Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Nature ; 588(7838): 521-525, 2020 12.
Article in English | MEDLINE | ID: mdl-33268893

ABSTRACT

Myosin II is the motor protein that enables muscle cells to contract and nonmuscle cells to move and change shape1. The molecule has two identical heads attached to an elongated tail, and can exist in two conformations: 10S and 6S, named for their sedimentation coefficients2,3. The 6S conformation has an extended tail and assembles into polymeric filaments, which pull on actin filaments to generate force and motion. In 10S myosin, the tail is folded into three segments and the heads bend back and interact with each other and the tail3-7, creating a compact conformation in which ATPase activity, actin activation and filament assembly are all highly inhibited7,8. This switched-off structure appears to function as a key energy-conserving storage molecule in muscle and nonmuscle cells9-12, which can be activated to form functional filaments as needed13-but the mechanism of its inhibition is not understood. Here we have solved the structure of smooth muscle 10S myosin by cryo-electron microscopy with sufficient resolution to enable improved understanding of the function of the head and tail regions of the molecule and of the key intramolecular contacts that cause inhibition. Our results suggest an atomic model for the off state of myosin II, for its activation and unfolding by phosphorylation, and for understanding the clustering of disease-causing mutations near sites of intramolecular interaction.


Subject(s)
Cryoelectron Microscopy , Myosin Type II/antagonists & inhibitors , Myosin Type II/ultrastructure , Animals , Binding Sites , Models, Molecular , Muscle, Smooth/chemistry , Mutation , Myosin Type II/chemistry , Myosin Type II/genetics , Phosphorylation , Protein Binding , Protein Conformation , Protein Unfolding , Turkeys
2.
J Am Chem Soc ; 146(26): 17747-17756, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38889317

ABSTRACT

Unveiling molecular mechanisms that dominate protein phase dynamics has been a pressing need for deciphering the intricate intracellular modulation machinery. While ions and biomacromolecules have been widely recognized for modulating protein phase separations, effects of small molecules that essentially constitute the cytosolic chemical atmosphere on the protein phase behaviors are rarely understood. Herein, we report that vitamin C (VC), a key small molecule for maintaining a reductive intracellular atmosphere, drives reentrant phase transitions of myosin II/F-actin (actomyosin) cytoskeletons. The actomyosin bundle condensates dissemble in the low-VC regime and assemble in the high-VC regime in vitro or inside neuronal cells, through a concurrent myosin II protein aggregation-dissociation process with monotonic VC concentration increase. Based on this finding, we employ in situ single-cell and single-vesicle electrochemistry to demonstrate the quantitative modulation of catecholamine transmitter vesicle exocytosis by intracellular VC atmosphere, i.e., exocytotic release amount increases in the low-VC regime and decreases in the high-VC regime. Furthermore, we show how VC regulates cytomembrane-vesicle fusion pore dynamics through counteractive or synergistic effects of actomyosin phase transitions and the intracellular free calcium level on membrane tensions. Our work uncovers the small molecule-based reversive protein phase regulatory mechanism, paving a new way to chemical neuromodulation and therapeutic repertoire expansion.


Subject(s)
Actins , Ascorbic Acid , Exocytosis , Ascorbic Acid/chemistry , Ascorbic Acid/pharmacology , Exocytosis/drug effects , Actins/metabolism , Actins/chemistry , Phase Transition , Animals , Myosin Type II/metabolism , Myosin Type II/antagonists & inhibitors , Electrochemical Techniques , Actomyosin/metabolism , Actomyosin/chemistry , Rats
3.
Development ; 147(3)2020 02 04.
Article in English | MEDLINE | ID: mdl-31964776

ABSTRACT

Directional cell intercalations of epithelial cells during gastrulation has, in several organisms, been shown to be associated with a planar cell polarity in the organisation of the actin-myosin cytoskeleton and is postulated to reflect directional tension that drives oriented cell intercalations. We have characterised and applied a recently introduced non-destructive optical manipulation technique to measure the tension in individual epithelial cell junctions of cells in various locations and orientations in the epiblast of chick embryos in the early stages of primitive streak formation. Junctional tension of mesendoderm precursors in the epiblast is higher in junctions oriented in the direction of intercalation than in junctions oriented perpendicular to the direction of intercalation and higher than in junctions of other cells in the epiblast. The kinetic data fit best with a simple viscoelastic Maxwell model, and we find that junctional tension, and to a lesser extent viscoelastic relaxation time, are dependent on myosin activity.


