Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters










Publication year range
1.
Biosens Bioelectron ; 93: 305-314, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-27591903

ABSTRACT

The actin-myosin system, responsible for muscle contraction, is also the force-generating element in dynamic nanodevices operating with surface-immobilized motor proteins. These devices require materials that are amenable to micro- and nano-fabrication, but also preserve the bioactivity of molecular motors. The complexity of the protein-surface systems is greatly amplified by those of the polymer-fluid interface; and of the structure and function of molecular motors, making the study of these interactions critical to the success of molecular motor-based nanodevices. We measured the density of the adsorbed motor protein (heavy meromyosin, HMM) using quartz crystal microbalance; and motor bioactivity with ATPase assay, on a set of model surfaces, i.e., nitrocellulose, polystyrene, poly(methyl methacrylate), and poly(butyl methacrylate), poly(tert-butyl methacrylate). A higher hydrophobicity of the adsorbing material translates in a higher total number of HMM molecules per unit area, but also in a lower uptake of water, and a lower ratio of active per total HMM molecules per unit area. We also measured the motility characteristics of actin filaments on the model surfaces, i.e., velocity, smoothness and deflection of movement, determined via in vitro motility assays. The filament velocities were found to be controlled by the relative number of active HMM per total motors, rather than their absolute surface density. The study allowed the formulation of the general engineering principles for the selection of polymeric materials for the manufacturing of dynamic nanodevices using protein molecular motors.


Subject(s)
Biosensing Techniques , Myosin Subfragments/chemistry , Nanotechnology , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/physiology , Myosin Subfragments/physiology , Myosins/chemistry , Myosins/physiology , Polymers/chemistry , Quartz Crystal Microbalance Techniques , Surface Properties
2.
Proc Natl Acad Sci U S A ; 112(34): 10703-7, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26261319

ABSTRACT

Constituents of living or synthetic active matter have access to a local energy supply that serves to keep the system out of thermal equilibrium. The statistical properties of such fluctuating active systems differ from those of their equilibrium counterparts. Using the actin filament gliding assay as a model, we studied how nonthermal distributions emerge in active matter. We found that the basic mechanism involves the interplay between local and random injection of energy, acting as an analog of a thermal heat bath, and nonequilibrium energy dissipation processes associated with sudden jump-like changes in the system's dynamic variables. We show here how such a mechanism leads to a nonthermal distribution of filament curvatures with a non-Gaussian shape. The experimental curvature statistics and filament relaxation dynamics are reproduced quantitatively by stochastic computer simulations and a simple kinetic model.


Subject(s)
Actin Cytoskeleton/physiology , Stochastic Processes , Actin Cytoskeleton/ultrastructure , Animals , Computer Simulation , Elasticity , Energy Transfer , Microscopy, Fluorescence , Models, Biological , Models, Theoretical , Motion , Myosin Subfragments/physiology , Statistical Distributions , Thermal Diffusion
3.
Anim Sci J ; 86(4): 459-67, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25410124

ABSTRACT

In skeletal muscle cells, myofibrillar proteins are highly organized into sarcomeres in which thick filaments interdigitate with thin filaments to generate contractile force. The size of thick filaments, which consist mainly of myosin molecules, is strictly controlled. However, little is known about the mechanisms by which myosin molecules assemble into thick filaments. Here, we assessed the ability of each domain of myosin heavy chain (Myh) to form thick filaments. We showed that exogenously expressed subfragment 2 (S2) + light meromyosin (LMM) of Myh was efficiently incorporated into thick filaments in muscle cells, although neither solely expressed S2 nor LMM targeted to thick filaments properly. In nonmuscle COS7 cells, S2+LMM formed more enlarged filaments/speckles than LMM. These results suggest that Myh filament formation is induced by S2 accompanying LMM. We further examined the effects of Myh C-terminus on thick filament assembly. C-terminal deletion mutants were incorporated not into entire thick filaments but rather into restricted regions of thick filaments. Our findings suggest that the elongation of myosin filaments to form thick filaments is regulated by S2 as well as C-terminus of LMM.


