1
|
Chavali SS, Carman PJ, Shuman H, Ostap EM, Sindelar CV. High resolution structures of Myosin-IC reveal a unique actin-binding orientation, ADP release pathway, and power stroke trajectory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632429. [PMID: 39829824 PMCID: PMC11741418 DOI: 10.1101/2025.01.10.632429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Myosin-IC (myo1c) is a class-I myosin that supports transport and remodeling of the plasma membrane and membrane-bound vesicles. Like other members of the myosin family, its biochemical kinetics are altered in response to changes in mechanical loads that resist the power stroke. However, myo1c is unique in that the primary force-sensitive kinetic transition is the isomerization that follows ATP binding, not ADP release as in other slow myosins. Myo1c also powers actin gliding along curved paths, propelling actin filaments in leftward circles. To understand the origins of this unique force-sensing and motile behavior, we solved actin-bound myo1c cryo-EM structures in the presence and absence of ADP. Our structures reveal that in contrast with other myosins, the myo1c lever arm swing is skewed, partly due to a different actin interface that reorients the motor domain on actin. The structures also reveal unique nucleotide-dependent behavior of both the nucleotide pocket as well as an element called the N-terminal extension. We incorporate these observations into a model that explains why force primarily regulates ATP binding in myo1c, rather than ADP release as in other myosins. Integrating our cryo-EM data with available crystallography structures allows the modeling of full-length myo1c during force generation, supplying insights into its role in membrane remodeling. These results highlight how relatively minor sequence differences in members of the myosin superfamily can significantly alter power stroke geometry and force-sensing properties, with important implications for biological function. Significance Statement Myosin-IC (myo1c) uses an ATP-driven 'power-stroke' to support slow intracellular membrane and vesicle transport. We used cryo-electron microscopy to understand adaptations of myo1c to perform its unique roles. We discovered an altered interface between myo1c and actin compared with the closely related myo1b, which repositions the motor domain and alters the trajectory of its lever arm swing compared to other myosins. This explains why myo1c propels actin filaments in a leftward circular path. We also discovered a unique role in force sensing for a structural element called the N-terminal extension and built a full-length atomic model for the myo1c power-stroke. Our findings highlight how myosins can tune their power-stroke geometries and force-sensing properties to adapt to diverse cellular functions.
Collapse
|
2
|
Stehle R. Phosphate rebinding induces force reversal via slow backward cycling of cross-bridges. Front Physiol 2025; 15:1476876. [PMID: 39839531 PMCID: PMC11747208 DOI: 10.3389/fphys.2024.1476876] [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: 08/06/2024] [Accepted: 11/28/2024] [Indexed: 01/23/2025] Open
Abstract
Objective Previous studies on muscle fibers, myofibrils, and myosin revealed that the release of inorganic phosphate (Pi) and the force-generating step(s) are reversible, with cross-bridges also cycling backward through these steps by reversing force-generating steps and rebinding Pi. The aim was to explore the significance of force redevelopment kinetics (rate constant k TR) in cardiac myofibrils for the coupling between the Pi binding induced force reversal and the rate-limiting transition f - for backward cycling of cross-bridges from force-generating to non-force-generating states. Methods k TR and force generation of cardiac myofibrils from guinea pigs were investigated at 0.015-20 mM Pi. The observed force-[Pi], force-log [Pi], k TR-[Pi], and k TR-force relations were assessed with various single-pathway models of the cross-bridge cycle that differed in sequence and kinetics of reversible Pi release, reversible force-generating step and reversible rate-limiting transition. Based on the interpretation that k TR reflects the sum of rate-limiting transitions in the cross-bridge cycle, an indicator, the coupling strength, was defined to quantify the contribution of Pi binding induced force reversal to the rate-limiting transition f - from the [Pi]-modulated k TR-force relation. Results Increasing [Pi] decreased force by a bi-linear force-log [Pi] relation, increased k TR in a slightly downward curved dependence with [Pi], and altered k TR almost reciprocally to force reflected by the k TR-force relation. Force-[Pi] and force-log [Pi] relations provided less selectivity for the exclusion of models than the k TR-[Pi] and k TR-force relations. The k TR-force relation observed in experiments with cardiac myofibrils yielded the coupling strength +0.84 ± 0.08 close to 1, the maximum coupling strength expected for the reciprocal k TR-force relationship. Single pathway models consisting of fast reversible force generation before or after rapid reversible Pi release failed to describe the observed k TR-force relation. Single pathway models consistent with the observed k TR-force relation had either slow Pi binding or slow force reversal, i.e., in the consistent single pathway models, f - was assigned to the rate of either Pi binding or force reversal. Conclusion Backward flux of cross-bridges from force-generating to non-force-generating states is limited by the rates of Pi binding or force reversal ruling out other rate-limiting steps uncoupled from Pi binding induced force reversal.
Collapse
Affiliation(s)
- Robert Stehle
- Institute of Vegetative Physiology, University of Cologne, Köln, Germany
| |
Collapse
|
3
|
Rassier DE, Månsson A. Mechanisms of myosin II force generation: insights from novel experimental techniques and approaches. Physiol Rev 2025; 105:1-93. [PMID: 38451233 DOI: 10.1152/physrev.00014.2023] [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: 03/16/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
Myosin II is a molecular motor that converts chemical energy derived from ATP hydrolysis into mechanical work. Myosin II isoforms are responsible for muscle contraction and a range of cell functions relying on the development of force and motion. When the motor attaches to actin, ATP is hydrolyzed and inorganic phosphate (Pi) and ADP are released from its active site. These reactions are coordinated with changes in the structure of myosin, promoting the so-called "power stroke" that causes the sliding of actin filaments. The general features of the myosin-actin interactions are well accepted, but there are critical issues that remain poorly understood, mostly due to technological limitations. In recent years, there has been a significant advance in structural, biochemical, and mechanical methods that have advanced the field considerably. New modeling approaches have also allowed researchers to understand actomyosin interactions at different levels of analysis. This paper reviews recent studies looking into the interaction between myosin II and actin filaments, which leads to power stroke and force generation. It reviews studies conducted with single myosin molecules, myosins working in filaments, muscle sarcomeres, myofibrils, and fibers. It also reviews the mathematical models that have been used to understand the mechanics of myosin II in approaches focusing on single molecules to ensembles. Finally, it includes brief sections on translational aspects, how changes in the myosin motor by mutations and/or posttranslational modifications may cause detrimental effects in diseases and aging, among other conditions, and how myosin II has become an emerging drug target.
Collapse
Affiliation(s)
- Dilson E Rassier
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Alf Månsson
- Physiology, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
4
|
Hei B, Tardiff JC, Schwartz SD. Human cardiac β-myosin powerstroke energetics: Thin filament, Pi displacement, and mutation effects. Biophys J 2024; 123:3133-3142. [PMID: 39001604 PMCID: PMC11427785 DOI: 10.1016/j.bpj.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
The powerstroke of human cardiac β-myosin is an important stage of the cross-bridge cycle that generates force for muscle contraction. However, the starting structure of this process has never been resolved, and the relative timing of the powerstroke and inorganic phosphate (Pi) release is still controversial. In this study, we generated an atomistic model of myosin on the thin filament and utilized metadynamics simulations to predict the absent starting structure of the powerstroke. We demonstrated that the displacement of Pi from the active site during the powerstroke is likely necessary, reducing the energy barrier of the conformation change. The effects of the presence of the thin filament, the hypertrophic cardiomyopathy mutation R712L, and the binding of mavacamten on the powerstroke process were also investigated.
Collapse
Affiliation(s)
- Bai Hei
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona
| | - Jil C Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Steven D Schwartz
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona.
| |
Collapse
|
5
|
Shen K, Chen T, Xiao M. MYH9-related inherited thrombocytopenia: the genetic spectrum, underlying mechanisms, clinical phenotypes, diagnosis, and management approaches. Res Pract Thromb Haemost 2024; 8:102552. [PMID: 39309229 PMCID: PMC11415342 DOI: 10.1016/j.rpth.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 09/25/2024] Open
Abstract
Inherited thrombocytopenias have been considered exceedingly rare for a long time, but recent advances have facilitated diagnosis and greatly enabled the discovery of new causative genes. MYH9-related disease (MYH9-RD) represents one of the most frequent forms of inherited thrombocytopenia, usually presenting with nonspecific clinical manifestations, which renders it difficult to establish an accurate diagnosis. MYH9-RD is an autosomal dominant-inherited thrombocytopenia caused by deleterious variants in the MYH9 gene encoding the heavy chain of nonmuscle myosin IIA. Patients with MYH9-RD usually present with thrombocytopenia and platelet macrocytosis at birth or in infancy, and most of them may develop one or more extrahematologic manifestations of progressive nephritis, sensorial hearing loss, presenile cataracts, and elevated liver enzymatic levels during childhood and adult life. Here, we have reviewed recent advances in the study of MYH9-RD, which aims to provide an updated and comprehensive summary of the current knowledge and improve our understanding of the genetic spectrum, underlying mechanisms, clinical phenotypes, diagnosis, and management approaches of this rare disease. Importantly, our goal is to enable physicians to better understand this rare disease and highlight the critical role of genetic etiologic analysis in ensuring accurate diagnosis, clinical management, and genetic counseling while avoiding ineffective and potentially harmful therapies for MYH9-RD patients.
Collapse
Affiliation(s)
- Kefeng Shen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Chen
- Department of Ophthalmology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Bodt SML, Ge J, Ma W, Rasicci DV, Desetty R, McCammon JA, Yengo CM. Dilated cardiomyopathy mutation in beta-cardiac myosin enhances actin activation of the power stroke and phosphate release. PNAS NEXUS 2024; 3:pgae279. [PMID: 39108304 PMCID: PMC11302452 DOI: 10.1093/pnasnexus/pgae279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/25/2024] [Indexed: 08/13/2024]
Abstract
Inherited mutations in human beta-cardiac myosin (M2β) can lead to severe forms of heart failure. The E525K mutation in M2β is associated with dilated cardiomyopathy (DCM) and was found to stabilize the interacting heads motif (IHM) and autoinhibited super-relaxed (SRX) state in dimeric heavy meromyosin. However, in monomeric M2β subfragment 1 (S1) we found that E525K enhances (threefold) the maximum steady-state actin-activated ATPase activity (k cat) and decreases (eightfold) the actin concentration at which ATPase is one-half maximal (K ATPase). We also found a twofold to fourfold increase in the actin-activated power stroke and phosphate release rate constants at 30 μM actin, which overall enhanced the duty ratio threefold. Loaded motility assays revealed that the enhanced intrinsic motor activity translates to increased ensemble force in M2β S1. Glutamate 525, located near the actin binding region in the so-called activation loop, is highly conserved and predicted to form a salt bridge with another conserved residue (lysine 484) in the relay helix. Enhanced sampling molecular dynamics simulations predict that the charge reversal mutation disrupts the E525-K484 salt bridge, inducing conformations with a more flexible relay helix and a wide phosphate release tunnel. Our results highlight a highly conserved allosteric pathway associated with actin activation of the power stroke and phosphate release and suggest an important feature of the autoinhibited IHM is to prevent this region of myosin from interacting with actin. The ability of the E525K mutation to stabilize the IHM likely overrides the enhanced intrinsic motor properties, which may be key to triggering DCM pathogenesis.