Subject(s)
Epithelial Cells/metabolism , Gastrulation/physiology , Intercellular Junctions/metabolism , Optical Tweezers , Primitive Streak/growth & development , Animals , Animals, Genetically Modified , Cell Movement/physiology , Cell Polarity/physiology , Chick Embryo , Gastrula/metabolism , Germ Layers/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hydrocarbons, Chlorinated/pharmacology , Microscopy, Fluorescence/methods , Myosin Type I/antagonists & inhibitors , Myosin Type I/metabolism , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Pyrroles/pharmacology , Signal Transduction/physiology
4.
EMBO Rep ; 20(8): e47047, 2019 08.
Article in English | MEDLINE | ID: mdl-31379129

ABSTRACT

We identify a novel endothelial membrane behaviour in transgenic zebrafish. Cerebral blood vessels extrude large transient spherical structures that persist for an average of 23 min before regressing into the parent vessel. We term these structures "kugeln", after the German for sphere. Kugeln are only observed arising from the cerebral vessels and are present as late as 28 days post fertilization. Kugeln do not communicate with the vessel lumen and can form in the absence of blood flow. They contain little or no cytoplasm, but the majority are highly positive for nitric oxide reactivity. Kugeln do not interact with brain lymphatic endothelial cells (BLECs) and can form in their absence, nor do they perform a scavenging role or interact with macrophages. Inhibition of actin polymerization, Myosin II, or Notch signalling reduces kugel formation, while inhibition of VEGF or Wnt dysregulation (either inhibition or activation) increases kugel formation. Kugeln represent a novel Notch-dependent NO-containing endothelial organelle restricted to the cerebral vessels, of currently unknown function.


Subject(s)
Blood Vessels/cytology , Brain/cytology , Endothelial Cells/ultrastructure , Gene Expression Regulation, Developmental , Neovascularization, Physiologic/genetics , Zebrafish/embryology , Actins/antagonists & inhibitors , Actins/genetics , Actins/metabolism , Animals , Animals, Genetically Modified , Blood Vessels/embryology , Blood Vessels/metabolism , Blood Vessels/ultrastructure , Brain/blood supply , Brain/embryology , Brain/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cerebrovascular Circulation/genetics , Embryo, Nonmammalian , Endothelial Cells/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Myosin Type II/antagonists & inhibitors , Myosin Type II/genetics , Myosin Type II/metabolism , Nitric Oxide/metabolism , Organelles/metabolism , Organelles/ultrastructure , Polymerization/drug effects , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Thiazolidines/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 115(9): E1991-E2000, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29444861

ABSTRACT

Electron microscope studies have shown that the switched-off state of myosin II in muscle involves intramolecular interaction between the two heads of myosin and between one head and the tail. The interaction, seen in both myosin filaments and isolated molecules, inhibits activity by blocking actin-binding and ATPase sites on myosin. This interacting-heads motif is highly conserved, occurring in invertebrates and vertebrates, in striated, smooth, and nonmuscle myosin IIs, and in myosins regulated by both Ca2+ binding and regulatory light-chain phosphorylation. Our goal was to determine how early this motif arose by studying the structure of inhibited myosin II molecules from primitive animals and from earlier, unicellular species that predate animals. Myosin II from Cnidaria (sea anemones, jellyfish), the most primitive animals with muscles, and Porifera (sponges), the most primitive of all animals (lacking muscle tissue) showed the same interacting-heads structure as myosins from higher animals, confirming the early origin of the motif. The social amoeba Dictyostelium discoideum showed a similar, but modified, version of the motif, while the amoeba Acanthamoeba castellanii and fission yeast (Schizosaccharomyces pombe) showed no head-head interaction, consistent with the different sequences and regulatory mechanisms of these myosins compared with animal myosin IIs. Our results suggest that head-head/head-tail interactions have been conserved, with slight modifications, as a mechanism for regulating myosin II activity from the emergence of the first animals and before. The early origins of these interactions highlight their importance in generating the inhibited (relaxed) state of myosin in muscle and nonmuscle cells.


