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










Publication year range
1.
PLoS One ; 7(6): e39422, 2012.
Article in English | MEDLINE | ID: mdl-22761792

ABSTRACT

The application of rapidly applied length steps to actively contracting muscle is a classic method for synchronizing the response of myosin cross-bridges so that the average response of the ensemble can be measured. Alternatively, electron tomography (ET) is a technique that can report the structure of the individual members of the ensemble. We probed the structure of active myosin motors (cross-bridges) by applying 0.5% changes in length (either a stretch or a release) within 2 ms to isometrically contracting insect flight muscle (IFM) fibers followed after 5-6 ms by rapid freezing against a liquid helium cooled copper mirror. ET of freeze-substituted fibers, embedded and thin-sectioned, provides 3-D cross-bridge images, sorted by multivariate data analysis into ~40 classes, distinct in average structure, population size and lattice distribution. Individual actin subunits are resolved facilitating quasi-atomic modeling of each class average to determine its binding strength (weak or strong) to actin. ~98% of strong-binding acto-myosin attachments present after a length perturbation are confined to "target zones" of only two actin subunits located exactly midway between successive troponin complexes along each long-pitch helical repeat of actin. Significant changes in the types, distribution and structure of actin-myosin attachments occurred in a manner consistent with the mechanical transients. Most dramatic is near disappearance, after either length perturbation, of a class of weak-binding cross-bridges, attached within the target zone, that are highly likely to be precursors of strong-binding cross-bridges. These weak-binding cross-bridges were originally observed in isometrically contracting IFM. Their disappearance following a quick stretch or release can be explained by a recent kinetic model for muscle contraction, as behaviour consistent with their identification as precursors of strong-binding cross-bridges. The results provide a detailed model for contraction in IFM that may be applicable to contraction in other types of muscle.


Subject(s)
Flight, Animal/physiology , Heteroptera/physiology , Isometric Contraction/physiology , Muscle, Skeletal/physiology , Actins/metabolism , Animals , Models, Biological , Troponin/metabolism
2.
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
3.
PLoS One ; 6(4): e18497, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21541028

ABSTRACT

Drosophila melanogaster is emerging as a powerful model system for the study of cardiac disease. Establishing peptide and protein maps of the Drosophila heart is central to implementation of protein network studies that will allow us to assess the hallmarks of Drosophila heart pathogenesis and gauge the degree of conservation with human disease mechanisms on a systems level. Using a gel-LC-MS/MS approach, we identified 1228 protein clusters from 145 dissected adult fly hearts. Contractile, cytostructural and mitochondrial proteins were most abundant consistent with electron micrographs of the Drosophila cardiac tube. Functional/Ontological enrichment analysis further showed that proteins involved in glycolysis, Ca(2+)-binding, redox, and G-protein signaling, among other processes, are also over-represented. Comparison with a mouse heart proteome revealed conservation at the level of molecular function, biological processes and cellular components. The subsisting peptidome encompassed 5169 distinct heart-associated peptides, of which 1293 (25%) had not been identified in a recent Drosophila peptide compendium. PeptideClassifier analysis was further used to map peptides to specific gene-models. 1872 peptides provide valuable information about protein isoform groups whereas a further 3112 uniquely identify specific protein isoforms and may be used as a heart-associated peptide resource for quantitative proteomic approaches based on multiple-reaction monitoring. In summary, identification of excitation-contraction protein landmarks, orthologues of proteins associated with cardiovascular defects, and conservation of protein ontologies, provides testimony to the heart-like character of the Drosophila cardiac tube and to the utility of proteomics as a complement to the power of genetics in this growing model of human heart disease.