Collapse
Affiliation(s)
- Skylar M L Bodt
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - Jinghua Ge
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - Wen Ma
- Department of Physics, University of Vermont, 149 Beaumont Avenue, Burlington, VT 05405, USA
| | - David V Rasicci
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University School of Medicine, 64 Medical Center Dr, Morgantown, WV 26506, USA
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| | - J Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, 500 University Dr, Hershey, PA 17033, USA
| |
Collapse
|
7
|
Freedman H, Tuszynski JA. Study of the Myosin Relay Helix Peptide by Molecular Dynamics Simulations, Pump-Probe and 2D Infrared Spectroscopy. Int J Mol Sci 2024; 25:6406. [PMID: 38928112 PMCID: PMC11203622 DOI: 10.3390/ijms25126406] [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: 03/25/2024] [Revised: 05/07/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
The Davydov model was conjectured to describe how an amide I excitation created during ATP hydrolysis in myosin might be significant in providing energy to drive myosin's chemomechanical cycle. The free energy surfaces of the myosin relay helix peptide dissolved in 2,2,2-trifluoroethanol (TFE), determined by metadynamics simulations, demonstrate local minima differing in free energy by only ~2 kT, corresponding to broken and stabilized hydrogen bonds, respectively. Experimental pump-probe and 2D infrared spectroscopy were performed on the peptide dissolved in TFE. The relative heights of two peaks seen in the pump-probe data and the corresponding relative volumes of diagonal peaks seen in the 2D-IR spectra at time delays between 0.5 ps and 1 ps differ noticeably from what is seen at earlier or later time delays or in the linear spectrum, indicating that a vibrational excitation may influence the conformational state of this helix. Thus, it is possible that the presence of an amide I excitation may be a direct factor in the conformational state taken on by the myosin relay helix following ATP hydrolysis in myosin.
Collapse
Affiliation(s)
- Holly Freedman
- Center for Molecular Spectroscopy and Dynamics, Institute for Basic Science (IBS), Seoul 02841, Republic of Korea
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, 2000 East 30 South Skaggs 306, Salt Lake City, UT 84112, USA
| | - Jack A. Tuszynski
- Department of Physics, University of Alberta, 11335 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada;
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, I-1029 Turin, Italy
- Department of Data Science and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland
| |
Collapse
|
8
|
Auguin D, Robert-Paganin J, Réty S, Kikuti C, David A, Theumer G, Schmidt AW, Knölker HJ, Houdusse A. Omecamtiv mecarbil and Mavacamten target the same myosin pocket despite opposite effects in heart contraction. Nat Commun 2024; 15:4885. [PMID: 38849353 PMCID: PMC11161628 DOI: 10.1038/s41467-024-47587-9] [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: 10/30/2023] [Accepted: 04/03/2024] [Indexed: 06/09/2024] Open
Abstract
Inherited cardiomyopathies are common cardiac diseases worldwide, leading in the late stage to heart failure and death. The most promising treatments against these diseases are small molecules directly modulating the force produced by β-cardiac myosin, the molecular motor driving heart contraction. Omecamtiv mecarbil and Mavacamten are two such molecules that completed phase 3 clinical trials, and the inhibitor Mavacamten is now approved by the FDA. In contrast to Mavacamten, Omecamtiv mecarbil acts as an activator of cardiac contractility. Here, we reveal by X-ray crystallography that both drugs target the same pocket and stabilize a pre-stroke structural state, with only few local differences. All-atom molecular dynamics simulations reveal how these molecules produce distinct effects in motor allostery thus impacting force production in opposite way. Altogether, our results provide the framework for rational drug development for the purpose of personalized medicine.
Collapse
Affiliation(s)
- Daniel Auguin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
- Laboratoire de Physiologie, Ecologie et Environnement (P2E), UPRES EA 1207/USC INRAE-1328, UFR Sciences et Techniques, Université d'Orléans, Orléans, France
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | - Stéphane Réty
- Laboratoire de Biologie et Modélisation de la Cellule, ENS de Lyon, CNRS, UMR 5239, Inserm, U1293, Université Claude Bernard Lyon 1, Lyon, France
| | - Carlos Kikuti
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | - Amandine David
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France
| | | | | | | | - Anne Houdusse
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, Paris, 75248, France.
| |
Collapse
|
9
|
Kawai M, Iorga B. Oscillatory work and the step that generates force in single myofibrils from rabbit psoas. Pflugers Arch 2024; 476:949-962. [PMID: 38558187 DOI: 10.1007/s00424-024-02935-y] [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: 11/08/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
The elementary molecular step that generates force by cross-bridges (CBs) in active muscles has been under intense investigation in the field of muscle biophysics. It is known that an increase in the phosphate (Pi) concentration diminishes isometric force in active fibers, indicating a tight coupling between the force generation step and the Pi release step. The question asked here is whether the force generation occurs before Pi release or after release. We investigated the effect of Pi on oscillatory work production in single myofibrils and found that Pi-attached state(s) to CBs is essential for its production. Oscillatory work is the mechanism that allows an insect to fly by beating its wings, and it also has been observed in skeletal and cardiac muscle fibers, implying that it is an essential feature of all striated muscle types. With our studies, oscillatory work disappears in the absence of Pi in experiments using myofibrils. This suggests that force is generated during a transition between steps of oscillatory work production, and that the states involved in force production must have Pi attached. With sinusoidal analysis, we obtained the kinetic constants around the Pi release steps, established a CB scheme, and evaluated force generated (and supported) by each CB state. Our results demonstrate that force is generated before Pi is released, and the same force is maintained after Pi is released. Stretch activation and/or delayed tension can also be explained with this CB scheme and forms the basis of force generation and oscillatory work production.
Collapse
Affiliation(s)
- Masataka Kawai
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, 52242, USA.
| | - Bogdan Iorga
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Bucharest, Romania
- Department of Molecular and Cell Physiology, Hannover Medical School, 30625, Hannover, Germany
| |
Collapse
|
10
|
Rynkiewicz MJ, Childers MC, Karpicheva OE, Regnier M, Geeves MA, Lehman W. Myosin's powerstroke transitions define atomic scale movement of cardiac thin filament tropomyosin. J Gen Physiol 2024; 156:e202413538. [PMID: 38607351 PMCID: PMC11010328 DOI: 10.1085/jgp.202413538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/27/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
Dynamic interactions between the myosin motor head on thick filaments and the actin molecular track on thin filaments drive the myosin-crossbridge cycle that powers muscle contraction. The process is initiated by Ca2+ and the opening of troponin-tropomyosin-blocked myosin-binding sites on actin. The ensuing recruitment of myosin heads and their transformation from pre-powerstroke to post-powerstroke conformation on actin produce the force required for contraction. Cryo-EM-based atomic models confirm that during this process, tropomyosin occupies three different average positions on actin. Tropomyosin pivoting on actin away from a TnI-imposed myosin-blocking position accounts for part of the Ca2+ activation observed. However, the structure of tropomyosin on thin filaments that follows pre-powerstroke myosin binding and its translocation during myosin's pre-powerstroke to post-powerstroke transition remains unresolved. Here, we approach this transition computationally in silico. We used the myosin helix-loop-helix motif as an anchor to dock models of pre-powerstroke cardiac myosin to the cleft between neighboring actin subunits along cardiac thin filaments. We then performed targeted molecular dynamics simulations of the transition between pre- and post-powerstroke conformations on actin in the presence of cardiac troponin-tropomyosin. These simulations show Arg 369 and Glu 370 on the tip of myosin Loop-4 encountering identically charged residues on tropomyosin. The charge repulsion between residues causes tropomyosin translocation across actin, thus accounting for the final regulatory step in the activation of the thin filament, and, in turn, facilitating myosin movement along the filament. We suggest that during muscle activity, myosin-induced tropomyosin movement is likely to result in unencumbered myosin head interactions on actin at low-energy cost.
Collapse
Affiliation(s)
- Michael J. Rynkiewicz
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | | | - Olga E. Karpicheva
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - William Lehman
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
11
|
Tan X, Qu S, Wang G, Zhang G, Liu T, Ling F, Wang G. Structure-based discovery of potent myosin inhibitors to guide antiparasite drug development. Eur J Med Chem 2024; 269:116338. [PMID: 38522112 DOI: 10.1016/j.ejmech.2024.116338] [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: 12/07/2023] [Revised: 02/27/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Monogenea, a prevalent parasite in aquaculture, poses significant threats to the industry, leading to substantial losses. Current preventive measures have proven insufficient, necessitating the development of novel and effective anti-parasitic drugs. In this investigation, we obtained the full-length myosin cDNA sequence by analyzing three-generation transcriptome data, revealing a 5817-base sequence encoding 1938 amino acids. Subsequently, we modeled and analyzed the characteristics of the secondary and tertiary of myosin, pinpointing the crucial functional region within the motor domain (amino acids 1-768). The prokaryotic expression of this domain yielded a protein of 87.44 kDa, confirmed as myosin by Western Blotting. Molecular docking identified ASN439 as the key amino acid residue involved in arctigenin and myosin binding, a result corroborated by site-directed mutagenesis, affirming the active cavity of this interaction. Chalcone and shikonin were chosen from a virtual sieve of molecular library of natural drugs based on the active cavity. Chalcone and shikonin exhibited EC50 values of 1.085 mg/L and 0.371 mg/L, respectively, with corresponding IC50 values for myosin of 0.44 mM and 0.14 mM. Given its superior activity and structure, shikonin was selected for further optimization of drug molecule design, culminating in the discovery of 1,4-naphthoquinone as a potent antiparasitic agent. This compound demonstrated an EC50 of 0.047 mg/L, LC50 of 0.23 mg/L, and a TI index of 4.893. These findings collectively highlight the potential of shikonin and 1,4-naphthoquinone as alternative compounds to control Gyrodactylus infections. Further optimization of medicinal chemistry holds promise for the development of more potent 1,4-naphthoquinone analogues, offering prospects for future anthelmintic control through combinatorial or replacement strategies.
Collapse
Affiliation(s)
- Xiaoping Tan
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Shenye Qu
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Guangshuo Wang
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Gengrong Zhang
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Tianqiang Liu
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Fei Ling
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China.
| | - Gaoxue Wang
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
12
|
Li M, Hu Y, Wang Q. Exploring the Super-Relaxed State of Human Cardiac β-Myosin by Molecular Dynamics Simulations. J Phys Chem B 2024; 128:3113-3120. [PMID: 38516963 DOI: 10.1021/acs.jpcb.3c07956] [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: 03/23/2024]
Abstract
Human β-cardiac myosin plays a critical role in generating the mechanical forces necessary for cardiac muscle contraction. This process relies on a delicate dynamic equilibrium between the disordered relaxed state (DRX) and the super-relaxed state (SRX) of myosin. Disruptions in this equilibrium due to mutations can lead to heart diseases. However, the structural characteristics of SRX and the molecular mechanisms underlying pathogenic mutations have remained elusive. To bridge this gap, we conducted molecular dynamics simulations and free energy calculations to explore the conformational changes in myosin. Our findings indicate that the size of the phosphate-binding pocket can serve as a valuable metric for characterizing the transition from the DRX to SRX state. Importantly, we established a global dynamic coupling network within the myosin motor head at the residue level, elucidating how the pathogenic mutation E483K impacts the equilibrium between SRX and DRX through allosteric effects. Our work illuminates molecular details of SRX and offers valuable insights into disease treatment through the regulation of SRX.