Subject(s)
Cytoskeleton/metabolism , Cytoskeleton/physiology , Muscle, Skeletal/cytology , Myosin Subfragments/metabolism , Skeletal Muscle Myosins/metabolism , Animals , Cells, Cultured , Mice , Mutant Proteins/metabolism , Myosin Subfragments/genetics , Myosin Subfragments/physiology , Sarcomeres
4.
Biochemistry ; 53(24): 4015-24, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24873380

ABSTRACT

The actin cytoskeleton carries out cellular functions, including division, migration, adhesion, and intracellular transport, that require a variety of actin binding proteins, including myosins. Our focus here is on class II nonmuscle myosin isoforms, NMIIA, NMIIB, and NMIIC, and their regulation by the actin binding protein, tropomyosin. NMII myosins are localized to different populations of stress fibers and the contractile ring, structures involved in force generation required for cell migration, adhesion, and cytokinesis. The stress fibers and contractile ring that contain NMII myosins also contain tropomyosin. Four mammalian genes encode more than 40 tropomyosins. Tropomyosins inhibit or activate actomyosin MgATPase and motility depending on the myosin and tropomyosin isoform. In vivo, tropomyosins play a role in cell migration, adhesion, cytokinesis, and NMII isoform localization in an isoform-specific manner. We postulate that the isoform-specific tropomyosin localization and effect on NMII isoform localization reflect modulation of NMII actomyosin kinetics and motile function. In this study, we compare the ability of different tropomyosin isoforms to support actin filament motility with NMIIA, NMIIB, and NMIIC as well as skeletal muscle myosin. Tropomyosins activated, inhibited, or had no effect on motility depending on the myosin, indicating that the myosin isoform is the primary determinant of the isoform-specific effect of tropomyosin on actomyosin regulation. Activation of motility of nonmuscle tropomyosin-actin filaments by NMII myosin correlates with an increased Vmax of the myosin MgATPase, implying a direct effect on the myosin MgATPase, in contrast to the skeletal tropomyosin-actin filament that has no effect on the Vmax or maximal filament velocity.


Subject(s)
Myosin Type II/metabolism , Tropomyosin/physiology , Actins/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cell Movement , Humans , Myosin Subfragments/physiology , Rats , Tropomyosin/chemistry
5.
Proc Natl Acad Sci U S A ; 111(18): E1833-42, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24753602

ABSTRACT

Myosin-10 is an actin-based molecular motor that participates in essential intracellular processes such as filopodia formation/extension, phagocytosis, cell migration, and mitotic spindle maintenance. To study this motor protein's mechano-chemical properties, we used a recombinant, truncated form of myosin-10 consisting of the first 936 amino acids, followed by a GCN4 leucine zipper motif, to force dimerization. Negative-stain electron microscopy reveals that the majority of molecules are dimeric with a head-to-head contour distance of ∼50 nm. In vitro motility assays show that myosin-10 moves actin filaments smoothly with a velocity of ∼310 nm/s. Steady-state and transient kinetic analysis of the ATPase cycle shows that the ADP release rate (∼13 s(-1)) is similar to the maximum ATPase activity (∼12-14 s(-1)) and therefore contributes to rate limitation of the enzymatic cycle. Single molecule optical tweezers experiments show that under intermediate load (∼0.5 pN), myosin-10 interacts intermittently with actin and produces a power stroke of ∼17 nm, composed of an initial 15-nm and subsequent 2-nm movement. At low optical trap loads, we observed staircase-like processive movements of myosin-10 interacting with the actin filament, consisting of up to six ∼35-nm steps per binding interaction. We discuss the implications of this load-dependent processivity of myosin-10 as a filopodial transport motor.


Subject(s)
Actins/physiology , Myosin Heavy Chains/physiology , Actins/chemistry , Adenosine Diphosphate/metabolism , Amino Acid Sequence , Animals , Biomechanical Phenomena , Cattle , In Vitro Techniques , Kinetics , Microscopy, Electron , Microscopy, Fluorescence , Models, Biological , Models, Molecular , Molecular Sequence Data , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Myosin Subfragments/chemistry , Myosin Subfragments/genetics , Myosin Subfragments/physiology , Optical Tweezers , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Pseudopodia/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
J Mol Biol ; 414(4): 477-84, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22037585