Subject(s)
Aging/metabolism , Drosophila melanogaster/metabolism , Myocardium/metabolism , Proteome/metabolism , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/classification , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/ultrastructure , Humans , Mass Spectrometry , Mice , Molecular Sequence Annotation , Myocardium/cytology , Myocardium/ultrastructure , Peptides/metabolism , Species Specificity
4.
PLoS One ; 5(9)2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20844746

ABSTRACT

BACKGROUND: Isometric muscle contraction, where force is generated without muscle shortening, is a molecular traffic jam in which the number of actin-attached motors is maximized and all states of motor action are trapped with consequently high heterogeneity. This heterogeneity is a major limitation to deciphering myosin conformational changes in situ. METHODOLOGY: We used multivariate data analysis to group repeat segments in electron tomograms of isometrically contracting insect flight muscle, mechanically monitored, rapidly frozen, freeze substituted, and thin sectioned. Improved resolution reveals the helical arrangement of F-actin subunits in the thin filament enabling an atomic model to be built into the thin filament density independent of the myosin. Actin-myosin attachments can now be assigned as weak or strong by their motor domain orientation relative to actin. Myosin attachments were quantified everywhere along the thin filament including troponin. Strong binding myosin attachments are found on only four F-actin subunits, the "target zone", situated exactly midway between successive troponin complexes. They show an axial lever arm range of 77°/12.9 nm. The lever arm azimuthal range of strong binding attachments has a highly skewed, 127° range compared with X-ray crystallographic structures. Two types of weak actin attachments are described. One type, found exclusively in the target zone, appears to represent pre-working-stroke intermediates. The other, which contacts tropomyosin rather than actin, is positioned M-ward of the target zone, i.e. the position toward which thin filaments slide during shortening. CONCLUSION: We present a model for the weak to strong transition in the myosin ATPase cycle that incorporates azimuthal movements of the motor domain on actin. Stress/strain in the S2 domain may explain azimuthal lever arm changes in the strong binding attachments. The results support previous conclusions that the weak attachments preceding force generation are very different from strong binding attachments.


Subject(s)
Actins/chemistry , Actins/metabolism , Insect Proteins/chemistry , Insect Proteins/metabolism , Insecta/physiology , Myosins/chemistry , Myosins/metabolism , Animals , Cryopreservation , Crystallography, X-Ray , Electron Microscope Tomography , Flight, Animal , Insecta/chemistry , Isometric Contraction , Models, Molecular , Muscles/chemistry , Muscles/physiology , Protein Binding , Protein Structure, Tertiary , Tissue Fixation
5.
J Struct Biol ; 168(3): 485-502, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19698791

ABSTRACT

During active muscle contraction, tension is generated through many simultaneous, independent interactions between the molecular motor myosin and the actin filaments. The ensemble of myosin motors displays heterogeneous conformations reflecting different mechanochemical steps of the ATPase pathway. We used electron tomography of actively contracting insect flight muscle fast-frozen, freeze substituted, Araldite embedded, thin-sectioned and stained, to obtain 3D snapshots of the multiplicity of actin-attached myosin structures. We describe procedures for alignment of the repeating lattice of sub-volumes (38.7 nm cross-bridge repeats bounded by troponin) and multivariate data analysis to identify self-similar repeats for computing class averages. Improvements in alignment and classification of repeat sub-volumes reveals (for the first time in active muscle images) the helix of actin subunits in the thin filament and the troponin density with sufficient clarity that a quasiatomic model of the thin filament can be built into the class averages independent of the myosin cross-bridges. We show how quasiatomic model building can identify both strong and weak myosin attachments to actin. We evaluate the accuracy of image classification to enumerate the different types of actin-myosin attachments.


Subject(s)
Electron Microscope Tomography/methods , Insecta/ultrastructure , Muscle Contraction/physiology , Animals , Muscles/pathology
6.
J Struct Biol ; 168(2): 240-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19635572

ABSTRACT

X-ray diffraction of the indirect flight muscle (IFM) in living Drosophila at rest and electron microscopy of intact and glycerinated IFM was used to compare the effects of mutations in the regulatory light chain (RLC) on sarcomeric structure. Truncation of the RLC N-terminal extension (Dmlc2(Delta2-46)) or disruption of the phosphorylation sites by substituting alanines (Dmlc2(S66A, S67A)) decreased the equatorial intensity ratio (I(20)/I(10)), indicating decreased myosin mass associated with the thin filaments. Phosphorylation site disruption (Dmlc2(S66A, S67A)), but not N-terminal extension truncation (Dmlc2(Delta2-46)), decreased the 14.5nm reflection intensity, indicating a spread of the axial distribution of the myosin heads. The arrangement of thick filaments and myosin heads in electron micrographs of the phosphorylation mutant (Dmlc2(S66A, S67A)) appeared normal in the relaxed and rigor states, but when calcium activated, fewer myosin heads formed cross-bridges. In transgenic flies with both alterations to the RLC (Dmlc2(Delta2-46; S66A, S67A)), the effects of the dual mutation were additive. The results suggest that the RLC N-terminal extension serves as a "tether" to help pre-position the myosin heads for attachment to actin, while phosphorylation of the RLC promotes head orientations that allow optimal interactions with the thin filament.


Subject(s)
Drosophila melanogaster/metabolism , Drosophila melanogaster/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myosin Light Chains/metabolism , Myosin Light Chains/ultrastructure , Animals , Microscopy, Electron , Myosin Light Chains/chemistry , Phosphorylation , X-Ray Diffraction
7.
J Mol Biol ; 381(3): 519-28, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18588896

ABSTRACT

The structure and function of myosin crossbridges in asynchronous insect flight muscle (IFM) have been elucidated in situ using multiple approaches. These include generating "atomic" models of myosin in multiple contractile states by rebuilding the crystal structure of chicken subfragment 1 (S1) to fit IFM crossbridges in lower-resolution electron microscopy tomograms and by "mapping" the functional effects of genetically substituted, isoform-specific domains, including the converter domain, in chimeric IFM myosin to sequences in the crystal structure of chicken S1. We prepared helical reconstructions (approximately 25 A resolution) to compare the structural characteristics of nucleotide-free myosin0 S1 bound to actin (acto-S1) isolated from chicken skeletal muscle (CSk) and the flight muscles of Lethocerus (Leth) wild-type Drosophila (wt Dros) and a Drosophila chimera (IFI-EC) wherein the converter domain of the indirect flight muscle myosin isoform has been replaced by the embryonic skeletal myosin converter domain. Superimposition of the maps of the frozen-hydrated acto-S1 complexes shows that differences between CSk and IFM S1 are limited to the azimuthal curvature of the lever arm: the regulatory light-chain (RLC) region of chicken skeletal S1 bends clockwise (as seen from the pointed end of actin) while those of IFM S1 project in a straight radial direction. All the IFM S1s are essentially identical other than some variation in the azimuthal spread of density in the RLC region. This spread is most pronounced in the IFI-EC S1, consistent with proposals that the embryonic converter domain increases the compliance of the IFM lever arm affecting the function of the myosin motor. These are the first unconstrained models of IFM S1 bound to actin and the first direct comparison of the vertebrate and invertebrate skeletal myosin II classes, the latter for which, data on the structure of discrete acto-S1 complexes, are not readily available.


Subject(s)
Actins/chemistry , Molecular Motor Proteins/chemistry , Muscle, Skeletal/chemistry , Myosin Subfragments/chemistry , Animals , Chickens , Drosophila , Flight, Animal , Heteroptera , Models, Biological , Muscle Fibers, Skeletal/chemistry , Protein Binding , Protein Isoforms/chemistry
8.
RNA ; 14(3): 445-53, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18192611

