-adrenergic system is an important mechanism to elevate cardiac output to meet hemodynamic demands and this process is depressed in failing hearts. While increased contractility involves augmented myoplasmic calcium transients, the myofilaments also adapt to boost the transduction of the calcium signal. Accordingly, ventricular contractility was found to be tightly correlated with PKA-mediated phosphorylation of two myofibrillar proteins, cardiac myosin binding protein-C (cMyBP-C) and cardiac troponin I (cTnI), implicating these two proteins as important transducers of hemodynamics to the cardiac sarcomere. Consistent with this, we have previously found that phosphorylation of myofilament proteins by PKA (a downstream signaling molecule of the beta-adrenergic system) increased force, slowed force development rates, sped loaded shortening, and increased power output in rat skinned cardiac myocyte preparations. Here, we sought to define molecule-specific mechanisms by which PKA-mediated phosphorylation regulates these contractile properties. Regarding cTnI, the incorporation of thin filaments with unphosphorylated cTnI decreased isometric force production and these changes were reversed by PKA-mediated phosphorylation in skinned cardiac myocytes. Further, incorporation of unphosphorylated cTnI sped rates of force development, which suggests less cooperative thin filament activation and reduced recruitment of non-cycling cross-bridges into the pool of cycling cross-bridges, a process that would tend to depress both myocyte force and power. Regarding MyBP-C, PKA treatment of slow-twitch skeletal muscle fibers caused phosphorylation of MyBP-C (but not slow skeletal TnI (ssTnI)) and yielded faster loaded shortening velocity and ∼30% increase in power output. These results add novel insight into the molecular specificity by which the β-adrenergic system regulates myofibrillar contractility and how attenuation of PKA-induced phosphorylation of cMyBP-C and cTnI may contribute to ventricular pump failure.
Collapse
Affiliation(s)
- Laurin M Hanft
- Department of Medical Pharmacology & Physiology, School of Medicine University of Missouri, Columbia, MO 65212, USA
| | - Timothy D Cornell
- Department of Medical Pharmacology & Physiology, School of Medicine University of Missouri, Columbia, MO 65212, USA
| | - Colin A McDonald
- Department of Medical Pharmacology & Physiology, School of Medicine University of Missouri, Columbia, MO 65212, USA
| | - Michael J Rovetto
- Department of Medical Pharmacology & Physiology, School of Medicine University of Missouri, Columbia, MO 65212, USA
| | - Craig A Emter
- Department of Biomedical Sciences, College of Veterinary Medicine University of Missouri, Columbia, MO 65211, USA
| | - Kerry S McDonald
- Department of Medical Pharmacology & Physiology, School of Medicine University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
50
|
Lindert S, Cheng Y, Kekenes-Huskey P, Regnier M, McCammon JA. Effects of HCM cTnI mutation R145G on troponin structure and modulation by PKA phosphorylation elucidated by molecular dynamics simulations. Biophys J 2015; 108:395-407. [PMID: 25606687 DOI: 10.1016/j.bpj.2014.11.3461] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/21/2014] [Accepted: 11/21/2014] [Indexed: 10/24/2022] Open
Abstract
Cardiac troponin (cTn) is a key molecule in the regulation of human cardiac muscle contraction. The N-terminal cardiac-specific peptide of the inhibitory subunit of troponin, cTnI (cTnI(1-39)), is a target for phosphorylation by protein kinase A (PKA) during β-adrenergic stimulation. We recently presented evidence indicating that this peptide interacts with the inhibitory peptide (cTnl(137-147)) when S23 and S24 are phosphorylated. The inhibitory peptide is also the target of the point mutation cTnI-R145G, which is associated with hypertrophic cardiomyopathy (HCM), a disease associated with sudden death in apparently healthy young adults. It has been shown that both phosphorylation and this mutation alter the cTnC-cTnI (C-I) interaction, which plays a crucial role in modulating contractile activation. However, little is known about the molecular-level events underlying this modulation. Here, we computationally investigated the effects of the cTnI-R145G mutation on the dynamics of cTn, cTnC Ca(2+) handling, and the C-I interaction. Comparisons were made with the cTnI-R145G/S23D/S24D phosphomimic mutation, which has been used both experimentally and computationally to study the cTnI N-terminal specific effects of PKA phosphorylation. Additional comparisons between the phosphomimic mutations and the real phosphorylations were made. For this purpose, we ran triplicate 150 ns molecular dynamics simulations of cTnI-R145G Ca(2+)-bound cTnC(1-161)-cTnI(1-172)-cTnT(236-285), cTnI-R145G/S23D/S24D Ca(2+)-bound cTnC(1-161)-cTnI(1-172)-cTnT(236-285), and cTnI-R145G/PS23/PS24 Ca(2+)-bound cTnC(1-161)-cTnI(1-172)-cTnT(236-285), respectively. We found that the cTnI-R145G mutation did not impact the overall dynamics of cTn, but stabilized crucial Ca(2+)-coordinating interactions. However, the phosphomimic mutations increased overall cTn fluctuations and destabilized Ca(2+) coordination. Interestingly, cTnI-R145G blunted the intrasubunit interactions between the cTnI N-terminal extension and the cTnI inhibitory peptide, which have been suggested to play a crucial role in modulating troponin function during β-adrenergic stimulation. These findings offer a molecular-level explanation for how the HCM mutation cTnI-R145G reduces the modulation of cTn by phosphorylation of S23/S24 during β-adrenergic stimulation.
Collapse
Affiliation(s)
- Steffen Lindert
- Department of Pharmacology, University of California San Diego, La Jolla, California; NSF Center for Theoretical Biological Physics, La Jolla, California.
| | - Yuanhua Cheng
- Department of Bioengineering, University of Washington, Seattle, Washington
| | - Peter Kekenes-Huskey
- Department of Pharmacology, University of California San Diego, La Jolla, California; Department of Chemistry, University of Kentucky, Lexington, Kentucky
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, Washington
| | - J Andrew McCammon
- Department of Pharmacology, University of California San Diego, La Jolla, California; Howard Hughes Medical Institute, University of California San Diego, La Jolla, California; Department of Chemistry and Biochemistry, National Biomedical Computation Resource, University of California San Diego, La Jolla, California; NSF Center for Theoretical Biological Physics, La Jolla, California
| |
Collapse
|