ABSTRACT

While mutations in the myosin subfragment 1 motor domain can directly disrupt the generation and transmission of force along myofibrils and lead to myopathy, the mechanism whereby mutations in the myosin rod influences mechanical function is less clear. Here, we used a combination of various imaging techniques and molecular dynamics simulations to test the hypothesis that perturbations in the myosin rod can disturb normal sarcomeric uniformity and, like motor domain lesions, would influence force production and propagation. We show that disrupting the rod can alter its nanomechanical properties and, in vivo, can drive asymmetric myofilament and sarcomere formation. Our imaging results indicate that myosin rod mutations likely disturb production and/or propagation of contractile force. This provides a unifying theory where common pathological cascades accompany both myosin motor and specific rod domain mutations. Finally, we suggest that sarcomeric inhomogeneity, caused by asymmetric thick filaments, could be a useful index of myopathic dysfunction.


Subject(s)
Motor Endplate/physiology , Muscular Diseases/physiopathology , Myosin Subfragments/physiology , Sarcomeres/physiology , Humans , Models, Molecular , Motor Endplate/genetics , Muscle Contraction , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutation , Myosin Subfragments/chemistry , Myosin Subfragments/genetics , Myosin Subfragments/ultrastructure , Sarcomeres/chemistry , Sarcomeres/genetics , Sarcomeres/ultrastructure
7.
Biochem Biophys Res Commun ; 405(4): 651-6, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21281603

ABSTRACT

Muscle contraction results from an attachment-detachment cycle between the myosin heads extending from myosin filaments and the sites on actin filaments. The myosin head first attaches to actin together with the products of ATP hydrolysis, performs a power stroke associated with release of hydrolysis products, and detaches from actin upon binding with new ATP. The detached myosin head then hydrolyses ATP, and performs a recovery stroke to restore its initial position. The strokes have been suggested to result from rotation of the lever arm domain around the converter domain, while the catalytic domain remains rigid. To ascertain the validity of the lever arm hypothesis in muscle, we recorded ATP-induced movement at different regions within individual myosin heads in hydrated myosin filaments, using the gas environmental chamber attached to the electron microscope. The myosin head were position-marked with gold particles using three different site-directed antibodies. The amplitude of ATP-induced movement at the actin binding site in the catalytic domain was similar to that at the boundary between the catalytic and converter domains, but was definitely larger than that at the regulatory light chain in the lever arm domain. These results are consistent with the myosin head lever arm mechanism in muscle contraction if some assumptions are made.


Subject(s)
Muscle Contraction , Myosin Subfragments/chemistry , Myosin Subfragments/physiology , Adenosine Triphosphate/chemistry , Animals , Microscopy, Electron , Protein Conformation , Rabbits
8.
Science ; 329(5992): 686-9, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20689017

ABSTRACT

Using optical trapping and fluorescence imaging techniques, we measured the step size and stiffness of single skeletal myosins interacting with actin filaments and arranged on myosin-rod cofilaments that approximate myosin mechanics during muscle contraction. Stiffness is dramatically lower for negatively compared to positively strained myosins, consistent with buckling of myosin's subfragment 2 rod domain. Low stiffness minimizes drag of negatively strained myosins during contraction at loaded conditions. Myosin's elastic portion is stretched during active force generation, reducing apparent step size with increasing load, even though the working stroke is approximately constant at about 8 nanometers. Taking account of the nonlinear nature of myosin elasticity is essential to relate myosin's internal structural changes to physiological force generation and filament sliding.


Subject(s)
Actin Cytoskeleton/physiology , Muscle Contraction , Myosins/physiology , Actomyosin/chemistry , Actomyosin/physiology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Compliance , Elasticity , Models, Biological , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal , Myosin Subfragments/physiology , Myosins/chemistry , Quantum Dots , Rabbits
9.
Biochim Biophys Acta ; 1794(11): 1599-605, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19635597

ABSTRACT

To understand the importance of selected regions of the regulatory light chain (RLC) for phosphorylation-dependent regulation of smooth muscle myosin (SMM), we expressed three heavy meromyosins (HMMs) containing the following RLC mutants; K12E in a critical region of the phosphorylation domain, GTDP(95-98)/AAAA in the central hinge, and R160C a putative binding residue for phosphorylated S19. Single-turnover actin-activated Mg(2+)-ATPase (V(max) and K(ATPase)) and in vitro actin-sliding velocities were examined for both unphosphorylated (up-) and phosphorylated (p-) states. Turnover rates for the up-state (0.007-0.030 s(-1)) and velocities (no motion) for all constructs were not significantly different from the up-wild type (WT) indicating that they were completely turned off. The apparent binding constants for actin in the presence of ATP (K(ATPase)) were too weak to measure as expected for fully regulated constructs. For p-HMM containing GTDP/AAAA, we found that both ATPase and motility were normal. The data suggest that the native sequence in the central hinge between the two lobes of the RLC is not required for turning the HMM off and on both kinetically and mechanically. For p-HMM containing R160C, all parameters were normal, suggesting that R160C is not involved in coordination of the phosphorylated S19. For p-HMM containing K12E, the V(max) was 64% and the actin-sliding velocity was approximately 50% of WT, suggesting that K12 is an important residue for the ability to sense or to promote the conformational changes required for kinetic and mechanical activation.


Subject(s)
Myosin Light Chains/physiology , Smooth Muscle Myosins/physiology , Amino Acid Substitution , Animals , Kinetics , Molecular Motor Proteins/genetics , Myosin Light Chains/genetics , Myosin Subfragments/genetics , Myosin Subfragments/physiology , Phosphorylation , Protein Structure, Tertiary , Smooth Muscle Myosins/genetics
10.
Front Biosci ; 13: 5732-54, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508618

ABSTRACT

In many types of biophysical studies of both single molecules and ensembles of molecular motors the motors are adsorbed to artificial surfaces. Some of the most important assay systems of this type (in vitro motility assays and related single molecule techniques) will be briefly described together with an account of breakthroughs in the understanding of actomyosin function that have resulted from their use. A poorly characterized, but potentially important, entity in these studies is the mechanism of motor adsorption to surfaces and the effects of motor surface interactions on experimental results. A better understanding of these phenomena is also important for the development of commercially viable nanotechnological applications powered by molecular motors. Here, we will consider several aspects of motor surface interactions with a particular focus on heavy meromyosin (HMM) from skeletal muscle. These aspects will be related to heavy meromyosin structure and relevant parts of the vast literature on protein-surface interactions for non-motor proteins. An overview of methods for studying motor-surface interactions will also be given. The information is used as a basis for further development of a model for HMM-surface interactions and is discussed in relation to experiments where nanopatterning has been employed for in vitro reconstruction of actomyosin order. The challenges and potentials of this approach in biophysical studies, compared to the use of self-assembly of biological components into supramolecular protein aggregates (e.g. myosin filaments) will be considered. Finally, this review will consider the implications for further developments of motor-powered lab-on-a-chip devices.


Subject(s)
Molecular Motor Proteins/physiology , Myosin Subfragments/physiology , Adsorption , Biomechanical Phenomena , Biophysics/methods , Kinetics , Molecular Motor Proteins/chemistry , Myosin Subfragments/chemistry , Peptide Fragments/chemistry , Static Electricity , Surface Properties
11.
J Biomech ; 41(7): 1611-5, 2008.
Article in English | MEDLINE | ID: mdl-18387614

ABSTRACT

When a muscle is stretched while activated, its steady-state isometric force following stretch is greater than the corresponding purely isometric force. This so-called residual force enhancement (RFE) has been observed for half a century, yet its mechanism remains unknown. Recent experiments suggest that RFE is not caused by non-uniformities in sarcomere lengths, as had been assumed for a long time, and cannot be explained primarily with increases in passive force, but is directly related to the kinetics of the cross-bridge cycle. Specifically, it has been suggested that stretching an attached cross-bridge increases its dwell time and duty ratio; therefore, the proportion of attached cross-bridges in a muscle would be increased by stretch, thereby causing RFE. A three bead laser trap setup was used for testing single cross-bridge (myosin II) interactions with actin. Upon attachment of a cross-bridge, a stretch or shortening of the cross-bridge was applied with a force of about 1.0 pN. The hypothesis that stretching a single cross-bridge increases its dwell time and duty ratio was rejected. However, stretching caused an increase in the average steady-state force per cross-bridge (3.4+/-0.4 pN; n=433) compared to shortening (1.9+/-0.3 pN; n=689). Therefore, based on the results of this study, RFE cannot be explained by an increased duty ratio and the associated increase in proportion of attached cross-bridges, but might be associated with an increased force per cross-bridge.