ABSTRACT

The process of mRNA localization typically utilizes cis-targeting elements and trans-recognition factors to direct the compartmental organization of translationally suppressed mRNAs. mRNA localization to the endoplasmic reticulum (ER), in contrast, occurs via a co-translational, signal sequence/signal recognition particle (SRP)-dependent mechanism. We have utilized cell fractionation/cDNA microarray analysis, shRNA-mediated suppression of SRP expression, and mRNA reporter construct studies to define the role of the SRP pathway in ER-directed mRNA localization. Cell fractionation studies of mRNA partitioning between the cytosol and ER demonstrated the expected enrichment of cytosolic/nucleoplasmic protein-encoding mRNAs and secretory/integral membrane protein-encoding mRNAs in the cytosol and ER fractions, respectively, and identified a subpopulation of cytosolic/nucleoplasmic protein-encoding mRNAs in the membrane-bound mRNA pool. The latter finding suggests a signal sequence-independent pathway of ER-directed mRNA localization. Extending from these findings, mRNA partitioning was examined in stable SRP54 shRNA knockdown HeLa cell lines. shRNA-directed reductions in SRP did not globally alter mRNA partitioning patterns, although defects in membrane protein processing were observed, further suggesting the existence of multiple pathways for mRNA localization to the ER. ER localization of GRP94-encoding mRNA was observed when translation was disabled by mutation of the start codon/insertion of a 5'UTR stem-loop structure or upon deletion of the encoded signal sequence. Combined, these data indicate that the mRNA localization to the ER can be conferred independent of the signal sequence/SRP pathway and suggest that mRNA localization to the ER may utilize cis-encoded targeting information.


Subject(s)
Endoplasmic Reticulum/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Recognition Particle/genetics , Signal Recognition Particle/metabolism , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , DNA Primers/genetics , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis , Subcellular Fractions/metabolism
9.
Muscle Nerve ; 36(1): 71-80, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17455272

ABSTRACT

Because previous studies of three-dimensional skeletal muscle cultures have shown limited differentiation, the goal of this study was to establish conditions that would produce mature sarcomeres in a mammalian-derived skeletal muscle construct. We evaluated the differentiation of bioartificial muscles generated from C(2)C(12) myoblasts in a collagen gel cultured under steady, passive tension for up to 36 days. Staining for alpha-actinin, myosin, and F-actin indicated the presence of striated fibers as early as 6 days post-differentiation. Electron microscopy at 16 days post-differentiation revealed multinucleated myotubes with ordered, striated myofibers. At 33 days, the cultures contained collagen fibers and showed localization of paxillin at the fiber termini, suggesting that myotendinous junctions were forming. The present study demonstrates mature muscle synthesis in a three-dimensional system using a pure mammalian myoblast cell line. Our results suggest that this culture model can be used to evaluate the effects of various mechanical and biochemical cues on muscle development under normal and pathological conditions.


Subject(s)
Cell Differentiation/physiology , Collagen/physiology , Muscle, Skeletal/cytology , Sarcomeres/ultrastructure , Animals , Cell Culture Techniques , Cells, Cultured , Imaging, Three-Dimensional , Mice , Mice, Inbred C3H , Microscopy, Electron, Transmission/methods
11.
J Mol Biol ; 362(4): 844-60, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16949613

ABSTRACT

Subfragment 2 (S2), the segment that links the two myosin heads to the thick filament backbone, may serve as a swing-out adapter allowing crossbridge access to actin, as the elastic component of crossbridges and as part of a phosphorylation-regulated on-off switch for crossbridges in smooth muscle. Low-salt expansion increases interfilament spacing (from 52 nm to 67 nm) of rigor insect flight muscle fibers and exposes a tethering segment of S2 in many crossbridges. Docking an actoS1 atomic model into EM tomograms of swollen rigor fibers identifies in situ for the first time the location, length and angle assignable to a segment of S2. Correspondence analysis of 1831 38.7 nm crossbridge repeats grouped self-similar forms from which class averages could be computed. The full range of the variability in angles and lengths of exposed S2 was displayed by using class averages for atomic fittings of acto-S1, while S2 was modeled by fitting a length of coiled-coil to unaveraged individual repeats. This hybrid modeling shows that the average length of S2 tethers along the thick filament (except near the tapered ends) is approximately 10 nm, or 16% of S2's total length, with an angular range encompassing 90 degrees axially and 120 degrees azimuthally. The large range of S2 angles indicates that some rigor bridges produce positive force that must be balanced by others producing drag force. The short tethering segment clarifies constraints on the function of S2 in accommodating variable myosin head access to actin. We suggest that the short length of S2 may also favor intermolecular head-head interactions in IFM relaxed thick filaments.