Collapse
Affiliation(s)
- Mingwei Li
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yao Hu
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Qian Wang
- Department of Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
13
|
Niu F, Li L, Wang L, Xiao J, Xu S, Liu Y, Lin L, Yu C, Wei Z. Autoinhibition and activation of myosin VI revealed by its cryo-EM structure. Nat Commun 2024; 15:1187. [PMID: 38331992 PMCID: PMC10853514 DOI: 10.1038/s41467-024-45424-7] [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: 09/15/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Myosin VI is the only molecular motor that moves towards the minus end along actin filaments. Numerous cellular processes require myosin VI and tight regulations of the motor's activity. Defects in myosin VI activity are known to cause genetic diseases such as deafness and cardiomyopathy. However, the molecular mechanisms underlying the activity regulation of myosin VI remain elusive. Here, we determined the high-resolution cryo-electron microscopic structure of myosin VI in its autoinhibited state. Our structure reveals that autoinhibited myosin VI adopts a compact, monomeric conformation via extensive interactions between the head and tail domains, orchestrated by an elongated single-α-helix region resembling a "spine". This autoinhibited structure effectively blocks cargo binding sites and represses the motor's ATPase activity. Certain cargo adaptors such as GIPC can release multiple inhibitory interactions and promote motor activity, pointing to a cargo-mediated activation of the processive motor. Moreover, our structural findings allow rationalization of disease-associated mutations in myosin VI. Beyond the activity regulation mechanisms of myosin VI, our study also sheds lights on how activities of other myosin motors such as myosin VII and X might be regulated.
Collapse
Affiliation(s)
- Fengfeng Niu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China
| | - Lingxuan Li
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lei Wang
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jinman Xiao
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
| | - Shun Xu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yong Liu
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Leishu Lin
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Cong Yu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Zhiyi Wei
- Department of Neuroscience and Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
14
|
Auguin D, Robert-Paganin J, Réty S, Kikuti C, David A, Theumer G, Schmidt AW, Knölker HJ, Houdusse A. Omecamtiv mecarbil and Mavacamten target the same myosin pocket despite antagonistic effects in heart contraction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.15.567213. [PMID: 38014327 PMCID: PMC10680719 DOI: 10.1101/2023.11.15.567213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Inherited cardiomyopathies are amongst the most common cardiac diseases worldwide, leading in the late-stage to heart failure and death. The most promising treatments against these diseases are small-molecules directly modulating the force produced by β-cardiac myosin, the molecular motor driving heart contraction. Two of these molecules that produce antagonistic effects on cardiac contractility have completed clinical phase 3 trials: the activator Omecamtiv mecarbil and the inhibitor Mavacamten. In this work, we reveal by X-ray crystallography that both drugs target the same pocket and stabilize a pre-stroke structural state, with only few local differences. All atoms molecular dynamics simulations reveal how these molecules can have antagonistic impact on the allostery of the motor by comparing β-cardiac myosin in the apo form or bound to Omecamtiv mecarbil or Mavacamten. Altogether, our results provide the framework for rational drug development for the purpose of personalized medicine.
Collapse
Affiliation(s)
- Daniel Auguin
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258 Paris cedex 05, France
- Laboratoire de Biologie des Ligneux et des Grandes Cultures, Université d'Orléans, UPRES EA 1207, INRAE- USC1328, F-45067 Orléans, France
| | - Julien Robert-Paganin
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258 Paris cedex 05, France
| | - Stéphane Réty
- Laboratoire de Biologie et Modélisation de la Cellule, ENS de Lyon, University Claude Bernard, CNRS UMR 5239, INSERM U1210, 46 Allée d'Italie Site Jacques Monod, F-69007 Lyon, France
| | - Carlos Kikuti
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258 Paris cedex 05, France
| | - Amandine David
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258 Paris cedex 05, France
| | - Gabriele Theumer
- Faculty of Chemistry, TU Dresden, Bergstraße 66, 01069 Dresden, Germany
| | - Arndt W Schmidt
- Faculty of Chemistry, TU Dresden, Bergstraße 66, 01069 Dresden, Germany
| | | | - Anne Houdusse
- Structural Motility, UMR 144 CNRS/Curie Institute, PSL Research University, 26 rue d'Ulm, 75258 Paris cedex 05, France
| |
Collapse
|
15
|
Bodt SML, Ge J, Ma W, Rasicci DV, Desetty R, McCammon JA, Yengo CM. Dilated cardiomyopathy mutation in beta-cardiac myosin enhances actin activation of the power stroke and phosphate release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566646. [PMID: 38014187 PMCID: PMC10680644 DOI: 10.1101/2023.11.10.566646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Inherited mutations in human beta-cardiac myosin (M2β) can lead to severe forms of heart failure. The E525K mutation in M2β is associated with dilated cardiomyopathy (DCM) and was found to stabilize the interacting heads motif (IHM) and autoinhibited super-relaxed (SRX) state in dimeric heavy meromyosin. However, in monomeric M2β subfragment 1 (S1) we found that E525K enhances (3-fold) the maximum steady-state actin-activated ATPase activity (kcat) and decreases (6-fold) the actin concentration at which ATPase is one-half maximal (KATPase). We also found a 3 to 4-fold increase in the actin-activated power stroke and phosphate release rate constants at 30 μM actin, which overall enhanced the duty ratio 3-fold. Loaded motility assays revealed that the enhanced intrinsic motor activity translates to increased ensemble force in M2β S1. Glutamate 525, located near the actin binding region in the so-called activation loop, is highly conserved and predicted to form a salt-bridge with another conserved residue (lysine 484) in the relay helix. Enhanced sampling molecular dynamics simulations predict that the charge reversal mutation disrupts the E525-K484 salt-bridge, inducing conformations with a more flexible relay helix and a wide phosphate release tunnel. Our results highlight a highly conserved allosteric pathway associated with actin activation of the power stroke and phosphate release and suggest an important feature of the autoinhibited IHM is to prevent this region of myosin from interacting with actin. The ability of the E525K mutation to stabilize the IHM likely overrides the enhanced intrinsic motor properties, which may be key to triggering DCM pathogenesis.
Collapse
Affiliation(s)
- Skylar M. L. Bodt
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jinghua Ge
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Wen Ma
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California
| | - David V. Rasicci
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
16
|
Månsson A, Ušaj M, Moretto L, Matusovsky O, Velayuthan LP, Friedman R, Rassier DE. New paradigms in actomyosin energy transduction: Critical evaluation of non-traditional models for orthophosphate release. Bioessays 2023; 45:e2300040. [PMID: 37366639 DOI: 10.1002/bies.202300040] [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: 02/27/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023]
Abstract
Release of the ATP hydrolysis product ortophosphate (Pi) from the active site of myosin is central in chemo-mechanical energy transduction and closely associated with the main force-generating structural change, the power-stroke. Despite intense investigations, the relative timing between Pi-release and the power-stroke remains poorly understood. This hampers in depth understanding of force production by myosin in health and disease and our understanding of myosin-active drugs. Since the 1990s and up to today, models that incorporate the Pi-release either distinctly before or after the power-stroke, in unbranched kinetic schemes, have dominated the literature. However, in recent years, alternative models have emerged to explain apparently contradictory findings. Here, we first compare and critically analyze three influential alternative models proposed previously. These are either characterized by a branched kinetic scheme or by partial uncoupling of Pi-release and the power-stroke. Finally, we suggest critical tests of the models aiming for a unified picture.
Collapse
Affiliation(s)
- Alf Månsson
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Marko Ušaj
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Luisa Moretto
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Oleg Matusovsky
- Department of Kinesiology and Physical Education, McGill University, Montreal, Québec, Canada
| | - Lok Priya Velayuthan
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Ran Friedman
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Dilson E Rassier
- Department of Kinesiology and Physical Education, McGill University, Montreal, Québec, Canada
| |
Collapse
|
17
|
Lehman SJ, Meller A, Solieva SO, Lotthammer JM, Greenberg L, Langer SJ, Greenberg MJ, Tardiff JC, Bowman GR, Leinwand L. Divergent Molecular Phenotypes in Point Mutations at the Same Residue in Beta-Myosin Heavy Chain Lead to Distinct Cardiomyopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547580. [PMID: 37461648 PMCID: PMC10349964 DOI: 10.1101/2023.07.03.547580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
In genetic cardiomyopathies, a frequently described phenomenon is how similar mutations in one protein can lead to discrete clinical phenotypes. One example is illustrated by two mutations in beta myosin heavy chain (β-MHC) that are linked to hypertrophic cardiomyopathy (HCM) (Ile467Val, I467V) and left ventricular non-compaction (LVNC) (Ile467Thr, I467T). To investigate how these missense mutations lead to independent diseases, we studied the molecular effects of each mutation using recombinant human β-MHC Subfragment 1 (S1) in in vitro assays. Both HCM-I467V and LVNC-I467T S1 mutations exhibited similar mechanochemical function, including unchanged ATPase and enhanced actin velocity but had opposing effects on the super-relaxed (SRX) state of myosin. HCM-I467V S1 showed a small reduction in the SRX state, shifting myosin to a more actin-available state that may lead to the "gain-of-function" phenotype commonly described in HCM. In contrast, LVNC-I467T significantly increased the population of myosin in the ultra-slow SRX state. Interestingly, molecular dynamics simulations reveal that I467T allosterically disrupts interactions between ADP and the nucleotide-binding pocket, which may result in an increased ADP release rate. This predicted change in ADP release rate may define the enhanced actin velocity measured in LVNC-I467T, but also describe the uncoupled mechanochemical function for this mutation where the enhanced ADP release rate may be sufficient to offset the increased SRX population of myosin. These contrasting molecular effects may lead to contractile dysregulation that initiates LVNC-associated signaling pathways that progress the phenotype. Together, analysis of these mutations provides evidence that phenotypic complexity originates at the molecular level and is critical to understanding disease progression and developing therapies.
Collapse
Affiliation(s)
- Sarah J Lehman
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| | - Artur Meller
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
- Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO, USA
| | - Shahlo O Solieva
- University of Pennsylvania, Department of Biochemistry and Biophysics, Philadelphia, PA, USA
| | - Jeffrey M Lotthammer
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Lina Greenberg
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Stephen J Langer
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| | - Michael J Greenberg
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Jil C Tardiff
- University of Arizona, Department of Biomedical Engineering, Tucson, AZ, USA
| | - Gregory R Bowman
- University of Pennsylvania, Department of Biochemistry and Biophysics, Philadelphia, PA, USA
| | - Leslie Leinwand
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| |
Collapse
|
18
|
Grinzato A, Auguin D, Kikuti C, Nandwani N, Moussaoui D, Pathak D, Kandiah E, Ruppel KM, Spudich JA, Houdusse A, Robert-Paganin J. Cryo-EM structure of the folded-back state of human β-cardiac myosin. Nat Commun 2023; 14:3166. [PMID: 37258552 PMCID: PMC10232470 DOI: 10.1038/s41467-023-38698-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/11/2023] [Indexed: 06/02/2023] Open
Abstract
To save energy and precisely regulate cardiac contractility, cardiac muscle myosin heads are sequestered in an 'off' state that can be converted to an 'on' state when exertion is increased. The 'off' state is equated with a folded-back structure known as the interacting-heads motif (IHM), which is a regulatory feature of all class-2 muscle and non-muscle myosins. We report here the human β-cardiac myosin IHM structure determined by cryo-electron microscopy to 3.6 Å resolution, providing details of all the interfaces stabilizing the 'off' state. The structure shows that these interfaces are hot spots of hypertrophic cardiomyopathy mutations that are thought to cause hypercontractility by destabilizing the 'off' state. Importantly, the cardiac and smooth muscle myosin IHM structures dramatically differ, providing structural evidence for the divergent physiological regulation of these muscle types. The cardiac IHM structure will facilitate development of clinically useful new molecules that modulate IHM stability.
Collapse
Affiliation(s)
- Alessandro Grinzato
- CM01 beamline. European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Daniel Auguin
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France
- Laboratoire de Biologie des Ligneux et des Grandes Cultures, Université d'Orléans, UPRES EA 1207, INRA-USC1328, F-45067, Orléans, France
| | - Carlos Kikuti
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France
| | - Neha Nandwani
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dihia Moussaoui
- BM29 BIOSAXS beamline, European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Divya Pathak
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Eaazhisai Kandiah
- CM01 beamline. European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Kathleen M Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France.
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, F-75005, Paris, France.
| |
Collapse
|
19
|
Marang C, Scott B, Chambers J, Gunther LK, Yengo CM, Debold EP. A mutation in switch I alters the load-dependent kinetics of myosin Va. Nat Commun 2023; 14:3137. [PMID: 37253724 PMCID: PMC10229639 DOI: 10.1038/s41467-023-38535-0] [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: 09/19/2022] [Accepted: 05/05/2023] [Indexed: 06/01/2023] Open
Abstract
Myosin Va is the molecular motor that drives intracellular vesicular transport, powered by the transduction of chemical energy from ATP into mechanical work. The coupling of the powerstroke and phosphate (Pi) release is key to understanding the transduction process, and crucial details of this process remain unclear. Therefore, we determined the effect of elevated Pi on the force-generating capacity of a mini-ensemble of myosin Va S1 (WT) in a laser trap assay. By increasing the stiffness of the laser trap we determined the effect of increasing resistive loads on the rate of Pi-induced detachment from actin, and quantified this effect using the Bell approximation. We observed that WT myosin generated higher forces and larger displacements at the higher laser trap stiffnesses in the presence of 30 mM Pi, but binding event lifetimes decreased dramatically, which is most consistent with the powerstroke preceding the release of Pi from the active site. Repeating these experiments using a construct with a mutation in switch I of the active site (S217A) caused a seven-fold increase in the load-dependence of the Pi-induced detachment rate, suggesting that the S217A region of switch I may help mediate the load-dependence of Pi-rebinding.
Collapse
Affiliation(s)
- Christopher Marang
- Department of Kinesiology, University of Massachusetts, Amherst, MA, 01003, USA
| | - Brent Scott
- Department of Kinesiology, University of Massachusetts, Amherst, MA, 01003, USA
| | - James Chambers
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Laura K Gunther
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Edward P Debold
- Department of Kinesiology, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
20
|
Doran MH, Rynkiewicz MJ, Rasicci D, Bodt SM, Barry ME, Bullitt E, Yengo CM, Moore JR, Lehman W. Conformational changes linked to ADP release from human cardiac myosin bound to actin-tropomyosin. J Gen Physiol 2023; 155:e202213267. [PMID: 36633586 PMCID: PMC9859928 DOI: 10.1085/jgp.202213267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/11/2022] [Accepted: 12/14/2022] [Indexed: 01/13/2023] Open
Abstract
Following binding to the thin filament, β-cardiac myosin couples ATP-hydrolysis to conformational rearrangements in the myosin motor that drive myofilament sliding and cardiac ventricular contraction. However, key features of the cardiac-specific actin-myosin interaction remain uncertain, including the structural effect of ADP release from myosin, which is rate-limiting during force generation. In fact, ADP release slows under experimental load or in the intact heart due to the afterload, thereby adjusting cardiac muscle power output to meet physiological demands. To further elucidate the structural basis of this fundamental process, we used a combination of cryo-EM reconstruction methodologies to determine structures of the human cardiac actin-myosin-tropomyosin filament complex at better than 3.4 Å-resolution in the presence and in the absence of Mg2+·ADP. Focused refinements of the myosin motor head and its essential light chains in these reconstructions reveal that small changes in the nucleotide-binding site are coupled to significant rigid body movements of the myosin converter domain and a 16-degree lever arm swing. Our structures provide a mechanistic framework to understand the effect of ADP binding and release on human cardiac β-myosin, and offer insights into the force-sensing mechanism displayed by the cardiac myosin motor.
Collapse
Affiliation(s)
- Matthew H. Doran
- School of Medicine, Department of Physiology and Biophysics, Boston University, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Michael J. Rynkiewicz
- School of Medicine, Department of Physiology and Biophysics, Boston University, Boston, MA, USA
| | - David Rasicci
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Skylar M.L. Bodt
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Meaghan E. Barry
- Department of Biological Science, University of Massachusetts Lowell, Lowell, MA, USA
| | - Esther Bullitt
- School of Medicine, Department of Physiology and Biophysics, Boston University, Boston, MA, USA
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Jeffrey R. Moore
- Department of Biological Science, University of Massachusetts Lowell, Lowell, MA, USA
| | - William Lehman
- School of Medicine, Department of Physiology and Biophysics, Boston University, Boston, MA, USA
| |
Collapse
|
21
|
Ma W, You S, Regnier M, McCammon JA. Integrating comparative modeling and accelerated simulations reveals conformational and energetic basis of actomyosin force generation. Proc Natl Acad Sci U S A 2023; 120:e2215836120. [PMID: 36802417 PMCID: PMC9992861 DOI: 10.1073/pnas.2215836120] [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: 09/23/2022] [Accepted: 01/15/2023] [Indexed: 02/23/2023] Open
Abstract
Muscle contraction is performed by arrays of contractile proteins in the sarcomere. Serious heart diseases, such as cardiomyopathy, can often be results of mutations in myosin and actin. Direct characterization of how small changes in the myosin-actin complex impact its force production remains challenging. Molecular dynamics (MD) simulations, although capable of studying protein structure-function relationships, are limited owing to the slow timescale of the myosin cycle as well as a lack of various intermediate structures for the actomyosin complex. Here, employing comparative modeling and enhanced sampling MD simulations, we show how the human cardiac myosin generates force during the mechanochemical cycle. Initial conformational ensembles for different myosin-actin states are learned from multiple structural templates with Rosetta. This enables us to efficiently sample the energy landscape of the system using Gaussian accelerated MD. Key myosin loop residues, whose substitutions are related to cardiomyopathy, are identified to form stable or metastable interactions with the actin surface. We find that the actin-binding cleft closure is allosterically coupled to the myosin motor core transitions and ATP-hydrolysis product release from the active site. Furthermore, a gate between switch I and switch II is suggested to control phosphate release at the prepowerstroke state. Our approach demonstrates the ability to link sequence and structural information to motor functions.
Collapse
Affiliation(s)
- Wen Ma
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA92093
| | - Shengjun You
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA98109
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA92093
- Department of Pharmacology, University of California, San Diego, La Jolla, CA92093
| |
Collapse
|
22
|
Lewis CTA, Ochala J. Myosin Heavy Chain as a Novel Key Modulator of Striated Muscle Resting State. Physiology (Bethesda) 2023; 38:0. [PMID: 36067133 DOI: 10.1152/physiol.00018.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
After years of intense research using structural, biological, and biochemical experimental procedures, it is clear that myosin molecules are essential for striated muscle contraction. However, this is just the tip of the iceberg of their function. Interestingly, it has been shown recently that these molecules (especially myosin heavy chains) are also crucial for cardiac and skeletal muscle resting state. In the present review, we first overview myosin heavy chain biochemical states and how they influence the consumption of ATP. We then detail how neighboring partner proteins including myosin light chains and myosin binding protein C intervene in such processes, modulating the ATP demand in health and disease. Finally, we present current experimental drugs targeting myosin ATP consumption and how they can treat muscle diseases.
Collapse
Affiliation(s)
| | - Julien Ochala
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Lee LA, Barrick SK, Meller A, Walklate J, Lotthammer JM, Tay JW, Stump WT, Bowman G, Geeves MA, Greenberg MJ, Leinwand LA. Functional divergence of the sarcomeric myosin, MYH7b, supports species-specific biological roles. J Biol Chem 2022; 299:102657. [PMID: 36334627 PMCID: PMC9800208 DOI: 10.1016/j.jbc.2022.102657] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/14/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Myosin heavy chain 7b (MYH7b) is an evolutionarily ancient member of the sarcomeric myosin family, which typically supports striated muscle function. However, in mammals, alternative splicing prevents MYH7b protein production in cardiac and most skeletal muscles and limits expression to a subset of specialized muscles and certain nonmuscle environments. In contrast, MYH7b protein is abundant in python cardiac and skeletal muscles. Although the MYH7b expression pattern diverges in mammals versus reptiles, MYH7b shares high sequence identity across species. So, it remains unclear how mammalian MYH7b function may differ from that of other sarcomeric myosins and whether human and python MYH7b motor functions diverge as their expression patterns suggest. Thus, we generated recombinant human and python MYH7b protein and measured their motor properties to investigate any species-specific differences in activity. Our results reveal that despite having similar working strokes, the MYH7b isoforms have slower actin-activated ATPase cycles and actin sliding velocities than human cardiac β-MyHC. Furthermore, python MYH7b is tuned to have slower motor activity than human MYH7b because of slower kinetics of the chemomechanical cycle. We found that the MYH7b isoforms adopt a higher proportion of myosin heads in the ultraslow, super-relaxed state compared with human cardiac β-MyHC. These findings are supported by molecular dynamics simulations that predict MYH7b preferentially occupies myosin active site conformations similar to those observed in the structurally inactive state. Together, these results suggest that MYH7b is specialized for slow and energy-conserving motor activity and that differential tuning of MYH7b orthologs contributes to species-specific biological roles.
Collapse
Affiliation(s)
- Lindsey A. Lee
- Molecular, Cellular, and Developmental Biology Department, Boulder, Colorado, USA,BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
| | - Samantha K. Barrick
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Artur Meller
- The Center for Science and Engineering of Living Systems, Washington University in St Louis, St Louis, Missouri, USA
| | - Jonathan Walklate
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Jeffrey M. Lotthammer
- The Center for Science and Engineering of Living Systems, Washington University in St Louis, St Louis, Missouri, USA
| | - Jian Wei Tay
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
| | - W. Tom Stump
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Gregory Bowman
- The Center for Science and Engineering of Living Systems, Washington University in St Louis, St Louis, Missouri, USA,Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael A. Geeves
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Leslie A. Leinwand
- Molecular, Cellular, and Developmental Biology Department, Boulder, Colorado, USA,BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA,For correspondence: Leslie A. Leinwand
| |
Collapse
|
24
|
Månsson A, Rassier DE. Insights into Muscle Contraction Derived from the Effects of Small-Molecular Actomyosin-Modulating Compounds. Int J Mol Sci 2022; 23:ijms232012084. [PMID: 36292937 PMCID: PMC9603234 DOI: 10.3390/ijms232012084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/25/2022] [Accepted: 10/03/2022] [Indexed: 01/10/2023] Open
Abstract
Bottom-up mechanokinetic models predict ensemble function of actin and myosin based on parameter values derived from studies using isolated proteins. To be generally useful, e.g., to analyze disease effects, such models must also be able to predict ensemble function when actomyosin interaction kinetics are modified differently from normal. Here, we test this capability for a model recently shown to predict several physiological phenomena along with the effects of the small molecular compound blebbistatin. We demonstrate that this model also qualitatively predicts effects of other well-characterized drugs as well as varied concentrations of MgATP. However, the effects of one compound, amrinone, are not well accounted for quantitatively. We therefore systematically varied key model parameters to address this issue, leading to the increased amplitude of the second sub-stroke of the power stroke from 1 nm to 2.2 nm, an unchanged first sub-stroke (5.3−5.5 nm), and an effective cross-bridge attachment rate that more than doubled. In addition to better accounting for the effects of amrinone, the modified model also accounts well for normal physiological ensemble function. Moreover, a Monte Carlo simulation-based version of the model was used to evaluate force−velocity data from small myosin ensembles. We discuss our findings in relation to key aspects of actin−myosin operation mechanisms causing a non-hyperbolic shape of the force−velocity relationship at high loads. We also discuss remaining limitations of the model, including uncertainty of whether the cross-bridge elasticity is linear or not, the capability to account for contractile properties of very small actomyosin ensembles (<20 myosin heads), and the mechanism for requirements of a higher cross-bridge attachment rate during shortening compared to during isometric contraction.
Collapse
Affiliation(s)
- Alf Månsson
- Department of Chemistry and Biomedical Sciences, Linnaeus University, 391 82 Kalmar, Sweden
- Correspondence: ; Tel.: +46-708-866243
| | - Dilson E. Rassier
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC H2W 1S4, Canada
| |
Collapse
|
25
|
Kozlova MI, Shalaeva DN, Dibrova DV, Mulkidjanian AY. Common Mechanism of Activated Catalysis in P-loop Fold Nucleoside Triphosphatases-United in Diversity. Biomolecules 2022; 12:1346. [PMID: 36291556 PMCID: PMC9599734 DOI: 10.3390/biom12101346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/20/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
To clarify the obscure hydrolysis mechanism of ubiquitous P-loop-fold nucleoside triphosphatases (Walker NTPases), we analysed the structures of 3136 catalytic sites with bound Mg-NTP complexes or their analogues. Our results are presented in two articles; here, in the second of them, we elucidated whether the Walker A and Walker B sequence motifs-common to all P-loop NTPases-could be directly involved in catalysis. We found that the hydrogen bonds (H-bonds) between the strictly conserved, Mg-coordinating Ser/Thr of the Walker A motif ([Ser/Thr]WA) and aspartate of the Walker B motif (AspWB) are particularly short (even as short as 2.4 ångströms) in the structures with bound transition state (TS) analogues. Given that a short H-bond implies parity in the pKa values of the H-bond partners, we suggest that, in response to the interactions of a P-loop NTPase with its cognate activating partner, a proton relocates from [Ser/Thr]WA to AspWB. The resulting anionic [Ser/Thr]WA alkoxide withdraws a proton from the catalytic water molecule, and the nascent hydroxyl attacks the gamma phosphate of NTP. When the gamma-phosphate breaks away, the trapped proton at AspWB passes by the Grotthuss relay via [Ser/Thr]WA to beta-phosphate and compensates for its developing negative charge that is thought to be responsible for the activation barrier of hydrolysis.
Collapse
Affiliation(s)
- Maria I. Kozlova
- School of Physics, Osnabrueck University, D-49069 Osnabrueck, Germany
| | - Daria N. Shalaeva
- School of Physics, Osnabrueck University, D-49069 Osnabrueck, Germany
| | - Daria V. Dibrova
- School of Physics, Osnabrueck University, D-49069 Osnabrueck, Germany
| | - Armen Y. Mulkidjanian
- School of Physics, Osnabrueck University, D-49069 Osnabrueck, Germany
- Center of Cellular Nanoanalytics, Osnabrueck University, D-49069 Osnabrueck, Germany
| |
Collapse
|
26
|
Multistep orthophosphate release tunes actomyosin energy transduction. Nat Commun 2022; 13:4575. [PMID: 35931685 PMCID: PMC9356070 DOI: 10.1038/s41467-022-32110-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 07/13/2022] [Indexed: 11/29/2022] Open
Abstract
Muscle contraction and a range of critical cellular functions rely on force-producing interactions between myosin motors and actin filaments, powered by turnover of adenosine triphosphate (ATP). The relationship between release of the ATP hydrolysis product ortophosphate (Pi) from the myosin active site and the force-generating structural change, the power-stroke, remains enigmatic despite its central role in energy transduction. Here, we present a model with multistep Pi-release that unifies current conflicting views while also revealing additional complexities of potential functional importance. The model is based on our evidence from kinetics, molecular modelling and single molecule fluorescence studies of Pi binding outside the active site. It is also consistent with high-speed atomic force microscopy movies of single myosin II molecules without Pi at the active site, showing consecutive snapshots of pre- and post-power stroke conformations. In addition to revealing critical features of energy transduction by actomyosin, the results suggest enzymatic mechanisms of potentially general relevance. Release of the ATP hydrolysis product orthophosphate (Pi) from the myosin active site is central in force generation but is poorly understood. Here, Moretto et al. present evidence for multistep Pi-release reconciling apparently contradictory results.
Collapse
|
27
|
Yang Z, Chen J, Li H, Lin Y. Genotype-Phenotype Associations with Restrictive Cardiomyopathy Induced by Pathogenic Genetic Mutations. Rev Cardiovasc Med 2022; 23:185. [PMID: 39077162 PMCID: PMC11273878 DOI: 10.31083/j.rcm2306185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 07/31/2024] Open
Abstract
Restrictive cardiomyopathy (RCM) is an uncommon cardiac muscle disease characterized by impaired ventricular filling and severe diastolic dysfunction with or without systolic dysfunction. The patients with RCM present poor prognosis and high prevalence of sudden cardiac death, especially in the young. The etiology of RCM may be idiopathic, familial or acquired predispositions from various systemic diseases. The genetic background of familial RCM is often caused by mutations in genes encoding proteins of sarcomeres and a significant minority by mutations in non-sarcomeric proteins and transthyretin proteins. It is important to identify the associations between genotype and phenotype to guide clinical diagnosis and treatment. Here, we have summarized the reported index cases with RCM involving genetic etiology to date and highlighted the most significant phenotype results.
Collapse
Affiliation(s)
- Zhe Yang
- The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710 Dongguan, Guangdong, China
- Department of Endocrinology and Metabolism, Zhuhai Hospital Affiliated to Jinan University; The First Hospital Affiliated to Medical College of Macao University of Science and Technology, 519000 Zhuhai, Guangdong, China
| | - Jia Chen
- The Second Department of Cardiology, The Second People's Hospital of Guangdong Province, 510310 Guangzhou, Guangdong, China
| | - Hong Li
- The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710 Dongguan, Guangdong, China
| | - Yubi Lin
- The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710 Dongguan, Guangdong, China
| |
Collapse
|
28
|
Debold EP. Mini‐ review: Recent insights into the relative timing of myosin’s powerstroke and release of phosphate. Cytoskeleton (Hoboken) 2022; 78:448-458. [DOI: 10.1002/cm.21695] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Edward P. Debold
- Department of Kinesiology University of Massachusetts Amherst Massachusetts
| |
Collapse
|
29
|
Pepper I, Galkin VE. Actomyosin Complex. Subcell Biochem 2022; 99:421-470. [PMID: 36151385 PMCID: PMC9710302 DOI: 10.1007/978-3-031-00793-4_14] [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] [Indexed: 01/03/2023]
Abstract
Formation of cross-bridges between actin and myosin occurs ubiquitously in eukaryotic cells and mediates muscle contraction, intracellular cargo transport, and cytoskeletal remodeling. Myosin motors repeatedly bind to and dissociate from actin filaments in a cycle that transduces the chemical energy from ATP hydrolysis into mechanical force generation. While the general layout of surface elements within the actin-binding interface is conserved among myosin classes, sequence divergence within these motifs alters the specific contacts involved in the actomyosin interaction as well as the kinetics of mechanochemical cycle phases. Additionally, diverse lever arm structures influence the motility and force production of myosin molecules during their actin interactions. The structural differences generated by myosin's molecular evolution have fine-tuned the kinetics of its isoforms and adapted them for their individual cellular roles. In this chapter, we will characterize the structural and biochemical basis of the actin-myosin interaction and explain its relationship with myosin's cellular roles, with emphasis on the structural variation among myosin isoforms that enables their functional specialization. We will also discuss the impact of accessory proteins, such as the troponin-tropomyosin complex and myosin-binding protein C, on the formation and regulation of actomyosin cross-bridges.
Collapse
Affiliation(s)
- Ian Pepper
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
30
|
Heissler SM, Arora AS, Billington N, Sellers JR, Chinthalapudi K. Cryo-EM structure of the autoinhibited state of myosin-2. SCIENCE ADVANCES 2021; 7:eabk3273. [PMID: 34936462 PMCID: PMC8694606 DOI: 10.1126/sciadv.abk3273] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/05/2021] [Indexed: 05/20/2023]
Abstract
We solved the near-atomic resolution structure of smooth muscle myosin-2 in the autoinhibited state (10S) using single-particle cryo–electron microscopy. The 3.4-Å structure reveals the precise molecular architecture of 10S and the structural basis for myosin-2 regulation. We reveal the position of the phosphorylation sites that control myosin autoinhibition and activation by phosphorylation of the regulatory light chain. Further, we present a previously unidentified conformational state in myosin-2 that traps ADP and Pi produced by the hydrolysis of ATP in the active site. This noncanonical state represents a branch of the myosin enzyme cycle and explains the autoinhibition of the enzyme function of 10S along with its reduced affinity for actin. Together, our structure defines the molecular mechanisms that drive 10S formation, stabilization, and relief by phosphorylation of the regulatory light chain.
Collapse
Affiliation(s)
- Sarah M. Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Amandeep S. Arora
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Neil Billington
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - James R. Sellers
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
31
|
Doran MH, Lehman W. The Central Role of the F-Actin Surface in Myosin Force Generation. BIOLOGY 2021; 10:1221. [PMID: 34943138 PMCID: PMC8698748 DOI: 10.3390/biology10121221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 11/17/2022]
Abstract
Actin is one of the most abundant and versatile proteins in eukaryotic cells. As discussed in many contributions to this Special Issue, its transition from a monomeric G-actin to a filamentous F-actin form plays a critical role in a variety of cellular processes, including control of cell shape and cell motility. Once polymerized from G-actin, F-actin forms the central core of muscle-thin filaments and acts as molecular tracks for myosin-based motor activity. The ATP-dependent cross-bridge cycle of myosin attachment and detachment drives the sliding of myosin thick filaments past thin filaments in muscle and the translocation of cargo in somatic cells. The variation in actin function is dependent on the variation in muscle and non-muscle myosin isoform behavior as well as interactions with a plethora of additional actin-binding proteins. Extensive work has been devoted to defining the kinetics of actin-based force generation powered by the ATPase activity of myosin. In addition, over the past decade, cryo-electron microscopy has revealed the atomic-evel details of the binding of myosin isoforms on the F-actin surface. Most accounts of the structural interactions between myosin and actin are described from the perspective of the myosin molecule. Here, we discuss myosin-binding to actin as viewed from the actin surface. We then describe conserved structural features of actin required for the binding of all or most myosin isoforms while also noting specific interactions unique to myosin isoforms.
Collapse
Affiliation(s)
- Matthew H. Doran
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| | - William Lehman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
32
|
Pospich S, Sweeney HL, Houdusse A, Raunser S. High-resolution structures of the actomyosin-V complex in three nucleotide states provide insights into the force generation mechanism. eLife 2021; 10:e73724. [PMID: 34812732 PMCID: PMC8735999 DOI: 10.7554/elife.73724] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
The molecular motor myosin undergoes a series of major structural transitions during its force-producing motor cycle. The underlying mechanism and its coupling to ATP hydrolysis and actin binding are only partially understood, mostly due to sparse structural data on actin-bound states of myosin. Here, we report 26 high-resolution cryo-EM structures of the actomyosin-V complex in the strong-ADP, rigor, and a previously unseen post-rigor transition state that binds the ATP analog AppNHp. The structures reveal a high flexibility of myosin in each state and provide valuable insights into the structural transitions of myosin-V upon ADP release and binding of AppNHp, as well as the actomyosin interface. In addition, they show how myosin is able to specifically alter the structure of F-actin.
Collapse
Affiliation(s)
- Sabrina Pospich
- Department of Structural Biochemistry, Max Planck Institute of Molecular PhysiologyDortmundGermany
| | - H Lee Sweeney
- Department of Pharmacology and Therapeutics and the Myology Institute, University of FloridaGainesvilleUnited States
| | - Anne Houdusse
- Structural Motility, Institut Curie, Centre National de la Recherche ScientifiqueParisFrance
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular PhysiologyDortmundGermany
| |
Collapse
|
33
|
Barrick SK, Greenberg MJ. Cardiac myosin contraction and mechanotransduction in health and disease. J Biol Chem 2021; 297:101297. [PMID: 34634306 PMCID: PMC8559575 DOI: 10.1016/j.jbc.2021.101297] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
Cardiac myosin is the molecular motor that powers heart contraction by converting chemical energy from ATP hydrolysis into mechanical force. The power output of the heart is tightly regulated to meet the physiological needs of the body. Recent multiscale studies spanning from molecules to tissues have revealed complex regulatory mechanisms that fine-tune cardiac contraction, in which myosin not only generates power output but also plays an active role in its regulation. Thus, myosin is both shaped by and actively involved in shaping its mechanical environment. Moreover, these studies have shown that cardiac myosin-generated tension affects physiological processes beyond muscle contraction. Here, we review these novel regulatory mechanisms, as well as the roles that myosin-based force generation and mechanotransduction play in development and disease. We describe how key intra- and intermolecular interactions contribute to the regulation of myosin-based contractility and the role of mechanical forces in tuning myosin function. We also discuss the emergence of cardiac myosin as a drug target for diseases including heart failure, leading to the discovery of therapeutics that directly tune myosin contractility. Finally, we highlight some of the outstanding questions that must be addressed to better understand myosin's functions and regulation, and we discuss prospects for translating these discoveries into precision medicine therapeutics targeting contractility and mechanotransduction.
Collapse
Affiliation(s)
- Samantha K Barrick
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
34
|
Malär AA, Wili N, Völker LA, Kozlova MI, Cadalbert R, Däpp A, Weber ME, Zehnder J, Jeschke G, Eckert H, Böckmann A, Klose D, Mulkidjanian AY, Meier BH, Wiegand T. Spectroscopic glimpses of the transition state of ATP hydrolysis trapped in a bacterial DnaB helicase. Nat Commun 2021; 12:5293. [PMID: 34489448 PMCID: PMC8421360 DOI: 10.1038/s41467-021-25599-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
The ATP hydrolysis transition state of motor proteins is a weakly populated protein state that can be stabilized and investigated by replacing ATP with chemical mimics. We present atomic-level structural and dynamic insights on a state created by ADP aluminum fluoride binding to the bacterial DnaB helicase from Helicobacter pylori. We determined the positioning of the metal ion cofactor within the active site using electron paramagnetic resonance, and identified the protein protons coordinating to the phosphate groups of ADP and DNA using proton-detected 31P,1H solid-state nuclear magnetic resonance spectroscopy at fast magic-angle spinning > 100 kHz, as well as temperature-dependent proton chemical-shift values to prove their engagements in hydrogen bonds. 19F and 27Al MAS NMR spectra reveal a highly mobile, fast-rotating aluminum fluoride unit pointing to the capture of a late ATP hydrolysis transition state in which the phosphoryl unit is already detached from the arginine and lysine fingers.
Collapse
Affiliation(s)
| | - Nino Wili
- Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | | | - Maria I Kozlova
- Department of Physics, Osnabrück University, Osnabrück, Germany
| | | | | | | | | | | | - Hellmut Eckert
- Institut für Physikalische Chemie, WWU Münster, Münster, Germany
- Instituto de Física de Sao Carlos, Universidade de Sao Paulo, Sao Carlos, SP, Brazil
| | - Anja Böckmann
- Molecular Microbiology and Structural Biochemistry UMR 5086 CNRS/Université de Lyon, Lyon, France
| | - Daniel Klose
- Physical Chemistry, ETH Zürich, Zürich, Switzerland.
| | - Armen Y Mulkidjanian
- Department of Physics, Osnabrück University, Osnabrück, Germany.
- School of Bioengineering and Bioinformatics and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.
| | - Beat H Meier
- Physical Chemistry, ETH Zürich, Zürich, Switzerland.
| | - Thomas Wiegand
- Physical Chemistry, ETH Zürich, Zürich, Switzerland.
- Max-Planck-Institute for Chemical Energy Conversion, Mülheim an der Ruhr, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen, Aachen, Germany.
| |
Collapse
|
35
|
Scott B, Marang C, Woodward M, Debold EP. Myosin's powerstroke occurs prior to the release of phosphate from the active site. Cytoskeleton (Hoboken) 2021; 78:185-198. [PMID: 34331410 DOI: 10.1002/cm.21682] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 02/06/2023]
Abstract
Myosins are a family of motor proteins responsible for various forms of cellular motility, including muscle contraction and vesicular transport. The most fundamental aspect of myosin is its ability to transduce the chemical energy from the hydrolysis of ATP into mechanical work, in the form of force and/or motion. A key unanswered question of the transduction process is the timing of the force-generating powerstroke relative to the release of phosphate (Pi ) from the active site. We examined the ability of single-headed myosin Va to generate a powerstroke in a single molecule laser trap assay while maintaining Pi in its active site, by either elevating Pi in solution or by introducing a mutation in myosin's active site (S217A) to slow Pi -release from the active site. Upon binding to the actin filament, WT myosin generated a powerstoke rapidly (≥500 s-1 ) and without a detectable delay, both in the absence and presence of 30 mM Pi . The elevated levels of Pi did, however, affect event lifetime, eliminating the longest 25% of binding events, confirming that Pi rebound to myosin's active site and accelerated detachment. The S217A construct also generated a powerstroke similar in size and rate upon binding to actin despite the slower Pi release rate. These findings provide direct evidence that myosin Va generates a powerstroke with Pi still in its active site. Therefore, the findings are most consistent with a model in which the powerstroke occurs prior to the release of Pi from the active site.
Collapse
Affiliation(s)
- Brent Scott
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Christopher Marang
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Mike Woodward
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Edward P Debold
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
36
|
Frohne A, Koenighofer M, Liu DT, Laccone F, Neesen J, Gstoettner W, Schoefer C, Lucas T, Frei K, Parzefall T. High Prevalence of MYO6 Variants in an Austrian Patient Cohort With Autosomal Dominant Hereditary Hearing Loss. Otol Neurotol 2021; 42:e648-e657. [PMID: 33710140 DOI: 10.1097/mao.0000000000003076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Genetic hearing loss (HL) is often monogenic. Whereas more than half of autosomal recessive (AR) cases in Austria are caused by mutations in a single gene, no disproportionately frequent contributing genetic factor has been identified in cases of autosomal dominant (AD) HL. The genetic characterization of HL continues to improve diagnosis, genetic counseling, and lays a foundation for the development of personalized medicine approaches. METHODS Diagnostic HL panel screening was performed in an Austrian multiplex family with AD HL, and segregation was tested with polymerase chain reaction and Sanger sequencing. In an independent approach, 18 unrelated patients with AD HL were screened for causative variants in all known HL genes to date and segregation was tested if additional family members were available. The pathogenicity of novel variants was assessed based on previous literature and bioinformatic tools such as prediction software and protein modeling. RESULTS In six of the 19 families under study, candidate pathogenic variants were identified in MYO6, including three novel variants (p.Gln441Pro, p.Ser612Tyr, and p.Gln650ValfsTer7). Some patients carried more than one likely pathogenic variant in known deafness genes. CONCLUSION These results suggest a potential high prevalence of MYO6 variants in Austrian cases of AD HL. The presence of multiple rare HL variants in some patients highlights the relevance of considering multiple-hit diagnoses for genetic counseling and targeted therapy design.
Collapse
Affiliation(s)
- Alexandra Frohne
- Department of Otorhinolaryngology, Head and Neck Surgery
- Department for Cell and Developmental Biology, Center for Anatomy and Cell Biology
| | | | | | - Franco Laccone
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Juergen Neesen
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | | | - Christian Schoefer
- Department for Cell and Developmental Biology, Center for Anatomy and Cell Biology
| | - Trevor Lucas
- Department for Cell and Developmental Biology, Center for Anatomy and Cell Biology
| | - Klemens Frei
- Department of Otorhinolaryngology, Head and Neck Surgery
| | | |
Collapse
|
37
|
A reverse stroke characterizes the force generation of cardiac myofilaments, leading to an understanding of heart function. Proc Natl Acad Sci U S A 2021; 118:2011659118. [PMID: 34088833 DOI: 10.1073/pnas.2011659118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Changes in the molecular properties of cardiac myosin strongly affect the interactions of myosin with actin that result in cardiac contraction and relaxation. However, it remains unclear how myosin molecules work together in cardiac myofilaments and which properties of the individual myosin molecules impact force production to drive cardiac contractility. Here, we measured the force production of cardiac myofilaments using optical tweezers. The measurements revealed that stepwise force generation was associated with a higher frequency of backward steps at lower loads and higher stall forces than those of fast skeletal myofilaments. To understand these unique collective behaviors of cardiac myosin, the dynamic responses of single cardiac and fast skeletal myosin molecules, interacting with actin filaments, were evaluated under load. The cardiac myosin molecules switched among three distinct conformational positions, ranging from pre- to post-power stroke positions, in 1 mM ADP and 0 to 10 mM phosphate solution. In contrast to cardiac myosin, fast skeletal myosin stayed primarily in the post-power stroke position, suggesting that cardiac myosin executes the reverse stroke more frequently than fast skeletal myosin. To elucidate how the reverse stroke affects the force production of myofilaments and possibly heart function, a simulation model was developed that combines the results from the single-molecule and myofilament experiments. The results of this model suggest that the reversal of the cardiac myosin power stroke may be key to characterizing the force output of cardiac myosin ensembles and possibly to facilitating heart contractions.
Collapse
|
38
|
Ward MD, Zimmerman MI, Meller A, Chung M, Swamidass SJ, Bowman GR. Deep learning the structural determinants of protein biochemical properties by comparing structural ensembles with DiffNets. Nat Commun 2021; 12:3023. [PMID: 34021153 PMCID: PMC8140102 DOI: 10.1038/s41467-021-23246-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/16/2021] [Indexed: 12/05/2022] Open
Abstract
Understanding the structural determinants of a protein's biochemical properties, such as activity and stability, is a major challenge in biology and medicine. Comparing computer simulations of protein variants with different biochemical properties is an increasingly powerful means to drive progress. However, success often hinges on dimensionality reduction algorithms for simplifying the complex ensemble of structures each variant adopts. Unfortunately, common algorithms rely on potentially misleading assumptions about what structural features are important, such as emphasizing larger geometric changes over smaller ones. Here we present DiffNets, self-supervised autoencoders that avoid such assumptions, and automatically identify the relevant features, by requiring that the low-dimensional representations they learn are sufficient to predict the biochemical differences between protein variants. For example, DiffNets automatically identify subtle structural signatures that predict the relative stabilities of β-lactamase variants and duty ratios of myosin isoforms. DiffNets should also be applicable to understanding other perturbations, such as ligand binding.
Collapse
Affiliation(s)
- Michael D Ward
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for the Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, USA
| | - Maxwell I Zimmerman
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for the Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, USA
| | - Artur Meller
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for the Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, USA
| | - Moses Chung
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for the Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, USA
| | - S J Swamidass
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory R Bowman
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA.
- Center for the Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
39
|
Cardiomyopathy mutations impact the actin-activated power stroke of human cardiac myosin. Biophys J 2021; 120:2222-2236. [PMID: 33864791 DOI: 10.1016/j.bpj.2021.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/02/2021] [Accepted: 04/01/2021] [Indexed: 11/24/2022] Open
Abstract
Cardiac muscle contraction is driven by the molecular motor myosin, which uses the energy from ATP hydrolysis to generate a power stroke when interacting with actin filaments, although it is unclear how this mechanism is impaired by mutations in myosin that can lead to heart failure. We have applied a fluorescence resonance energy transfer (FRET) strategy to investigate structural changes in the lever arm domain of human β-cardiac myosin subfragment 1 (M2β-S1). We exchanged the human ventricular regulatory light chain labeled at a single cysteine (V105C) with Alexa 488 onto M2β-S1, which served as a donor for Cy3ATP bound to the active site. We monitored the FRET signal during the actin-activated product release steps using transient kinetic measurements. We propose that the fast phase measured with our FRET probes represents the macroscopic rate constant associated with actin-activated rotation of the lever arm during the power stroke in M2β-S1. Our results demonstrated M2β-S1 has a slower actin-activated power stroke compared with fast skeletal muscle myosin and myosin V. Measurements at different temperatures comparing the rate constants of the actin-activated power stroke and phosphate release are consistent with a model in which the power stroke occurs before phosphate release and the two steps are tightly coupled. We suggest that the actin-activated power stroke is highly reversible but followed by a highly irreversible phosphate release step in the absence of load and free phosphate. We demonstrated that hypertrophic cardiomyopathy (R723G)- and dilated cardiomyopathy (F764L)-associated mutations both reduced actin activation of the power stroke in M2β-S1. We also demonstrate that both mutations alter in vitro actin gliding in the presence and absence of load. Thus, examining the structural kinetics of the power stroke in M2β-S1 has revealed critical mutation-associated defects in the myosin ATPase pathway, suggesting these measurements will be extremely important for establishing structure-based mechanisms of contractile dysfunction.
Collapse
|
40
|
Gunther LK, Rohde JA, Tang W, Cirilo JA, Marang CP, Scott BD, Thomas DD, Debold EP, Yengo CM. FRET and optical trapping reveal mechanisms of actin activation of the power stroke and phosphate release in myosin V. J Biol Chem 2021; 295:17383-17397. [PMID: 33453985 DOI: 10.1074/jbc.ra120.015632] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/06/2020] [Indexed: 11/06/2022] Open
Abstract
Myosins generate force and motion by precisely coordinating their mechanical and chemical cycles, but the nature and timing of this coordination remains controversial. We utilized a FRET approach to examine the kinetics of structural changes in the force-generating lever arm in myosin V. We directly compared the FRET results with single-molecule mechanical events examined by optical trapping. We introduced a mutation (S217A) in the conserved switch I region of the active site to examine how myosin couples structural changes in the actin- and nucleotide-binding regions with force generation. Specifically, S217A enhanced the maximum rate of lever arm priming (recovery stroke) while slowing ATP hydrolysis, demonstrating that it uncouples these two steps. We determined that the mutation dramatically slows both actin-induced rotation of the lever arm (power stroke) and phosphate release (≥10-fold), whereas our simulations suggest that the maximum rate of both steps is unchanged by the mutation. Time-resolved FRET revealed that the structure of the pre- and post-power stroke conformations and mole fractions of these conformations were not altered by the mutation. Optical trapping results demonstrated that S217A does not dramatically alter unitary displacements or slow the working stroke rate constant, consistent with the mutation disrupting an actin-induced conformational change prior to the power stroke. We propose that communication between the actin- and nucleotide-binding regions of myosin assures a proper actin-binding interface and active site have formed before producing a power stroke. Variability in this coupling is likely crucial for mediating motor-based functions such as muscle contraction and intracellular transport.
Collapse
Affiliation(s)
- Laura K Gunther
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania, USA
| | - John A Rohde
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Wanjian Tang
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania, USA
| | - Joseph A Cirilo
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania, USA
| | - Christopher P Marang
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Brent D Scott
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Edward P Debold
- Department of Kinesiology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
41
|
Mugnai ML, Thirumalai D. Step-Wise Hydration of Magnesium by Four Water Molecules Precedes Phosphate Release in a Myosin Motor. J Phys Chem B 2021; 125:1107-1117. [PMID: 33481593 DOI: 10.1021/acs.jpcb.0c10004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular motors, such as myosin, kinesin, and dynein, convert the energy released by the hydrolysis of ATP into mechanical work, thus allowing them to undergo directional motion on cytoskeletal tracks. A pivotal step in the chemomechanical transduction in myosin motors occurs after they bind to the actin filament, which triggers the release of phosphate (Pi, product of ATP hydrolysis) and the rotation of the lever arm. Here, we investigate the mechanism of phosphate release in myosin VI using extensive molecular dynamics simulations involving multiple trajectories of several μs. Because the escape of phosphate is expected to occur on time-scales on the order of milliseconds or more in myosin VI, we observed Pi release only if the trajectories were initiated with a rotated phosphate inside the nucleotide binding pocket. We discovered that although Pi populates the traditional "back door" route, phosphate exits through various other gateways, thus establishing the heterogeneity in the escape routes. Remarkably, we observed that the release of phosphate is preceded by a stepwise hydration of the ADP-bound magnesium ion. The release of the anion occurred only after four water molecules hydrated the cation (Mg2+). By performing comparative structural analyses, we show that hydration of magnesium is the key step in the phosphate release in a number of ATPases and GTPases. Nature may have evolved hydration of Mg2+ as a general molecular switch for Pi release, which is a universal step in the catalytic cycle of many machines that share little sequence or structural similarity.
Collapse
Affiliation(s)
- Mauro Lorenzo Mugnai
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - D Thirumalai
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
42
|
Sun S, Karki C, Xie Y, Xian Y, Guo W, Gao BZ, Li L. Hybrid method for representing ions in implicit solvation calculations. Comput Struct Biotechnol J 2021; 19:801-811. [PMID: 33598096 PMCID: PMC7847951 DOI: 10.1016/j.csbj.2021.01.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 12/16/2022] Open
Abstract
Fast and accurate calculations of the electrostatic features of highly charged biomolecules such as DNA, RNA, and highly charged proteins are crucial and challenging tasks. Traditional implicit solvent methods calculate the electrostatic features quickly, but these methods are not able to balance the high net biomolecular charges effectively. Explicit solvent methods add unbalanced ions to neutralize the highly charged biomolecules in molecular dynamic simulations, which require more expensive computing resources. Here we report developing a novel method, Hybridizing Ions Treatment (HIT), which hybridizes the implicit solvent method with an explicit method to realistically calculate the electrostatic potential for highly charged biomolecules. HIT utilizes the ionic distribution from an explicit method to predict the bound ions. The bound ions are then added in the implicit solvent method to perform the electrostatic potential calculations. In this study, two training sets were developed to optimize parameters for HIT. The performance on the testing set demonstrates that HIT significantly improves the electrostatic calculations. Results on molecular motors myosin and kinesin reveal some mechanisms and explain some previous experimental findings. HIT can be widely used to study highly charged biomolecules, including DNA, RNA, molecular motors, and other highly charged biomolecules. The HIT package is available at http://compbio.utep.edu/static/downloads/download_hit.zip.
Collapse
Affiliation(s)
- Shengjie Sun
- Computational Science Program, University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Chitra Karki
- Computational Science Program, University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Yixin Xie
- Computational Science Program, University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Yuejiao Xian
- Department of Chemistry, University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Wenhan Guo
- Computational Science Program, University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| | - Bruce Z Gao
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
| | - Lin Li
- Computational Science Program, University of Texas at El Paso, 500 W University Ave, TX 79968, USA.,Department of Physics, University of Texas at El Paso, 500 W University Ave, TX 79968, USA
| |
Collapse
|
43
|
Walker BC, Walczak CE, Cochran JC. Switch-1 instability at the active site decouples ATP hydrolysis from force generation in myosin II. Cytoskeleton (Hoboken) 2021; 78:3-13. [PMID: 33381891 PMCID: PMC7986744 DOI: 10.1002/cm.21650] [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: 09/10/2020] [Revised: 12/21/2020] [Accepted: 12/28/2020] [Indexed: 11/24/2022]
Abstract
Myosin active site elements (i.e., switch‐1) bind both ATP and a divalent metal to coordinate ATP hydrolysis. ATP hydrolysis at the active site is linked via allosteric communication to the actin polymer binding site and lever arm movement, thus coupling the free energy of ATP hydrolysis to force generation. How active site motifs are functionally linked to actin binding and the power stroke is still poorly understood. We hypothesize that destabilizing switch‐1 movement at the active site will negatively affect the tight coupling of the ATPase catalytic cycle to force production. Using a metal‐switch system, we tested the effect of interfering with switch‐1 coordination of the divalent metal cofactor on force generation. We found that while ATPase activity increased, motility was inhibited. Our results demonstrate that a single atom change that affects the switch‐1 interaction with the divalent metal directly affects actin binding and productive force generation. Even slight modification of the switch‐1 divalent metal coordination can decouple ATP hydrolysis from motility. Switch‐1 movement is therefore critical for both structural communication with the actin binding site, as well as coupling the energy of ATP hydrolysis to force generation.
Collapse
Affiliation(s)
- Benjamin C Walker
- Department of Molecular & Cellular Biochemistry, Indiana University, Bloomington, Indiana, USA
| | - Claire E Walczak
- Medical Sciences, Indiana University School of Medicine-Bloomington, Bloomington, Indiana, USA
| | - Jared C Cochran
- Department of Molecular & Cellular Biochemistry, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
44
|
Franz P, Ewert W, Preller M, Tsiavaliaris G. Unraveling a Force-Generating Allosteric Pathway of Actomyosin Communication Associated with ADP and P i Release. Int J Mol Sci 2020; 22:ijms22010104. [PMID: 33374308 PMCID: PMC7795666 DOI: 10.3390/ijms22010104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 11/16/2022] Open
Abstract
The actomyosin system generates mechanical work with the execution of the power stroke, an ATP-driven, two-step rotational swing of the myosin-neck that occurs post ATP hydrolysis during the transition from weakly to strongly actin-bound myosin states concomitant with Pi release and prior to ADP dissociation. The activating role of actin on product release and force generation is well documented; however, the communication paths associated with weak-to-strong transitions are poorly characterized. With the aid of mutant analyses based on kinetic investigations and simulations, we identified the W-helix as an important hub coupling the structural changes of switch elements during ATP hydrolysis to temporally controlled interactions with actin that are passed to the central transducer and converter. Disturbing the W-helix/transducer pathway increased actin-activated ATP turnover and reduced motor performance as a consequence of prolonged duration of the strongly actin-attached states. Actin-triggered Pi release was accelerated, while ADP release considerably decelerated, both limiting maximum ATPase, thus transforming myosin-2 into a high-duty-ratio motor. This kinetic signature of the mutant allowed us to define the fractional occupancies of intermediate states during the ATPase cycle providing evidence that myosin populates a cleft-closure state of strong actin interaction during the weak-to-strong transition with bound hydrolysis products before accomplishing the power stroke.
Collapse
Affiliation(s)
- Peter Franz
- Cellular Biophysics, Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany;
| | - Wiebke Ewert
- Structural Bioinformatics and Chemical Biology, Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany; (W.E.); (M.P.)
| | - Matthias Preller
- Structural Bioinformatics and Chemical Biology, Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany; (W.E.); (M.P.)
- Department of Natural Sciences, University of Applied Sciences Bonn-Rhein-Sieg, 53757 Sankt Augustin, Germany
| | - Georgios Tsiavaliaris
- Cellular Biophysics, Institute for Biophysical Chemistry, Hannover Medical School, 30625 Hannover, Germany;
- Correspondence:
| |
Collapse
|
45
|
Zheng G, Zhang C, Zhong C. Identification of potential prognostic biomarkers for breast cancer using WGCNA and PPI integrated techniques. Ann Diagn Pathol 2020; 50:151675. [PMID: 33291061 DOI: 10.1016/j.anndiagpath.2020.151675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/26/2020] [Indexed: 12/17/2022]
Abstract
In this study, we aimed to detect promising prognostic factors of breast cancer and interpreted the relevant mechanisms using an integrated bioinformatics analysis. RNA sequencing profile of breast cancer was downloaded from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases, which were combined as a group (TCGA_GTEx). GSE70947 dataset was from Gene Expression Omnibus. Blue and turquoise modules, respectively identified in TCGA_GTEx database and GSE70947 dataset using weighted co-expression network analysis (WGCNA), were both notably associated with breast cancer. By comparing genes in the two significant modules with differentially expressed genes (DEGs), we obtained a set of 40 shared genes, which were mainly enriched in chromosome segregation and mismatch repair pathway. After protein-protein interaction (PPI) network and overall survival analysis, two hub genes EXO1 and KIF4A were extracted from the set of 40 shared genes, which were up-regulated and associated with the dismal outcome of breast cancer patients. There was a notable negative correlation between EXO1 and KIF4A expression and age of breast cancer patients, whereas a positive relationship with two another clinical traits stage and tumor category was detected. Univariate and multivariate Cox regression analysis revealed that the two hub genes could be independent prognostic factors of breast cancer. Mechanistically, gene correlation analysis suggested that EXO1 and KIF4A exerted their oncogenic role via promoting breast cancer cell proliferation. Overall, our findings identify two promising individual prognostic predictors of breast cancer and pave the new way for diagnosis and therapy strategy of breast cancer.
Collapse
Affiliation(s)
- Guili Zheng
- Department of Oncology, No. 960 Hospital of PLA, Jinan, China
| | - Cong Zhang
- Department of Radiation Oncology, No. 960 Hospital of PLA, Jinan, China
| | - Chen Zhong
- Department of Oncology, No. 960 Hospital of PLA, Jinan, China.
| |
Collapse
|
46
|
Hypothesis: Single Actomyosin Properties Account for Ensemble Behavior in Active Muscle Shortening and Isometric Contraction. Int J Mol Sci 2020; 21:ijms21218399. [PMID: 33182367 PMCID: PMC7664901 DOI: 10.3390/ijms21218399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 11/17/2022] Open
Abstract
Muscle contraction results from cyclic interactions between myosin II motors and actin with two sets of proteins organized in overlapping thick and thin filaments, respectively, in a nearly crystalline lattice in a muscle sarcomere. However, a sarcomere contains a huge number of other proteins, some with important roles in muscle contraction. In particular, these include thin filament proteins, troponin and tropomyosin; thick filament proteins, myosin binding protein C; and the elastic protein, titin, that connects the thin and thick filaments. Furthermore, the order and 3D organization of the myofilament lattice may be important per se for contractile function. It is possible to model muscle contraction based on actin and myosin alone with properties derived in studies using single molecules and biochemical solution kinetics. It is also possible to reproduce several features of muscle contraction in experiments using only isolated actin and myosin, arguing against the importance of order and accessory proteins. Therefore, in this paper, it is hypothesized that “single molecule actomyosin properties account for the contractile properties of a half sarcomere during shortening and isometric contraction at almost saturating Ca concentrations”. In this paper, existing evidence for and against this hypothesis is reviewed and new modeling results to support the arguments are presented. Finally, further experimental tests are proposed, which if they corroborate, at least approximately, the hypothesis, should significantly benefit future effective analysis of a range of experimental studies, as well as drug discovery efforts.
Collapse
|
47
|
Ewert W, Franz P, Tsiavaliaris G, Preller M. Structural and Computational Insights into a Blebbistatin-Bound Myosin•ADP Complex with Characteristics of an ADP-Release Conformation along the Two-Step Myosin Power Stoke. Int J Mol Sci 2020; 21:ijms21197417. [PMID: 33049993 PMCID: PMC7582316 DOI: 10.3390/ijms21197417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/06/2020] [Indexed: 01/13/2023] Open
Abstract
The motor protein myosin drives a wide range of cellular and muscular functions by generating directed movement and force, fueled through adenosine triphosphate (ATP) hydrolysis. Release of the hydrolysis product adenosine diphosphate (ADP) is a fundamental and regulatory process during force production. However, details about the molecular mechanism accompanying ADP release are scarce due to the lack of representative structures. Here we solved a novel blebbistatin-bound myosin conformation with critical structural elements in positions between the myosin pre-power stroke and rigor states. ADP in this structure is repositioned towards the surface by the phosphate-sensing P-loop, and stabilized in a partially unbound conformation via a salt-bridge between Arg131 and Glu187. A 5 Å rotation separates the mechanical converter in this conformation from the rigor position. The crystallized myosin structure thus resembles a conformation towards the end of the two-step power stroke, associated with ADP release. Computationally reconstructing ADP release from myosin by means of molecular dynamics simulations further supported the existence of an equivalent conformation along the power stroke that shows the same major characteristics in the myosin motor domain as the resolved blebbistatin-bound myosin-II·ADP crystal structure, and identified a communication hub centered on Arg232 that mediates chemomechanical energy transduction.
Collapse
Affiliation(s)
- Wiebke Ewert
- Institute for Biophysical Chemistry, Structural Bioinformatics and Chemical Biology, Hannover Medical School, 30625 Hannover, Germany;
| | - Peter Franz
- Institute for Biophysical Chemistry, Cellular Biophysics, Hannover Medical School, 30625 Hannover, Germany; (P.F.); (G.T.)
| | - Georgios Tsiavaliaris
- Institute for Biophysical Chemistry, Cellular Biophysics, Hannover Medical School, 30625 Hannover, Germany; (P.F.); (G.T.)
| | - Matthias Preller
- Institute for Biophysical Chemistry, Structural Bioinformatics and Chemical Biology, Hannover Medical School, 30625 Hannover, Germany;
- Department of Natural Sciences, University of Applied Sciences Bonn-Rhein-Sieg, 53359 Rheinbach, Germany
- Correspondence: ; Tel.: +49-511-532-2804
| |
Collapse
|
48
|
Offer G, Ranatunga K. The Location and Rate of the Phosphate Release Step in the Muscle Cross-Bridge Cycle. Biophys J 2020; 119:1501-1512. [DOI: 10.1016/j.bpj.2020.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 11/26/2022] Open
|
49
|
Abstract
Directed movements on actin filaments within the cell are powered by molecular motors of the myosin superfamily. On actin filaments, myosin motors convert the energy from ATP into force and movement. Myosin motors power such diverse cellular functions as cytokinesis, membrane trafficking, organelle movements, and cellular migration. Myosin generates force and movement via a number of structural changes associated with hydrolysis of ATP, binding to actin, and release of the ATP hydrolysis products while bound to actin. Herein we provide an overview of those structural changes and how they relate to the actin-myosin ATPase cycle. These structural changes are the basis of chemo-mechanical transduction by myosin motors.
Collapse
|
50
|
Zhou Y, Zhou XE, Gong Y, Zhu Y, Cao X, Brunzelle JS, Xu HE, Zhou M, Melcher K, Zhang F. Structural basis of Fusarium myosin I inhibition by phenamacril. PLoS Pathog 2020; 16:e1008323. [PMID: 32163521 PMCID: PMC7100991 DOI: 10.1371/journal.ppat.1008323] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/27/2020] [Accepted: 01/16/2020] [Indexed: 11/26/2022] Open
Abstract
Fusarium is a genus of filamentous fungi that includes species that cause devastating diseases in major staple crops, such as wheat, maize, rice, and barley, resulting in severe yield losses and mycotoxin contamination of infected grains. Phenamacril is a novel fungicide that is considered environmentally benign due to its exceptional specificity; it inhibits the ATPase activity of the sole class I myosin of only a subset of Fusarium species including the major plant pathogens F. graminearum, F. asiaticum and F. fujikuroi. To understand the underlying mechanisms of inhibition, species specificity, and resistance mutations, we have determined the crystal structure of phenamacril-bound F. graminearum myosin I. Phenamacril binds in the actin-binding cleft in a new allosteric pocket that contains the central residue of the regulatory Switch 2 loop and that is collapsed in the structure of a myosin with closed actin-binding cleft, suggesting that pocket occupancy blocks cleft closure. We have further identified a single, transferable phenamacril-binding residue found exclusively in phenamacril-sensitive myosins to confer phenamacril selectivity. Phenamacril is a recently identified myosin I inhibitor that is a potent and highly species-specific and myosin subtype-selective fungicide. We report the high-resolution structure of the phenamacril-bound myosin I motor domain of the major crop pathogen Fusarium graminearum, providing insight into the molecular mechanism of phenamacril action and resistance. These results are of broad significance for understanding the mode of actions of myosin-based fungicides and for designing novel myosin I inhibitors for crop protection and for treatment of human myosin dysfunction diseases.
Collapse
Affiliation(s)
- Yuxin Zhou
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
- Center of Cancer and Cell Biology, Program for Structural Biology, Van Andel Institute, Grand Rapids, Michigan, United States of America
| | - X. Edward Zhou
- Center of Cancer and Cell Biology, Program for Structural Biology, Van Andel Institute, Grand Rapids, Michigan, United States of America
| | - Yuanping Gong
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Yuanye Zhu
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Xiaoman Cao
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Joseph S. Brunzelle
- Northwestern University Synchrotron Research Center, Life Sciences Collaborative Access Team, Northwestern University, Argonne, Illinois, United States of America
| | - H. Eric Xu
- Center of Cancer and Cell Biology, Program for Structural Biology, Van Andel Institute, Grand Rapids, Michigan, United States of America
- Center for Structure and Function of Drug Targets, The CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mingguo Zhou
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
- * E-mail: (MZ); (KM); (FZ)
| | - Karsten Melcher
- Center of Cancer and Cell Biology, Program for Structural Biology, Van Andel Institute, Grand Rapids, Michigan, United States of America
- * E-mail: (MZ); (KM); (FZ)
| | - Feng Zhang
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
- * E-mail: (MZ); (KM); (FZ)
| |
Collapse
|