Subject(s)
Myosin Type II/antagonists & inhibitors , Actins/chemistry , Adenosine Triphosphate/chemistry , Amino Acid Motifs , Animals , Biological Evolution , Calcium/chemistry , Cell Line , Computational Biology , Cryoelectron Microscopy , Dictyostelium , Image Processing, Computer-Assisted , Insecta , Microscopy, Electron , Myosin Type II/chemistry , Phosphorylation , Porifera , Protein Binding , Schizosaccharomyces , Scyphozoa , Sea Anemones , Turkeys
6.
Proc Natl Acad Sci U S A ; 114(36): 9529-9534, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827333

ABSTRACT

Active transport in the cytoplasm plays critical roles in living cell physiology. However, the mechanical resistance that intracellular compartments experience, which is governed by the cytoplasmic material property, remains elusive, especially its dependence on size and speed. Here we use optical tweezers to drag a bead in the cytoplasm and directly probe the mechanical resistance with varying size a and speed V We introduce a method, combining the direct measurement and a simple scaling analysis, to reveal different origins of the size- and speed-dependent resistance in living mammalian cytoplasm. We show that the cytoplasm exhibits size-independent viscoelasticity as long as the effective strain rate V/a is maintained in a relatively low range (0.1 s-1 < V/a < 2 s-1) and exhibits size-dependent poroelasticity at a high effective strain rate regime (5 s-1 < V/a < 80 s-1). Moreover, the cytoplasmic modulus is found to be positively correlated with only V/a in the viscoelastic regime but also increases with the bead size at a constant V/a in the poroelastic regime. Based on our measurements, we obtain a full-scale state diagram of the living mammalian cytoplasm, which shows that the cytoplasm changes from a viscous fluid to an elastic solid, as well as from compressible material to incompressible material, with increases in the values of two dimensionless parameters, respectively. This state diagram is useful to understand the underlying mechanical nature of the cytoplasm in a variety of cellular processes over a broad range of speed and size scales.


Subject(s)
Cytoplasm/chemistry , Cytoplasm/physiology , Adenosine Triphosphate/metabolism , Animals , Biomechanical Phenomena , Cytoplasm/drug effects , Cytoskeleton/chemistry , Elasticity , Epithelial Cells/cytology , HeLa Cells/cytology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Kidney/cytology , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Optical Tweezers , Rats , Viscosity
7.
Biochemistry ; 56(41): 5526-5538, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28898058

ABSTRACT

SM22α, also named transgelin, is an actin filament-associated protein in smooth muscle and fibroblasts. Three decades after its discovery, the biological function of SM22α remains under investigation. Here we report a novel finding that the expression and degradation of SM22α/transgelin are regulated by mechanical tension. Following a mass spectrometry identification of SM22α degradation in isolated and tension-unloaded mouse aorta, we developed specific monoclonal antibodies to study the regulation of SM22α in human fetal lung myofibroblast line MRC-5 and primary cultures of neonatal mouse skin fibroblasts. The level of SM22α is positively related to the mechanical tension in the cytoskeleton produced by the myosin II motor in response to the stiffness of the culture matrix. Quantitative reverse transcription polymerase chain reaction demonstrated that the expression of SM22α is regulated at the transcriptional level. This mechanical regulation resembles that of calponin 2, another actin filament-associated protein. Immunofluorescent staining co-localized SM22α with F-actin, myosin, and calponin 2 in mouse skin fibroblasts. The close phylogenetic relationship between SM22α and the calponin family supports that SM22α is a calponin-like regulatory protein. The level of SM22α is decreased in skin fibroblasts isolated from calponin 2 knockout mice, suggesting interrelated regulation and function of the two proteins. On the other hand, SM22α expression was maximized at a matrix stiffness higher than that for calponin 2 in the same cell type, indicating differentiated regulation and tension responsiveness. The novel mechanoregulation of SM22α/transgelin lays the groundwork for understanding its cellular functions.


Subject(s)
Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Keratinocytes/metabolism , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Myofibroblasts/metabolism , Animals , Biomarkers/metabolism , Calcium-Binding Proteins , Calpain/metabolism , Cell Line , Cells, Cultured , Cytoskeleton/chemistry , Cytoskeleton/drug effects , Enzyme Inhibitors/pharmacology , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects , Gene Expression Regulation/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Muscle Proteins/genetics , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Organ Specificity , Pliability , Protein Transport/drug effects , Calponins
8.
Eur J Neurosci ; 46(2): 1751-1757, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28452088

ABSTRACT

In neurons, axons possess a molecularly defined and highly organised proximal region - the axon initial segment (AIS) - that is a key regulator of both electrical excitability and cellular polarity. Despite existing as a large, dense structure with specialised cytoskeletal architecture, the AIS is surprisingly plastic, with sustained alterations in neuronal activity bringing about significant alterations to its position, length or molecular composition. However, although the upstream activity-dependent signalling pathways that lead to such plasticity have begun to be elucidated, the downstream mechanisms that produce structural changes at the AIS are completely unknown. Here, we use dissociated cultures of rat hippocampus to show that two forms of AIS plasticity in dentate granule cells - long-term relocation, and more rapid shortening - are completely blocked by treatment with blebbistatin, a potent and selective myosin II ATPase inhibitor. These data establish a link between myosin II and AIS function, and suggest that myosin II's primary role at the structure may be to effect activity-dependent morphological alterations.


Subject(s)
Axon Initial Segment/metabolism , Myosin Type II/metabolism , Neuronal Plasticity/physiology , Animals , Axon Initial Segment/drug effects , Calcineurin/metabolism , Cells, Cultured , Central Nervous System Agents/pharmacology , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Endocytosis/drug effects , Endocytosis/physiology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Myosin Type II/antagonists & inhibitors , Neuronal Plasticity/drug effects , Rats, Wistar
9.
Biochem Biophys Res Commun ; 490(3): 849-854, 2017 08 26.
Article in English | MEDLINE | ID: mdl-28648599

ABSTRACT

2,3-Butandione 2-monoxime (BDM) is a widely used myosin inhibitor with an unclear mode of action. In this report, we investigated the mechanism of BDM oxime group nucleophilic reactivity on the phosphoester bond of ATP. BDM increased the ATPase activity of skeletal myosin subfragment 1 (S1) under conditions in which ATP cleavage is the rate-limiting step (K+, EDTA-ATPase activity of native S1 and Mg2+-ATPase activity of trinitrophenylated S1 and partially unfolded S1). Furthermore, the effect of BDM on the S1-bound adenosine 5'-(ß,γ-imido) triphosphate (AMPPNP) 31P NMR spectrum suggests that BDM changes the microenvironment around the phosphorus atoms of myosin-bound nucleotide. A computational search for the BDM-binding site in the adenosine 5'-[γ-thio] triphosphate (myosin-ATPγS) complex predicted that BDM is located adjacent to the nucleotide on myosin. Therefore, we propose that the BDM oxime group catalytically assists in ATP cleavage, thereby enhancing the ATPase activity of myosin in a manner analogous to pralidoxime-mediated reactivation of organophosphate-inactivated acetylcholinesterase. This is the first study suggesting that oxime provides catalytic assistance for ATP cleavage by an ATP-hydrolyzing enzyme.


Subject(s)
Adenosine Triphosphate/metabolism , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Oximes/chemistry , Oximes/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Ca(2+) Mg(2+)-ATPase/metabolism , Molecular Docking Simulation , Myosin Subfragments/metabolism , Rabbits
10.
Bioorg Med Chem Lett ; 27(13): 2986-2989, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28512027

ABSTRACT

Myosin II is an interesting target for therapeutic intervention, as it is involved in a large number of motility-based diseases. (S)-Blebbistatin is a known micromolar inhibitor of this protein. A new series of (S)-blebbistatin derivatives with a modified A-ring was synthesized and the myosin II inhibitory properties were evaluated in vitro. In this way, we gained insight into the influence of structural modifications in this part of the scaffold on myosin II inhibitory potency. Our results indicate there are few possibilities for potency enhancement via ring A modification of the blebbistatin scaffold.


Subject(s)
Dictyostelium/enzymology , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Myosin Type II/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemistry , Molecular Structure , Myosin Type II/metabolism , Structure-Activity Relationship
11.
Org Biomol Chem ; 15(9): 2104-2118, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28220174

ABSTRACT

In search of myosin II inhibitors with superior research tool properties, a chemical optimization campaign of the blebbistatin scaffold was conducted in this paper. (S)-Blebbistatin is the best known small-molecule inhibitor of myosin II ATPase activity. Unfortunately, as a research tool this compound has several deficiencies: it is photolabile and (photo)toxic, has low water solubility, and its (fluorescent) precipitates interfere in (fluorescence) readouts. In view of obtaining tool compounds with improved properties, both enantiomers of a series of D-ring modified polar analogs were prepared. We identified (S)-3'-hydroxyblebbistatin (S)-2 and (S)-3'-aminoblebbistatin (S)-3 as two myosin II inhibitors with a 30-fold higher water solubility than (S)-blebbistatin. These molecules furthermore do not cause interference in (fluorescence) readouts. (S)-2 and (S)-3 thus are superior alternatives to (S)-blebbistatin as research tools to study myosin II.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Myosin Type II/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Molecular Structure , Myosin Type II/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
12.
Proc Natl Acad Sci U S A ; 111(48): 17176-81, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404288

ABSTRACT

Rigidity sensing and durotaxis are thought to be important elements in wound healing, tissue formation, and cancer treatment. It has been challenging, however, to study the underlying mechanism due to difficulties in capturing cells during the transient response to a rigidity interface. We have addressed this problem by developing a model experimental system that confines cells to a micropatterned area with a rigidity border. The system consists of a rigid domain of one large adhesive island, adjacent to a soft domain of small adhesive islands grafted on a nonadhesive soft gel. This configuration allowed us to test rigidity sensing away from the cell body during probing and spreading. NIH 3T3 cells responded to the micropatterned rigidity border similarly to cells at a conventional rigidity border, by showing a strong preference for staying on the rigid side. Furthermore, cells used filopodia extensions to probe substrate rigidity at a distance in front of the leading edge and regulated their responses based on the strain of the intervening substrate. Soft substrates inhibited focal adhesion maturation and promoted cell retraction, whereas rigid substrates allowed stable adhesions and cell spreading. Myosin II was required for not only the generation of probing forces but also the retraction in response to soft substrates. We suggest that a myosin II-driven, filopodia-based probing mechanism ahead of the leading edge allows cells to migrate efficiently, by sensing physical characteristics before moving over a substrate to avoid backtracking.


Subject(s)
Cell Movement/physiology , Fibroblasts/physiology , Focal Adhesions/physiology , Pseudopodia/physiology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/drug effects , Cell Physiological Phenomena , Cellular Microenvironment/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Focal Adhesions/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Hydrogels , Mice , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Models, Biological , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , NIH 3T3 Cells , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Surface Properties , Time-Lapse Imaging/methods
13.
J Neurosci ; 35(4): 1380-9, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25632116

ABSTRACT

Activity-dependent bulk endocytosis allows neurons to internalize large portions of the plasma membrane in response to stimulation. However, whether this critical type of compensatory endocytosis is unique to neurons or also occurs in other excitable cells is currently unknown. Here we used fluorescent 70 kDa dextran to demonstrate that secretagogue-induced bulk endocytosis also occurs in bovine chromaffin cells. The relatively large size of the bulk endosomes found in this model allowed us to investigate how the neck of the budding endosomes constricts to allow efficient recruitment of the fission machinery. Using time-lapse imaging of Lifeact-GFP-transfected chromaffin cells in combination with fluorescent 70 kDa dextran, we detected acto-myosin II rings surrounding dextran-positive budding endosomes. Importantly, these rings were transient and contracted before disappearing, suggesting that they might be involved in restricting the size of the budding endosome neck. Based on the complete recovery of dextran fluorescence after photobleaching, we demonstrated that the actin ring-associated budding endosomes were still connected with the extracellular fluid. In contrast, no such recovery was observed following the constriction and disappearance of the actin rings, suggesting that these structures were pinched-off endosomes. Finally, we showed that the rings were initiated by a circular array of phosphatidylinositol(4,5)bisphosphate microdomains, and that their constriction was sensitive to both myosin II and dynamin inhibition. The acto-myosin II rings therefore play a key role in constricting the neck of budding bulk endosomes before dynamin-dependent fission from the plasma membrane of neurosecretory cells.


Subject(s)
Actins/metabolism , Chromaffin Cells/physiology , Chromaffin Cells/ultrastructure , Endocytosis/physiology , Endosomes/metabolism , Myosin Type II/metabolism , Adrenal Glands/cytology , Animals , Biological Transport/drug effects , Cattle , Cell Membrane/metabolism , Cells, Cultured , Chromaffin Cells/drug effects , Dextrans/metabolism , Dynamins/antagonists & inhibitors , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/ultrastructure , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Hydrazones/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Myosin Type II/antagonists & inhibitors , Naphthols/pharmacology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Rhodamines/metabolism , Time Factors , Transfection
14.
Am J Physiol Cell Physiol ; 310(1): C89-98, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26491051

ABSTRACT

Filamin B (FLNB) is a dimeric actin-binding protein that orchestrates the reorganization of the actin cytoskeleton. Congenital mutations of FLNB at the actin-binding domain (ABD) are known to cause abnormalities of skeletal development, such as atelosteogenesis types I and III and Larsen's syndrome, although the underlying mechanisms are poorly understood. Here, using fluorescence microscopy, we characterized the reorganization of the actin cytoskeleton in cells expressing each of six pathological FLNB mutants that have been linked to skeletal abnormalities. The subfractionation assay showed a greater accumulation of the FLNB ABD mutants W148R and E227K than the wild-type protein to the cytoskeleton. Ectopic expression of FLNB-W148R and, to a lesser extent, FLNB-E227K induced prominent F-actin accumulations and the consequent rearrangement of focal adhesions, myosin II, and septin filaments and results in a delayed directional migration of the cells. The W148R protein-induced cytoskeletal rearrangement was partially attenuated by the inhibition of myosin II, p21-activated protein kinase, or Rho-associated protein kinase. The expression of a single-head ABD fragment with the mutations partially mimicked the rearrangement induced by the dimer. The F-actin clustering through the interaction with the mutant FLNB ABD may limit the cytoskeletal reorganization, preventing normal skeletal development.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Movement , Filamins/genetics , Filamins/metabolism , Mutation, Missense , Actin Cytoskeleton/drug effects , Animals , Cell Movement/drug effects , Focal Adhesions/metabolism , Genotype , HEK293 Cells , Humans , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Rats , Time Factors , Transfection , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
15.
Hum Mol Genet ; 23(7): 1677-86, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24203699

ABSTRACT

Mutations in the dysferlin gene resulting in dysferlin-deficiency lead to limb-girdle muscular dystrophy 2B and Myoshi myopathy in humans. Dysferlin has been proposed as a critical regulator of vesicle-mediated membrane resealing in muscle fibers, and localizes to muscle fiber wounds following sarcolemma damage. Studies in fibroblasts and urchin eggs suggest that trafficking and fusion of intracellular vesicles with the plasma membrane during resealing requires the intracellular cytoskeleton. However, the contribution of dysferlin-containing vesicles to resealing in muscle and the role of the cytoskeleton in regulating dysferlin-containing vesicle biology is unclear. Here, we use live-cell imaging to examine the behavior of dysferlin-containing vesicles following cellular wounding in muscle cells and examine the role of microtubules and kinesin in dysferlin-containing vesicle behavior following wounding. Our data indicate that dysferlin-containing vesicles move along microtubules via the kinesin motor KIF5B in muscle cells. Membrane wounding induces dysferlin-containing vesicle-vesicle fusion and the formation of extremely large cytoplasmic vesicles, and this response depends on both microtubules and functional KIF5B. In non-muscle cell types, lysosomes are critical mediators of membrane resealing, and our data indicate that dysferlin-containing vesicles are capable of fusing with lysosomes following wounding which may contribute to formation of large wound sealing vesicles in muscle cells. Overall, our data provide mechanistic evidence that microtubule-based transport of dysferlin-containing vesicles may be critical for resealing, and highlight a critical role for dysferlin-containing vesicle-vesicle and vesicle-organelle fusion in response to wounding in muscle cells.


Subject(s)
Cell Membrane/pathology , Cytoplasmic Vesicles/pathology , Kinesins/metabolism , Membrane Fusion/physiology , Membrane Proteins/genetics , Microtubules/metabolism , Muscle Proteins/genetics , Animals , Cell Line , Green Fluorescent Proteins , Heterocyclic Compounds, 4 or More Rings/pharmacology , Kinesins/genetics , Lysosomes/metabolism , Muscle Cells/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/genetics , Myosin Type II/antagonists & inhibitors , Nocodazole/pharmacology , Rats , Tubulin Modulators/pharmacology
16.
Development ; 140(15): 3198-209, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23804498

ABSTRACT

Epithelial-to-mesenchymal transitions (EMTs) are crucial for morphogenesis and carcinoma metastasis, yet mechanisms controlling the underlying cell behaviors are poorly understood. RhoGTPase signaling has been implicated in EMT; however, previous studies have yielded conflicting results regarding Rho function, and its role in EMT remains poorly understood. Elucidation of precise Rho functions has been challenging because Rho signaling is highly context dependent and its activity is tightly regulated spatiotemporally within the cell. To date, few studies have examined how Rho affects cell motility in intact organisms, and the pattern of Rho activity during motile cell behaviors of EMT has not been determined in any system. Here, we image endogenous active Rho during EMT in vivo, and analyze effects of Rho and Rho-kinase (ROCK) manipulation on cell motility in vivo. We show that Rho is activated in a discrete apical region of premigratory neural crest cells during EMT, and Rho-ROCK signaling is essential for apical detachment and generation of motility within the neuroepithelium, a process that has been poorly understood. Furthermore, we find that Arhgap1 restricts Rho activation to apical areas, and this restriction is necessary for detachment. Our results provide new insight into mechanisms controlling local Rho activation and how it affects dynamic cell behaviors and actomyosin contraction during key steps of EMT in an intact living organism.


Subject(s)
GTPase-Activating Proteins/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Epithelial-Mesenchymal Transition , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , Gene Knockdown Techniques , Models, Neurological , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Neural Crest/cytology , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
17.
Stem Cells ; 33(3): 674-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25346532

ABSTRACT

Neural crest is a population of multipotent progenitor cells that form at the border of neural and non-neural ectoderm in vertebrate embryos, and undergo epithelial-mesenchymal transition and migration. According to the traditional view, the neural crest is specified in early embryos by signaling molecules including BMP, FGF, and Wnt proteins. Here, we identify a novel signaling pathway leading to neural crest specification, which involves Rho-associated kinase (ROCK) and its downstream target nonmuscle Myosin II. We show that ROCK inhibitors promote differentiation of human embryonic stem cells (hESCs) into neural crest-like progenitors (NCPs) that are characterized by specific molecular markers and ability to differentiate into multiple cell types, including neurons, chondrocytes, osteocytes, and smooth muscle cells. Moreover, inhibition of Myosin II was sufficient for generating NCPs at high efficiency. Whereas Myosin II has been previously implicated in the self-renewal and survival of hESCs, we demonstrate its role in neural crest development during ESC differentiation. Inhibition of this pathway in Xenopus embryos expanded neural crest in vivo, further indicating that neural crest specification is controlled by ROCK-dependent Myosin II activity. We propose that changes in cell morphology in response to ROCK and Myosin II inhibition initiate mechanical signaling leading to neural crest fates.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Myosin Type II/antagonists & inhibitors , Myosins/antagonists & inhibitors , Neural Crest/cytology , Neural Crest/metabolism , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Embryonic Stem Cells/drug effects , Humans , Myosin Type II/metabolism , Myosins/genetics , Neural Crest/drug effects , Pyridines/pharmacology , Xenopus laevis , rho-Associated Kinases/genetics
18.
Nature ; 465(7296): 373-7, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20485438

ABSTRACT

Crawling locomotion of eukaryotic cells is achieved by a process dependent on the actin cytoskeleton: protrusion of the leading edge requires assembly of a network of actin filaments, which must be disassembled at the cell rear for sustained motility. Although ADF/cofilin proteins have been shown to contribute to actin disassembly, it is not clear how activity of these locally acting proteins could be coordinated over the distance scale of the whole cell. Here we show that non-muscle myosin II has a direct role in actin network disassembly in crawling cells. In fish keratocytes undergoing motility, myosin II is concentrated in regions at the rear with high rates of network disassembly. Activation of myosin II by ATP in detergent-extracted cytoskeletons results in rear-localized disassembly of the actin network. Inhibition of myosin II activity and stabilization of actin filaments synergistically impede cell motility, suggesting the existence of two disassembly pathways, one of which requires myosin II activity. Our results establish the importance of myosin II as an enzyme for actin network disassembly; we propose that gradual formation and reorganization of an actomyosin network provides an intrinsic destruction timer, enabling long-range coordination of actin network treadmilling in motile cells.