Subject(s)
Flight, Animal , Insecta/ultrastructure , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure , Myosins/chemistry , Myosins/ultrastructure , Tomography , Animals , Models, Molecular , Muscle Rigidity/pathology , Protein Structure, Tertiary
12.
J Mol Biol ; 361(5): 823-38, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16887144

ABSTRACT

Low-angle X-ray diffraction patterns from relaxed fruitfly (Drosophila) flight muscle recorded on the BioCat beamline at the Argonne Advanced Photon Source (APS) show many features similar to such patterns from the "classic" insect flight muscle in Lethocerus, the giant water bug, but there is a characteristically different pattern of sampling of the myosin filament layer-lines, which indicates the presence of a superlattice of myosin filaments in the Drosophila A-band. We show from analysis of the structure factor for this lattice that the sampling pattern is exactly as expected if adjacent four-stranded myosin filaments, of repeat 116 nm, are axially shifted in the hexagonal A-band lattice by one-third of the 14.5 nm axial spacing between crowns of myosin heads. In addition, electron micrographs of Drosophila and other flies (e.g. the house fly (Musca) and the flesh fly (Sarcophaga)) combined with image processing confirm that the same A-band superlattice occurs in all of these flies; it may be a general property of the Diptera. The different A-band organisation in flies compared with Lethocerus, which operates at a much lower wing beat frequency (approximately 30 Hz) and requires a warm-up period, may be a way of optimising the myosin and actin filament geometry needed both for stretch activation at the higher wing beat frequencies (50 Hz to 1000 Hz) of flies and their need for a rapid escape response.


Subject(s)
Actin Cytoskeleton/chemistry , Drosophila/metabolism , Flight, Animal/physiology , Muscles/chemistry , Myosins/chemistry , Animals , Computer Simulation , Female , Models, Biological , Muscles/ultrastructure , Structure-Activity Relationship , X-Ray Diffraction
13.
Proc Natl Acad Sci U S A ; 103(5): 1394-9, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16432241

ABSTRACT

Drosophila melanogaster genetics provides the advantage of molecularly defined P-element insertions and deletions that span the entire genome. Although Drosophila has been extensively used as a model system to study heart development, it has not been used to dissect the genetics of adult human heart disease because of an inability to phenotype the adult fly heart in vivo. Here we report the development of a strategy to measure cardiac function in awake adult Drosophila that opens the field of Drosophila genetics to the study of human dilated cardiomyopathies. Through the application of optical coherence tomography, we accurately distinguish between normal and abnormal cardiac function based on measurements of internal cardiac chamber dimensions in vivo. Normal Drosophila have a fractional shortening of 87 +/- 4%, whereas cardiomyopathic flies that contain a mutation in troponin I or tropomyosin show severe impairment of systolic function. To determine whether the fly can be used as a model system to recapitulate human dilated cardiomyopathy, we generated transgenic Drosophila with inducible cardiac expression of a mutant of human delta-sarcoglycan (deltasg(S151A)), which has previously been associated with familial dilated cardiomyopathy. Compared to transgenic flies overexpressing wild-type deltasg, or the standard laboratory strain w(1118), Drosophila expressing deltasg(S151A) developed marked impairment of systolic function and significantly enlarged cardiac chambers. These data illustrate the utility of Drosophila as a model system to study dilated cardiomyopathy and the applicability of the vast genetic resources available in Drosophila to systematically study the genetic mechanisms responsible for human cardiac disease.


Subject(s)
Cardiomyopathy, Dilated/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Heart Diseases/genetics , Animals , Cardiomyopathy, Dilated/pathology , Crosses, Genetic , DNA, Complementary/metabolism , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Female , Heart/physiology , Homozygote , Male , Microscopy, Electron , Models, Biological , Models, Genetic , Mutation , Myocardium/ultrastructure , Point Mutation , Sarcoplasmic Reticulum/metabolism , Time Factors , Tomography , Transgenes , Tropomyosin/genetics , Troponin I/metabolism , Wings, Animal
14.
J Struct Biol ; 147(3): 268-82, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15450296

ABSTRACT

As a first step toward freeze-trapping and 3-D modeling of the very rapid load-induced structural responses of active myosin heads, we explored the conformational range of longer lasting force-dependent changes in rigor crossbridges of insect flight muscle (IFM). Rigor IFM fibers were slam-frozen after ramp stretch (1000 ms) of 1-2% and freeze-substituted. Tomograms were calculated from tilt series of 30 nm longitudinal sections of Araldite-embedded fibers. Modified procedures of alignment and correspondence analysis grouped self-similar crossbridge forms into 16 class averages with 4.5 nm resolution, revealing actin protomers and myosin S2 segments of some crossbridges for the first time in muscle thin sections. Acto-S1 atomic models manually fitted to crossbridge density required a range of lever arm adjustments to match variably distorted rigor crossbridges. Some lever arms were unchanged compared with low tension rigor, while others were bent and displaced M-ward by up to 4.5 nm. The average displacement was 1.6 +/- 1.0 nm. "Map back" images that replaced each unaveraged 39 nm crossbridge motif by its class average showed an ordered mix of distorted and unaltered crossbridges distributed along the 116 nm repeat that reflects differences in rigor myosin head loading even before stretch.


Subject(s)
Myosins/chemistry , Animals , Cryoelectron Microscopy/instrumentation , Cryoelectron Microscopy/methods , Flight, Animal , Insecta , Models, Molecular , Protein Conformation , Stress, Mechanical , Synchrotrons , Tomography/methods , X-Ray Diffraction/methods
15.
Biophys J ; 87(2): 1101-11, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15298914

ABSTRACT

Asynchronous insect flight muscle is specialized for myogenic oscillatory work, but can also produce isometric tetanic contraction. In skinned insect flight muscle fibers from Lethocerus, with sarcomere length monitored by a striation follower, we determined the relation between isometric force (F(0)) at serial increments of [Ca(2+)] and the additional active force recruited at each [Ca(2+)] by a stretch of approximately 12 nm per half-sarcomere (F(SA)). The isometric force-pCa relation shows that 1.5-2 units of pCa are necessary to raise isometric force from its threshold (pCa approximately 6.5) to its maximum (F(0,max)). The amplitude of F(SA) depends only on the preceding baseline level of isometric force, which must reach at least 0.05 F(0,max) to enable stretch-activation. F(SA) rises very steeply to its maximum as F(0) reaches approximately 0.2 F(0,max), then decreases as F(0) increases so as to produce a constant sum (F(0) + F(SA)) = F(max). Thus Ca- and stretch-activation are complementary pathways that trigger a common process of cross-bridge attachment and force production. We suggest that stretch-induced distortion of attached cross-bridges relieves the steric blocking by tropomyosin of additional binding sites on actin, thereby enabling maximum force even at low [Ca(2+)].


Subject(s)
Calcium/pharmacology , Flight, Animal/physiology , Heteroptera/physiology , Isometric Contraction/physiology , Mechanotransduction, Cellular/physiology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Heteroptera/drug effects , Isometric Contraction/drug effects , Magnesium/pharmacology , Mechanotransduction, Cellular/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Physical Stimulation/methods , Stress, Mechanical
16.
Biophys J ; 86(5): 3009-19, 2004 May.
Article in English | MEDLINE | ID: mdl-15111415

ABSTRACT

Electron micrographic tomograms of isometrically active insect flight muscle, freeze substituted after rapid freezing, show binding of single myosin heads at varying angles that is largely restricted to actin target zones every 38.7 nm. To quantify the parameters that govern this pattern, we measured the number and position of attached myosin heads by tracing cross-bridges through the three-dimensional tomogram from their origins on 14.5-nm-spaced shelves along the thick filament to their thin filament attachments in the target zones. The relationship between the probability of cross-bridge formation and axial offset between the shelf and target zone center was well fitted by a Gaussian distribution. One head of each myosin whose origin is close to an actin target zone forms a cross-bridge most of the time. The probability of cross-bridge formation remains high for myosin heads originating within 8 nm axially of the target zone center and is low outside 12 nm. We infer that most target zone cross-bridges are nearly perpendicular to the filaments (60% within 11 degrees ). The results suggest that in isometric contraction, most cross-bridges maintain tension near the beginning of their working stroke at angles near perpendicular to the filament axis. Moreover, in the absence of filament sliding, cross-bridges cannot change tilt angle while attached nor reach other target zones while detached, so may cycle repeatedly on and off the same actin target monomer.


Subject(s)
Actins/chemistry , Muscle Fibers, Skeletal/cytology , Actins/metabolism , Adenosine Triphosphate/chemistry , Animals , Calcium/metabolism , Flight, Animal , Hemiptera , Image Processing, Computer-Assisted , Microscopy, Electron , Muscle Contraction , Muscles/metabolism , Myosin Subfragments/chemistry , Myosins/chemistry , Normal Distribution
17.
RNA ; 9(9): 1123-37, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12923260

ABSTRACT

In eukaryotic cells, it is generally accepted that protein synthesis is compartmentalized; soluble proteins are synthesized on free ribosomes, whereas secretory and membrane proteins are synthesized on endoplasmic reticulum (ER)-bound ribosomes. The partitioning of mRNAs that accompanies such compartmentalization arises early in protein synthesis, when ribosomes engaged in the translation of mRNAs encoding signal-sequence-bearing proteins are targeted to the ER. In this report, we use multiple cell fractionation protocols, in combination with cDNA microarray, nuclease protection, and Northern blot analyses, to assess the distribution of mRNAs between free and ER-bound ribosomes. We find a broad representation of mRNAs encoding soluble proteins in the ER fraction, with a subset of such mRNAs displaying substantial ER partitioning. In addition, we present evidence that membrane-bound ribosomes engage in the translation of mRNAs encoding soluble proteins. Single-cell in situ hybridization analysis of the subcellular distribution of mRNAs encoding ER-localized and soluble proteins identify two overall patterns of mRNA distribution in the cell-endoplasmic reticular and cytosolic. However, both partitioning patterns include a distinct perinuclear component. These results identify previously unappreciated roles for membrane-bound ribosomes in the subcellular compartmentalization of protein synthesis and indicate possible functions for the perinuclear membrane domain in mRNA sorting in the cell.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Ribosomes/metabolism , Cell Fractionation , DNA, Complementary/genetics , Humans , Jurkat Cells , Microscopy, Electron , Oligonucleotide Array Sequence Analysis
18.
Am J Physiol Heart Circ Physiol ; 284(5): H1827-38, 2003 May.
Article in English | MEDLINE | ID: mdl-12543641

ABSTRACT

Tropomodulins are a family of proteins that cap the slow-growing end of actin filaments. Erythrocyte tropomodulin (E-Tmod) stabilizes short actin protofilaments in erythrocytes and caps longer sarcomeric actin filaments in striated muscles. We report the knockin of the beta-galactosidase gene (LacZ) under the control of the endogenous E-Tmod promoter and the knockout of E-Tmod in mouse embryonic stem cells. E-Tmod(-/-) embryos die around embryonic day 10 and exhibit a noncontractile heart tube with disorganized myofibrils and underdevelopment of the right ventricle, accumulation of mechanically weakened primitive erythroid cells in the yolk sac, and failure of primary capillary plexuses to remodel into vitelline vessels, all required to establish blood circulation between the yolk sac and the embryo proper. We propose a hemodynamic "plexus channel selection" mechanism as the basis for vitelline vascular remodeling. The defects in cardiac contractility, vitelline circulation, and hematopoiesis reflect an essential role for E-Tmod capping of the actin filaments in both assembly of cardiac sarcomeres and of the membrane skeleton in erythroid cells that is not compensated for by other proteins.


Subject(s)
Actin Cytoskeleton/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Erythroid Precursor Cells/metabolism , Microfilament Proteins , Neovascularization, Physiologic/physiology , Animals , Biomarkers , Embryo, Mammalian/blood supply , Embryo, Mammalian/physiology , Erythrocytes/physiology , Gene Expression Regulation, Developmental , Heart/embryology , Heart/physiology , Hematopoiesis/genetics , Lac Operon , Mice , Mice, Knockout , Microscopy, Electron , Myocardial Contraction/genetics , Myocytes, Cardiac/ultrastructure , Phenotype , Rotation , Stress, Mechanical , Tropomodulin , Yolk Sac/blood supply , Yolk Sac/physiology
19.
J Struct Biol ; 138(1-2): 92-104, 2002.
Article in English | MEDLINE | ID: mdl-12160705

ABSTRACT

Electron tomography, correspondence analysis, molecular model building, and real-space refinement provide detailed 3-D structures for in situ myosin crossbridges in the nucleotide-free state (rigor), thought to represent the end of the power stroke. Unaveraged tomograms from a 25-nm longitudinal section of insect flight muscle preserved native structural variation. Recurring crossbridge motifs that repeat every 38.7 nm along the actin filament were extracted from the tomogram and classified by correspondence analysis into 25 class averages, which improved the signal to noise ratio. Models based on the atomic structures of actin and of myosin subfragment 1 were rebuilt to fit 11 class averages. A real-space refinement procedure was applied to quantitatively fit the reconstructions and to minimize steric clashes between domains introduced during the fitting. These combined procedures show that no single myosin head structure can fit all the in situ crossbridges. The validity of the approach is supported by agreement of these atomic models with fluorescent probe data from vertebrate muscle as well as with data from regulatory light chain crosslinking between heads of smooth muscle heavy meromyosin when bound to actin.


Subject(s)
Models, Molecular , Muscle, Skeletal/chemistry , Tomography, X-Ray Computed , Actomyosin/chemistry , Actomyosin/ultrastructure , Animals , Flight, Animal , Hemiptera , Insecta , Microscopy, Electron , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/chemistry , Muscle Proteins/ultrastructure , Muscle, Skeletal/ultrastructure
20.
J Biol Chem ; 277(29): 26642-51, 2002 Jul 19.
Article in English | MEDLINE | ID: mdl-12011079

ABSTRACT

Endocytosis of ligand-activated receptors requires dynamin-mediated GTP hydrolysis, which is regulated by dynamin self-assembly. Here, we demonstrate that phosphorylation of dynamin I by c-Src induces its self-assembly and increases its GTPase activity. Electron microscopic analyses reveal that tyrosine-phosphorylated dynamin I spontaneously self-assembles into large stacks of rings. Tyrosine 597 was identified as being phosphorylated both in vitro and in cultured cells following epidermal growth factor receptor stimulation. The replacement of tyrosine 597 with phenylalanine impairs Src kinase-induced dynamin I self-assembly and GTPase activity in vitro. Expression of Y597F dynamin I in cells attenuates agonist-driven epidermal growth factor receptor internalization. Thus, c-Src-mediated tyrosine phosphorylation is required for the function of dynamin in ligand-induced signaling receptor internalization.


Subject(s)
Endocytosis , ErbB Receptors/metabolism , GTP Phosphohydrolases/metabolism , src-Family Kinases/metabolism , Animals , COS Cells , CSK Tyrosine-Protein Kinase , Dynamin I , Dynamins , Ligands , Phosphorylation , Protein Conformation , Protein-Tyrosine Kinases/metabolism , Rats , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...