Subject(s)
Actin Cytoskeleton/physiology , Isometric Contraction/physiology , Muscle Relaxation/physiology , Myosin Subfragments/physiology , Sarcomeres/physiology , Animals , Rabbits
12.
Langmuir ; 23(22): 11147-56, 2007 Oct 23.
Article in English | MEDLINE | ID: mdl-17696458

ABSTRACT

The in vitro motility assay is valuable for fundamental studies of actomyosin function and has recently been combined with nanostructuring techniques for the development of nanotechnological applications. However, the limited understanding of the interaction mechanisms between myosin motor fragments (heavy meromyosin, HMM) and artificial surfaces hampers the development as well as the interpretation of fundamental studies. Here we elucidate the HMM-surface interaction mechanisms for a range of negatively charged surfaces (silanized glass and SiO2), which is relevant both to nanotechnology and fundamental studies. The results show that the HMM-propelled actin filament sliding speed (after a single injection of HMM, 120 microg/mL) increased with the contact angle of the surfaces (in the range of 20-80 degrees). However, quartz crystal microbalance (QCM) studies suggested a reduction in the adsorption of HMM (with coupled water) under these conditions. This result and actin filament binding data, together with previous measurements of the HMM density (Sundberg, M.; Balaz, M.; Bunk, R.; Rosengren-Holmberg, J. P.; Montelius, L.; Nicholls, I. A.; Omling, P.; Tågerud, S.; Månsson, A. Langmuir 2006, 22, 7302-7312. Balaz, M.; Sundberg, M.; Persson, M.; Kvassman, J.; Månsson, A. Biochemistry 2007, 46, 7233-7251), are consistent with (1) an HMM monolayer and (2) different HMM configurations at different contact angles of the surface. More specifically, the QCM and in vitro motility assay data are consistent with a model where the molecules are adsorbed either via their flexible C-terminal tail part (HMMC) or via their positively charged N-terminal motor domain (HMMN) without other surface contact points. Measurements of zeta potentials suggest that an increased contact angle is correlated with a reduced negative charge of the surfaces. As a consequence, the HMMC configuration would be the dominant configuration at high contact angles but would be supplemented with electrostatically adsorbed HMM molecules (HMMN configuration) at low contact angles. This would explain the higher initial HMM adsorption (from probability arguments) under the latter conditions. Furthermore, because the HMMN mode would have no actin binding it would also account for the lower sliding velocity at low contact angles. The results are compared to previous studies of the microtubule-kinesin system and are also discussed in relation to fundamental studies of actomyosin and nanotechnological developments and applications.


Subject(s)
Molecular Motor Proteins/chemistry , Molecular Motor Proteins/physiology , Myosin Subfragments/chemistry , Myosin Subfragments/physiology , Actomyosin/chemistry , Actomyosin/physiology , Adsorption , Animals , Biophysical Phenomena , Biophysics , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Kinesins/physiology , Microscopy, Atomic Force , Microtubules/physiology , Models, Molecular , Nanotechnology , Quartz , Rabbits , Silicon Dioxide , Static Electricity , Surface Plasmon Resonance , Surface Properties , Trimethylsilyl Compounds
13.
J Mol Biol ; 367(5): 1312-29, 2007 Apr 13.
Article in English | MEDLINE | ID: mdl-17316684

ABSTRACT

Muscle myosin heavy chain (MHC) rod domains intertwine to form alpha-helical coiled-coil dimers; these subsequently multimerize into thick filaments via electrostatic interactions. The subfragment 2/light meromyosin "hinge" region of the MHC rod, located in the C-terminal third of heavy meromyosin, may form a less stable coiled-coil than flanking regions. Partial "melting" of this region has been proposed to result in a helix to random-coil transition. A portion of the Drosophila melanogaster MHC hinge is encoded by mutually exclusive alternative exons 15a and 15b, the use of which correlates with fast (hinge A) or slow (hinge B) muscle physiological properties. To test the functional significance of alternative hinge regions, we constructed transgenic fly lines in which fast muscle isovariant hinge A was switched for slow muscle hinge B in the MHC isoforms of indirect flight and jump muscles. Substitution of the slow muscle hinge B impaired flight ability, increased sarcomere lengths by approximately 13% and resulted in minor disruption to indirect flight muscle sarcomeric structure compared with a transgenic control. With age, residual flight ability decreased rapidly and myofibrils developed peripheral defects. Computational analysis indicates that hinge B has a greater coiled-coil propensity and thus reduced flexibility compared to hinge A. Intriguingly, the MHC rod with hinge B was approximately 5 nm longer than myosin with hinge A, consistent with the more rigid coiled-coil conformation predicted for hinge B. Our study demonstrates that hinge B cannot functionally substitute for hinge A in fast muscle types, likely as a result of differences in the molecular structure of the rod, subtle changes in myofibril structure and decreased ability to maintain sarcomere structure in indirect flight muscle myofibrils. Thus, alternative hinges are important in dictating the distinct functional properties of myosin isoforms and the muscles in which they are expressed.


Subject(s)
Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Myosin Subfragments/genetics , Myosin Subfragments/physiology , Alternative Splicing , Amino Acid Sequence , Animals , Animals, Genetically Modified , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Models, Biological , Molecular Sequence Data , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/chemistry , Muscle, Skeletal/ultrastructure , Myosin Heavy Chains/genetics , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transgenes
14.
Tsitologiia ; 48(7): 554-9, 2006.
Article in Russian | MEDLINE | ID: mdl-17087146

ABSTRACT

The effect of caldesmon (CaD) on conformational changes in F-actin modified by fluorescent probe TRITC-phalloidin was investigated by polarized fluorimetry. Changes were induced by a subfragment-1 (S-1) of myosin in the absence or presence of CaD in ghost muscle fibers obtained from intact and denervated slow (SOL) and fast (EDL) skeletal muscles of rats. S-1 binding to actin of both SOL and EDL muscles was shown to cause changes in polarized parameters of TRITC-phalloidin typical for a strong actin-myosin binding as well as of transition ofactin subunits from "off" to "on" state. CaD inhibits this significantly. Denervation atrophy inhibits the effect of S-1 as well but does not affect the capability of CaD decreasing the formation of strong binding in actomyosin complex. It is supposed that CaD "freezes" F-actin structure in "off" state. The denervation atrophy has no effect on CaD responsibility to bind thin filaments and to switch "off" actin monomers.


Subject(s)
Actins/metabolism , Calmodulin-Binding Proteins/physiology , Muscle, Skeletal/physiology , Myosins/metabolism , Actins/chemistry , Animals , Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/isolation & purification , Calmodulin-Binding Proteins/pharmacology , Male , Muscle Contraction , Muscle Denervation , Muscle, Skeletal/innervation , Myosin Subfragments/isolation & purification , Myosin Subfragments/metabolism , Myosin Subfragments/physiology , Protein Binding , Protein Conformation/drug effects , Rats , Rats, Wistar , Time Factors
15.
J Physiol ; 577(Pt 3): 935-44, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17008370

ABSTRACT

Myosin heavy chain (MHC) isoforms in vertebrate striated muscles are distinguished functionally by differences in chemomechanical kinetics. These kinetic differences may influence the cross-bridge-dependent co-operativity of thin filament Ca(2+) activation. To determine whether Ca(2+) sensitivity of unloaded thin filament sliding depends upon MHC isoform kinetics, we performed in vitro motility assays with rabbit skeletal heavy meromyosin (rsHMM) or porcine cardiac myosin (pcMyosin). Regulated thin filaments were reconstituted with recombinant human cardiac troponin (rhcTn) and alpha-tropomyosin (rhcTm) expressed in Escherichia coli. All three subunits of rhcTn were coexpressed as a functional complex using a novel construct with a glutathione S-transferase (GST) affinity tag at the N-terminus of human cardiac troponin T (hcTnT) and an intervening tobacco etch virus (TEV) protease site that allows purification of rhcTn without denaturation, and removal of the GST tag without proteolysis of rhcTn subunits. Use of this highly purified rhcTn in our motility studies resulted in a clear definition of the regulated motility profile for both fast and slow MHC isoforms. Maximum sliding speed (pCa 5) of regulated thin filaments was roughly fivefold faster with rsHMM compared with pcMyosin, although speed was increased by 1.6- to 1.9-fold for regulated over unregulated actin with both MHC isoforms. The Ca(2+) sensitivity of regulated thin filament sliding speed was unaffected by MHC isoform. Our motility results suggest that the cellular changes in isoform expression that result in regulation of myosin kinetics can occur independently of changes that influence thin filament Ca(2+) sensitivity.


Subject(s)
Actin Cytoskeleton/physiology , Calcium/metabolism , Heart/physiology , Myocardial Contraction/physiology , Myocardium/metabolism , Myosins/physiology , Animals , Cardiac Myosins/physiology , Humans , Isoenzymes/physiology , Kinetics , Myosin Subfragments/physiology , Rabbits , Recombinant Proteins/metabolism , Swine , Tropomyosin/metabolism , Troponin/metabolism , Troponin T/metabolism
16.
Curr Opin Struct Biol ; 16(2): 204-12, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16563742

ABSTRACT

Myosin filaments interact with actin to generate muscle contraction and many forms of cell motility. X-ray and electron microscopy (EM) studies have revealed the general organization of myosin molecules in relaxed filaments, but technical difficulties have prevented a detailed description. Recent studies using improved ultrastructural and image analysis techniques are overcoming these problems. Three-dimensional reconstructions using single-particle methods have provided many new insights into the organization of the myosin heads and tails. Docking of atomic structures into cryo-EM density maps suggests how regulated myosin filaments are 'switched off', bringing about muscle relaxation. Additionally, sequence analysis suggests probable interactions between myosin tails in the backbone, whereas crystallographic and EM studies are starting to reveal tail interactions directly in three dimensions.


Subject(s)
Actin Cytoskeleton/physiology , Myosins/physiology , Actin Cytoskeleton/ultrastructure , Animals , Models, Biological , Muscle Contraction , Muscle, Skeletal/physiology , Muscle, Skeletal/ultrastructure , Muscle, Smooth/physiology , Muscle, Smooth/ultrastructure , Myosin Subfragments/physiology , Myosin Subfragments/ultrastructure , Myosins/ultrastructure
17.
J Mol Biol ; 355(3): 432-42, 2006 Jan 20.
Article in English | MEDLINE | ID: mdl-16325202

ABSTRACT

In rat skeletal muscle the unloaded shortening velocity (Vo) is defined by the myosin isoform expressed in the muscle fibre. In 2001 we suggested that ADP release from actomyosin in solution (controlled by k(-AD)) was of the right size to limit Vo. However, to compare mechanical and solution kinetic data required a series of corrections to compensate for the differences in experimental conditions (0.5 M KCl, 22 degrees C for kinetic assays of myosin, 200 mM ionic strength, 12 degrees C to measure Vo). Here, a method was developed to prepare heavy meromyosin (HMM) from pure myosin isoforms isolated from single muscle fibres and to study k(-AD) (determined from the affinity of the acto-myosin complex for ADP, KAD) and the rate of ATP-induced acto-HMM dissociation (controlled by K1k+2) under the same experimental condition used to measure Vo). In fast-muscle myosin isolated from a wide range of mammalian muscles, k(-AD) was found to be too fast to limit Vo, whereas K1k+2 was of the right magnitude for ATP-induced dissociation of the cross-bridge to limit shortening velocity. The result was unexpected and prompted further experiments using the stopped-flow approach on myosin subfragment-1 (S1) and HMM obtained from bulk preparations of rabbit and rat muscle. These confirmed that the rate of cross-bridge dissociation by ATP limits the velocity of contraction for fast myosin II isoforms at 12 degrees C, while k(-AD) limits the velocity of slow myosin II isoforms. Extrapolating our data to 37 degrees C suggests that at physiological temperature the rate of ADP dissociation may limit Vo for both isoforms.


Subject(s)
Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/physiology , Muscle, Skeletal/physiology , Myosin Subfragments/physiology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , In Vitro Techniques , Kinetics , Mice , Models, Biological , Myosin Type II/physiology , Osmolar Concentration , Protein Isoforms/physiology , Rabbits , Rats , Swine , Temperature
18.
J Muscle Res Cell Motil ; 26(1): 31-7, 2005.
Article in English | MEDLINE | ID: mdl-16075160

ABSTRACT

The myosin II motor from Dictyostelium discoideum has been engineered to contain single tryptophan residues at strategic locations to probe movements of switch 1 and switch 2. The tryptophan residue at W501 probes movement of the relay helix and indirectly reports on switch 2 movement. This probe suggests that there is an equilibrium between the switch 2 open- and closed-states when the gamma-phosphate position is occupied. Actin does not appear to greatly affect this equilibrium directly, but has indirect influence via switch 1. The latter region has been probed by introducing tryptophan residues at positions 239 and 242. The kinetics of the actomyosin ATPase in solution is discussed with respect to recent crossbridge models based on high-resolution crystal structures.


Subject(s)
Movement/physiology , Myosin Subfragments/physiology , Actins/chemistry , Actins/physiology , Animals , Kinetics , Muscle, Skeletal/chemistry , Muscle, Skeletal/physiology , Myosin Subfragments/chemistry , Myosin Subfragments/ultrastructure , Time Factors , Tryptophan/chemistry , Tryptophan/physiology
19.
J Gerontol A Biol Sci Med Sci ; 60(4): 425-31, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15933379

ABSTRACT

To understand the molecular basis of the functional decline in aging muscle, we examined the functional (actomyosin ATPase) and chemical (cysteine content) changes in actin and myosin purified from the muscles of young (4- to 12-month-old) and old (27- to 35-month-old) Fisher 344 rats. Using the soluble, catalytically active myosin fragment, heavy meromyosin (HMM), we determined the maximum rate (V(max)) and actin concentration at half V(max) (K(m)) of the actomyosin ATPase, using four combinations of actin and HMM from old and young rats. V(max) and K(m) were significantly lower when both actin and HMM were obtained from old rats than when both proteins were obtained from young rats. The number of reactive cysteines in HMM significantly decreased with age, but no change was detected in the number of reactive cysteines in actin. We conclude that aging results in chemical changes in myosin (probably oxidation of cysteines) that have inhibitory effects on the actin-activated myosin ATPase.


Subject(s)
Actomyosin/physiology , Aging/physiology , Actins/analysis , Actomyosin/analysis , Aging/metabolism , Animals , Cysteine/analysis , Female , Male , Muscle Contraction/physiology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myosin Subfragments/physiology , Myosins/analysis , Myosins/physiology , Oxidation-Reduction , Rats , Rats, Inbred F344 , Structure-Activity Relationship
20.
Langmuir ; 21(8): 3213-6, 2005 Apr 12.
Article in English | MEDLINE | ID: mdl-15807553

ABSTRACT

The capability of assembling biomotors onto specific locations of solid substrates is a key for development of biomotor-based nanomechanical systems. We developed a method to direct the assembly of the heavy meromyosin fragment from rabbit skeletal muscle myosin onto specific locations of Au substrates utilizing surface molecular patterns. In this strategy, chemically directed patterns of streptavidin were achieved to direct highly specific assembly of biotinylated heavy meromyosin on the substrates--a strategy applicable for patterning a variety of biotinylated molecules--while BSA was utilized to avoid nonspecific adsorption. In vitro motility assays of filament sliding were used to confirm functionality of assembled actomyosin.


Subject(s)
Actomyosin/physiology , Gold/chemistry , Nanotechnology/methods , Actin Cytoskeleton/metabolism , Actomyosin/ultrastructure , Adsorption , Animals , Biological Assay , Biotinylation , Kinetics , Microscopy, Fluorescence , Movement , Myosin Subfragments/chemistry , Myosin Subfragments/physiology , Myosin Subfragments/ultrastructure , Rabbits , Serum Albumin, Bovine/pharmacology , Skeletal Muscle Myosins/chemistry , Skeletal Muscle Myosins/physiology , Skeletal Muscle Myosins/ultrastructure , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...