Subject(s)
Actins/chemistry , Actins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Myosin Type II/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Movement/drug effects , Cichlids , Cytoskeleton/chemistry , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Depsipeptides/pharmacology , Detergents , Heterocyclic Compounds, 4 or More Rings/pharmacology , Myosin Type II/antagonists & inhibitors , Protein Binding/drug effects , Protein Transport
19.
Ann Plast Surg ; 77(3): e46-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-25144422

ABSTRACT

BACKGROUND: Scar contracture (SC) is one of the most common complications resulting from major burn injuries. Numerous treatments are currently available but they do not always yield excellent therapeutic results. Recent reports suggest that botulinum toxin type A (BTXA) is effective at reducing SC clinically, but the molecular mechanism for this action is unknown. α-Smooth muscle actin (α-SMA) and myosin II are the main components of stress fibers, which are the contractile structures of fibroblasts. The effects of BTXA on α-SMA and myosin II in SC are still unknown. This study aimed to explore the effect of BTXA on α-SMA and myosin II expression in fibroblasts derived from SC and to elucidate its actual mechanism further. METHODS: Fibroblasts were isolated from tissue specimens of SC. Fibroblasts were cultured in Dulbecco modified Eagle medium with different concentrations of BTXA and their proliferation was analyzed through the tetrazolium-based colorimetric method at 1, 4, and 7 days. Proteins of α-SMA and myosin II were checked using Western blot in fibroblasts treated with different concentrations of BTXA at 1, 4, and 7 days. RESULTS: Fibroblasts without BTXA treatment had a higher proliferation than that in other groups, which indicated that the proliferation of fibroblasts was significantly inhibited by BTXA (P < 0.05). Proteins of α-SMA and myosin II between fibroblasts with BTXA and fibroblasts without BTXA are statistically significant (P < 0.05). CONCLUSIONS: These results suggest that BTXA effectively inhibited the growth of fibroblasts derived from SC and reduced the expression of α-SMA and myosin II, which provided theoretical support for the application of BTXA to control SC.


Subject(s)
Actins/antagonists & inhibitors , Botulinum Toxins, Type A/pharmacology , Cicatrix/drug therapy , Contracture/drug therapy , Dermatologic Agents/pharmacology , Fibroblasts/drug effects , Myosin Type II/antagonists & inhibitors , Biomarkers/metabolism , Botulinum Toxins, Type A/therapeutic use , Burns/complications , Cell Proliferation/drug effects , Cells, Cultured , Cicatrix/etiology , Cicatrix/metabolism , Contracture/etiology , Contracture/metabolism , Dermatologic Agents/therapeutic use , Fibroblasts/metabolism , Humans
20.
J Cell Sci ; 126(Pt 17): 3873-83, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23843614

ABSTRACT

Astrocytes exhibit a complex, branched morphology, allowing them to functionally interact with numerous blood vessels, neighboring glial processes and neuronal elements, including synapses. They also respond to central nervous system (CNS) injury by a process known as astrogliosis, which involves morphological changes, including cell body hypertrophy and thickening of major processes. Following severe injury, astrocytes exhibit drastically reduced morphological complexity and collectively form a glial scar. The mechanistic details behind these morphological changes are unknown. Here, we investigate the regulation of the actin-nucleating Arp2/3 complex in controlling dynamic changes in astrocyte morphology. In contrast to other cell types, Arp2/3 inhibition drives the rapid expansion of astrocyte cell bodies and major processes. This intervention results in a reduced morphological complexity of astrocytes in both dissociated culture and in brain slices. We show that this expansion requires functional myosin II downstream of ROCK and RhoA. Knockdown of the Arp2/3 subunit Arp3 or the Arp2/3 activator N-WASP by siRNA also results in cell body expansion and reduced morphological complexity, whereas depleting WAVE2 specifically reduces the branching complexity of astrocyte processes. By contrast, knockdown of the Arp2/3 inhibitor PICK1 increases astrocyte branching complexity. Furthermore, astrocyte expansion induced by ischemic conditions is delayed by PICK1 knockdown or N-WASP overexpression. Our findings identify a new morphological outcome for Arp2/3 activation in restricting rather than promoting outwards movement of the plasma membrane in astrocytes. The Arp2/3 regulators PICK1, and N-WASP and WAVE2 function antagonistically to control the complexity of astrocyte branched morphology, and this mechanism underlies the morphological changes seen in astrocytes during their response to pathological insult.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Astrocytes/metabolism , Carrier Proteins/metabolism , Central Nervous System/metabolism , Nuclear Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Actin-Related Protein 2-3 Complex/genetics , Amides/pharmacology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Carrier Proteins/genetics , Cells, Cultured , Colforsin/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts , HEK293 Cells , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Mice , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Nuclear Proteins/genetics , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering , Rats , Thiazoles/pharmacology , Thiones/pharmacology , Uracil/analogs & derivatives , Uracil/pharmacology , Vasodilator Agents/pharmacology , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL