Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
1.
Proc Natl Acad Sci U S A ; 121(13): e2401625121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38507449

ABSTRACT

Molecular motors employ chemical energy to generate unidirectional mechanical output against a track while navigating a chaotic cellular environment, potential disorder on the track, and against Brownian motion. Nevertheless, decades of nanometer-precise optical studies suggest that myosin-5a, one of the prototypical molecular motors, takes uniform steps spanning 13 subunits (36 nm) along its F-actin track. Here, we use high-resolution interferometric scattering microscopy to reveal that myosin takes strides spanning 22 to 34 actin subunits, despite walking straight along the helical actin filament. We show that cumulative angular disorder in F-actin accounts for the observed proportion of each stride length, akin to crossing a river on variably spaced stepping stones. Electron microscopy revealed the structure of the stepping molecule. Our results indicate that both motor and track are soft materials that can adapt to function in complex cellular conditions.


Subject(s)
Actins , Myosin Type V , Actins/chemistry , Myosins/chemistry , Actin Cytoskeleton/chemistry , Motion , Myosin Type V/chemistry
2.
Cell ; 135(3): 535-48, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18984164

ABSTRACT

Learning-related plasticity at excitatory synapses in the mammalian brain requires the trafficking of AMPA receptors and the growth of dendritic spines. However, the mechanisms that couple plasticity stimuli to the trafficking of postsynaptic cargo are poorly understood. Here we demonstrate that myosin Vb (MyoVb), a Ca2+-sensitive motor, conducts spine trafficking during long-term potentiation (LTP) of synaptic strength. Upon activation of NMDA receptors and corresponding Ca2+ influx, MyoVb associates with recycling endosomes (REs), triggering rapid spine recruitment of endosomes and local exocytosis in spines. Disruption of MyoVb or its interaction with the RE adaptor Rab11-FIP2 abolishes LTP-induced exocytosis from REs and prevents both AMPA receptor insertion and spine growth. Furthermore, induction of tight binding of MyoVb to actin using an acute chemical genetic strategy eradicates LTP in hippocampal slices. Thus, Ca2+-activated MyoVb captures and mobilizes REs for AMPA receptor insertion and spine growth, providing a mechanistic link between the induction and expression of postsynaptic plasticity.


Subject(s)
Endosomes/metabolism , Long-Term Potentiation , Myosin Type V/metabolism , Neuronal Plasticity , Receptors, AMPA/metabolism , Animals , Calcium/metabolism , Cell Line , Dendrites/metabolism , Dendritic Spines/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Mice, Inbred Strains , Myosin Type V/chemistry , Neurons/metabolism , Rats , Synapses/metabolism , rab GTP-Binding Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 116(40): 19777-19785, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31506355

ABSTRACT

Two mechanisms have been proposed for the function of motor proteins: The power stroke and the Brownian ratchet. The former refers to generation of a large downhill free energy gradient over which the motor protein moves nearly irreversibly in making a step, whereas the latter refers to biasing or rectifying the diffusive motion of the motor. Both mechanisms require input of free energy, which generally involves the processing of an ATP (adenosine 5'-triphosphate) molecule. Recent advances in experiments that reveal the details of the stepping motion of motor proteins, together with computer simulations of atomistic structures, have provided greater insights into the mechanisms. Here, we compare the various models of the power stroke and the Brownian ratchet that have been proposed. The 2 mechanisms are not mutually exclusive, and various motor proteins employ them to different extents to perform their biological function. As examples, we discuss linear motor proteins Kinesin-1 and myosin-V, and the rotary motor F1-ATPase, all of which involve a power stroke as the essential element of their stepping mechanism.


Subject(s)
Adenosine Triphosphate/chemistry , Kinesins/chemistry , Myosin Type V/chemistry , Myosins/chemistry , Proton-Translocating ATPases/chemistry , Adenosine Diphosphate/chemistry , Animals , Computer Simulation , Dyneins/chemistry , Humans , Hydrolysis , Models, Biological , Molecular Conformation , Molecular Motor Proteins/chemistry , Motion , Pectinidae , Protein Conformation , Sheep , Static Electricity , Stress, Mechanical
4.
J Biol Chem ; 294(5): 1554-1567, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30518549

ABSTRACT

Myosins are molecular motors that use a conserved ATPase cycle to generate force. We investigated two mutations in the converter domain of myosin V (R712G and F750L) to examine how altering specific structural transitions in the motor ATPase cycle can impair myosin mechanochemistry. The corresponding mutations in the human ß-cardiac myosin gene are associated with hypertrophic and dilated cardiomyopathy, respectively. Despite similar steady-state actin-activated ATPase and unloaded in vitro motility-sliding velocities, both R712G and F750L were less able to overcome frictional loads measured in the loaded motility assay. Transient kinetic analysis and stopped-flow FRET demonstrated that the R712G mutation slowed the maximum ATP hydrolysis and recovery-stroke rate constants, whereas the F750L mutation enhanced these steps. In both mutants, the fast and slow power-stroke as well as actin-activated phosphate release rate constants were not significantly different from WT. Time-resolved FRET experiments revealed that R712G and F750L populate the pre- and post-power-stroke states with similar FRET distance and distance distribution profiles. The R712G mutant increased the mole fraction in the post-power-stroke conformation in the strong actin-binding states, whereas the F750L decreased this population in the actomyosin ADP state. We conclude that mutations in key allosteric pathways can shift the equilibrium and/or alter the activation energy associated with key structural transitions without altering the overall conformation of the pre- and post-power-stroke states. Thus, therapies designed to alter the transition between structural states may be able to rescue the impaired motor function induced by disease mutations.


Subject(s)
Mechanotransduction, Cellular , Motor Activity , Mutation , Myosin Type V/chemistry , Myosin Type V/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Chickens , Models, Molecular , Myosin Type V/genetics , Protein Binding , Protein Conformation , Protein Domains , Sequence Homology
5.
J Biol Chem ; 294(15): 5896-5906, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30804213

ABSTRACT

Class V myosins are actin-dependent motors, which recognize numerous cellular cargos mainly via the C-terminal globular tail domain (GTD). Myo2, a yeast class V myosin, can transport a broad range of organelles. However, little is known about the capacity of Myo2-GTD to recognize such a diverse array of cargos specifically at the molecular level. Here, we solved crystal structures of Myo2-GTD (at 1.9-3.1 Å resolutions) in complex with three cargo adaptor proteins: Smy1 (for polarization of secretory vesicles), Inp2 (for peroxisome transport), and Mmr1 (for mitochondria transport). The structures of Smy1- and Inp2-bound Myo2-GTD, along with site-directed mutagenesis experiments, revealed a binding site in subdomain-I having a hydrophobic groove with high flexibility enabling Myo2-GTD to accommodate different protein sequences. The Myo2-GTD-Mmr1 complex structure confirmed and complemented a previously identified mitochondrion/vacuole-specific binding region. Moreover, differences between the conformations and locations of cargo-binding sites identified here for Myo2 and those reported for mammalian MyoVA (MyoVA) suggest that class V myosins potentially have co-evolved with their specific cargos. Our structural and biochemical analysis not only uncovers a molecular mechanism that explains the diverse cargo recognition by Myo2-GTD, but also provides structural information useful for future functional studies of class V myosins in cargo transport.


Subject(s)
Myosin Heavy Chains/chemistry , Myosin Heavy Chains/metabolism , Myosin Type V/chemistry , Myosin Type V/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Binding Sites , Evolution, Molecular , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Protein Domains , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
6.
J Cell Sci ; 131(3)2018 02 07.
Article in English | MEDLINE | ID: mdl-29222113

ABSTRACT

Myosins are critical motor proteins that contribute to the secretory pathway, polarized growth, and cytokinesis. The globular tail domains of class V myosins have been shown to be important for cargo binding and actin cable organization. Additionally, phosphorylation plays a role in class V myosin cargo choice. Our previous studies on the class V myosin MyoE in the fungal pathogen Aspergillus fumigatus confirmed its requirement for normal morphology and virulence. However, the domains and molecular mechanisms governing the functions of MyoE remain unknown. Here, by analyzing tail mutants, we demonstrate that the tail is required for radial growth, conidiation, septation frequency and MyoE's location at the septum. Furthermore, MyoE is phosphorylated at multiple residues in vivo; however, alanine substitution mutants revealed that no single phosphorylated residue was critical. Importantly, in the absence of the phosphatase calcineurin, an additional residue was phosphorylated in its tail domain. Mutation of this tail residue led to mislocalization of MyoE from the septa. This work reveals the importance of the MyoE tail domain and its phosphorylation/dephosphorylation in the growth and morphology of A. fumigatus.


Subject(s)
Aspergillus fumigatus/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Hyphae/growth & development , Myosin Type V/chemistry , Myosin Type V/metabolism , Acetylation , Actins/metabolism , Calcineurin/metabolism , Conserved Sequence , Microtubules/metabolism , Models, Biological , Mutant Proteins/metabolism , Phenotype , Phosphorylation , Protein Domains , Protein Subunits/metabolism , Protein Transport , Sequence Deletion , Spores, Fungal/metabolism , Structure-Activity Relationship
7.
Biochem Biophys Res Commun ; 527(1): 232-237, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446373

ABSTRACT

The human fungal pathogen Aspergillus fumigatus causes life-threatening invasive aspergillosis in immunocompromised individuals. Adaptation to the host environment is integral to survival of A. fumigatus and requires the coordination of short- and long-distance vesicular transport to move essential components throughout the fungus. We previously reported the importance of MyoE, the only class V myosin, for hyphal growth and virulence of A. fumigatus. Class V myosins are actin-based, cargo-carrying motor proteins that contain unique binding sites for specific cargo. Specific cargo carried by myosin V has not been identified in any fungus, and previous studies have only identified single components that interact with class V myosins. Here we utilized a mass spectrometry-based whole proteomic approach to identify MyoE interacting proteins in A. fumigatus for the first time. Several proteins previously shown to interact with myosin V through physical and genetic approaches were confirmed, validating our proteomic analysis. Importantly, we identified novel MyoE-interacting proteins, including members of the cytoskeleton network, cell wall synthesis, calcium signaling and a group of coat protein complex II (COPII) proteins involved in the endoplasmic reticulum (ER) to Golgi transport. Furthermore, we analyzed the localization patterns of the COPII proteins, UsoA (Uso1), SrgE (Sec31), and SrgF (Sec23), which suggested a potential role for MyoE in ER to Golgi trafficking.


Subject(s)
Aspergillus fumigatus/chemistry , COP-Coated Vesicles/chemistry , Myosin Type V/chemistry , Biological Transport , COP-Coated Vesicles/metabolism , Humans , Microscopy, Fluorescence , Myosin Type V/isolation & purification , Myosin Type V/metabolism
8.
Proc Natl Acad Sci U S A ; 114(9): 2259-2264, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193897

ABSTRACT

The detailed dynamics of the cycle of myosin-V are explored by simulation approaches, examining the nature of the energy-driven motion. Our study started with Langevin dynamics (LD) simulations on a very coarse landscape with a single rate-limiting barrier and reproduced the stall force and the hand-over-hand dynamics. We then considered a more realistic landscape and used time-dependent Monte Carlo (MC) simulations that allowed trajectories long enough to reproduce the force/velocity characteristic sigmoidal correlation, while also reproducing the hand-over-hand motion. Overall, our study indicated that the notion of a downhill lever-up to lever-down process (popularly known as the powerstroke mechanism) is the result of the energetics of the complete myosin-V cycle and is not the source of directional motion or force generation on its own. The present work further emphasizes the need to use well-defined energy landscapes in studying molecular motors in general and myosin in particular.


Subject(s)
Actins/chemistry , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Myosin Type V/chemistry , Phosphates/chemistry , Actins/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Biomechanical Phenomena , Humans , Kinetics , Molecular Dynamics Simulation , Monte Carlo Method , Myosin Type V/metabolism , Phosphates/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Thermodynamics
9.
Proc Natl Acad Sci U S A ; 113(13): E1844-52, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976594

ABSTRACT

Molecular motors produce force when they interact with their cellular tracks. For myosin motors, the primary force-generating state has MgADP tightly bound, whereas myosin is strongly bound to actin. We have generated an 8-Å cryoEM reconstruction of this state for myosin V and used molecular dynamics flexed fitting for model building. We compare this state to the subsequent state on actin (Rigor). The ADP-bound structure reveals that the actin-binding cleft is closed, even though MgADP is tightly bound. This state is accomplished by a previously unseen conformation of the ß-sheet underlying the nucleotide pocket. The transition from the force-generating ADP state to Rigor requires a 9.5° rotation of the myosin lever arm, coupled to a ß-sheet rearrangement. Thus, the structure reveals the detailed rearrangements underlying myosin force generation as well as the basis of strain-dependent ADP release that is essential for processive myosins, such as myosin V.


Subject(s)
Actins/metabolism , Adenosine Diphosphate/metabolism , Myosin Type V/chemistry , Myosin Type V/metabolism , Actins/chemistry , Binding Sites , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation
10.
Proc Natl Acad Sci U S A ; 113(40): E5812-E5820, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647889

ABSTRACT

The motor function of vertebrate myosin-5a is inhibited by its tail in a Ca2+-dependent manner. We previously demonstrated that the calmodulin (CaM) bound to the first isoleucine-glutamine (IQ) motif (IQ1) of myosin-5a is responsible for the Ca2+-dependent regulation of myosin-5a. We have solved the crystal structure of a truncated myosin-5a containing the motor domain and IQ1 (MD-IQ1) complexed with Ca2+-bound CaM (Ca2+-CaM) at 2.5-Å resolution. Compared with the structure of the MD-IQ1 complexed with essential light chain (an equivalent of apo-CaM), MD-IQ1/Ca2+-CaM displays large conformational differences in IQ1/CaM and little difference in the motor domain. In the MD-IQ1/Ca2+-CaM structure, the N-lobe and the C-lobe of Ca2+-CaM adopt an open conformation and grip the C-terminal and the N-terminal portions of the IQ1, respectively. Remarkably, the interlobe linker of CaM in IQ1/Ca2+-CaM is in a position opposite that in IQ1/apo-CaM, suggesting that CaM flip-flops relative to the IQ1 during the Ca2+ transition. We demonstrated that CaM continuously associates with the IQ1 during the Ca2+ transition and that the binding of CaM to IQ1 increases Ca2+ affinity and substantially changes the kinetics of the Ca2+ transition, suggesting that the IQ1/CaM complex functions as an intact Ca2+ sensor responding to distinct calcium signals.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Myosin Type V/chemistry , Myosin Type V/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Calmodulin/chemistry , Calorimetry , Crystallography, X-Ray , Kinetics , Mice , Models, Biological , Protein Domains , Rabbits , Spectrometry, Fluorescence , Structural Homology, Protein , Tryptophan/metabolism
11.
J Biol Chem ; 292(26): 10998-11008, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28476885

ABSTRACT

Myosin Vc (myoVc) is unique among vertebrate class V myosin isoforms in that it requires teams of motors to move continuously on single actin filaments. Single molecules of myoVc cannot take multiple hand-over-hand steps from one actin-binding site to the next without dissociating, in stark contrast to the well studied myosin Va (myoVa) isoform. At low salt, single myoVc motors can, however, move processively on actin bundles, and at physiologic ionic strength, even teams of myoVc motors require actin bundles to sustain continuous motion. Here, we linked defined numbers of myoVc or myoVa molecules to DNA nanostructures as synthetic cargos. Using total internal reflectance fluorescence microscopy, we compared the stepping behavior of myoVc versus myoVa ensembles and myoVc stepping patterns on single actin filaments versus actin bundles. Run lengths of both myoVc and myoVa teams increased with motor number, but only multiple myoVc motors showed a run-length enhancement on actin bundles compared with actin filaments. By resolving the stepping behavior of individual myoVc motors with a quantum dot bound to the motor domain, we found that coupling of two myoVc motors significantly decreased the futile back and side steps that were frequently observed for single myoVc motors. Changes in the inter-motor distance between two coupled myoVc motors affected stepping dynamics, suggesting that mechanical tension coordinates the stepping behavior of two myoVc motors for efficient directional motion. Our study provides a molecular basis to explain how teams of myoVc motors are suited to transport cargos such as zymogen granules on actin bundles.


Subject(s)
Actin Cytoskeleton/chemistry , Myosin Heavy Chains/chemistry , Myosin Type V/chemistry , Quantum Dots/chemistry , Secretory Vesicles/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Biological Transport, Active , Mice , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Type V/genetics , Myosin Type V/metabolism , Secretory Vesicles/genetics , Secretory Vesicles/metabolism
12.
J Biol Chem ; 292(44): 18372-18385, 2017 11 03.
Article in English | MEDLINE | ID: mdl-28882893

ABSTRACT

Myosin-5B is a ubiquitous molecular motor that transports cargo vesicles of the endomembrane system in intracellular recycling pathways. Myosin-5B malfunction causes the congenital enteropathy microvillus inclusion disease, underlining its importance in cellular homeostasis. Here we describe the interaction of myosin-5B with F-actin, nucleotides, and the pyrazolopyrimidine compound myoVin-1. We show that single-headed myosin-5B is an intermediate duty ratio motor with a kinetic ATPase cycle that is rate-limited by the release of phosphate. The presence of a second head generates strain and gating in the myosin-5B dimer that alters the kinetic signature by reducing the actin-activated ADP release rate to become rate-limiting. This kinetic transition into a high-duty ratio motor is a prerequisite for the proposed transport function of myosin-5B in cellular recycling pathways. Moreover, we show that the small molecule compound myoVin-1 inhibits the enzymatic and functional activity of myosin-5B in vitro Partial inhibition of the actin-activated steady-state ATPase activity and sliding velocity suggests that caution should be used when probing the effect of myoVin-1 on myosin-5-dependent transport processes in cells.


Subject(s)
Actin Cytoskeleton/metabolism , Malabsorption Syndromes/metabolism , Microvilli/pathology , Models, Molecular , Mucolipidoses/metabolism , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Actin Cytoskeleton/chemistry , Amino Acid Substitution , Binding Sites , Computational Biology , Dimerization , Enzyme Inhibitors/pharmacology , Expert Systems , Humans , Kinetics , Malabsorption Syndromes/genetics , Microvilli/genetics , Microvilli/metabolism , Molecular Docking Simulation , Mucolipidoses/genetics , Mutation , Myosin Heavy Chains/antagonists & inhibitors , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Myosin Type V/antagonists & inhibitors , Myosin Type V/chemistry , Myosin Type V/genetics , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Structural Homology, Protein
13.
J Biol Chem ; 292(45): 18500-18517, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28939769

ABSTRACT

Membrane tethering is a fundamental process essential for the compartmental specificity of intracellular membrane trafficking in eukaryotic cells. Rab-family small GTPases and specific sets of Rab-interacting effector proteins, including coiled-coil tethering proteins and multisubunit tethering complexes, are reported to be responsible for membrane tethering. However, whether and how these key components directly and specifically tether subcellular membranes remains enigmatic. Using chemically defined proteoliposomal systems reconstituted with purified human Rab proteins and synthetic liposomal membranes to study the molecular basis of membrane tethering, we established here that Rab-family GTPases have a highly conserved function to directly mediate membrane tethering, even in the absence of any types of Rab effectors such as the so-called tethering proteins. Moreover, we demonstrate that membrane tethering mediated by endosomal Rab11a is drastically and selectively stimulated by its cognate Rab effectors, class V myosins (Myo5A and Myo5B), in a GTP-dependent manner. Of note, Myo5A and Myo5B exclusively recognized and cooperated with the membrane-anchored form of their cognate Rab11a to support membrane tethering mediated by trans-Rab assemblies on opposing membranes. Our findings support the novel concept that Rab-family proteins provide a bona fide membrane tether to physically and specifically link two distinct lipid bilayers of subcellular membranes. They further indicate that Rab-interacting effector proteins, including class V myosins, can regulate these Rab-mediated membrane-tethering reactions.


Subject(s)
Endosomes/metabolism , Guanosine Triphosphate/metabolism , Intracellular Membranes/metabolism , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , rab GTP-Binding Proteins/agonists , Acylation , Endosomes/enzymology , Histidine/chemistry , Histidine/genetics , Histidine/metabolism , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/enzymology , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Liposomes , Lysine/analogs & derivatives , Lysine/chemistry , Lysine/metabolism , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Myosin Type V/chemistry , Myosin Type V/genetics , Oleic Acids/chemistry , Oleic Acids/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Prenylation , Protein Processing, Post-Translational , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Succinates/chemistry , Succinates/metabolism , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
14.
Phys Chem Chem Phys ; 20(7): 4752-4759, 2018 Feb 14.
Article in English | MEDLINE | ID: mdl-29379931

ABSTRACT

Based on the available experimental evidence, we present a simple and general model to describe the movement dynamics of molecular motors that can move processively on their linear tracks by using the chemical energy derived from ATP hydrolysis. An important aspect of the model is the non-tight coupling between the ATP hydrolysis and mechanical stepping, in contrast to the prevailing models presented in the literature that assume the tight chemomechanical coupling. With kinesin as an example, based on the current model, we study in detail its movement dynamics under a backward load, reproducing well the diverse available single-molecule experimental data such as the forward to backward step ratio, velocity, dwell time, randomness, run length, etc., versus the load. Moreover, predicted results are provided on the force-dependence of the mean number of ATP molecules consumed per mechanical step. Additionally, the theoretical data for the dynamics of myosin-V obtained based on the model are also in good agreement with the available experimental data.


Subject(s)
Models, Biological , Molecular Motor Proteins/chemistry , Adenosine Triphosphate/chemistry , Binding Sites , Biomechanical Phenomena , Hydrolysis , Kinetics , Motion , Myosin Type V/chemistry , Protein Binding
15.
Proc Natl Acad Sci U S A ; 112(47): 14593-8, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26553992

ABSTRACT

Myosins use a conserved structural mechanism to convert the energy from ATP hydrolysis into a large swing of the force-generating lever arm. The precise timing of the lever arm movement with respect to the steps in the actomyosin ATPase cycle has not been determined. We have developed a FRET system in myosin V that uses three donor-acceptor pairs to examine the kinetics of lever arm swing during the recovery and power stroke phases of the ATPase cycle. During the recovery stroke the lever arm swing is tightly coupled to priming the active site for ATP hydrolysis. The lever arm swing during the power stroke occurs in two steps, a fast step that occurs before phosphate release and a slow step that occurs before ADP release. Time-resolved FRET demonstrates a 20-Å change in distance between the pre- and postpower stroke states and shows that the lever arm is more dynamic in the postpower stroke state. Our results suggest myosin binding to actin in the ADP.Pi complex triggers a rapid power stroke that gates the release of phosphate, whereas a second slower power stroke may be important for mediating strain sensitivity.


Subject(s)
Myosin Type V/chemistry , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Catalysis , Catalytic Domain , Fluorescence Resonance Energy Transfer
16.
Biophys J ; 113(3): 558-571, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28793211

ABSTRACT

Allosteric effects often underlie the activity of proteins, and elucidating generic design aspects and functional principles unique to allosteric phenomena represent a major challenge. Here an approach consisting of the in silico design of synthetic structures, which, as the principal element of allostery, encode dynamical long-range coupling among two sites, is presented. The structures are represented by elastic networks, similar to coarse-grained models of real proteins. A strategy of evolutionary optimization was implemented to iteratively improve allosteric coupling. In the designed structures, allosteric interactions were analyzed in terms of strain propagation, and simple pathways that emerged during evolution were identified as signatures through which long-range communication was established. Moreover, robustness of allosteric performance with respect to mutations was demonstrated. As it turned out, the designed prototype structures reveal dynamical properties resembling those found in real allosteric proteins. Hence, they may serve as toy models of complex allosteric systems, such as proteins. Application of the developed modeling scheme to the allosteric transition in the myosin V molecular motor was also demonstrated.


Subject(s)
Elasticity , Models, Molecular , Myosin Type V/chemistry , Myosin Type V/metabolism , Allosteric Regulation , Evolution, Molecular , Protein Conformation
17.
J Biol Chem ; 291(15): 8241-50, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26912658

ABSTRACT

Vertebrates have three isoforms of class V myosin (Myo5), Myo5a, Myo5b, and Myo5c, which are involved in transport of multiple cargoes. It is well established that the motor functions of Myo5a and Myo5b are regulated by a tail inhibition mechanism. Here we found that the motor function of Myo5c was also inhibited by its globular tail domain (GTD), and this inhibition was abolished by high Ca(2+), indicating that the tail inhibition mechanism is conserved in vertebrate Myo5. Interestingly, we found that Myo5a-GTD and Myo5c-GTD were not interchangeable in terms of inhibition of motor function, indicating isoform-specific interactions between the GTD and the head of Myo5. To identify the isoform-specific interactions, we produced a number of Myo5 chimeras by swapping the corresponding regions of Myo5a and Myo5c. We found that Myo5a-GTD, with its H11-H12 loop being substituted with that of Myo5c, was able to inhibit the ATPase activity of Myo5c and that Myo5a-GTD was able to inhibit the ATPase activity of Myo5c-S1 and Myo5c-HMM only when their IQ1 motif was substituted with that of Myo5a. Those results indicate that the H11-H12 loop in the GTD and the IQ1 motif in the head dictate the isoform-specific interactions between the GTD and head of Myo5. Because the IQ1 motif is wrapped by calmodulin, whose conformation is influenced by the sequence of the IQ1 motif, we proposed that the calmodulin bound to the IQ1 motif interacts with the H11-H12 loop of the GTD in the inhibited state of Myo5.


Subject(s)
Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Calcium/metabolism , Humans , Mice , Molecular Sequence Data , Myosin Heavy Chains/chemistry , Myosin Type V/chemistry , Protein Conformation , Protein Interaction Maps , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Sequence Alignment
18.
J Biol Chem ; 291(26): 13571-9, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129208

ABSTRACT

Myosin-5a contains two heavy chains, which are dimerized via the coiled-coil regions. Thus, myosin-5a comprises two heads and two globular tail domains (GTDs). The GTD is the inhibitory domain that binds to the head and inhibits its motor function. Although the two-headed structure is essential for the processive movement of myosin-5a along actin filaments, little is known about the role of GTD dimerization. Here, we investigated the effect of GTD dimerization on its inhibitory activity. We found that the potent inhibitory activity of the GTD is dependent on its dimerization by the preceding coiled-coil regions, indicating synergistic interactions between the two GTDs and the two heads of myosin-5a. Moreover, we found that alanine mutations of the two conserved basic residues at N-terminal extension of the GTD not only weaken the inhibitory activity of the GTD but also enhance the activation of myosin-5a by its cargo-binding protein melanophilin (Mlph). These results are consistent with the GTD forming a head to head dimer, in which the N-terminal extension of the GTD interacts with the Mlph-binding site in the counterpart GTD. The Mlph-binding site at the GTD-GTD interface must be exposed prior to the binding of Mlph. We therefore propose that the inhibited Myo5a is equilibrated between the folded state, in which the Mlph-binding site is buried, and the preactivated state, in which the Mlph-binding site is exposed, and that Mlph is able to bind to the Myo5a in preactivated state and activates its motor function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Protein Folding , Protein Multimerization/physiology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Substitution , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/genetics , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , Mutation, Missense , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Myosin Type V/chemistry , Myosin Type V/genetics , Protein Domains , Protein Structure, Quaternary
19.
Proc Natl Acad Sci U S A ; 111(38): E3986-95, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25201964

ABSTRACT

Myosin Va is an actin-based molecular motor responsible for transport and positioning of a wide array of intracellular cargoes. Although myosin Va motors have been well characterized at the single-molecule level, physiological transport is carried out by ensembles of motors. Studies that explore the behavior of ensembles of molecular motors have used nonphysiological cargoes such as DNA linkers or glass beads, which do not reproduce one key aspect of vesicular systems--the fluid intermotor coupling of biological lipid membranes. Using a system of defined synthetic lipid vesicles (100- to 650-nm diameter) composed of either 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) (fluid at room temperature) or 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) (gel at room temperature) with a range of surface densities of myosin Va motors (32-125 motors per µm(2)), we demonstrate that the velocity of vesicle transport by ensembles of myosin Va is sensitive to properties of the cargo. Gel-state DPPC vesicles bound with multiple motors travel at velocities equal to or less than vesicles with a single myosin Va (∼450 nm/s), whereas surprisingly, ensembles of myosin Va are able to transport fluid-state DOPC vesicles at velocities significantly faster (>700 nm/s) than a single motor. To explain these data, we developed a Monte Carlo simulation that suggests that these reductions in velocity can be attributed to two distinct mechanisms of intermotor interference (i.e., load-dependent modulation of stepping kinetics and binding-site exclusion), whereas faster transport velocities are consistent with a model wherein the normal stepping behavior of the myosin is supplemented by the preferential detachment of the trailing motor from the actin track.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/analogs & derivatives , Membranes, Artificial , Myosin Heavy Chains/chemistry , Myosin Type V/chemistry , Phosphatidylcholines/chemistry , Transport Vesicles/chemistry , 1,2-Dipalmitoylphosphatidylcholine/chemistry , Animals , Biological Transport, Active , Mice , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Type V/genetics , Myosin Type V/metabolism , Transport Vesicles/genetics , Transport Vesicles/metabolism
20.
Biophys J ; 111(10): 2228-2240, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27851945

ABSTRACT

Myosin Va (myoVa) is a processive, actin-based molecular motor essential for intracellular cargo transport. When a cargo is transported by an ensemble of myoVa motors, each motor faces significant physical barriers and directional challenges created by the complex actin cytoskeleton, a network of actin filaments and actin bundles. The principles that govern the interaction of multiple motors attached to the same cargo are still poorly understood. To understand the mechanical interactions between multiple motors, we developed a simple in vitro model in which two individual myoVa motors labeled with different-colored Qdots are linked via a third Qdot that acts as a cargo. The velocity of this two-motor complex was reduced by 27% as compared to a single motor, whereas run length was increased by only 37%, much less than expected from multimotor transport models. Therefore, at low ATP, which allowed us to identify individual motor steps, we investigated the intermotor dynamics within the two-motor complex. The randomness of stepping leads to a buildup of tension in the linkage between motors-which in turn slows down the leading motor-and increases the frequency of backward steps and the detachment rate. We establish a direct relationship between the velocity reduction and the distribution of intermotor distances. The analysis of run lengths and dwell times for the two-motor complex, which has only one motor engaged with the actin track, reveals that half of the runs are terminated by almost simultaneous detachment of both motors. This finding challenges the assumptions of conventional multimotor models based on consecutive motor detachment. Similar, but even more drastic, results were observed with two-motor complexes on actin bundles, which showed a run length that was even shorter than that of a single motor.


Subject(s)
Actin Cytoskeleton/metabolism , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Actins/metabolism , Animals , Biological Transport , Kinetics , Mice , Models, Molecular , Myosin Heavy Chains/chemistry , Myosin Type V/chemistry , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL