cytochalasin-d has been researched along with jasplakinolide* in 105 studies
105 other study(ies) available for cytochalasin-d and jasplakinolide
Article | Year |
---|---|
Effect of jasplakinolide and cytochalasin D on cortical elements involved in the gliding motility of the eugregarine Gregarina garnhami (Apicomplexa).
Since apicomplexans represent exclusively parasitic unicellular organisms with medical and economic impacts, the principles of their motility have been studied intensively. By contrast, the movement in apicomplexan basal groups, such as gregarines, remains to be elucidated. The present study focuses on Gregarina garnhami parasitising the digestive tract of the locust Schistocerca gregaria, and investigates the involvement of cytoskeletal elements (the ectoplasmic network and myonemes) and the secretion of mucosubstances during eugregarine gliding motility. Combined microscopic analyses were used to verify the role of actin filaments and membranes' organisation in G. garnhami motility. A freeze-etching analysis of membranes revealed the size, density, and arrangement of intramembranous particles along with the distribution and size of pores and ducts. Experimental assays using actin-modifying drugs (jasplakinolide, cytochalasin D) confirmed that actin most likely plays a role in cell motility, principally in its filamentous form (=F-actin). Myonemes, localised in the border between the ectoplasm and endoplasm, correspond to the concentric bundles of F-actin. Microscopic analyses confirmed that changes in gamonts motility corresponding to the changes in the organisation and density of myonemes and the ectoplasmic network in drug-treated cells, suggesting that these structures might serve as contractile elements facilitating gliding motility in G. garnhami. Topics: Actins; Apicomplexa; Cytochalasin D; Depsipeptides; Insecticides; Movement; Nucleic Acid Synthesis Inhibitors | 2018 |
High-throughput cell mechanical phenotyping for label-free titration assays of cytoskeletal modifications.
The mechanical fingerprint of cells is inherently linked to the structure of the cytoskeleton and can serve as a label-free marker for cell homeostasis or pathologic states. How cytoskeletal composition affects the physical response of cells to external loads has been intensively studied with a spectrum of techniques, yet quantitative and statistically powerful investigations in the form of titration assays are hampered by the low throughput of most available methods. In this study, we employ real-time deformability cytometry (RT-DC), a novel microfluidic tool to examine the effects of biochemically modified F-actin and microtubule stability and nuclear chromatin structure on cell deformation in a human leukemia cell line (HL60). The high throughput of our method facilitates extensive titration assays that allow for significance assessment of the observed effects and extraction of half-maximal concentrations for most of the applied reagents. We quantitatively show that integrity of the F-actin cortex and microtubule network dominate cell deformation on millisecond timescales probed with RT-DC. Drug-induced alterations in the nuclear chromatin structure were not found to consistently affect cell deformation. The sensitivity of the high-throughput cell mechanical measurements to the cytoskeletal modifications we present in this study opens up new possibilities for label-free dose-response assays of cytoskeletal modifications. Topics: Actins; Biomechanical Phenomena; Chromatin; Computer Systems; Cytochalasin D; Cytoskeleton; Depsipeptides; High-Throughput Screening Assays; HL-60 Cells; Humans; Hydroxamic Acids; Microtubules; Nocodazole; Paclitaxel; Phenotype; Staining and Labeling | 2017 |
Cortical cytoskeleton dynamics regulates plasma membrane calcium ATPase isoform-2 (PMCA2) activity.
We have previously shown that purified actin can directly bind to human plasma membrane Ca Topics: Actin Cytoskeleton; Actins; Calcium; Calcium Signaling; Cell Membrane; Colchicine; Cytochalasin D; Depsipeptides; Gene Expression Regulation; HEK293 Cells; Humans; Microtubules; Plasma Membrane Calcium-Transporting ATPases; Time-Lapse Imaging | 2017 |
Pixuna virus modifies host cell cytoskeleton to secure infection.
Pixuna virus (PIXV) is an enzootic member of the Venezuelan Equine Encephalitis Virus complex and belongs to the New World cluster of alphaviruses. Herein we explore the role of the cellular cytoskeleton during PIXV replication. We first identified that PIXV undergoes an eclipse phase consisting of 4 h followed by 20 h of an exponential phase in Vero cells. The infected cells showed morphological changes due to structural modifications in actin microfilaments (MFs) and microtubules (MTs). Cytoskeleton-binding agents, that alter the architecture and dynamics of MFs and MTs, were used to study the role of cytoskeleton on PIXV replication. The virus production was significantly affected (p < 0.05) after treatment with paclitaxel or nocodazole due to changes in the MTs network. Interestingly, disassembly of MFs with cytochalasin D, at early stage of PIXV replication cycle, significantly increased the virus yields in the extracellular medium (p < 0.005). Furthermore, the stabilization of actin network with jasplakinolide had no effect on virus yields. Our results demonstrate that PIXV relies not only on intact MTs for the efficient production of virus, but also on a dynamic actin network during the early steps of viral replication. Topics: Alphavirus; Animals; Chlorocebus aethiops; Cytochalasin D; Cytoskeleton; Depsipeptides; Host-Pathogen Interactions; Microtubules; Nocodazole; Paclitaxel; Time Factors; Tubulin Modulators; Vero Cells; Virus Replication | 2017 |
Premature termination codon readthrough in human cells occurs in novel cytoplasmic foci and requires UPF proteins.
Nonsense-mutation-containing messenger ribonucleoprotein particles (mRNPs) transit through cytoplasmic foci called P-bodies before undergoing nonsense-mediated mRNA decay (NMD), a cytoplasmic mRNA surveillance mechanism. This study shows that the cytoskeleton modulates transport of nonsense-mutation-containing mRNPs to and from P-bodies. Impairing the integrity of cytoskeleton causes inhibition of NMD. The cytoskeleton thus plays a crucial role in NMD. Interestingly, disruption of actin filaments results in both inhibition of NMD and activation of premature termination codon (PTC) readthrough, while disruption of microtubules causes only NMD inhibition. Activation of PTC readthrough occurs concomitantly with the appearance of cytoplasmic foci containing UPF proteins and mRNAs with nonsense mutations but lacking the P-body marker DCP1a. These findings demonstrate that in human cells, PTC readthrough occurs in novel 'readthrough bodies' and requires the presence of UPF proteins. Topics: Actin Cytoskeleton; Cell Line; Codon, Nonsense; Cytochalasin D; Cytoplasm; Cytoskeleton; Depsipeptides; Down-Regulation; Humans; Nonsense Mediated mRNA Decay; Protein Biosynthesis; Ribonucleoproteins; RNA Helicases | 2017 |
Shear stress-mediated changes in the expression of complement regulatory protein CD59 on human endothelial progenitor cells by ECM-integrinα
Membrane regulatory proteins, such as CD46, CD55, and CD59, prevent excess complement activation and to protect cells from damage. Previous investigations confirmed that shear stress in the physiological range was more favorable for endothelial progenitor cells (EPCs) to repair injured vascular endothelial cells and operates mainly in atheroprotective actions. However, detailed events that contribute to shear stress-induced protection in EPCs, particularly the mechanisms of signal transduction, remain poorly understood. In this study, we observed shear stress-mediated changes in the expression of complement regulatory proteins CD46, CD55, and CD59 on human EPCs and focused on the mechanical transmission mechanism in transformed cells in response to the ECM-F-actin pathway in vitro. Shear stress was observed to promote the expression of complement regulatory protein CD59, but not CD46 or CD55, on EPCs. In addition, the shear stress-induced CD59 expression was confirmed to be associated with the ECM components and was alleviated in EPCs pretreated with GRGDSP, which inhibits ECM components-integrin interaction. Furthermore, shear stress also promotes the rearrangement and polymerization of F-actin. However, shear stress-induced CD59 expression was reduced when the F-actin stress fiber formation process was delayed by Gly-Arg-Gly-Asp-Ser-Pro (GRGDSP) or destroyed by cytochalasin D (Cyto D), while Jasplakinolide (JAS) reversed the expression of CD59 through promotion of F-actin polymerization and its stabilizing capacities. Our results indicates that shear stress is an important mediator in EPC expression of CD59 regulated by the ECM-F-actin pathway, which is a key factor in preventing membrane attack complex (MAC) -mediated cell autolysis. Topics: Actin Cytoskeleton; Actins; CD55 Antigens; CD59 Antigens; Complement Membrane Attack Complex; Cytochalasin D; Depsipeptides; Endothelial Progenitor Cells; Extracellular Matrix; Fetal Blood; Gene Expression Regulation; Humans; Integrin alphaVbeta3; Mechanotransduction, Cellular; Membrane Cofactor Protein; Oligopeptides; Primary Cell Culture; Stress, Mechanical | 2017 |
Effects of cytoskeletal drugs on actin cortex elasticity.
Mechanical properties of cells are known to be influenced by the actin cytoskeleton. In this article, the action of drugs that interact with the actin cortex is investigated by tether extraction and rheology experiments using optical tweezers. The influences of Blebbistatin, Cytochalasin D and Jasplakinolide on the cell mechanical properties are evaluated. The results, in contradiction to current views for Jasplakinolide, show that all three drugs and treatments destabilize the actin cytoskeleton, decreasing the cell membrane tension. The cell membrane bending modulus increased when the actin cytoskeleton was disorganized by Cytochalasin D. This effect was not observed for Blebbistatin and Jasplakinolide. All drugs decreased by two-fold the cell viscoelastic moduli, but only Cytochalasin D was able to alter the actin network into a more fluid-like structure. The results can be interpreted as the interplay between the actin network and the distribution of myosins as actin cross-linkers in the cytoskeleton. This information may contribute to a better understanding of how the membrane and cytoskeleton are involved in cell mechanical properties, underlining the role that each one plays in these properties. Topics: Actin Cytoskeleton; Animals; Biomechanical Phenomena; Cell Membrane; Cytochalasin D; Depsipeptides; Elasticity; Heterocyclic Compounds, 4 or More Rings; Humans; Mice; Myosins; NIH 3T3 Cells; Optical Tweezers; Rheology; Viscosity | 2017 |
Small G Rac1 is involved in replication cycle of dengue serotype 2 virus in EAhy926 cells via the regulation of actin cytoskeleton.
Bleeding is a clinical characteristic of severe dengue and may be due to increased vascular permeability. However, the pathogenesis of severe dengue remains unclear. In this study, we showed that the Rac1-microfilament signal pathway was involved in the process of DENV serotype 2 (DENV2) infection in EAhy926 cells. DENV2 infection induced dynamic changes in actin organization, and treatment with Cytochalasin D or Jasplakinolide disrupted microfilament dynamics, reduced DENV2 entry, and inhibited DENV2 assembly and maturation. Rac1 activities decreased during the early phase and gradually increased by the late phase of infection. Expression of the dominant-negative form of Rac1 promoted DENV2 entry but inhibited viral assembly, maturation and release. Our findings demonstrated that Rac1 plays an important role in the DENV2 life cycle by regulating actin reorganization in EAhy926 cells. This finding provides further insight into the pathogenesis of severe dengue. Topics: Actin Cytoskeleton; Aedes; Animals; Cell Line; Cytochalasin D; Dengue Virus; Depsipeptides; Membrane Fusion; rac1 GTP-Binding Protein; Virus Replication | 2016 |
TREK-1 Regulates Cytokine Secretion from Cultured Human Alveolar Epithelial Cells Independently of Cytoskeletal Rearrangements.
TREK-1 deficient alveolar epithelial cells (AECs) secrete less IL-6, more MCP-1, and contain less F-actin. Whether these alterations in cytokine secretion and F-actin content are related remains unknown. We now hypothesized that cytokine secretion from TREK-1-deficient AECs was regulated by cytoskeletal rearrangements.. We determined F-actin and α-tubulin contents of control, TREK-1-deficient and TREK-1-overexpressing human A549 cells by confocal microscopy and western blotting, and measured IL-6 and MCP-1 levels using real-time PCR and ELISA.. Cytochalasin D decreased the F-actin content of control cells. Jasplakinolide increased the F-actin content of TREK-1 deficient cells, similar to the effect of TREK-1 overexpression in control cells. Treatment of control and TREK-1 deficient cells with TNF-α, a strong stimulus for IL-6 and MCP-1 secretion, had no effect on F-actin structures. The combination of TNF-α+cytochalasin D or TNF-α+jasplakinolide had no additional effect on the F-actin content or architecture when compared to cytochalasin D or jasplakinolide alone. Although TREK-1 deficient AECs contained less F-actin at baseline, quantified biochemically, they contained more α-tubulin. Exposure to nocodazole disrupted α-tubulin filaments in control and TREK-1 deficient cells, but left the overall amount of α-tubulin unchanged. Although TNF-α had no effect on the F-actin or α-tubulin contents, it increased IL-6 and MCP-1 production and secretion from control and TREK-1 deficient cells. IL-6 and MCP-1 secretions from control and TREK-1 deficient cells after TNF-α+jasplakinolide or TNF-α+nocodazole treatment was similar to the effect of TNF-α alone. Interestingly, cytochalasin D decreased TNF-α-induced IL-6 but not MCP-1 secretion from control but not TREK-1 deficient cells.. Although cytochalasin D, jasplakinolide and nocodazole altered the F-actin and α-tubulin structures of control and TREK-1 deficient AEC, the changes in cytokine secretion from TREK-1 deficient cells cannot be explained by cytoskeletal rearrangements in these cells. Topics: Actins; Blotting, Western; Cell Line; Chemokine CCL2; Cytochalasin D; Cytoskeleton; Depsipeptides; Enzyme-Linked Immunosorbent Assay; Epithelial Cells; Humans; Interleukin-6; Microscopy, Confocal; Nocodazole; Potassium Channels, Tandem Pore Domain; Real-Time Polymerase Chain Reaction; Tubulin | 2015 |
Modulation of the extracellular matrix patterning of thrombospondins by actin dynamics and thrombospondin oligomer state.
Thrombospondins (TSPs) are evolutionarily-conserved, secreted glycoproteins that interact with cell surfaces and extracellular matrix (ECM) and have complex roles in cell interactions. Unlike the structural components of the ECM that form networks or fibrils, TSPs are deposited into ECM as arrays of nanoscale puncta. The cellular and molecular mechanisms for the patterning of TSPs in ECM are poorly understood. In the present study, we investigated whether the mechanisms of TSP patterning in cell-derived ECM involves actin cytoskeletal pathways or TSP oligomer state. From tests of a suite of pharmacological inhibitors of small GTPases, actomyosin-based contractility, or actin microfilament integrity and dynamics, cytochalasin D and jasplakinolide treatment of cells were identified to result in altered ECM patterning of a model TSP1 trimer. The strong effect of cytochalasin D indicated that mechanisms controlling puncta patterning depend on global F-actin dynamics. Similar spatial changes were obtained with endogenous TSPs after cytochalasin D treatment, implicating physiological relevance. Under matched experimental conditions with ectopically-expressed TSPs, the magnitude of the effect was markedly lower for pentameric TSP5 and Drosophila TSP, than for trimeric TSP1 or dimeric Ciona TSPA. To distinguish between the variables of protein sequence or oligomer state, we generated novel, chimeric pentamers of TSP1. These proteins accumulated within ECM at higher levels than TSP1 trimers, yet the effect of cytochalasin D on the spatial distribution of puncta was reduced. These findings introduce a novel concept that F-actin dynamics modulate the patterning of TSPs in ECM and that TSP oligomer state is a key determinant of this process. Topics: Actins; Animals; Cell Line, Tumor; Chlorocebus aethiops; COS Cells; Cytochalasin D; Cytoskeleton; Depsipeptides; Drosophila Proteins; Extracellular Matrix; Humans; Protein Multimerization; Rats; Recombinant Proteins; Thrombospondin 1; Thrombospondins | 2015 |
Feeling for Filaments: Quantification of the Cortical Actin Web in Live Vascular Endothelium.
Contact-mode atomic force microscopy (AFM) has been shown to reveal cortical actin structures. Using live endothelial cells, we visualized cortical actin dynamics simultaneously by AFM and confocal fluorescence microscopy. We present a method that quantifies dynamic changes in the mechanical ultrastructure of the cortical actin web. We argue that the commonly used, so-called error signal imaging in AFM allows a qualitative, but not quantitative, analysis of cortical actin dynamics. The approach we used comprises fast force-curve-based topography imaging and subsequent image processing that enhances local height differences. Dynamic changes in the organization of the cytoskeleton network can be observed and quantified by surface roughness calculations and automated morphometrics. Upon treatment with low concentrations of the actin-destabilizing agent cytochalasin D, the cortical cytoskeleton network is thinned out and the average mesh size increases. In contrast, jasplakinolide, a drug that enhances actin polymerization, consolidates the cytoskeleton network and reduces the average mesh area. In conclusion, cortical actin dynamics can be quantified in live cells. To our knowledge, this opens a new pathway for conducting quantitative structure-function analyses of the endothelial actin web just beneath the apical plasma membrane. Topics: Actins; Animals; Antineoplastic Agents; Aorta; Calcium; Cattle; Cells, Cultured; Cytochalasin D; Depsipeptides; Endothelium, Vascular; Microscopy, Atomic Force; Microscopy, Confocal; Microscopy, Fluorescence; Nucleic Acid Synthesis Inhibitors | 2015 |
Endothelial actin depolymerization mediates NADPH oxidase-superoxide production during flow reversal.
Slow moving blood flow and changes in flow direction, e.g., negative wall shear stress, can cause increased superoxide (O2(·-)) production in vascular endothelial cells. The mechanism by which shear stress increases O2(·-) production, however, is not well established. We tested the hypothesis that actin depolymerization, which occurs during flow reversal, mediates O2(·-) production in vascular endothelial cells via NADPH oxidase, and more specifically, the subunit p47(phox). Using a swine model, we created complete blood flow reversal in one carotid artery, while the contralateral vessel maintained forward blood flow as control. We measured actin depolymerization, NADPH oxidase activity, and reactive oxygen species (ROS) production in the presence of various inhibitors. Flow reversal was found to induce actin depolymerization and a 3.9 ± 1.0-fold increase in ROS production as compared with forward flow. NADPH oxidase activity was 1.4 ± 0.2 times higher in vessel segments subjected to reversed blood flow when measured by a direct enzyme assay. The NADPH oxidase subunits gp91(phox) (Nox2) and p47(phox) content in the vessels remained unchanged after 4 h of flow reversal. In contrast, p47(phox) phosphorylation was increased in vessels with reversed flow. The response caused by reversed flow was reduced by in vivo treatment with jasplakinolide, an actin stabilizer (only a 1.7 ± 0.3-fold increase). Apocynin (an antioxidant) prevented reversed flow-induced ROS production when the animals were treated in vivo. Cytochalasin D mimicked actin depolymerization in vitro and caused a 5.2 ± 3.0-fold increase in ROS production. These findings suggest that actin filaments play an important role in negative shear stress-induced ROS production by potentiating NADPH oxidase activity, and more specifically, the p47(phox) subunit in vascular endothelium. Topics: Acetophenones; Actin Cytoskeleton; Actin Depolymerizing Factors; Actins; Animals; Antioxidants; Carotid Arteries; Coronary Circulation; Cytochalasin D; Depsipeptides; Endothelium, Vascular; NADPH Oxidases; Polymerization; Superoxides; Swine | 2014 |
F‑actin cytoskeleton reorganization is associated with hepatic stellate cell activation.
The activation of hepatic stellate cells (HSCs) is involved in the development of hepatic fibrosis. Previous studies have indicated that the acquisition of certain properties by activated HSCs is highly dependent on the reorganization of the actin cytoskeleton. However, direct evidence showing that the reorganization of the actin cytoskeleton is responsible for HSC activation is lacking. The aim of the present study was to investigate the role of cytoskeletal reorganization during HSC activation and to clarify the underlying mechanism. HSC-T6 cells were treated either with the F-actin stabilizer jasplakinolide (Jas) or the depolymerizer cytochalasin D (Cyto D). The actin cytoskeleton was evaluated via assessment of stress fiber formation. Furthermore, the activation properties of HSCs, including proliferation, adhesion, migration and the expression of α-smooth muscle actin (α-SMA) and collagen 1, were investigated in vitro. The results showed that Jas and Cyto D affected the actin distribution in HSC-T6 cells. Treatment with Jas resulted in thick actin bundles and a patchy appearance in the cytoplasm in HSC-T6 cells. In parallel, polymerization of actin microfilaments induced by Jas upregulated the expression of α-SMA and collagen 1, and also enhanced the migration and adhesion properties of HSC-T6 cells. Furthermore, the activation of HSC-T6 cells induced by the reorganization of the actin cytoskeleton was associated with the p38 mitogen-activated protein kinase (p38 MAPK) pathway. In conclusion, the present study suggests that the reorganization of the F-actin cytoskeleton is associated with HSC activation and that the p38 MAPK pathway is involved in this process. The inhibition of F-actin reorganization may thus be a potential key factor or molecular target for the control of liver fibrosis or cirrhosis. Topics: Actin Cytoskeleton; Actins; Animals; Cell Line; Cytochalasin D; Depsipeptides; Hepatic Stellate Cells; Liver Cirrhosis; MAP Kinase Signaling System; p38 Mitogen-Activated Protein Kinases; Rats | 2014 |
Modulation of TMEM16A-channel activity as Ca²⁺ activated Cl⁻ conductance via the interaction with actin cytoskeleton in murine portal vein.
TMEM16A is a major component of Ca(2+)-activated Cl(-) channel (CaCC) conductance in murine portal vein smooth muscle cells (mPVSMCs). Here, the regulation of CaCC activity by the actin cytoskeleton was examined in mPVSMCs. Actin disruption by cytochalasin D did not affect the current density, but increased the deactivation time constant in mPVSMCs. The elongated deactivation was recovered by jasplakinolide. When murine TMEM16A was transfected into HEK293 cells that have a poorly developed actin cytoskeleton, electrophysiological properties of CaCC currents were not changed by cytochalasin D. In conclusion, the CaCC activity in mPVSMCs is modified by the interaction of TMEM16A with abundant actin cytoskeleton. Topics: Actin Cytoskeleton; Animals; Anoctamin-1; Cells, Cultured; Chloride Channels; Cytochalasin D; Depsipeptides; Electrophysiological Phenomena; HEK293 Cells; Humans; Mice; Muscle, Smooth, Vascular; Portal Vein; Transfection | 2014 |
The role of RhoA and cytoskeleton in myofibroblast transformation in hyperoxic lung fibrosis.
Myofibroblast transformation is a key process in the pathogenesis of lung fibrosis. We have previously reported that hyperoxia induces RhoA activation in HFL-1 lung fibroblasts and RhoA mediates collagen synthesis in hyperoxic lung fibrosis. In this study, we investigated the role of RhoA and actin cytoskeleton in hyperoxia-induced myofibroblast transformation. Exposure of HFL-1 lung fibroblasts to hyperoxia stimulated actin filament formation, shift of G-actin to F-actin, nuclear colocalization of myocardin-related transcription factor-A (MRTF-A), recruitment of MRTF-A to the α-smooth muscle actin (α-SMA) gene promoter, myofibroblast transformation, and collagen-I synthesis. Inhibition of RhoA by C3 transferase CT-04 or dominant-negative RhoA mutant T19N, and inhibition of ROCK by Y27632, prevented myofibroblast transformation and collagen-I synthesis. Moreover, inhibition of RhoA by CT-04 prevented hyperoxia-induced actin filament formation, shift of G-actin to F-actin, and nuclear colocalization of MRTF-A. In addition, disrupting actin filaments with cytochalasin D or scavenging reactive oxygen species (ROS) with tiron attenuated actin filament formation, nuclear colocalization of MRTF-A, myofibroblast transformation, and collagen-I synthesis. Furthermore, overexpression of constitutively active RhoA mutant Q63L or stabilization of actin filaments recapitulated the effects of hyperoxia on the actin cytoskeleton and nuclear colocalization of MRTF-A, myofibroblast transformation, and collagen-I synthesis. Interestingly, knocking down MRTF-A prevented hyperoxia-induced increase in the recruitment of MRTF-A to the serum response factor transcriptional complex on the α-SMA gene promoter, myofibroblast transformation, and collagen-I synthesis. Finally, Y27632 and tiron attenuated hyperoxia-induced increases in α-SMA and collagen-I in mouse lungs. Together, these results indicate that the actin cytoskeletal reorganization due to the ROS/RhoA-ROCK pathway mediates myofibroblast transformation and collagen synthesis in lung fibrosis of oxygen toxicity. MRTF-A contributes to the regulatory effect of the actin cytoskeleton on myofibroblast transformation during hyperoxia. Topics: 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt; Actins; Active Transport, Cell Nucleus; Animals; Collagen; Cytochalasin D; Cytoskeleton; Depsipeptides; Free Radical Scavengers; Hyperoxia; Male; Mice; Myofibroblasts; NADPH Oxidase 4; NADPH Oxidases; Promoter Regions, Genetic; Pulmonary Fibrosis; Reactive Oxygen Species; rho-Associated Kinases; rhoA GTP-Binding Protein; Trans-Activators | 2013 |
Actin retrograde flow and actomyosin II arc contraction drive receptor cluster dynamics at the immunological synapse in Jurkat T cells.
Actin retrograde flow and actomyosin II contraction have both been implicated in the inward movement of T cell receptor (TCR) microclusters and immunological synapse formation, but no study has integrated and quantified their relative contributions. Using Jurkat T cells expressing fluorescent myosin IIA heavy chain and F-tractin-a novel reporter for F-actin-we now provide direct evidence that the distal supramolecular activation cluster (dSMAC) and peripheral supramolecular activation cluster (pSMAC) correspond to lamellipodial (LP) and lamellar (LM) actin networks, respectively, as hypothesized previously. Our images reveal concentric and contracting actomyosin II arcs/rings at the LM/pSMAC. Moreover, the speeds of centripetally moving TCR microclusters correspond very closely to the rates of actin retrograde flow in the LP/dSMAC and actomyosin II arc contraction in the LM/pSMAC. Using cytochalasin D and jasplakinolide to selectively inhibit actin retrograde flow in the LP/dSMAC and blebbistatin to selectively inhibit actomyosin II arc contraction in the LM/pSMAC, we demonstrate that both forces are required for centripetal TCR microcluster transport. Finally, we show that leukocyte function-associated antigen 1 clusters accumulate over time at the inner aspect of the LM/pSMAC and that this accumulation depends on actomyosin II contraction. Thus actin retrograde flow and actomyosin II arc contraction coordinately drive receptor cluster dynamics at the immunological synapse. Topics: Actins; Actomyosin; Cytochalasin D; Depsipeptides; Green Fluorescent Proteins; Heterocyclic Compounds, 4 or More Rings; Humans; Immunological Synapses; Jurkat Cells; Lymphocyte Function-Associated Antigen-1; Nonmuscle Myosin Type IIA; Receptors, Antigen, T-Cell; T-Lymphocytes | 2012 |
Defective erythroid maturation in gelsolin mutant mice.
During late differentiation, erythroid cells undergo profound changes involving actin filament remodeling. One of the proteins controlling actin dynamics is gelsolin, a calcium-activated actin filament severing and capping protein. Gelsolin-null (Gsn(-/-)) mice generated in a C57BL/6 background are viable and fertile.1. We analyzed the functional roles of gelsolin in erythropoiesis by: (i) evaluating gelsolin expression in murine fetal liver cells at different stages of erythroid differentiation (using reverse transcription polymerase chain reaction analysis and immunohistochemistry), and (ii) characterizing embryonic and adult erythropoiesis in Gsn(-/-) BALB/c mice (morphology and erythroid cultures).. In the context of a BALB/c background, the Gsn(-/-) mutation causes embryonic death. Gsn(-/-) embryos show defective erythroid maturation with persistence of circulating nucleated cells. The few Gsn(-/-) mice reaching adulthood fail to recover from phenylhydrazine-induced acute anemia, revealing an impaired response to stress erythropoiesis. In in vitro differentiation assays, E13.5 fetal liver Gsn(-/-) cells failed to undergo terminal maturation, a defect partially rescued by Cytochalasin D, and mimicked by administration of Jasplakinolide to the wild-type control samples.. In BALB/c mice, gelsolin deficiency alters the equilibrium between erythrocyte actin polymerization and depolymerization, causing impaired terminal maturation. We suggest a non-redundant role for gelsolin in terminal erythroid differentiation, possibly contributing to the Gsn(-/-) mice lethality observed in mid-gestation. Topics: Actins; Anemia; Animals; Biomarkers; Cell Differentiation; Cytochalasin D; Depsipeptides; Embryo, Mammalian; Embryonic Stem Cells; Erythrocytes; Erythropoiesis; Fetus; Gelsolin; Gene Expression Regulation, Developmental; Liver; Mice; Mice, Inbred BALB C; Mice, Knockout; Phenylhydrazines | 2012 |
Molecular assembly and dynamics of fluorescent protein-tagged single KCa1.1 channel in expression system and vascular smooth muscle cells.
The large-conductance Ca(2+)-activated K(+) (K(Ca)1.1, BK) channel has pivotal roles in the regulation of vascular tone. To clarify the molecular dynamics of BK channels and their functionally coupled protein on the membrane surface, we examined single-molecule imaging of fluorescent-labeled BK subunits in the plasma membrane using total internal reflection fluorescence (TIRF) microscopy. The dynamic mobility of yellow fluorescent protein (YFP)-tagged BKα subunit (BKα-YFP) expressed in human embryo kidney 293 (HEK) cells was detected in TIRF regions at the level of individual channels and their clusters on the plasma membrane with a diffusion coefficient of 6.7 × 10(3) nm(2)/s. When BKα-YFP was coexpressed with cyan fluorescent protein (CFP)-tagged BKβ1 subunit (BKβ1-CFP) in HEK cells, the mobility was reduced by ∼50%. Fluorescent image analyses suggest that green fluorescent protein (GFP)-tagged BKα subunit (BKα-GFP) expressed in vascular smooth muscle cells (VSMCs), at low density, preferentially formed a heterotetrameric molecular assembly with native BKα subunits, rather than homotetrameric BKα-GFP. Movement of BKα-YFP in VSMCs (0.29 × 10(3) nm(2)/s) was far more restricted than BKα-YFP/BKβ1-CFP in HEK cells (2.5 × 10(3) nm(2)/s). Actin disruption by pretreatment with cytochalasin D in VSMCs appeared to increase the mobile behavior of BKα-YFP, which was then significantly reduced by addition of jasplakinolide. Most BKα-YFP colocalized with caveolin 1 (Cav1)-CFP in VSMCs, but unexpectedly not frequently in HEK cells. Fluorescence resonance energy transfer analyses showed the direct interaction between BKα-YFP and Cav1-CFP, particularly in VSMCs. These results, obtained by single molecule imaging in living cells, indicate that the dynamics of BKα molecules on the membrane surface are strongly restricted or regulated by its auxiliary β-subunit, cytoskeleton, and direct interaction with Cav1 in VSMCs. Topics: Actins; Animals; Aorta, Thoracic; Caveolin 1; Cell Membrane; Cells, Cultured; Cytochalasin D; Cytoskeleton; Depsipeptides; Fluorescence Resonance Energy Transfer; Fluorescent Dyes; HEK293 Cells; Humans; Large-Conductance Calcium-Activated Potassium Channels; Male; Microscopy, Fluorescence; Muscle, Smooth, Vascular; Myocytes, Smooth Muscle; Protein Subunits; Rats | 2012 |
p57(KIP2) control of actin cytoskeleton dynamics is responsible for its mitochondrial pro-apoptotic effect.
p57 (Kip2, cyclin-dependent kinase inhibitor 1C), often found downregulated in cancer, is reported to hold tumor suppressor properties. Originally described as a cyclin-dependent kinase (cdk) inhibitor, p57(KIP2) has since been shown to influence other cellular processes, beyond cell cycle regulation, including cell death and cell migration. Inhibition of cell migration by p57(KIP2) is attributed to the stabilization of the actin cytoskeleton through the activation of LIM domain kinase-1 (LIMK-1). Furthermore, p57(KIP2) is able to enhance mitochondrial-mediated apoptosis. Here, we report that the cell death promoting effect of p57(KIP2) is linked to its effect on the actin cytoskeleton. Indeed, whereas Jasplakinolide, an actin cytoskeleton-stabilizing agent, mimicked p57(KIP2)'s pro-apoptotic effect, destabilizing the actin cytoskeleton with cytochalsin D reversed p57(KIP2)'s pro-apoptotic function. Conversely, LIMK-1, the enzyme mediating p57(KIP2)'s effect on the actin cytoskeleton, was required for p57(KIP2)'s death promoting effect. Finally, p57(KIP2-)mediated stabilization of the actin cytoskeleton was associated with the displacement of hexokinase-1, an inhibitor of the mitochondrial voltage-dependent anion channel, from the mitochondria, providing a possible mechanism for the promotion of the mitochondrial apoptotic cell death pathway. Altogether, our findings link together two tumor suppressor properties of p57(KIP2), by showing that the promotion of cell death by p57(KIP2) requires its actin cytoskeleton stabilization function. Topics: Actin Cytoskeleton; Apoptosis; Cell Movement; Cyclin-Dependent Kinase Inhibitor p57; Cytochalasin D; Depsipeptides; HeLa Cells; Hexokinase; Humans; Lim Kinases; Mitochondria; Staurosporine; Voltage-Dependent Anion Channel 1 | 2012 |
Actin polymerization controls the activation of multidrug efflux at fertilization by translocation and fine-scale positioning of ABCB1 on microvilli.
Fertilization changes the structure and function of the cell surface. In sea urchins, these changes include polymerization of cortical actin and a coincident, switch-like increase in the activity of the multidrug efflux transporter ABCB1a. However, it is not clear how cortical reorganization leads to changes in membrane transport physiology. In this study, we used three-dimensional superresolution fluorescence microscopy to resolve the fine-scale movements of the transporter along polymerizing actin filaments, and we show that efflux activity is established after ABCB1a translocates to the tips of the microvilli. Inhibition of actin polymerization or bundle formation prevents tip localization, resulting in the patching of ABCB1a at the cell surface and decreased efflux activity. In contrast, enhanced actin polymerization promotes tip localization. Finally, interference with Rab11, a regulator of apical recycling, inhibits activation of efflux activity in embryos. Together our results show that actin-mediated, short-range traffic and positioning of transporters at the cell surface regulates multidrug efflux activity and highlight the multifaceted roles of microvilli in the spatial distribution of membrane proteins. Topics: Actins; Animals; ATP Binding Cassette Transporter, Subfamily B, Member 1; Biological Transport; Blotting, Western; Bridged Bicyclo Compounds, Heterocyclic; Cell Membrane; Cytochalasin D; Depsipeptides; Embryo, Nonmammalian; Female; Fertilization; Male; Microscopy, Confocal; Microscopy, Fluorescence; Microvilli; Ovum; Phylogeny; Polymerization; Protein Transport; rab GTP-Binding Proteins; Sea Urchins; Thiazolidines | 2012 |
Regulation of retinoschisin secretion in Weri-Rb1 cells by the F-actin and microtubule cytoskeleton.
Retinoschisin is encoded by the gene responsible for X-linked retinoschisis (XLRS), an early onset macular degeneration that results in a splitting of the inner layers of the retina and severe loss in vision. Retinoschisin is predominantly expressed and secreted from photoreceptor cells as a homo-oligomer protein; it then associates with the surface of retinal cells and maintains the retina cellular architecture. Many missense mutations in the XLRS1 gene are known to cause intracellular retention of retinoschisin, indicating that the secretion process of the protein is a critical step for its normal function in the retina. However, the molecular mechanisms underlying retinoschisin's secretion remain to be fully elucidated. In this study, we investigated the role of the F-actin cytoskeleton in the secretion of retinoschisin by treating Weri-Rb1 cells, which are known to secrete retinoschisin, with cytochalasin D, jasplakinolide, Y-27632, and dibutyryl cGMP. Our results show that cytochalasin D and jasplakinolide inhibit retinoschisin secretion, whereas Y-27632 and dibutyryl cGMP enhance secretion causing F-actin alterations. We also demonstrate that high concentrations of taxol, which hyperpolymerizes microtubules, inhibit retinoschisin secretion. Our data suggest that retinoschisin secretion is regulated by the F-actin cytoskeleton, that cGMP or inhibition of ROCK alters F-actin structure enhancing the secretion, and that the microtubule cytoskeleton is also involved in this process. Topics: Actins; Amides; Blotting, Western; Cell Line, Tumor; Cytochalasin D; Cytoskeleton; Depsipeptides; Dibutyryl Cyclic GMP; Eye Proteins; Humans; Immunohistochemistry; Microtubules; Paclitaxel; Pyridines; Retinoschisis; Tubulin Modulators | 2011 |
Identification of human P2X1 receptor-interacting proteins reveals a role of the cytoskeleton in receptor regulation.
P2X1 receptors are ATP-gated ion channels expressed by smooth muscle and blood cells. Carboxyl-terminally His-FLAG-tagged human P2X1 receptors were stably expressed in HEK293 cells and co-purified with cytoskeletal proteins including actin. Disruption of the actin cytoskeleton with cytochalasin D inhibited P2X1 receptor currents with no effect on the time course of the response or surface expression of the receptor. Stabilization of the cytoskeleton with jasplakinolide had no effect on P2X1 receptor currents but decreased receptor mobility. P2X2 receptor currents were unaffected by cytochalasin, and P2X1/2 receptor chimeras were used to identify the molecular basis of actin sensitivity. These studies showed that the intracellular amino terminus accounts for the inhibitory effects of cytoskeletal disruption similar to that shown for lipid raft/cholesterol sensitivity. Stabilization of the cytoskeleton with jasplakinolide abolished the inhibitory effects of cholesterol depletion on P2X1 receptor currents, suggesting that lipid rafts may regulate the receptor through stabilization of the cytoskeleton. These studies show that the cytoskeleton plays an important role in P2X1 receptor regulation. Topics: Actins; Adenosine Triphosphate; Animals; Cytochalasin D; Cytoskeleton; Depsipeptides; Electrophysiology; Gene Expression Regulation; HEK293 Cells; Humans; Membrane Microdomains; Microscopy, Confocal; Models, Biological; Myocytes, Smooth Muscle; Protein Kinase C; Protein Structure, Tertiary; Rats; Receptors, Purinergic P2X1 | 2011 |
Submembranous septins as relatively stable components of actin-based membrane skeleton.
The cell cortex is organized by the dynamic interplay between the plasma membrane, membrane proteins, and the cytoskeleton. Despite the cortical localization of septin heteropolymers in vivo and their direct interaction with phospholipid membranes in vitro, their behavior and roles remain elusive. This study characterizes the major cortical septin assembly found in mammalian tissue culture cells by fluorescence recovery after photobleaching analysis. GFP-tagged septin subunits, which colocalized with cortical actin, exhibited slower turnover than some other cortical proteins that were analyzed (e.g., actin, syntaxin-1A and a glutamate aspartate transporter [GLAST]). Perturbation of actin turnover by cytochalasin D or jasplakinolide retarded the cortical septin turnover, while septin depletion by RNAi did not recognizably affect cortical actin turnover. These phenomena are compatibly interpreted by septins' selective association with a subset of actin-based membrane skeleton, as revealed by rapid-freeze deep-etch immuno-replica electron microscopy. We applied the assay system to test septins' presumptive scaffold function on their physiological binding partners. Septin filament destabilization by RNAi-mediated subunit depletion facilitated the turnover of GLAST, depending on the carboxyl-terminal 29 residues, while a septin filament-stabilizing drug forchlorfenuron restrained more GLAST in the unexchangeable fraction. These data indicate that cortical septin heteropolymers are components of the actin-based membrane skeleton providing scaffolds for their interacting partners probably by impeding their lateral diffusion. We predict that diverse submembranous septin clusters found in vivo may serve as scaffolds or reserve pools for specific membrane-bound proteins. Topics: Actins; Animals; Antineoplastic Agents; Cell Line, Tumor; Cell Membrane; Cytochalasin D; Cytoskeleton; Depsipeptides; Membrane Proteins; Mice; Nucleic Acid Synthesis Inhibitors; Phenylurea Compounds; Pyridines; Septins | 2011 |
Actin cytoskeleton modulates ADMA-induced NF-kappaB nuclear translocation and ICAM-1 expression in endothelial cells.
Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase (NOS) inhibitor, increases the activity of NF-κB (NF-κB) and then induces the expression of intercellular adhesion molecule-1 (ICAM-1). However, the mechanisms regulating ADMA-induced NF-κB activation are unknown. This study investigated the function of actin cytoskeleton for ADMA-induced NF-κB activation and ICAM-1 expression in endothelial cells.. Human umbilical vein endothelial cells (HUVEC) were cultured and left untreated or challenged for 24 h with 100 µM ADMA in the absence and presence of 5 µM cytochalasin D (Cyt D), or 1 µM Jasplakinolide (Jas). The form of actin cytoskeleton, the translocation of NF-κB, NF-κB DNA binding activity, and the expression of ICAM-1 were determined.. ADMA increased the formation of stress fiber in endothelial cells, and Cyt D clearly induced destabilization of the actin filaments. Either stabilizing or destabilizing the actin cytoskeleton prevented ADMA-induced NF-κB activation. It also showed that the inhibition of NF-κB activity was due to the impaired NF-κB nuclear translocation. Further, stabilizing or destabilizing the actin cytoskeleton inhibited the expression of the NF-κB target protein, ICAM-1.. Actin cytoskeleton may be engaged in modulated ADMA-induced NF-κB activation and thereby ICAM-1 expression in endothelial cells. Topics: Actin Cytoskeleton; Arginine; Cell Nucleus; Cytochalasin D; Depsipeptides; DNA; Human Umbilical Vein Endothelial Cells; Humans; Intercellular Adhesion Molecule-1; NF-kappa B; Protein Binding; Protein Transport; Stress Fibers; Transcription Factor RelA | 2011 |
Abolishing myofibroblast arrhythmogeneicity by pharmacological ablation of α-smooth muscle actin containing stress fibers.
Myofibroblasts typically appear in the myocardium after insults to the heart like mechanical overload and infarction. Apart from contributing to fibrotic remodeling, myofibroblasts induce arrhythmogenic slow conduction and ectopic activity in cardiomyocytes after establishment of heterocellular electrotonic coupling in vitro. So far, it is not known whether α-smooth muscle actin (α-SMA) containing stress fibers, the cytoskeletal components that set myofibroblasts apart from resident fibroblasts, are essential for myofibroblasts to develop arrhythmogenic interactions with cardiomyocytes.. We investigated whether pharmacological ablation of α-SMA containing stress fibers by actin-targeting drugs affects arrhythmogenic myofibroblast-cardiomyocyte cross-talk.. Experiments were performed with patterned growth cell cultures of neonatal rat ventricular cardiomyocytes coated with cardiac myofibroblasts. The preparations exhibited slow conduction and ectopic activity under control conditions. Exposure to actin-targeting drugs (Cytochalasin D, Latrunculin B, Jasplakinolide) for 24 hours led to disruption of α-SMA containing stress fibers. In parallel, conduction velocities increased dose-dependently to values indistinguishable from cardiomyocyte-only preparations and ectopic activity measured continuously over 24 hours was completely suppressed. Mechanistically, antiarrhythmic effects were due to myofibroblast hyperpolarization (Cytochalasin D, Latrunculin B) and disruption of heterocellular gap junctional coupling (Jasplakinolide), which caused normalization of membrane polarization of adjacent cardiomyocytes.. The results suggest that α-SMA containing stress fibers importantly contribute to myofibroblast arrhythmogeneicity. After ablation of this cytoskeletal component, cells lose their arrhythmic effects on cardiomyocytes, even if heterocellular electrotonic coupling is sustained. The findings identify α-SMA containing stress fibers as a potential future target of antiarrhythmic therapy in hearts undergoing structural remodeling. Topics: Actins; Action Potentials; Animals; Animals, Newborn; Anti-Arrhythmia Agents; Arrhythmias, Cardiac; Bridged Bicyclo Compounds, Heterocyclic; Cell Communication; Cell Shape; Cells, Cultured; Coculture Techniques; Cytochalasin D; Depsipeptides; Dose-Response Relationship, Drug; Gap Junctions; Myocytes, Cardiac; Myofibroblasts; Phenotype; Rats; Rats, Wistar; Stress Fibers; Thiazolidines; Time Factors | 2011 |
Actin cytoskeleton regulates stretch-activated Ca2+ influx in human pulmonary microvascular endothelial cells.
During high tidal volume mechanical ventilation in patients with acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), regions of the lung are exposed to excessive stretch, causing inflammatory responses and further lung damage. In this study, the effects of mechanical stretch on intracellular Ca(2+) concentration ([Ca(2+)](i)), which regulates a variety of endothelial properties, were investigated in human pulmonary microvascular endothelial cells (HPMVECs). HPMVECs grown on fibronectin-coated silicon chambers were exposed to uniaxial stretching, using a cell-stretching apparatus. After stretching and subsequent unloading, [Ca(2+)](i), as measured by fura-2 fluorescence, was transiently increased in a strain amplitude-dependent manner. The elevation of [Ca(2+)](i) induced by stretch was not evident in the Ca(2+)-free solution and was blocked by Gd(3+), a stretch-activated channel inhibitor, or ruthenium red, a transient receptor potential vanilloid inhibitor. The disruption of actin polymerization with cytochalasin D inhibited the stretch-induced elevation of [Ca(2+)](i). In contrast, increases in [Ca(2+)](i) induced by thapsigargin or thrombin were not affected by cytochalasin D. Increased actin polymerization with sphingosine-1-phosphate or jasplakinolide enhanced the stretch-induced elevation of [Ca(2+)](i). A simple network model of the cytoskeleton was also developed in support of the notion that actin stress fibers are required for efficient force transmission to open stretch-activated Ca(2+) channels. In conclusion, mechanical stretch activates Ca(2+) influx via stretch-activated channels which are tightly regulated by the actin cytoskeleton different from other Ca(2+) influx pathways such as receptor-operated and store-operated Ca(2+) entries in HPMVECs. These results suggest that abnormal Ca(2+) homeostasis because of excessive mechanical stretch during mechanical ventilation may play a role in the progression of ALI/ARDS. Topics: Actins; Calcium; Cells, Cultured; Cytochalasin D; Cytoskeleton; Depsipeptides; Humans; Lung; Lysophospholipids; Microcirculation; Microscopy, Fluorescence; Models, Chemical; Sphingosine; Stress, Mechanical; Thapsigargin | 2010 |
The kinetics of force-induced cell reorganization depend on microtubules and actin.
The cytoskeleton is an important factor in the functional and structural adaption of cells to mechanical forces. In this study we investigated the impact of microtubules and the acto-myosin machinery on the kinetics of force-induced reorientation of NIH3T3 fibroblasts. These cells were subjected to uniaxial stretching forces that are known to induce cellular reorientation perpendicular to the stretch direction. We found that disruption of filamentous actin using cytochalasin D and latrunculin B as well as an induction of a massive unpolarized actin polymerization by jasplakinolide, inhibited the stretch-induced reorientation. Similarly, blocking of myosin II activity abolished the stretch-induced reorientation of cells but, interestingly, increased their motility under stretching conditions in comparison to myosin-inhibited nonstretched cells. Investigating the contribution of microtubules to the cellular reorientation, we found that, although not playing a significant role in reorientation itself, microtubule stability had a significant impact on the kinetics of this event. Overall, we conclude that acto-myosin, together with microtubules, regulate the kinetics of force-induced cell reorientation. Topics: Actins; Animals; Bridged Bicyclo Compounds, Heterocyclic; Cell Movement; Cell Polarity; Cells, Cultured; Cytochalasin D; Depsipeptides; Heterocyclic Compounds, 4 or More Rings; Kinetics; Mice; Microtubules; NIH 3T3 Cells; Nocodazole; Paclitaxel; Structure-Activity Relationship; Thiazolidines | 2010 |
Role of the cytoskeleton in Cd2+-induced death of mouse mesangial cells.
Cadmium induces apoptotic cell death in mouse mesangial cells that is in part dependent on reactive oxygen species (ROS). Cadmium also activates multiple kinases in these cells, including the Ca2+/calmodulin-dependent protein kinase II (CaMK-II) and p38 kinase, and also leads to disruption of the filamentous actin cytoskeleton. We investigated the role of the cytoskeleton in Cd2+-induced cell death. Cell viability was decreased by Cd2+ and two types of apoptotic death, defined by flow cytometry, were increased. Disruption of actin filaments with cytochalasin D was partially protective, whereas stabilization of the cytoskeleton with jasplakinolide was without effect, indicating that cytoskeletal disruption contributes to, but is not necessary for, induction of apoptosis. Inhibition of CaMK-II and p38 kinase, mitochondrial stabilization with cyclosporine A, and the antioxidant N-acetyl cysteine all protected against apoptosis and prevented disruption of the cytoskeleton. Cytochalasin D decreased Cd2+-dependent ROS production, reduced the decline in mitochondrial membrane potential, and decreased phosphorylation of p38 kinase. We conclude that Cd2+-dependent actin disruption is a downstream event facilitating apoptotic death. Cadmium-dependent cell death involves actin-dependent mitochondrial changes, ROS production, and p38 activation. Topics: Animals; Cadmium Chloride; Cell Death; Cell Survival; Cells, Cultured; Cytochalasin D; Cytoskeleton; Depsipeptides; Membrane Potential, Mitochondrial; Mesangial Cells; Mice; Reactive Oxygen Species | 2010 |
Statistical analysis of high-resolution light microscope images reveals effects of cytoskeleton-disrupting drugs on the membrane organization of the nicotinic acetylcholine receptor.
Extensive evidence supports the notion that the cytoskeleton participates in the immobilization and membrane clustering of the nicotinic acetylcholine receptor (AChR) at the neuromuscular junction. Stimulated emission depletion fluorescence microscopy has revealed the supramolecular organization of AChR nanoclusters at the surface of CHO-K1/A5 cells with subdiffraction resolution (Kellner et al., Neuroscience 144:135-143 2007). We studied the effect of two cytoskeletal-disrupting drugs (cytochalasin D and jasplakinolide) on the nanoscale distribution of muscle-type AChR expressed in these cells by means of mathematical and statistical analysis of images obtained with the same high-resolution microscopy. AChR nanoclusters were found to be randomly distributed in both controls and cells treated with either drug for distances larger than 500 nm. Treatments altered the distribution of AChR nanoclusters according to their brightness/size. Cytochalasin D and jasplakinolide produced a statistically significant increase in the proportion of medium-size nanoclusters and a diminution of small nanoclusters, indicating higher disrupting activity on the latter. This was further corroborated by the diminution of the brightness/diameter ratio of nanoclusters (a measure of the intracluster density of AChR molecules) and by Ripley's analysis applied to simulated patterns with intracluster aggregation of AChR molecules. The combined analytical tools bring out subtle changes in the two-dimensional organization of the AChR nanoaggregates on disruption of the cytoskeletal network and throw light on the possible link between the cytoskeleton and the distribution of the AChR at the cell surface. Topics: Animals; CHO Cells; Cricetinae; Cricetulus; Cytochalasin D; Depsipeptides; Microscopy, Confocal; Microscopy, Fluorescence; Receptors, Nicotinic | 2010 |
Actin reorganization contributes to loss of cell adhesion in pemphigus vulgaris.
In the human autoimmune blistering skin disease pemphigus vulgaris autoantibodies (PV-IgG), which are mainly directed against keratinocyte cell adhesion molecules desmoglein (Dsg) 3 and Dsg1, cause keratinocyte cell dissociation (acantholysis). Recent studies reported that loss of keratinocyte cell adhesion was accompanied by profound alterations of the actin cytoskeleton. Nevertheless, the relevance of actin reorganization in this process is unclear at present. In this study, we provide evidence for an important role of actin reorganization in pemphigus pathogenesis. In parallel to loss of cell adhesion and fragmentation of Dsg3 staining along cell borders, PV-IgG treatment resulted in striking changes in actin cytoskeleton organization. Moreover, in experiments using fluorescence recovery after photobleaching (FRAP), PV-IgG were detected to interfere with actin dynamics. Therefore, we investigated whether pharmacological manipulation of actin polymerization modulates pathogenic effects of PV-IgG. Pharmacological stabilization of actin filaments via jasplakinolide significantly blocked cell dissociation and Dsg3 fragmentation, whereas cytochalasin D-induced actin depolymerization strongly enhanced pathogenic effects of PV-IgG. To substantiate these findings, we studied whether the protective effects of Rho GTPases, which are potent regulators of the actin cytoskeleton and were shown to be involved in pemphigus pathogenesis, were dependent on modulation of actin dynamics. Cytotoxic necrotizing factor-1 (CNF-1)-mediated activation of Rho-GTPases enhanced the cortical junction-associated actin belt and blunted PV-IgG-induced cell dissociation. However, when actin polymerization was blocked under these conditions via addition of latrunculin B, the protective effects of CNF-1 were abrogated. Taken together, these experiments indicate that reorganization of cortical actin filaments is a critical step in PV-IgG-induced keratinocyte dissociation. Topics: Actins; Autoantibodies; Bacterial Toxins; Bridged Bicyclo Compounds, Heterocyclic; Cell Adhesion; Cell Line; Cytochalasin D; Depsipeptides; Desmoglein 3; Enzyme Activation; Escherichia coli Proteins; Fluorescence Recovery After Photobleaching; Humans; Immunoglobulin G; Keratinocytes; Pemphigus; rho GTP-Binding Proteins; Thiazolidines; Time Factors | 2010 |
Effects of exposure to a time-varying 1.5 T magnetic field on the neurotransmitter-activated increase in intracellular Ca(2+) in relation to actin fiber and mitochondrial functions in bovine adrenal chromaffin cells.
It has been reported that exposure to electromagnetic fields influences intracellular signal transduction. We studied the effects of exposure to a time-varying 1.5 T magnetic field on membrane properties, membrane cation transport and intracellular Ca(2+) mobilization in relation to signals. We also studied the mechanism of the effect of exposure to the magnetic field on intracellular Ca(2+) release from Ca(2+) stores in adrenal chromaffin cells.. We measured the physiological functions of ER, actin protein, and mitochondria with respect to a neurotransmitter-induced increase in Ca(2+) in chromaffin cells exposed to the time-varying 1.5 T magnetic field for 2h.. Exposure to the magnetic field significantly reduced the increase in [Ca(2+)]i. The exposure depolarized the mitochondria membrane and lowered oxygen uptake, but did not reduce the intracellular ATP content. Magnetic field-exposure caused a morphological change in intracellular F-actin. F-actin in exposed cells seemed to be less dense than in control cells, but the decrease was smaller than that in cytochalasin D-treated cells. The increase in G-actin (i.e., the decrease in F-actin) due to exposure was recovered by jasplakinolide, but inhibition of Ca(2+) release by the exposure was unaffected.. These results suggest that the magnetic field-exposure influenced both the ER and mitochondria, but the inhibition of Ca(2+) release from ER was not due to mitochondria inhibition. The effect of eddy currents induced in the culture medium may indirectly influence intracellular actin and suppress the transient increase in [Ca(2+)]i. Topics: Acetylcholine; Actin Cytoskeleton; Actins; Adenosine Triphosphate; Adrenal Glands; Animals; Calcium; Cattle; Cells, Cultured; Chromaffin Cells; Colchicine; Cytochalasin D; Depsipeptides; Electromagnetic Fields; Endoplasmic Reticulum; Immunoblotting; Intracellular Space; Membrane Potential, Mitochondrial; Microscopy, Confocal; Mitochondria; Neurotransmitter Agents; Nucleic Acid Synthesis Inhibitors; Oxygen Consumption; Time Factors; Tubulin Modulators | 2010 |
Reduced levels of intracellular calcium releasing in spermatozoa from asthenozoospermic patients.
Asthenozoospermia is one of the most common findings present in infertile males characterized by reduced or absent sperm motility, but its aetiology remains unknown in most cases. In addition, calcium is one of the most important ions regulating sperm motility. In this study we have investigated the progesterone-evoked intracellular calcium signal in ejaculated spermatozoa from men with normospermia or asthenozoospermia.. Human ejaculates were obtained from healthy volunteers and asthenospermic men by masturbation after 4-5 days of abstinence. For determination of cytosolic free calcium concentration, spermatozoa were loaded with the fluorescent ratiometric calcium indicator Fura-2.. Treatment of spermatozoa from normospermic men with 20 micromolar progesterone plus 1 micromolar thapsigargin in a calcium free medium induced a typical transient increase in cytosolic free calcium concentration due to calcium release from internal stores. Similar results were obtained when spermatozoa were stimulated with progesterone alone. Subsequent addition of calcium to the external medium evoked a sustained elevation in cytosolic free calcium concentration indicative of capacitative calcium entry. However, when progesterone plus thapsigargin were administered to spermatozoa from patients with asthenozoospermia, calcium signal and subsequent calcium entry was much smaller compared to normospermic patients. As expected, pretreatment of normospermic spermatozoa with both the anti-progesterone receptor c262 antibody and with progesterone receptor antagonist RU-38486 decreased the calcium release induced by progesterone. Treatment of spermatozoa with cytochalasin D or jasplakinolide decreased the calcium entry evoked by depletion of internal calcium stores in normospermic patients, whereas these treatments proved to be ineffective at modifying the calcium entry in patients with asthenozoospermia.. Our results suggest that spermatozoa from asthenozoospermic patients present a reduced responsiveness to progesterone. Topics: Actin Cytoskeleton; Asthenozoospermia; Biological Transport; Calcium; Calcium Signaling; Cytochalasin D; Cytosol; Depsipeptides; Enzyme Inhibitors; Humans; Male; Progesterone; Progestins; Spermatozoa; Thapsigargin | 2009 |
Role of cytoskeletal components in stress-relaxation behavior of adherent vascular smooth muscle cells.
A number of recent studies have demonstrated the effectiveness of atomic force microscopy (AFM) for characterization of cellular stress-relaxation behavior. However, this technique's recent development creates considerable need for exploration of appropriate mechanical models for analysis of the resultant data and of the roles of various cytoskeletal components responsible for governing stress-relaxation behavior. The viscoelastic properties of vascular smooth muscle cells (VSMCs) are of particular interest due to their role in the development of vascular diseases, including atherosclerosis and restenosis. Various cytoskeletal agents, including cytochalasin D, jasplakinolide, paclitaxel, and nocodazole, were used to alter the cytoskeletal architecture of the VSMCs. Stress-relaxation experiments were performed on the VSMCs using AFM. The quasilinear viscoelastic (QLV) reduced-relaxation function, as well as a simple power-law model, and the standard linear solid (SLS) model, were fitted to the resultant stress-relaxation data. Actin depolymerization via cytochalasin D resulted in significant increases in both rate of relaxation and percentage of relaxation; actin stabilization via jasplakinolide did not affect stress-relaxation behavior. Microtubule depolymerization via nocodazole resulted in nonsignificant increases in rate and percentage of relaxation, while microtubule stabilization via paclitaxel caused significant decreases in both rate and percentage of relaxation. Both the QLV reduced-relaxation function and the power-law model provided excellent fits to the data (R(2)=0.98), while the SLS model was less adequate (R(2)=0.91). Data from the current study indicate the important role of not only actin, but also microtubules, in governing VSMC viscoelastic behavior. Excellent fits to the data show potential for future use of both the QLV reduced-relaxation function and power-law models in conjunction with AFM stress-relaxation experiments. Topics: Actins; Animals; Aorta; Cell Adhesion; Cytochalasin D; Depsipeptides; Elasticity; Endothelium, Vascular; Linear Models; Male; Microtubules; Models, Biological; Muscle Relaxation; Myocytes, Smooth Muscle; Nocodazole; Paclitaxel; Rats; Rats, Sprague-Dawley; Reproducibility of Results; Stress, Mechanical; Tubulin Modulators; Viscosity | 2009 |
Participation of actin on Giardia lamblia growth and encystation.
Microfilaments play a determinant role in different cell processes such as: motility, cell division, phagocytosis and intracellular transport; however, these structures are poorly understood in the parasite Giardia lamblia.. By confocal microscopy using TRITC-phalloidin, we found structured actin distributed in the entire trophozoite, the label stand out at the ventral disc, median body, flagella and around the nuclei. During Giardia encystation, a sequence of morphological changes concurrent to modifications on the distribution of structured actin and in the expression of actin mRNA were observed. To elucidate whether actin participates actively on growth and encystation, cells were treated with Cytochalasin D, Latrunculin A and Jasplakinolide and analyzed by confocal and scanning electron microscopy. All drugs caused a growth reduction (27 to 45%) and changes on the distribution of actin. Besides, 60 to 80% of trophozoites treated with the drugs, exhibited damage at the caudal region, alterations in the flagella and wrinkles-like on the plasma membrane. The drugs also altered the cyst-yield and the morphology, scanning electron microscopy revealed diminished cytokinesis, cysts with damages in the wall and alterations in the size and on the intermembranal space. Furthermore, the drugs caused a significant reduction of the intensity of fluorescence-labeled CWP1 on ESV and on cyst wall, this was coincident with a reduction of CWP1 gene expression (34%).. All our results, indicated an important role of actin in the morphology, growth and encystation and indirectly suggested an actin role in gene expression. Topics: Actin Cytoskeleton; Actins; Animals; Bridged Bicyclo Compounds, Heterocyclic; Cytochalasin D; Depsipeptides; Flagella; Giardia lamblia; Mice; Mice, Inbred BALB C; Microscopy, Confocal; Microscopy, Electron, Scanning; Models, Biological; Phalloidine; Rats; Rats, Wistar; Rhodamines; Thiazolidines | 2009 |
Amplification of transducer gain by angiotensin II-mediated enhancement of cortical actin density in osmosensory neurons.
Osmosensory neurons transduce osmotic signals into a neural spike code that commands behavioral and endocrine responses that mediate body fluid homeostasis. Although changes in osmoregulatory reflex gain are known to occur under physiological and pathological conditions, the basis for this modulation is unknown. Here, we show that angiotensin II amplifies osmosensory transduction by enhancing the proportional relationship between osmolality, receptor potential, and action potential firing in rat supraoptic nucleus neurons. This effect is mediated by a phospholipase C- and protein kinase C-dependent increase in cellular mechanosensitivity that is associated with a rapid increase in cortical actin filament density. Preventing this increase with cytochalasin D eliminated the enhancement of mechanosensitivity, whereas enhancing actin filament density with jasplakinolide potentiated mechanosensitivity and occluded the effects of angiotensin II. These results indicate that a receptor-mediated increase in cortical actin density can enhance osmosensitivity in acutely isolated supraoptic neurons. Topics: Actins; Action Potentials; Angiotensin II; Animals; Antineoplastic Agents; Cytochalasin D; Cytoskeleton; Depsipeptides; Male; Mechanotransduction, Cellular; Neurons, Afferent; Nucleic Acid Synthesis Inhibitors; Protein Kinase C; Rats; Rats, Long-Evans; Supraoptic Nucleus; Type C Phospholipases; Water-Electrolyte Balance | 2008 |
Actin is required for endocytic trafficking in the malaria parasite Plasmodium falciparum.
The intra-erythrocytic stages of the malaria parasite endocytose large quantities of the surrounding erythrocyte cytoplasm and deliver it to a digestive food vacuole via endocytic vesicles. Digestion provides amino acids for parasite protein synthesis and is required to maintain the osmotic integrity of the host cell. The parasite endocytic pathway has been described morphologically by electron microscopy, but the molecular mechanisms that mediate and regulate it remain elusive. Given the involvement of actin in endocytosis in other eukaryotes, we have used actin inhibitors to assess the requirement for this protein in the endocytic pathway of the human malaria parasite, Plasmodium falciparum. Treatment of cultures with cytochalasin D did not affect haemoglobin levels in the parasites when co-administered with protease inhibitors, and neither did it affect the uptake of the endocytic tracer horseradish peroxidase, suggesting the absence of actin in the mechanism of endocytosis. However, in the absence of protease inhibitors, treated parasites contained increased levels of haemoglobin due to an accumulation of enlarged endocytic vesicles, as determined by immunofluorescence and electron microscopy, suggesting a role for actin in vesicle trafficking, possibly by mediating vesicle maturation and/or fusion to the digestive vacuole. In contrast to cytochalasin D, treatment with jasplakinolide led to an inhibition of endocytosis, an accumulation of vesicles closer to the plasma membrane and a marked concentration of actin in the parasite cortex. We propose that the stabilization of cortical actin filaments by jasplakinolide interferes with normal endocytic vesicle formation and migration from the cell periphery. Topics: Actin Cytoskeleton; Actins; Animals; Biological Transport; Cells, Cultured; Cytochalasin D; Depsipeptides; Endocytosis; Erythrocytes; Hemoglobins; Humans; Microscopy, Electron, Transmission; Microscopy, Fluorescence; Plasmodium falciparum; Protease Inhibitors; Transport Vesicles | 2008 |
Rho-kinase-dependent F-actin rearrangement is involved in the inhibition of PI3-kinase/Akt during ischemia-reperfusion-induced endothelial cell apoptosis.
Activation of cytoskeleton regulator Rho-kinase during ischemia-reperfusion (I/R) plays a major role in I/R injury and apoptosis. Since Rho-kinase is a negative regulator of the pro-survival phosphatidylinositol 3-kinase (PI3-kinase)/Akt pathway, we hypothesized that inhibition of Rho-kinase can prevent I/R-induced endothelial cell apoptosis by maintaining PI3-kinase/Akt activity and that protective effects of Rho-kinase inhibition are facilitated by prevention of F-actin rearrangement. Human umbilical vein endothelial cells were subjected to 1 h of simulated ischemia and 1 or 24 h of simulated reperfusion after treatment with Rho-kinase inhibitor Y-27632, PI3-kinase inhibitor wortmannin, F-actin depolymerizers cytochalasinD and latrunculinA and F-actin stabilizer jasplakinolide. Intracellular ATP levels decreased following I/R. Y-27632 treatment reduced I/R-induced apoptosis by 31% (P < 0.01) and maintained Akt activity. Both effects were blocked by co-treatment with wortmannin. Y-27632 treatment prevented the formation of F-actin bundles during I/R. Similar results were observed with cytochalasinD treatment. In contrast, latrunculinA and jasplakinolide treatment did not prevent the formation of F-actin bundles during I/R and had no effect on I/R-induced apoptosis. Apoptosis and Akt activity were inversely correlated (R (2) = 0.68, P < 0.05). In conclusion, prevention of F-actin rearrangement by Rho-kinase inhibition or by cytochalasinD treatment attenuated I/R-induced endothelial cell apoptosis by maintaining PI3-kinase and Akt activity. Topics: Actins; Amides; Androstadienes; Apoptosis; Bridged Bicyclo Compounds, Heterocyclic; Cells, Cultured; Cytochalasin D; Depsipeptides; Endothelium, Vascular; Humans; Phosphoinositide-3 Kinase Inhibitors; Proto-Oncogene Proteins c-akt; Pyridines; Reperfusion Injury; rho-Associated Kinases; Thiazolidines; Wortmannin | 2008 |
Latrunculin B facilitates Shiga toxin 1 transcellular transcytosis across T84 intestinal epithelial cells.
Shiga toxins (Stx), released into the intestinal lumen by enterohemorrhagic Escherichia coli (EHEC), are major virulence factors responsible for gastrointestinal and systemic illnesses. These pathologies are believed to be due to the action of the toxins on endothelial cells, which express the Stx receptor, the glycosphingolipid Gb3. To reach the endothelial cells, Stx must translocate across the intestinal epithelial monolayer. This process is poorly understood. We investigated Stx1 movement across the intestinal epithelial T84 cell model and the role of actin turnover in this transcytosis. We showed that changes in the actin cytoskeleton due to latrunculin B, but not cytochalasin D or jasplakinolide, significantly facilitate toxin transcytosis across T84 monolayers. This trafficking is transcellular and completely inhibited by tannic acid, a cell impermeable plasma membrane fixative. This indicates that actin turnover could play an important role in Stx1 transcellular transcytosis across intestinal epithelium in vitro. Since EHEC attachment to epithelial cells causes an actin rearrangement, this finding may be highly relevant to Stx-induced disease. Topics: Actins; Biological Transport; Bridged Bicyclo Compounds, Heterocyclic; Cell Line, Tumor; Cytochalasin D; Depsipeptides; Dextrans; Epithelial Cells; Fluorescent Antibody Technique; Humans; Immunoblotting; Intestinal Mucosa; Shiga Toxin 1; Thiazolidines | 2008 |
Role of nitric oxide in actin depolymerization and programmed cell death induced by fusicoccin in sycamore (Acer pseudoplatanus) cultured cells.
Programmed cell death (PCD) plays a vital role in plant development and is involved in defence mechanisms against biotic and abiotic stresses. Different forms of PCD have been described in plants on the basis of the cell organelle first involved. In sycamore (Acer pseudoplatanus L.) cultured cells, the phytotoxin fusicoccin (FC) induces cell death. However, only a fraction of the dead cells shows the typical hallmarks of animal apoptosis, including cell shrinkage, chromatin condensation, DNA fragmentation and release of cytochrome c from the mitochondrion. In this work, we show that the scavenging of nitric oxide (NO), produced in the presence of FC, by 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (cPTIO) and rutin inhibits cell death without affecting DNA fragmentation and cytochrome c release. In addition, we show that FC induces a massive depolymerization of actin filaments that is prevented by the NO scavengers. Finally, the addition of actin-depolymerizing drugs induces PCD in control cells and overcomes the inhibiting effect of cPTIO on FC-induced cell death. Vice versa, the addition of actin-stabilizing drugs to FC-treated cells partially inhibits the phytotoxin-induced PCD. These results suggest that besides an apoptotic-like form of PCD involving the release of cytochrome c, FC induces at least another form of cell death, likely mediated by NO and independent of cytochrome c release, and they make it tempting to speculate that changes in actin cytoskeleton are involved in this form of PCD. Topics: Acer; Actin Cytoskeleton; Actins; Apoptosis; Benzoates; Cell Nucleus; Cells, Cultured; Cytochalasin D; Cytochromes c; Cytoskeleton; Depsipeptides; DNA Fragmentation; Glycosides; Hydrogen Peroxide; Imidazoles; Nitric Oxide; Rutin | 2008 |
A new model for hemoglobin ingestion and transport by the human malaria parasite Plasmodium falciparum.
The current model for hemoglobin ingestion and transport by intraerythrocytic Plasmodium falciparum malaria parasites shares similarities with endocytosis. However, the model is largely hypothetical, and the mechanisms responsible for the ingestion and transport of host cell hemoglobin to the lysosome-like food vacuole (FV) of the parasite are poorly understood. Because actin dynamics play key roles in vesicle formation and transport in endocytosis, we used the actin-perturbing agents jasplakinolide and cytochalasin D to investigate the role of parasite actin in hemoglobin ingestion and transport to the FV. In addition, we tested the current hemoglobin trafficking model through extensive analysis of serial thin sections of parasitized erythrocytes (PE) by electron microscopy. We find that actin dynamics play multiple, important roles in the hemoglobin transport pathway, and that hemoglobin delivery to the FV via the cytostomes might be required for parasite survival. Evidence is provided for a new model, in which hemoglobin transport to the FV occurs by a vesicle-independent process. Topics: Actin Cytoskeleton; Actins; Animals; Antifungal Agents; Cytochalasin D; Depsipeptides; Endocytosis; Erythrocytes; Hemoglobins; Humans; Malaria, Falciparum; Microscopy, Electron, Transmission; Microtomy; Models, Biological; Nucleic Acid Synthesis Inhibitors; Plasmodium falciparum; Protein Transport; Transport Vesicles; Vacuoles | 2008 |
Participation of Rho, ROCK-2, and GAP activities during actin microfilament rearrangements in Entamoeba histolytica induced by fibronectin signaling.
In Entamoeba histolytica little is known about the microfilament rearrangements formed by actin and ABPs. Fibronectin regulates many aspects of cell behavior involving the actin cytoskeleton and members of the Rho family of small GTPases. Using trophozoites interacted with fibronectin and glass, we present evidence related with the formation and regulation of different microfilament rearrangements and their cellular distribution, the effect of actin affecting drugs on these arrangements, and on trophozoites adhesion; we also demonstrate that actin isoforms are induced after adhesion, and also the selective participation of specific actin binding proteins such as ABP-120 and phospho-paxillin, regarding their location in the different actin structures. In addition, we show results that confirm the participation of EhRho, ROCK-2, and GAP activities. We propose that fibronectin induced signaling in E. histolytica trophozoites have important consequences in the actin cytoskeleton that may affect its behavior during the invasive process in the host. Topics: Actin Cytoskeleton; Actins; Animals; Bridged Bicyclo Compounds, Heterocyclic; Cytochalasin D; Cytoskeleton; Depsipeptides; Entamoeba histolytica; Fibronectins; GTPase-Activating Proteins; Humans; Nucleic Acid Synthesis Inhibitors; Protein Isoforms; rho GTP-Binding Proteins; rho-Associated Kinases; Signal Transduction; Thiazolidines; Trophozoites | 2008 |
A key role for reverse Na+/Ca2+ exchange influenced by the actin cytoskeleton in store-operated Ca2+ entry in human platelets: evidence against the de novo conformational coupling hypothesis.
We have previously demonstrated a role for the reorganization of the actin cytoskeleton in store-operated calcium entry (SOCE) in human platelets and interpreted this as evidence for a de novo conformational coupling step in SOCE activation involving the type II IP(3) receptor and the platelet hTRPC1-containing store-operated channel (SOC). Here, we present evidence challenging this model. The actin polymerization inhibitors cytochalasin D or latrunculin A significantly reduced Ca2+ but not Mn2+ or Na+ entry into thapsigargin (TG)-treated platelets. Jasplakinolide, which induces actin polymerization, also inhibited Ca2+ but not Mn2+ or Na+ entry. However, an anti-hTRPC1 antibody inhibited TG-evoked entry of all three cations, indicating that they all permeate an hTRPC1-containing store-operated channel (SOC). These results indicate that the reorganization of the actin cytoskeleton is not involved in SOC activation. The inhibitors of the Na+/Ca2+ exchanger (NCX), KB-R7943 or SN-6, caused a dose-dependent inhibition of Ca2+ but not Mn2+ or Na+ entry into TG-treated platelets. The effects of the NCX inhibitors were not additive with those of actin polymerization inhibitors, suggesting a common point of action. These results indicate a role for two Ca2+ permeable pathways activated following Ca2+ store depletion in human platelets: A Ca2+-permeable, hTRPC1-containing SOC and reverse Na+/Ca2+ exchange, which is activated following Na+ entry through the SOC and requires a functional actin cytoskeleton. Topics: Actins; Benzyl Compounds; Blood Platelets; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Cytochalasin D; Cytoskeleton; Depsipeptides; Humans; Manganese; Potassium; Sodium; Sodium-Calcium Exchanger; Thapsigargin; Thiazolidines; Thiourea | 2007 |
Insulin-increased prolactin gene expression requires actin treadmilling: potential role for p21 activated kinase.
Insulin-increased prolactin gene transcription in GH4 cells was enhanced by binding on fibronectin. This was mediated by receptor-like protein tyrosine phosphatase alpha, which activated Src, Rho, and phosphatidylinositol 3-kinase. It suggested that insulin signaling to gene transcription was partly dependent on actin rearrangement. This was confirmed through studies using inhibitors of actin treadmilling. Cytochalasin D, jasplakinolide, latrunculin B, and swinholide A altered the actin cytoskeleton of GH4 cells, as assessed by Alexa Fluor phalloidin staining, and inhibited insulin-increased prolactin gene transcription. These reagents did not affect the controls. Nor was it due to a gross defect of insulin signaling because activation/translocation of glycogen synthase kinase 3beta and mammalian target of rapamycin were not affected. Expression of wild-type and mutant actin treadmilling agents, Cdc42, TC10, neuronal Wiskott-Aldrich syndrome protein, and Nck, indicated that they were essential to insulin-increased prolactin gene expression, and suggested that activation of p21 associated kinase (PAK) might also be essential to this process. PAK expression also increased and PAK mutants decreased prolactin promoter activity in insulin-treated cells. The activation of PAK in the presence of inhibitors was also consistent with a role in activation of insulin-increased prolactin gene expression. Finally, small interfering RNA-mediated reduction of PAK decreased the effect of insulin on prolactin gene expression. Thus, it is likely that insulin activation of actin treadmilling through Cdc42/TC10 and neuronal Wiskott-Aldrich syndrome protein activates PAK and prolactin gene transcription. Topics: Actins; Animals; Blotting, Western; Bridged Bicyclo Compounds, Heterocyclic; cdc42 GTP-Binding Protein; Cytochalasin D; Depsipeptides; Electrophoresis, Polyacrylamide Gel; Gene Expression; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Insulin; Marine Toxins; Microscopy, Fluorescence; Mutation; p21-Activated Kinases; PC12 Cells; Phosphorylation; Prolactin; Protein Kinases; Rats; rho GTP-Binding Proteins; RNA, Small Interfering; Signal Transduction; Thiazolidines; TOR Serine-Threonine Kinases; Transcription, Genetic; Transfection | 2007 |
Modulation of CCK-8-evoked intracellular Ca2+ waves by hydrogen peroxide in mouse pancreatic acinar cells.
In the present study we have employed single cell imaging analysis to monitor the propagation of cholecystokinin-evoked Ca(2+) waves in mouse pancreatic acinar cells. Stimulation of cells with 1 nM CCK-8 led to an initial Ca(2+) release at the luminal cell pole and subsequent spreading of the Ca(2+) signal towards the basolateral membrane in the form of a Ca(2+) wave. Inhibition of sarcoendoplasmic reticulum Ca(2+)-ATPase (SERCA) activity by 1 microM thapsigargin, preincubation in the presence of 100 microM H(2)O(2) or inhibition of PKC with either 5 microM Ro31-8220 or 3 microM GF-109203-X all led to a faster propagation of CCK-8-induced Ca(2+) signals. The propagation of CCK-8-evoked Ca(2+) signals was slowed down by activation of PKC with 1 microM PMA, and preincubation of cells in the presence of H(2)O(2) counteracted the effect of PKC inhibition. The protonophore FCCP (100 nM) and the inhibitor of the mitochondrial Ca(2+)-uniporter Ru360 (10 microM) led to an increase in the propagation rate of CCK-8-evoked Ca(2+) waves. Finally, depolymerisation of actin cytoskeleton with cytochalasin D (10 microM) led to a faster propagation of CCK-8-evoked Ca(2+) signals. Stabilization of actin cytoskeleton with jasplakinolide (10 microM) did not induce significant changes on CCK-8-evoked Ca(2+) waves. Preincubation of cells in the presence of H(2)O(2) counteracted the effect of cytochalasin D on CCK-8-evoked Ca(2+) wave propagation. Our results suggest that spreading of cytosolic Ca(2+) waves evoked by CCK-8 can be modulated by low levels of oxidants acting on multiple Ca(2+)-handling mechanisms. Topics: Animals; Benzimidazoles; Calcium Signaling; Carbocyanines; Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone; Cholecystokinin; Cytochalasin D; Cytoskeleton; Depsipeptides; Dose-Response Relationship, Drug; Hydrogen Peroxide; Indoles; Intracellular Fluid; Male; Maleimides; Membrane Potential, Mitochondrial; Mice; Mitochondria; Oligomycins; Organotin Compounds; Pancreas, Exocrine; Protein Kinase C; Ruthenium Compounds; Salicylates; Sarcoplasmic Reticulum Calcium-Transporting ATPases; Sincalide; Thapsigargin | 2007 |
Characterization of the activities of actin-affecting drugs on tumor cell migration.
Metastases kill 90% of cancer patients. It is thus a major challenge in cancer therapy to inhibit the spreading of tumor cells from primary tumor sites to those particular organs where metastases are likely to occur. Whereas the actin cytoskeleton is a key component involved in cell migration, agents targeting actin dynamics have been relatively poorly investigated. Consequently, valuable in vitro pharmacological tools are needed to selectively identify this type of agent. In response to the absence of any standardized process, the present work aims to develop a multi-assay strategy for screening actin-affecting drugs with anti-migratory potentials. To validate our approach, we used two cancer cell lines (MCF7 and A549) and three actin-affecting drugs (cytochalasin D, latrunculin A, and jasplakinolide). We quantified the effects of these drugs on the kinetics of actin polymerization in tubes (by means of spectrofluorimetry) and on the dynamics of actin cytoskeletons within whole cells (by means of fluorescence microscopy). Using quantitative videomicroscopy, we investigated the actual effects of the drugs on cell motility. Finally, the combined drug effects on cell motility and cell growth were evaluated by means of a scratch-wound assay. While our results showed concordant drug-induced effects on actin polymerization occurring in vitro in test tubes and within whole cells, the whole cell assay appeared more sensitive than the tube assay. The inhibition of actin polymerization induced by cytochalasin D was paralleled by a decrease in cell motility for both cell types. In the case of jasplakinolide, which induces actin polymerization, while it significantly enhanced the locomotion of the A549 cells, it significantly inhibited that of the MCF-7 ones. All these effects were confirmed by means of the scratch-wound assay except of the jasplakinolide-induced effects on MCF-7 cell motility. These later seemed compensated by an additional effect occurring during wound recolonization (possibly acting on the cell growth features). In conclusion, the use of multi-assays with different levels of sophistication and biological relevance is recommended in the screening of new actin-affecting drugs with potentially anti-migratory effects. Topics: Actins; Adenocarcinoma; Antineoplastic Agents; Breast Neoplasms; Bridged Bicyclo Compounds, Heterocyclic; Carcinoma, Non-Small-Cell Lung; Cell Line, Tumor; Cell Movement; Cytochalasin D; Cytoskeleton; Depsipeptides; Humans; Inhibitory Concentration 50; Lung Neoplasms; Neoplasm Invasiveness; Statistics, Nonparametric; Thiazoles; Thiazolidines | 2006 |
Growth and density-dependent regulation of NO synthase by the actin cytoskeleton in pulmonary artery endothelial cells.
We previously reported association of eNOS with actin increases eNOS activity. In the present study, regulation of activity of eNOS by actin cytoskeleton during endothelial growth was studied. We found eNOS activity in PAEC increased when cells grew from preconfluence to confluence. eNOS activity was much greater in PAEC in higher density than those in lower density, suggesting increase in eNOS activity during cell growth is caused by increase in cell density. Although eNOS protein contents were also increased when endothelial cells grew from preconfluence to confluence, magnitude of increase in eNOS activity was much higher than increase in eNOS protein content, suggesting posttranslational mechanisms played an important role in regulation of eNOS activity during endothelial growth. Confocal fluorescence microscopy revealed eNOS was colocalized with G-actin in preconfluent cells in perinuclear region, with both G-actin in perinuclear area and cortical F-actin in plasma membrane in confluent cells. There was more beta-actin coimmunoprecipitated with eNOS in Triton X-100-soluble fraction in confluent cells in later growth phase and in high density. Decrease in eNOS association with beta-actin by silencing beta-actin expression using beta-actin siRNA causes inhibition of eNOS activity, NO production, and endothelial monolayer wound repair in PAEC. Moreover, PAEC incubation with cytochalasin D and jasplakinolide resulted in increases in eNOS/actin association and in eNOS activity without changes in eNOS protein content. Yeast two-hybrid experiments suggested strong association between eNOS oxygenase domain and beta-actin. These results indicate increase in eNOS association with actin is responsible for greater eNOS activity in confluent PAEC. Topics: Actins; Animals; Cell Count; Cell Division; Cells, Cultured; Cytochalasin D; Cytoskeleton; Depsipeptides; Endothelial Cells; Gene Silencing; Humans; Immunoprecipitation; Nitric Oxide Synthase Type III; Nucleic Acid Synthesis Inhibitors; Pulmonary Artery; Swine; Tissue Distribution; Two-Hybrid System Techniques; Yeasts | 2006 |
Glutamate-induced Ca2+ influx in third-order neurons of salamander retina is regulated by the actin cytoskeleton.
Ligand-gated ion channels (ionotropic receptors) link to the cortical cytoskeleton via specialized scaffold proteins and thereby to appropriate signal transduction pathways in the cell. We studied the role of filamentous actin in the regulation of Ca influx through glutamate receptor-activated channels in third-order neurons of salamander retina. Staining by Alexa-Fluor 488-phalloidin, to visualize polymerized actin, we show localization of filamentous actin in neurites, and the membrane surrounding the cell soma. With Ca(2+) imaging we found that in dissociated neurons, depolymerization of filamentous actin by latrunculin A, or cytochalasin D significantly reduced glutamate-induced intracellular Ca(2+) accumulation to 53+/-7% of control value. Jasplakinolide, a stabilizer of filamentous actin, by itself slightly increased the glutamate-induced Ca(2+) signal and completely attenuated the inhibitory effect when applied in combination with actin depolymerizing agents. These results indicate that in salamander retinal neurons the actin cytoskeleton regulates Ca(2+) influx through ionotropic glutamate receptor-activated channels, suggesting regulatory roles for filamentous actin in a number of Ca(2+)-dependent physiological and pathological processes. Topics: Actins; Ambystoma; Animals; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Cells, Cultured; Cytochalasin D; Cytoskeleton; Data Interpretation, Statistical; Depsipeptides; Glutamic Acid; In Vitro Techniques; Microscopy, Confocal; Receptors, Glutamate; Retina; Retinal Ganglion Cells; Thiazoles; Thiazolidines | 2006 |
The state of the actin cytoskeleton determines its association with gephyrin: role of ena/VASP family members.
The role the cytoskeleton plays in generating and/or maintaining gephyrin-dependent receptor clusters at inhibitory synapses is poorly understood. Here, the effects of actin cytoskeleton disruption were investigated in eGFP-gephyrin-transfected cells and hippocampal neurons. While gephyrin was not associated with microfilaments in transfected cells, it colocalized with G-actin and cytochalasin-D-induced F-actin patches. The linker region between the MoeA and MogA homology domains of gephyrin was required for colocalization with F-actin patches and for the binding of gephyrin to ena/VASP, an actin anti-capping factor that, in vitro, caused gephyrin binding to polymerized actin. In hippocampal neurons, treatment with cytochalasin D resulted in the redistribution of the neuronal ena/VASP homologue Mena into actin patches and, at early stages of development, a reduction in the number of gephyrin clusters. Our data suggest that Mena binding to F-actin allows for gephyrin recruitment to the leading edge of uncapped actin filaments. Topics: Actin Cytoskeleton; Actins; Animals; Antineoplastic Agents; Bridged Bicyclo Compounds, Heterocyclic; Carrier Proteins; Cell Adhesion Molecules; Cells, Cultured; Cytochalasin D; Cytoskeleton; Depsipeptides; Hippocampus; Humans; Membrane Proteins; Microfilament Proteins; Neurons; Nucleic Acid Synthesis Inhibitors; Phosphoproteins; Protein Structure, Tertiary; Rats; Recombinant Fusion Proteins; Synapses; Thiazoles; Thiazolidines | 2006 |
Cytoskeletal architecture differentially controls post-transcriptional processing of IL-6 and IL-8 mRNA in airway epithelial-like cells.
Airway epithelial cells are critically dependent on an intact cytoskeleton for innate defense functions. There are various pathophysiological conditions that affect the cytoskeletal architecture. We studied the effect of cytoskeletal distortion in polarized airway epithelial-like NCI-H292 cells on inflammatory gene expression, exemplified by interleukin(IL)-6 and IL-8. Disruption of microtubule structure with vinblastin and of actin with cytochalasin D did not affect TNF-alpha-induced IL-6 and IL-8 gene transcription but stabilized IL-8 and IL-6 mRNA. In line with previous studies, IL-8 mRNA stabilization was paralleled by hyperresponsive IL-8 production, but surprisingly, IL-6 production was reduced despite IL-6 mRNA stabilization. Polysome profiling revealed that, in cells with a disrupted cytoskeleton, translational efficiency of IL-6 mRNA was reduced, whereas that of IL-8 mRNA remained unaffected. Our findings indicate that distortion of the cytoskeleton in airway epithelial cells differentially affects both degradation and translation of IL-6 and IL-8 mRNA, modifying inflammatory gene expression and thus their innate defense function. Topics: CCAAT-Enhancer-Binding Protein-beta; CCAAT-Enhancer-Binding Proteins; Cell Line, Tumor; Cytochalasin D; Cytoskeleton; Dactinomycin; Depsipeptides; Epithelial Cells; Humans; Interleukin-12; Interleukin-6; Lung; NF-kappa B; Paclitaxel; Proto-Oncogene Proteins c-fos; Proto-Oncogene Proteins c-jun; RNA Processing, Post-Transcriptional; RNA Stability; RNA, Messenger; Transcription Factor RelA; Tumor Necrosis Factor-alpha; Vinblastine | 2006 |
Tubulin polymerization modulates interleukin-2 receptor signal transduction in human T cells.
Few data exist on the modulation of cytokine receptor signaling by the actin or tubulin cytoskeleton. Therefore, we studied interleukin-2 receptor (IL-2R) signaling in phytohemagglutinine (PHA)-pretreated human T cells in the context of alterations in the cytoskeletal system induced by cytochalasin D (CyD), jasplaklinolide (Jas), taxol (Tax), or colchicine (Col). We found that changes in cytoskeletal tubulin polymerization altered the strength of several IL-2-triggered signals. Moreover, Tax-induced tubulin hyperpolymerization augmented the surface expression of the IL-2R ss -chain and enhanced the association of the IL-2R beta -chain with cytoskeletal tubulin. The IL-2R beta-chain, in turn, was constitutively associated with tubulin and, more weakly, actin. To exclude the possibility that these associations are artifacts caused by PHA, we confirmed them in T cells from TCR-transgenic DO 11.10 mice stimulated with their nominal antigen. We conclude that altered polymerization of cytoskeletal components, especially tubulin, is accompanied by modulation of IL-2 signaling at the receptor level. Topics: Actins; Animals; Antineoplastic Agents; Antineoplastic Agents, Phytogenic; Colchicine; Cytochalasin D; Cytoskeleton; Depsipeptides; Gout Suppressants; Humans; Interleukin-2; Mice; Mice, Transgenic; Mitogens; Nucleic Acid Synthesis Inhibitors; Paclitaxel; Phytohemagglutinins; Receptors, Interleukin-2; Signal Transduction; T-Lymphocytes; Tubulin | 2006 |
Actin polymerisation at the cytoplasmic face of eukaryotic nuclei.
There exists abundant molecular and ultra-structural evidence to suggest that cytoplasmic actin can physically interact with the nuclear envelope (NE) membrane system. However, this interaction has yet to be characterised in living interphase cells.. Using a fluorescent conjugate of the actin binding drug cytochalasin D (CD-BODIPY) we provide evidence that polymerising actin accumulates in vicinity to the NE. In addition, both transiently expressed fluorescent actin and cytoplasmic micro-injection of fluorescent actin resulted in accumulation of actin at the NE-membrane. Consistent with the idea that the cytoplasmic phase of NE-membranes can support this novel pool of perinuclear actin polymerisation we show that isolated, intact, differentiated primary hepatocyte nuclei support actin polymerisation in vitro. Further this phenomenon was inhibited by treatments hindering steric access to outer-nuclear-membrane proteins (e.g. wheat germ agglutinin, anti-nesprin and anti-nucleoporin antibodies).. We conclude that actin polymerisation occurs around interphase nuclei of living cells at the cytoplasmic phase of NE-membranes. Topics: Actins; Animals; Binding Sites; Biopolymers; Boron Compounds; Bridged Bicyclo Compounds, Heterocyclic; Circular Dichroism; Cytochalasin D; Depsipeptides; Fluorescent Dyes; HeLa Cells; Humans; Liver; Nuclear Envelope; Rabbits; Rats; Thiazoles; Thiazolidines | 2006 |
Store-mediated calcium entry in pleural mesothelial cells.
Store-mediated Ca2+ entry is thought as the main pathway for Ca2+ influx in non-excitable cells. Although a role for the actin cytoskeleton in store-mediated Ca2+ entry has been proposed in some cell types, the role of actin cytoskeleton in store-mediated Ca2+ entry is still a controversy. To address this question, the effects of cytoskeletal modifiers on store-mediated Ca2+ entry in pleural mesothelial cells were examined. Thapsigargin (1 microM) induced a sufficient signal for the activation of store-mediated Ca2+ entry in pleural mesothelial cells. In the absence of extracellular Ca2+, thapsigargin induced only a transient elevation of [Ca2+]i. Moreover, re-addition of Ca2+ increased the elevation of [Ca2+]i. Passive elevations in [Ca2+]i without thapsigargin, which is induced from Ca2+ containing solution switch to Ca2+ free solution and re-add Ca2+ containing solution, were not observed in pleural mesothelial cells. Thapsigargin-induced Ca2+ entry was still present after nifedipine (1 microM) treatment. However, SKF96365 (1 microM) blocked thapsigargin-induced Ca2+ entry. Mycalolide B (1 microM) completely disrupts actin cytoskeleton in pleural mesothelial cells, but thapsigargin-induced store-mediated Ca2+ entry was preserved. Jasplakinolide (3 microM) prevented thapsigargin-induced store-mediated Ca2+ entry. These results suggest that store-mediated Ca2+ entry in pleural mesothelial cells may be mediated by a recently proposed secretion-like coupling model for store-mediated Ca2+ entry. Topics: Actins; Animals; Calcium; Cell Shape; Cells, Cultured; Cytochalasin D; Cytoskeleton; Depsipeptides; Epithelial Cells; Imidazoles; Marine Toxins; Microscopy, Confocal; Nifedipine; Oxazoles; Pleura; Rats; Thapsigargin | 2006 |
Regulation of actin dynamics is critical for endothelial barrier functions.
We tested the hypothesis that the equilibrium between F- and G-actin in endothelial cells modulates the integrity of the actin cytoskeleton and is important for the maintenance of endothelial barrier functions in vivo and in vitro. We used the actin-depolymerizing agent cytochalasin D and jasplakinolide, an actin filament (F-actin) stabilizing and promoting substance, to modulate the actin cytoskeleton. Low doses of jasplakinolide (0.1 microM), which we have previously shown to reduce the permeability-increasing effect of cytochalasin D, had no influence on resting permeability of single-perfused mesenteric microvessels in vivo as well as on monolayer integrity. The F-actin content of cultured endothelial cells remained unchanged. In contrast, higher doses (10 microM) of jasplakinolide increased permeability (hydraulic conductivity) to the same extent as cytochalasin D and induced formation of intercellular gaps in cultured myocardial endothelial (MyEnd) cell monolayers. This was accompanied by a 34% increase of F-actin and pronounced disorganization of the actin cytoskeleton in MyEnd cells. Furthermore, we tested whether an increase of cAMP by forskolin and rolipram would prevent the cytochalasin D-induced barrier breakdown. Conditions that increase intracellular cAMP failed to block the cytochalasin D-induced permeability increase in vivo and the reduction of vascular endothelial cadherin-mediated adhesion in vitro. Taken together, these data support the hypothesis that the state of polymerization of the actin cytoskeleton is critical for maintenance of endothelial barrier functions and that both depolymerization by cytochalasin D and hyperpolymerization of actin by jasplakinolide resulted in an increase of microvessel permeability in vivo. However, cAMP, which is known to support endothelial barrier functions, seems to work by mechanisms other than stabilizing F-actin. Topics: Actin Cytoskeleton; Actins; Animals; Antigens, CD; Antineoplastic Agents; Cadherins; Cell Line, Transformed; Cyclic AMP; Cytochalasin D; Depsipeptides; Dose-Response Relationship, Drug; Endothelium, Vascular; Gap Junctions; Male; Mice; Microcirculation; Nucleic Acid Synthesis Inhibitors; Rats; Rats, Sprague-Dawley | 2005 |
Perturbation of actin dynamics induces NF-kappaB activation in myelomonocytic cells through an NADPH oxidase-dependent pathway.
Although several reports showed the effect of compounds disrupting microtubules on NF-kappaB (nuclear factor kappaB) activation, nothing is known about agents perturbing actin dynamics. In the present study, we have shown that actin cytoskeleton disruption induced by actin-depolymerizing agents such as cytochalasin D and latrunculin B and actin-polymerizing compounds such as jasplakinolide induced NF-kappaB activation in myelomonocytic cells. The transduction pathway involved the IkappaB (inhibitory kappaB) kinase complex and a degradation of IkappaBalpha. We have shown that NF-kappaB activation in response to the perturbation of actin dynamics required reactive oxygen species, as demonstrated by the effect of antioxidants. Actin cytoskeleton disruption by cytochalasin D induced O2- release from human monocytes, through the activation of the NADPH oxidase, as confirmed by the phosphorylation and by the membrane translocation of p47phox. NF-kappaB activation after actin cytoskeleton disruption could be physiologically relevant during monocyte activation and/or recruitment into injured tissues, where cellular attachment, migration and phagocytosis result in cyclic shifts in cytoskeletal organization and disorganization. Topics: Actins; Cell Line; Cytochalasin D; Cytoskeleton; Depsipeptides; Humans; I-kappa B Kinase; Monocytes; NADPH Oxidases; NF-kappa B; Oxygen; Protein Serine-Threonine Kinases; Signal Transduction | 2005 |
Disruption of the filamentous actin cytoskeleton is necessary for the activation of capacitative calcium entry in naive smooth muscle cells.
It has been proposed that cytoskeleton plays a key positive role in the activation of capacitative calcium entry (CCE), which supported the secretion-like hypothesis for the mechanisms underlying this process. However, its role on CCE in native smooth muscle is unknown. Here we demonstrate that CCE in isolated gallbladder myocytes was enhanced by cytochalasin D or latrunculin A treatments (agents that cause actin disassembly) whereas it was reduced by jasplakinolide treatment (which causes actin polymerization), suggesting that actin cytoskeleton acts as a barrier in CCE. In addition, we show for the first time that depletion of intracellular Ca2+ stores by thapsigargin and cholecystokinin in BAPTA-loaded cells induced a decrease in F-actin content that was consistent with a link between CCE and actin reorganization. In conclusion, these data suggest an active participation of actin reorganization in the implementation of CCE and support a conformational coupling model for this process in naive smooth muscle cells. Topics: Actin Cytoskeleton; Actins; Animals; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Cytochalasin D; Depsipeptides; Gallbladder; Guinea Pigs; Ion Transport; Male; Muscle, Smooth; Thiazoles; Thiazolidines | 2005 |
Involvement of actin in agonist-induced endocytosis of the G protein-coupled receptor for thromboxane A2: overcoming of actin disruption by arrestin-3 but not arrestin-2.
The role of actin in endocytosis of G protein-coupled receptors is poorly defined. In the present study, we demonstrate that agents that depolymerize (latrunculin B and cytochalasin D) or stabilize (jasplakinolide) the actin cytoskeleton blocked agonist-induced endocytosis of the beta isoform of the thromboxane A(2) receptor (TPbeta) in HEK293 cells. This suggests that endocytosis of TPbeta requires active remodeling of the actin cytoskeleton. On the other hand, disruption of microtubules with colchicine did not affect endocytosis of the receptor. Expression of wild-type and mutant forms of the small GTPases RhoA and Cdc42 potently inhibited endocytosis of TPbeta, further indicating a role for the dynamic regulation of the actin cytoskeleton in this pathway. Agonist treatment of TPbeta in HEK293 cells resulted in the formation of actin stress fibers through Galpha(q/11) signaling. Because we previously showed that endocytosis of TPbeta is dependent on arrestins, we decided to explore the relation between arrestin-2 and -3 and actin in endocytosis of this receptor. Interestingly, we show that the inhibition of TPbeta endocytosis by the actin toxins in HEK293 cells was overcome by the overexpression of arrestin-3, but not of arrestin-2. These results indicate that the actin cytoskeleton is not essential in arrestin-3-mediated endocytosis of TPbeta. However, arrestin-3 could not promote endocytosis of the TPbetaY339A and TPbetaI343A carboxyl-terminal mutants when the actin cytoskeleton was disrupted. Our data provide new evidence that the actin cytoskeleton plays an essential role in TPbeta endocytosis. Furthermore, our work suggests the existence of actin-dependent and -independent arrestin-mediated pathways of endocytosis. Topics: Actins; Antineoplastic Agents; Arrestins; Bridged Bicyclo Compounds, Heterocyclic; cdc42 GTP-Binding Protein; Cell Line; Clathrin; Cloning, Molecular; Colchicine; Cytochalasin D; Cytoskeleton; Depsipeptides; Endocytosis; Enzyme-Linked Immunosorbent Assay; GTP-Binding Protein alpha Subunits, Gq-G11; Humans; Marine Toxins; Microscopy, Fluorescence; Nucleic Acid Synthesis Inhibitors; Phosphoproteins; Plasmids; Protein Binding; Protein Structure, Tertiary; Receptors, G-Protein-Coupled; Receptors, Thromboxane A2, Prostaglandin H2; rhoA GTP-Binding Protein; Signal Transduction; Thiazoles; Thiazolidines; Time Factors; Transfection | 2005 |
Actin polymerization promotes the reversal of streaming in the apex of pollen tubes.
Actin polymerization is important in the control of pollen tube growth. Thus, treatment of pollen tubes with low concentrations of latrunculin B (Lat-B), which inhibits actin polymerization, permits streaming but reversibly blocks oscillatory growth. In the current study, we employ Jasplakinolide (Jas), a sponge cyclodepsipeptide that stabilizes actin microfilaments and promotes polymerization. Uniquely, Jas (2 microM) blocks streaming in the shank of the tube, but induces the formation of a toroidal-shaped domain in the swollen apex, of which longitudinal optical sections exhibit circles of motion. The polarity of this rotary motion is identical to that of reverse fountain motility in control pollen tubes, with the forward direction occurring at the edge of the cell and the rearward direction in the cell interior. Support for the idea that actin polymerization in the apical domain contributes to the formation of this rotary motility activity derives from the appearance therein of aggregates and flared cables of F-actin, using immunofluorescence, and by the reduction in G-actin as indicated with fluorescent DNAse. In addition, Jas reduces the tip-focused Ca2+ gradient. However, the alkaline band appears in the swollen apex and is spatially localized with the reverse fountain streaming activity. Taken together, our results support the idea that actin polymerization promotes reversal of streaming in the apex of the lily pollen tube. Topics: Actin Cytoskeleton; Actins; Antifungal Agents; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Calcium Signaling; Cell Movement; Cell Polarity; Cytochalasin D; Cytoplasm; Cytoplasmic Streaming; Deoxyribonucleases; Depsipeptides; Flowers; Germination; Lilium; Nucleic Acid Synthesis Inhibitors; Pollen; Polymers; Reproduction; Thiazoles; Thiazolidines | 2005 |
Neutrophil-bead collision assay: pharmacologically induced changes in membrane mechanics regulate the PSGL-1/P-selectin adhesion lifetime.
Visualization of flowing neutrophils colliding with adherent 1-mum-diameter beads presenting P-selectin allowed the simultaneous measurement of collision efficiency (epsilon), membrane tethering fraction (f), membrane tether growth dynamics, and PSGL-1/P-selectin binding lifetime. For 1391 collisions analyzed over venous wall shear rates from 25 to 200 s(-1), epsilon decreased from 0.17 to 0.004, whereas f increased from 0.15 to 0.70, and the average projected membrane tether length, L(tether)(m), increased from 0.35 mum to approximately 2.0 mum over this shear range. At all shear rates tested, adhesive collisions lacking membrane tethers had average bond lifetimes less than those observed for collisions with tethers. For adhesive collisions that failed to form membrane tethers, the regressed Bell parameters (consistent with single bond Monte Carlo simulation) were zero-stress off-rate, k(off)(0) = 0.56 s(-1) and reactive compliance, r = 0.10 nm, similar to published atomic force microscopy (AFM) measurements. For all adhesion events (+/- tethers), the bond lifetime distributions were more similar to those obtained by rolling assay and best simulated by Monte Carlo with the above Bell parameters and an average of 1.48 bonds (n = 1 bond (67%), n = 2 (22%), and n = 3-5 (11%)). For collisions at 100 s(-1), pretreatment of neutrophils with actin depolymerizing agents, latrunculin or cytochalasin D, had no effect on epsilon, but increased L(tether)(m) by 1.74- or 2.65-fold and prolonged the average tether lifetime by 1.41- or 1.65-fold, respectively. Jasplakinolide, an actin polymerizing agent known to cause blebbing, yielded results similar to the depolymerizing agents. Conversely, cholesterol-depletion with methyl-beta-cyclodextrin or formaldehyde fixation had no effect on epsilon, but reduced L(tether)(m) by 66% or 97% and reduced the average tether lifetime by 30% or 42%, respectively. The neutrophil-bead collision assay combines advantages of atomic force microscopy (small contact zone), aggregometry (discrete interactions), micropipette manipulation (tether visualization), and rolling assays (physiologic flow loading). Membrane tether growth can be enhanced or reduced pharmacologically with consequent effects on PSGL-1/P-selectin lifetimes. Topics: Actins; beta-Cyclodextrins; Biophysics; Bridged Bicyclo Compounds, Heterocyclic; Cell Adhesion; Cell Membrane; Cell Movement; Cholesterol; Cytochalasin D; Depsipeptides; Formaldehyde; Humans; Kinetics; Mechanics; Membrane Glycoproteins; Microscopy, Atomic Force; Microspheres; Models, Statistical; Monte Carlo Method; Neutrophils; P-Selectin; Protein Binding; Selectins; Stress, Mechanical; Thiazoles; Thiazolidines | 2005 |
Leptin-induced dynamic alterations in the actin cytoskeleton mediate the activation and synaptic clustering of BK channels.
Phosphoinositide 3-kinase (PI3-kinase) has been shown to link leptin receptor activation to stimulation of large conductance Ca2+-activated K+ (BK) channels and subsequent inhibition of hippocampal epileptiform-like activity. However, the downstream targets of PI3-kinase in this action of leptin are unknown. Here we show that BK channel activation by leptin is dependent on the actin cytoskeleton, as it is prevented by actin filament stabilization and mimicked by actin disruption. Fluorescent labeling of polymerized actin filaments revealed that leptin promotes the rapid rearrangement of actin filaments via activation of PI 3-kinase; an action paralleled by discrete increases in PtdIns(3,4,5)P3 immunoreactivity in close proximity to BK channels. After leptin exposure, there was also an actin-dependent increase in the association of BK channel immunoreactivity with synaptic markers. These data are consistent with the notion that leptin activates BK channels via PI 3-kinase-dependent reorganization of actin filaments and subsequent clustering of BK channels at synapses. Topics: Actins; Animals; Antineoplastic Agents; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Cytochalasin D; Cytoskeleton; Depsipeptides; Electrophysiology; Hippocampus; Image Processing, Computer-Assisted; Immunohistochemistry; Large-Conductance Calcium-Activated Potassium Channels; Leptin; Magnesium; Microscopy, Confocal; Microscopy, Fluorescence; Models, Biological; Neurons; Nocodazole; Nucleic Acid Synthesis Inhibitors; Phosphatidylinositol 3-Kinases; Porifera; Potassium Channels; Potassium Channels, Calcium-Activated; Rats; Synapses; Thiazoles; Thiazolidines; Time Factors; Tubulin Modulators | 2005 |
Deconstructing the cadherin-catenin-actin complex.
Spatial and functional organization of cells in tissues is determined by cell-cell adhesion, thought to be initiated through trans-interactions between extracellular domains of the cadherin family of adhesion proteins, and strengthened by linkage to the actin cytoskeleton. Prevailing dogma is that cadherins are linked to the actin cytoskeleton through beta-catenin and alpha-catenin, although the quaternary complex has never been demonstrated. We test this hypothesis and find that alpha-catenin does not interact with actin filaments and the E-cadherin-beta-catenin complex simultaneously, even in the presence of the actin binding proteins vinculin and alpha-actinin, either in solution or on isolated cadherin-containing membranes. Direct analysis in polarized cells shows that mobilities of E-cadherin, beta-catenin, and alpha-catenin are similar, regardless of the dynamic state of actin assembly, whereas actin and several actin binding proteins have higher mobilities. These results suggest that the linkage between the cadherin-catenin complex and actin filaments is more dynamic than previously appreciated. Topics: Actins; alpha Catenin; Animals; Antineoplastic Agents; beta Catenin; Cadherins; Cell Adhesion; Cell Line; Cell Membrane; Cytochalasin D; Cytoskeleton; Depsipeptides; Fluorescent Dyes; Mice; Microfilament Proteins; Multiprotein Complexes; Nucleic Acid Synthesis Inhibitors; Protein Binding; Protein Isoforms; Recombinant Fusion Proteins; Vinculin | 2005 |
Calcium transients regulate patterned actin assembly during myofibrillogenesis.
The highly ordered arrangement of sarcomeric myosin during striated muscle development requires spontaneous calcium (Ca(2+)) transients. Here, we show that blocking transients also compromises patterned assembly of actin thin filaments, titin, and capZ. Because a conserved temporal assembly pattern has been described for these proteins, selective inhibitors of either thick or thin filament formation were used to determine their relative temporal interdependencies. For example, inhibition of myosin light chain kinase (MLCK) by application of a specific inhibitory peptide or phorbol myistate acetate (PMA) disrupts myosin assembly without significantly affecting formation of actin bands. The MLCK inhibitor ML-7, however, disrupted actin as well as myosin. Surprisingly, agents that interfere with actin dynamics, such as cytochalasin D, produced only minor organizational disruptions in actin, capZ, and titin staining. However, cytochalasin D and other actin disrupting compounds significantly perturbed myosin organization. The results indicate that (1) Ca(2+) transients regulate one or more of the earliest steps in sarcomere formation, (2) mature actin filaments can assemble independently of myosin band formation, and (3) myosin thick filament assembly is extremely sensitive to disruption of either the actin or titin filament systems. Topics: Actin Cytoskeleton; Actins; Animals; Azepines; Bridged Bicyclo Compounds, Heterocyclic; Calcium Signaling; CapZ Actin Capping Protein; Connectin; Cytochalasin D; Depsipeptides; Microfilament Proteins; Muscle Development; Muscle Proteins; Myosin-Light-Chain Kinase; Myosins; Naphthalenes; Peptides, Cyclic; Protein Kinases; Sarcomeres; Tetradecanoylphorbol Acetate; Thiazoles; Thiazolidines; Xenopus | 2004 |
The role of the cytoskeleton in cellular adhesion molecule expression in tumor necrosis factor-stimulated endothelial cells.
Leukocyte infiltration is a hallmark of the atherosclerotic lesion. These cells are captured by cellular adhesion molecules (CAMs), including vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), platelet-endothelial cell adhesion molecule (PECAM), and E-selectin, on endothelial cells (EC). We examined the role of the actin cytoskeleton in tumor necrosis factor-alpha (TNF-alpha)-induced translocation of CAMs to the cell surface. Human aortic EC were grown on 96-well plates and an ELISA was used to assess surface expression of the CAMs. TNF-alpha increased VCAM-1, ICAM-1, and E-selectin by 4 h but had no affect on the expression of PECAM. A functioning actin cytoskeleton was important for VCAM-1 and ICAM-1 expression as both cytochalasin D, an actin filament disruptor, and jasplakinolide, an actin filament stabilizer, attenuated the expression of these CAMs. These compounds were ineffective in altering E-selectin surface expression. Myosin light chains are phosphorylated in response to TNF-alpha and this appears to be regulated by Rho kinase instead of myosin light chain kinase. However, the Rho kinase inhibitor, Y27632, had no affect on TNF-alpha-induced CAM expression. ML-7, a myosin light chain kinase inhibitor, had a modest inhibitory effect on the translocation of VCAM-1 but not on ICAM-1 or E-selectin. These data suggest that the surface expression of VCAM-1 and ICAM-1 is dependent on cycling of the actin cytoskeleton. Nevertheless, modulation of actin filaments via myosin light chain phosphorylation is not necessary. The regulation of E-selectin surface expression differs from that of the other CAMs. Topics: Actins; Amides; Azepines; Blotting, Western; Cell Adhesion Molecules; Cell Line; Cytochalasin D; Cytoskeleton; Depsipeptides; E-Selectin; Endothelial Cells; Enzyme-Linked Immunosorbent Assay; Humans; Immunohistochemistry; Intercellular Adhesion Molecule-1; Intracellular Signaling Peptides and Proteins; Microscopy, Fluorescence; Myosin Light Chains; Myosin-Light-Chain Kinase; Naphthalenes; Phosphorylation; Platelet Endothelial Cell Adhesion Molecule-1; Protein Serine-Threonine Kinases; Pyridines; rho-Associated Kinases; Tumor Necrosis Factor-alpha; Vascular Cell Adhesion Molecule-1 | 2004 |
Role of adhesion and contraction in Rac 1-regulated endothelial barrier function in vivo and in vitro.
We demonstrated previously that inhibition of the small GTPase Rac-1 by Clostridium sordellii lethal toxin (LT) increased the hydraulic conductivity (L(p)) of rat venular microvessels and induced gap formation in cultured myocardial endothelial cells (MyEnd). In MyEnd cells, we also demonstrated that both LT and cytochalasin D reduced cellular adhesion of vascular endothelial (VE)-cadherin-coated beads. Here we further evaluate the contribution of actin depolymerization, myosin-based contraction, and VE-cadherin linkage to the actin cytoskeleton to LT-induced permeability. The actin-depolymerizing agent cytochalasin D increased L(p) in single rat mesenteric microvessels to the same extent as LT over 80 min. However, whereas the actin-stabilizing agent jasplakinolide blunted the L(p) increase due to cytochalasin D by 78%, it had no effect on the LT response. This conforms to the hypothesis that the predominant mechanism whereby Rac-1 stabilizes the endothelial barrier in intact microvessels is separate from actin polymerization and likely at the level of the VE-cadherin linkage to the actin cytoskeleton. In intact vessels, neither inhibition of contraction (butanedione monoxime, an inhibitor of myosin ATPase) nor inhibition of Rho kinase (Y-27632) modified the response to LT, even though both inhibitors lowered resting L(p). In contrast butanedione monoxime and inhibition of myosin light chain kinase completely inhibited LT-induced intercellular gap formation and largely reduced the LT-induced permeability increase in MyEnd monolayers. These results support the hypothesis that the contractile mechanisms that contribute to the formation of large gaps between cultured endothelial cells exposed to inflammatory conditions do not significantly contribute to increased permeability in intact microvessels. Topics: Actins; Amides; Animals; Antigens, CD; Azepines; Bacterial Toxins; Cadherins; Capillary Permeability; Cell Adhesion; Cell Line, Transformed; Cytochalasin D; Cytoskeleton; Depsipeptides; Diacetyl; Endothelium, Vascular; Enzyme Inhibitors; Extracellular Space; Mice; Microcirculation; Myosins; Naphthalenes; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Pyridines; rac1 GTP-Binding Protein; Rats; Splanchnic Circulation; Vasoconstriction | 2004 |
Actin polymerization-driven molecular movement of mDia1 in living cells.
mDia1, a Rho effector, belongs to the Formin family of proteins, which shares the conserved tandem FH1-FH2 unit structure. Formins including mDia1 accelerate actin nucleation while interacting with actin filament fast-growing ends. Here our single-molecule imaging revealed fast directional movement of mDia1 FH1-FH2 for tens of microns in living cells. The movement of mDia1 FH1-FH2 was blocked by actin-perturbing drugs, and the speed of mDia1 FH1-FH2 movement appeared to correlate with actin elongation rates. In vitro, mDia1 FH1-FH2 associated persistently with the growing actin barbed end. mDia1 probably moves processively along the growing end of actin filaments in cells, and Formins may be a molecular motility machinery that is independent from motor proteins. Topics: Actin Cytoskeleton; Actins; Animals; Biopolymers; Bridged Bicyclo Compounds, Heterocyclic; Carrier Proteins; Cytochalasin D; Depsipeptides; Formins; Mice; Microtubules; Movement; Mutation; Myosins; Peptides, Cyclic; Recombinant Fusion Proteins; rhoA GTP-Binding Protein; Thiazoles; Thiazolidines; Xenopus | 2004 |
Two pathways for store-mediated calcium entry differentially dependent on the actin cytoskeleton in human platelets.
A major pathway for stimulated Ca(2+) entry in non-excitable cells is activated following depletion of intracellular Ca(2+) stores. Secretion-like coupling between elements in the plasma membrane (PM) and Ca(2+) stores has been proposed as the most likely mechanism to activate this store-mediated Ca(2+) entry (SMCE) in several cell types. Here we identify two mechanisms for SMCE in human platelets activated by depletion of two independent Ca(2+) pools, which are differentially modulated by the actin cytoskeleton. Ca(2+) entry induced by depletion of a 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ)-sensitive pool is increased by disassembly of the actin cytoskeleton and that induced by a TBHQ-insensitive pool is reduced. Stabilization of the actin cytoskeleton prevented Ca(2+) entry by both mechanisms. We propose that the membrane-associated actin network prevents constitutive Ca(2+) entry via both pathways. Reorganization of the actin cytoskeleton permits the activation of Ca(2+) entry via both mechanisms, but only SMCE activated by the TBHQ-insensitive pool requires new actin polymerization, which may support membrane trafficking toward the PM. Topics: Actins; Animals; Antineoplastic Agents; Biological Transport; Blood Platelets; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Calcium-Transporting ATPases; Cytochalasin D; Cytoskeleton; Depsipeptides; Enzyme Inhibitors; Humans; Hydroquinones; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; ras Proteins; Signal Transduction; Thiazoles; Thiazolidines | 2004 |
Two distinct actin networks drive the protrusion of migrating cells.
Cell migration initiates by extension of the actin cytoskeleton at the leading edge. Computational analysis of fluorescent speckle microscopy movies of migrating epithelial cells revealed this process is mediated by two spatially colocalized but kinematically, kinetically, molecularly, and functionally distinct actin networks. A lamellipodium network assembled at the leading edge but completely disassembled within 1 to 3 micrometers. It was weakly coupled to the rest of the cytoskeleton and promoted the random protrusion and retraction of the leading edge. Productive cell advance was a function of the second colocalized network, the lamella, where actomyosin contraction was integrated with substrate adhesion. Topics: Actin Cytoskeleton; Actins; Animals; Cell Line; Cell Movement; Cells, Cultured; Cytochalasin D; Depsipeptides; Epithelial Cells; Heterocyclic Compounds, 4 or More Rings; Kinetics; Macropodidae; Microscopy, Fluorescence; Motion Pictures; Peptides, Cyclic; Pseudopodia; Salamandridae | 2004 |
Presence of cytoskeleton proteins in parotid glands and their roles during secretion.
Amylase secretion is induced by the accumulation of cAMP in response to beta-adrenergic stimulation and by the augmentation of intracellular Ca2+ in response to muscarinic-cholinergic stimulation in rat parotid glands. The roles of cytoskeleton and motor proteins in the secretory process are not yet known. We examined the effects of cytoskeleton-modulating reagents on the amylase release induced by isoproterenol (IPR) and carbamylcholine (Cch) in rat parotid acinar cells. The amylase release induced by Cch was decreased by the microtubule-disrupting reagent colchicine (Colch) and the myosin ATPase inhibitor 2,3-butanediene monoxime (BDM), but the release induced by IPR was not. The actin filament-stabilizing reagent jasplakinolide (Jasp) and actin filament-disrupting reagent cytochalasin D (CytoD) decreased the amylase release induced by both the beta-adrenergic and the muscarinic-cholinergic stimulants. Pretreatment with CytoD affected the shape of the acinar cells, which showed an intermediate state between the fusion of the secretory granules with the apical membrane and the retrieval of the membranes only after stimulation with IPR. Myosin and Dynein/dynactin complex were detected in the secretory granule membrane fraction. We concluded from this study that the cytoskeleton played different roles in the beta-adrenergic and the muscarinic-cholinergic secretory processes. Topics: Actins; Adenosine Triphosphatases; Adrenergic beta-Agonists; Amylases; Animals; Carbachol; Cholinergic Agonists; Colchicine; Cytochalasin D; Cytoskeletal Proteins; Cytoskeleton; Depsipeptides; Diacetyl; Enzyme Inhibitors; Immunohistochemistry; Isoproterenol; Male; Parotid Gland; Rats; Rats, Wistar; Secretory Vesicles | 2004 |
Evidence for secretion-like coupling involving pp60src in the activation and maintenance of store-mediated Ca2+ entry in mouse pancreatic acinar cells.
Store-mediated Ca2+ entry (SMCE) is one of the main pathways for Ca2+ influx in non-excitable cells. Recent studies favour a secretion-like coupling mechanism to explain SMCE, where Ca2+ entry is mediated by an interaction of the endoplasmic reticulum (ER) with the plasma membrane (PM) and is modulated by the actin cytoskeleton. To explore this possibility further we have now investigated the role of the actin cytoskeleton in the activation and maintenance of SMCE in pancreatic acinar cells, a more specialized secretory cell type which might be an ideal cellular model to investigate further the properties of the secretion-like coupling model. In these cells, the cytoskeletal disrupters cytochalasin D and latrunculin A inhibited both the activation and maintenance of SMCE. In addition, stabilization of a cortical actin barrier by jasplakinolide prevented the activation, but not the maintenance, of SMCE, suggesting that, as for secretion, the actin cytoskeleton plays a double role in SMCE as a negative modulator of the interaction between the ER and PM, but is also required for this mechanism, since the cytoskeleton disrupters impaired Ca2+ entry. Finally, depletion of the intracellular Ca2+ stores induces cytoskeletal association and activation of pp60(src), which is independent on Ca2+ entry. pp60(src) activation requires the integrity of the actin cytoskeleton and participates in the initial phase of the activation of SMCE in pancreatic acinar cells. Topics: Actins; Animals; Biopolymers; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Cytochalasin D; Cytoskeleton; Depsipeptides; Ion Transport; Male; Mice; Pancreas; Peptides, Cyclic; Proto-Oncogene Proteins pp60(c-src); Spectrometry, Fluorescence; Thapsigargin; Thiazoles; Thiazolidines | 2003 |
Rho protein-mediated changes in the structure of the actin cytoskeleton regulate human inducible NO synthase gene expression.
Rho proteins (Rho, Rac, Cdc 42) are known to control the organization of the actin cytoskeleton as well as gene expression. Inhibition of Rho proteins by Clostridium difficile toxin B disrupted the F-actin cytoskeleton and enhanced cytokine-induced inducible nitric oxide synthase (iNOS) expression in human epithelial cells. Also specific inhibition by Y-27632 of p160ROCK, which mediates Rho effects on actin fibers, caused a disruption of the actin cytoskeleton and a superinduction of cytokine-induced iNOS expression. Accordingly, direct disruption of the actin cytoskeleton by cytochalasin D, latrunculin B, or jasplakinolide enhanced cytokine-induced iNOS expression. The transcription factor serum response factor (SRF) has been described as mediating actin cytoskeleton-dependent regulation of gene expression. Direct targets of SRF are activating protein 1 (AP1)-dependent genes. All compounds used inhibited SRF- and AP1-dependent reporter gene expression in DLD-1 cells. However, the enhancing effect of the actin cytoskeleton-disrupting compounds on human iNOS promoter activity was much less pronounced than the effect on iNOS mRNA expression. Therefore, besides transcriptional mechanisms, posttranscriptional effects seem to be involved in the regulation of iNOS expression by the above compounds. In conclusion, our data suggest that Rho protein-mediated changes of the actin cytoskeleton negatively modulate the expression of human iNOS. Topics: Actin Cytoskeleton; Bacterial Proteins; Bacterial Toxins; Bridged Bicyclo Compounds, Heterocyclic; Cytochalasin D; Cytokines; Cytoskeleton; Depsipeptides; Enzyme Inhibitors; Eukaryotic Cells; Gene Expression Regulation, Enzymologic; Humans; Intracellular Signaling Peptides and Proteins; Nitric Oxide; Nitric Oxide Synthase; Peptides, Cyclic; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; rho GTP-Binding Proteins; rho-Associated Kinases; RNA, Messenger; Serum Response Factor; Thiazoles; Thiazolidines; Transcription Factor AP-1; Transcription, Genetic; Tumor Cells, Cultured | 2003 |
Cell-specific behavior of P2X7 receptors in mouse parotid acinar and duct cells.
P2X7 receptors (P2X7Rs) affect many epithelial cell functions including transcellular ion transport, secretion, and cell death. Here we used parotid acinar and duct cells to reveal the unique cell-specific assembly and gating of the P2X7R channels. Immunolocalization indicated expression of P2X7Rs in the luminal membrane of both cell types. Stimulation with 5 mm ATP raised [Ca2+]i levels in a cell-specific manner and activated multiple currents. The current mediated by P2X7R was isolated by infusing the cells with high [EGTA]. The initial activation of acinar cell P2X7Rs by ATP was slow requiring approximately 2.5 min. Subsequent removal and addition of ATP, however, resulted in rapid inhibition and activation (gating) of the P2X7Rs. By contrast, P2X7Rs in duct cells displayed only rapid gating by ATP. Activation of P2X7Rs in both cell types was verified by (a) low Km for ATP, (b) sensitivity to external divalent ions, (c) lack of desensitization/inactivation, (d) permeability to Na+, and (e) inhibition by Brilliant Blue G, Cu2+, and pyridoxal phosphate-6-azophenyl-2',4'-disulfonic acid tetrasodium. The slow P2X7R activation in acinar cells was not affected by manipulation of exo-/endocytosis. Rather, disassembly or solidification of the actin cytoskeleton prior to incubation with ATP prevented channel assembly. Remarkably, after completion of the slow activation, manipulation of the actin cytoskeleton no longer affected gating by ATP. Accordingly, manipulation of the actin cytoskeleton had no effect on P2X7R gating by ATP in duct cells. We concluded that P2X7Rs are not active in resting acinar cells. On exposure to ATP, P2X7Rs are assembled into functional channels with the aid of the actin cytoskeleton. Once assembled, P2X7Rs are subject to rapid gating by ATP. Duct cell P2X7Rs are preassembled and therefore continually subject to rapid gating by ATP. This cell-specific behavior may reflect the specific function of P2X7Rs in the two cell types. Topics: Actins; Adenosine Triphosphate; Animals; Antineoplastic Agents; Benzenesulfonates; Calcium; Cell Death; Cell Membrane; Coloring Agents; Copper; Cytochalasin D; Cytoskeleton; Depsipeptides; Egtazic Acid; Electrophysiology; Guanosine 5'-O-(3-Thiotriphosphate); Guanosine Diphosphate; Immunohistochemistry; Indicators and Reagents; Ions; Kinetics; Mice; Nucleic Acid Synthesis Inhibitors; Parotid Gland; Peptides, Cyclic; Pyridoxal Phosphate; Receptors, Purinergic P2; Receptors, Purinergic P2X7; Reverse Transcriptase Polymerase Chain Reaction; RNA; Sodium; Thionucleotides; Time Factors | 2003 |
FcepsilonRI cross-linking-induced actin assembly mediates calcium signalling in RBL-2H3 mast cells.
1. To determine the role of actin assembly in the Ca(2+) signalling of mast cells activated by cross-linking of FcepsilonRI, we examined the effects of cytochalasin D, an inhibitor of actin polymerization. 2. In the RBL-2H3 cells, F-actin content was increased by sensitization with anti-dinitrophenol (DNP) IgE. In these cells, cytochalasin D induced oscillatory increases in cytosolic Ca(2+) ([Ca(2+)](i)); these increase were inhibited by jasplakinolide, a stabilizer of actin filaments. 3. In the IgE-sensitized RBL-2H3 cells, DNP-human serum albumin (DNP-HSA) augmented actin assembly. DNP-HSA also increased the production of IP(3), [Ca(2+)](i) and degranulation. Cytochalasin D enhanced all of these DNP-HSA-induced effects. 4. In a Ca(2+)-free solution, DNP-HSA induced a transient increase in [Ca(2+)](i), and this increase was accelerated by cytochalasin D. After cessation of the DNP-HSA-induced Ca(2+) release, the re-addition of Ca(2+) induced a sustained increase in [Ca(2+)](i) through capacitative Ca(2+) entry (CCE), and this increase was enhanced by cytochalasin D. 5 The effect of cytochalasin D in enhancing the CCE activity was prevented by xestospongin C. 6. In contrast, neither the Ca(2+) release nor the CCE activation that was induced by thapsigargin was affected by cytochalasin D. 7. These results suggest that actin de-polymerization stimulates the FcepsilonRI-mediated signalling to augment the release of Ca(2+) from the endoplasmic reticulum in RBL-2H3 cells. Topics: Actins; Analysis of Variance; Animals; Calcium Signaling; Cations, Divalent; Cell Degranulation; Cross-Linking Reagents; Cytochalasin D; Depsipeptides; Dinitrophenols; Humans; Immunoglobulin E; Inositol 1,4,5-Trisphosphate; Macrocyclic Compounds; Mast Cells; Oxazoles; Peptides, Cyclic; Rats; Receptors, IgE; Serum Albumin; Thapsigargin; Tumor Cells, Cultured | 2002 |
Participation of host cell actin filaments during interaction of trypomastigote forms of Trypanosoma cruzi with host cells.
The involvement of actin filaments from the host cell on the process of invasion of trypomastigote forms of Trypanosma cruzi was analyzed in seven different cell lines. Prior incubation of all cell lines with cytochalasin D, under conditions which interfere with actin filaments, markedly inhibited parasite internalization and increased parasite attachment. Attached parasites were readily ingested following washing of the drug-treated cells. Cytochalasin treatment interfered with the distribution of actin filaments of the host cell as evaluated by visualization of the filaments using confocal laser scanning microscopy of cells incubated in the presence of FITC-phalloidin. Concentration of actin filaments could be observed in most, but not all, parasites in the process of internalization. We also treated LLCMK 2 and macrophage cells with Jasplakinolide, a drug that stabilizes actin filaments, before interaction with the trypomastigote forms. This drug partially inhibits parasite invasion into the cells. Prior incubation of the host cells in the presence of colchicine, which interfere with microtubules, also inhibited parasite internalization into the cells. Topics: Actin Cytoskeleton; Animals; Antineoplastic Agents; Chagas Disease; Chlorocebus aethiops; Colchicine; Cytochalasin D; Depsipeptides; Fluorescent Antibody Technique; Host-Parasite Interactions; Macrophages; Microscopy, Electron, Scanning; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Pseudopodia; Trypanosoma cruzi; Vero Cells | 2002 |
CD2-associated protein directly interacts with the actin cytoskeleton.
CD2-associated protein (CD2AP) is an adapter protein associating with several membrane proteins, including nephrin, mutated in congenital nephrotic syndrome of the Finnish type, and polycystin-2, mutated in type 2 autosomal dominant polycystic kidney disease. Both proteins have critical roles in the maintenance of the integrity of the nephrons. Previous studies have suggested a role for CD2AP in the regulation of the organization of the actin cytoskeleton, but it has not been known whether the postulated association between CD2AP and actin is direct or mediated by other proteins. In this study, we address this question by using various cellular and biochemical approaches. We show that CD2AP and F-actin partially colocalize in cultured cells and that disruption of the actin cytoskeleton results in disorganization of endogenous CD2AP. Using cytoskeletal fractionation by differential centrifugation, we demonstrate that a proportion of CD2AP associates with the actin cytoskeleton. Furthermore, using pure actin and purified CD2AP fusion proteins in an F-actin coprecipitation assay, we show that CD2AP directly associates with filamentous actin and that this interaction is mediated by means of the COOH terminus of CD2AP. The present results suggest that CD2AP is involved in the regulation of the actin cytoskeleton and indicate that CD2AP may act as a direct adapter between the actin cytoskeleton and cell membrane proteins, such as nephrin and polycystin-2. Alterations in these interactions could explain some of the pathophysiological changes in congenital nephrotic syndrome and polycystic kidney disease. Topics: Actins; Adaptor Proteins, Signal Transducing; Animals; Cell Membrane; Cytochalasin D; Cytoskeletal Proteins; Cytoskeleton; Depsipeptides; Epithelial Cells; Immunohistochemistry; Kidney Tubules, Collecting; Membrane Proteins; Mice; Microscopy, Immunoelectron; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Proteins; TRPP Cation Channels | 2002 |
Involvement of calcium signaling and the actin cytoskeleton in the membrane block to polyspermy in mouse eggs.
This study examines the effects of actin microfilament-disrupting drugs on events of fertilization, with emphasis on gamete membrane interactions. Mouse eggs, freed of their zonae pellucidae, were treated with drugs that perturb the actin cytoskeleton by different mechanisms (cytochalasin B, cytochalasin D, jasplakinolide, latrunculin B) and then inseminated. Cytochalasin B, jasplakinolide, and latrunculin B treatments resulted in a decrease in the percentage of eggs fertilized and the average number of sperm fused per egg. However, cytochalasin D treatment resulted in an increase in the average number of sperm fused per egg and the percentage of polyspermic eggs. This increase in polyspermy occurred despite the observation that cytochalasin D treatment caused a decrease in sperm-egg binding and did not affect spontaneous acrosome reactions or sperm motility. This suggested that cytochalasin D-treated eggs had an impaired ability to establish a block to polyspermy at the level of the plasma membrane. The effect of cytochalasin D on the block to polyspermy was not due to a general disruption of egg activation because sperm-induced calcium oscillations and cortical granule exocytosis were similar in cytochalasin D-treated and control eggs. However, buffering of intracellular calcium levels with the calcium chelator BAPTA-AM resulted in an increase in polyspermy. Together, these data suggest that a postfertilization decrease in egg membrane receptivity to sperm requires functions of the egg actin cytoskeleton that are disrupted by cytochalasin D. Furthermore, egg activation-associated increased intracellular calcium levels are necessary but not sufficient to affect postfertilization membrane dynamics that contribute to a membrane block to polyspermy. Topics: Acrosome; Acrosome Reaction; Actin Cytoskeleton; Animals; Bridged Bicyclo Compounds, Heterocyclic; Calcimycin; Calcium; Cytochalasin B; Cytochalasin D; Cytoskeleton; Depsipeptides; Exocytosis; Female; Fertilization in Vitro; Male; Mice; Oocytes; Peptides, Cyclic; Signal Transduction; Sperm Motility; Spermatozoa; Thiazoles; Thiazolidines | 2002 |
Endoplasmic reticulum calcium release is modulated by actin polymerization.
Intracellular calcium ions regulate the structure and functions of cytoskeletal proteins. On the other hand, recent studies have shown that the cytoskeleton, and actin filaments in particular, can modulate calcium influx through plasma membrane ligand- and voltage-gated channels. We now report that calcium release from inositol trisphosphate (IP3) and ryanodine-sensitive endoplasmic reticulum (ER) stores is modulated by polymerization and depolymerization of actin filaments in cultured hippocampal neurons. Depolymerization of actin filaments with cytochalasin D attenuates calcium release induced by carbamylcholine (CCh; a muscarinic agonist for IP3 pathway), caffeine (a ryanodine receptor agonist) and thapsigargin (an inhibitor of the ER calcium- ATPase) in both the presence and absence of extracellular calcium. Conversely, the actin polymerizing agent jasplakinolide potentiates calcium release induced by CCh, caffeine and thapsigargin. Cytochalasin D attenuated, while jasplakinolide augmented, thapsigargin-induced JNK activation and neuronal cell death. Our data show that the actin cytoskeleton regulates ER calcium release, suggesting roles for actin in the various physiological and pathological processes that involve calcium release. Topics: Actin Cytoskeleton; Actins; Animals; Biopolymers; Caffeine; Calcium; Calcium-Transporting ATPases; Carbachol; Cell Death; Cells, Cultured; Cytochalasin D; Depsipeptides; Endoplasmic Reticulum; Enzyme Inhibitors; Hippocampus; Inositol Phosphates; Muscarinic Agonists; Neurons; Peptides, Cyclic; Protein Binding; Rats; Ryanodine; Ryanodine Receptor Calcium Release Channel | 2002 |
A functional link between the actin cytoskeleton and lipid rafts during budding of filamentous influenza virions.
Morphogenesis of influenza virus is a poorly understood process that produces two types of enveloped virion: approximately 100-nm spheres and similar diameter filaments that reach 20 microm in length. Spherical particles assemble at plasma membrane lipid rafts in a process independent of microfilaments. The budding site of filamentous virions is hitherto uncharacterised but their formation involves the actin cytoskeleton. We confirm microfilament involvement in filamentous budding and show that after disruption of cortical actin by jasplakinolide, HA, NP, and M1 redistributed around beta-actin clusters to form novel annular membrane structures. HA in filamentous virions and jasplakinolide-induced annuli was detergent insoluble at 4 degrees C. Furthermore, in both cases HA partitioned into low buoyant density detergent-insoluble glycolipid domains, indicating that filamentous virions and annuli contain reorganised lipid rafts. We propose that the actin cytoskeleton is required to maintain the correct organisation of lipid rafts for incorporation into budding viral filaments. Topics: Actins; Animals; Cell Line; Cytochalasin D; Cytoskeleton; Depsipeptides; Dogs; Hemagglutinin Glycoproteins, Influenza Virus; Humans; Influenza A virus; Intracellular Fluid; Membrane Microdomains; Nucleocapsid Proteins; Nucleoproteins; Peptides, Cyclic; RNA-Binding Proteins; Viral Core Proteins; Viral Matrix Proteins; Virion; Virus Assembly | 2002 |
Rapid turnover of actin in dendritic spines and its regulation by activity.
Dendritic spines are motile structures that contain high concentrations of filamentous actin. Using hippocampal neurons expressing fluorescent actin and the method of fluorescence recovery after photobleaching, we found that 85 +/- 2% of actin in the spine was dynamic, with a turnover time of 44.2 +/- 4.0 s. The rapid turnover is not compatible with current models invoking a large population of stable filaments and static coupling of filaments to postsynaptic components. Low-frequency stimulation known to induce long-term depression in these neurons stabilized nearly half the dynamic actin in the spine. This effect depended on the activation of N-methyl-D-aspartate (NMDA) receptors and the influx of calcium. In neurons from mice lacking gelsolin, a calcium-dependent actin-binding protein, activity-dependent stabilization of actin was impaired. Our studies provide new information on the kinetics of actin turnover in spines, its regulation by neural activity and the mechanisms involved in this regulation. Topics: Actins; Animals; Animals, Newborn; Calcium; Cells, Cultured; Cytochalasin D; Dendrites; Depsipeptides; Gelsolin; Green Fluorescent Proteins; Hippocampus; Indicators and Reagents; Luminescent Proteins; Mice; Mice, Inbred BALB C; Mice, Transgenic; Microscopy, Fluorescence; Neurons; Patch-Clamp Techniques; Peptides, Cyclic; Rats; Receptors, N-Methyl-D-Aspartate; Recombinant Fusion Proteins; Synapses | 2002 |
Actin filament turnover removes bundles from Drosophila bristle cells.
Drosophila bristle cells form enormous extensions that are supported by equally impressive scaffolds of modular, polarized and crosslinked actin filament bundles. As the cell matures and support is taken over by the secreted cuticle, the actin scaffold is completely removed. This removal begins during cell elongation and proceeds via an orderly series of steps that operate on each module. Using confocal and electron microscopy, we found that the approximately 500-filament modules are fractured longitudinally into 25-50-filament subbundles, indicating that module breakdown is the reverse of assembly. Time-lapse confocal analysis of GFP-decorated bundles in live cells showed that modules were shortened by subunit removal from filament barbed ends, again indicating that module breakdown is the reverse of assembly. Module shortening takes place at a fairly slow rate of approximately 1microm/hour, implying that maximally crosslinked modules are not rapidly depolymerized. Barbed-end depolymerization was prevented with jasplakinolide and accelerated with cycloheximide, indicating that barbed-end maintenance requires continuous protein synthesis. Subbundle adhesion was lost in the presence of cytochalasin, indicating that continuous actin polymerization is required. Thus, these polarized actin filament bundles are dynamic structures that require continuous maintenance owing to protein and actin filament turnover. We propose that after cell elongation, maintenance falls behind turnover, resulting in the removal of this modular cytoskeleton. Topics: Actin Cytoskeleton; Actins; Animals; Cell Surface Extensions; Cycloheximide; Cytochalasin D; Depsipeptides; Drosophila melanogaster; Insecticides; Microscopy, Confocal; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Protein Synthesis Inhibitors; Pupa | 2002 |
Cholangiocytes exhibit dynamic, actin-dependent apical membrane turnover.
The present studies of cholangiocytes used complementary histological, biochemical, and electrophysiological methods to identify a dense population of subapical vesicles, quantify the rates of vesicular trafficking, and assess the contribution of the actin cytoskeleton to membrane trafficking. FM 1-43 fluorescence measured significant basal rates of total exocytosis (1.33 +/- 0.16% plasma membrane/min) in isolated cholangiocytes and apical exocytosis in cholangiocyte monolayers. Cell surface area remained unchanged, indicating that there was a concurrent, equivalent rate of endocytosis. FM 1-43 washout studies showed that 36% of the endocytosed membrane was recycled to the plasma membrane. 8-(4-Chlorophenylthio)adenosine 3',5'-cyclic monophosphate (CPT-cAMP; cAMP analog) increased exocytosis by 71 +/- 31%, whereas the Rp diastereomer of adenosine 3',5'-cyclic monophosphothioate (Rp-cAMPS; protein kinase A inhibitor) diminished basal exocytosis by 53 +/- 11%. A dense population of 140-nm subapical vesicles arose, in part, from apical membrane endocytosis. Phalloidin staining showed that a subpopulation of the endocytosed vesicles was encapsulated by F-actin. Furthermore, membrane trafficking was inhibited by disrupting the actin cytoskeleton with cytochalasin D (51 +/- 13% of control) or jasplakinolide (58 +/- 9% of control). These studies indicate that there is a high rate of vesicular trafficking at the apical membrane of cholangiocytes and suggest that both cAMP and the actin cytoskeleton contribute importantly to these events. Topics: Actins; Animals; Antineoplastic Agents; Bile Ducts; Biological Transport; Cell Line; Cell Membrane; Cell Polarity; Cyclic AMP; Cytochalasin D; Cytoskeleton; Depsipeptides; Endocytosis; Exocytosis; Fluorescent Dyes; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Rats; Transport Vesicles | 2002 |
Smooth muscle differentiation marker gene expression is regulated by RhoA-mediated actin polymerization.
Smooth muscle cell (SMC) differentiation is regulated by a complex array of local environmental cues, but the intracellular signaling pathways and the transcription mechanisms that regulate this process are largely unknown. We and others have shown that serum response factor (SRF) contributes to SMC-specific gene transcription, and because the small GTPase RhoA has been shown to regulate SRF, the goal of the present study was to test the hypothesis that RhoA signaling is a critical mechanism for regulating SMC differentiation. Coexpression of constitutively active RhoA in rat aortic SMC cultures significantly increased the activity of the SMC-specific promoters, SM22 and SM alpha-actin, whereas coexpression of C3 transferase abolished the activity of these promoters. Inhibition of either stress fiber formation with the Rho kinase inhibitor Y-27632 (10 microm) or actin polymerization with latrunculin B (0.5 microm) significantly decreased the activity of SM22 and SM alpha-actin promoters. In contrast, increasing actin polymerization with jasplakinolide (0.5 microm) increased SM22 and SM alpha-actin promoter activity by 22-fold and 13-fold, respectively. The above interventions had little or no effect on the transcription of an SRF-dependent c-fos promoter or on a minimal thymidine kinase promoter that is not SRF-dependent. Taken together, the results of these studies indicate that in SMC, RhoA-dependent regulation of the actin cytoskeleton selectively regulates SMC differentiation marker gene expression by modulating SRF-dependent transcription. The results also suggest that RhoA signaling may serve as a convergence point for the multiple signaling pathways that regulate SMC differentiation. Topics: Actins; Amides; Animals; Aorta; Biomarkers; Biopolymers; Bridged Bicyclo Compounds, Heterocyclic; Cell Differentiation; Cells, Cultured; Cytochalasin D; Depsipeptides; DNA-Binding Proteins; Fluorescent Antibody Technique; Gene Expression Regulation; Genes, Reporter; Muscle, Smooth, Vascular; Nuclear Proteins; Peptides, Cyclic; Promoter Regions, Genetic; Pyridines; Rats; rhoA GTP-Binding Protein; Serum Response Factor; Signal Transduction; Stress Fibers; Thiazoles; Thiazolidines; Transcription, Genetic; Transfection | 2001 |
Diverse regulation of microfilament assembly, production of TNF-alpha, and reactive oxygen intermediates by actin modulating substances and inhibitors of ADP-ribosylation in human monocytes stimulated with LPS.
Lipopolysaccharide (LPS), a potent activator of human monocytes, induced F-actin polymerization in a concentration- and time-dependent manner. To test whether cytoskeletal events participate in the control of the LPS-induced ROI and TNF-alpha production, three natural occurring actin-modulating substances, cytochalasin D (Cyt D), latrunculin B (Lat B), and jasplakinolide (JK), were used. Here we show that treatment of monocytes with Cyt D, Lat B, or JK led to a rearrangement of the actin cytoskeleton, which upon addition of LPS was further modified. Cyt D and Lat B induced generation of ROI in the absence of LPS and enhanced the LPS-triggered respiratory burst. JK also proved to be a potent activator of ROI-production but only in the presence of LPS. TNF-alpha production was hardly affected by the three substances. There was no correlation between a specific state of Cyt D-, Lat B-, or JK-modified actin polymerization and ROI-production. Inhibitors of ADP-ribosylation proved to be activators of F-actin polymerization. They were shown to prevent ROI- and TNF-alpha production and to reduce the capability of LPS to mediate maximal F-actin assembly. At concentrations at which inhibition was greatest, maximal blockage of ROI and TNF-alpha production was observed. These findings may argue for a role of ADP-ribosylation in the transduction pathways mediating the biological responses, with involvement in the assembly of actin-containing cytoskeletal microfilaments. Topics: Actin Cytoskeleton; Actins; Adenosine Diphosphate Ribose; Antineoplastic Agents; Bridged Bicyclo Compounds, Heterocyclic; Cytochalasin D; Depsipeptides; Dose-Response Relationship, Drug; Humans; In Vitro Techniques; Lipopolysaccharides; Monocytes; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Phosphorylation; Polymers; Reactive Oxygen Species; Thiazoles; Thiazolidines; Tumor Necrosis Factor-alpha | 2001 |
Role of LPS-induced microfilament depolymerization in MIP-2 production from rat pneumocytes.
We have previously demonstrated that lipopolysaccharide (LPS) induces production of macrophage inflammatory protein-2 (MIP-2), a C-X-C chemokine for neutrophil recruitment and activation, in primary cultured rat lung alveolar epithelial cells. We have also demonstrated that LPS depolymerizes microfilaments in rat alveolar epithelial cells. To determine whether the polymerization status of microfilaments affects LPS-induced MIP-2 production, we treated rat alveolar epithelial cells with cytochalasin D (CytoD), a microfilament-disrupting agent, before and during LPS stimulation. A lower concentration (0.1 microM) of CytoD inhibited LPS-induced MIP-2 production without affecting microfilament polymerization. In contrast, LPS-induced MIP-2 production was enhanced by a higher concentration (10 microM) of CytoD, which disrupted the filamentous structure of actin. Jasplakinolide (1 nM to 1 microM), a polymerizing agent for microfilaments, decreased LPS-induced MIP-2 secretion. Jasplakinolide (1 microM) also blocked LPS-induced depolymerization of microfilaments. These results suggest that, in alveolar epithelial cells, LPS-induced MIP-2 production is at least partially regulated by microfilament depolymerization. Topics: Actin Cytoskeleton; Actins; Animals; Chemokine CXCL2; Chemokines; Cytochalasin D; Depsipeptides; Dose-Response Relationship, Drug; Gene Expression; Lipopolysaccharides; Male; Peptides, Cyclic; Polymers; Pulmonary Alveoli; Rats; Rats, Sprague-Dawley | 2001 |
Initiation of apoptosis by actin cytoskeletal derangement in human airway epithelial cells.
Changes in epithelial cell shape can lead to cell death and detachment. Actin filaments are cleaved during apoptosis, but whether disruption in the actin cytoskeletal network, as one manifestation of cell shape change, can itself induce apoptosis is not known. We tested this hypothesis in the airway epithelial cell line 1HAEo(-) and in primary airway epithelial cells by preventing actin filament elongation with cytochalasin D or by aggregating actin filaments with jasplakinolide. Disruption of actin filament integrity promptly induced apoptosis in adherent epithelial cells within 5 h. Jasplakinolide-induced apoptosis did not disrupt focal adhesions, whereas cytochalasin D-induced apoptosis decreased focal adhesion protein expression and occurred despite ligation of the fibronectin receptor. Death induction was abrogated by the caspase inhibitors z-VAD-fmk and Ac-DEVD-cho but not by blocking the Fas (CD95) receptor. Whereas cytochalasin D--induced apoptosis was associated with cleavage of pro-caspase-8, jasplakinolide-induced apoptosis was not. Both agents induced formation of a death-inducing signaling complex. These data demonstrate that disruption of actin filament integrity with either cytochalasin D or jasplakinolide induces apoptosis in airway epithelial cells but by different mechanisms, and suggest that actin may be an early modulator of apoptotic commitment. Topics: Actins; Animals; Antineoplastic Agents; Apoptosis; Caspase 8; Caspase 9; Caspases; Cell Adhesion; Cell Division; Cell Line; Cytochalasin D; Cytoskeletal Proteins; Cytoskeleton; Depsipeptides; Dogs; Focal Adhesions; Humans; Kidney; Kinetics; Paxillin; Peptides, Cyclic; Phosphoproteins; Receptors, Fibronectin; Respiratory Mucosa; Vinculin | 2001 |
Effects of the actin-stabilizing drug, jasplakinolide, on pigment granule motility in isolated retinal pigment epithelial (RPE) cells of green sunfish, Lepomis cyanellus.
The retinal pigment epithelium (RPE) of teleosts contains pigment granules that migrate in response to changes in light condition. Dissociated, cultured RPE cells in vitro can be triggered to aggregate or disperse pigment granules by the application of cAMP or dopamine, respectively. Previous research using the actin-disrupting drug, cytochalasin D, suggested that pigment granule motility is actin dependent. To further examine the role of actin in pigment granule motility, we tested the effects of the actin-stabilizing drug, jasplakinolide, on pigment granule motility. Pigment granules in previously dispersed RPE cells remained dispersed after jasplakinolide exposure (0.1-1 microM), but the drug halted movement of most pigment granules and stimulated rapid bi-directional movements in a small subset of granules. Jasplakinolide also blocked net pigment granule aggregation and interfered with the maintenance of full aggregation. Although jasplakinolide did not block pigment granule dispersion, it did alter the motility of dispersing granules compared to control cells; rather than the normal saltatory, primarily centrifugal movements, granules of jasplakinolide-treated cells demonstrated slow, creeping centrifugal movements and more rapid bi-directional movements. Jasplakinolide also altered cell morphology; the length and thickness of apical projections increased, and enlarged, paddle-like structures, which contained F-actin appeared at the tips of projections. Actin antibody labeling of jasplakinolide-treated cells revealed a more reticulated network of actin compared to antibody-labeled control cells. These results indicate that jasplakinolide-induced disruption of the actin network compromises normal pigment granule dispersion and aggregation in isolated RPE cells, thus providing further evidence that these movements are actin dependent. Topics: Actins; Animals; Antineoplastic Agents; Cardiotonic Agents; Cell Movement; Cells, Cultured; Cyclic AMP; Cytochalasin D; Depsipeptides; Dopamine; Dose-Response Relationship, Drug; Fluorescent Dyes; Microscopy, Video; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Perciformes; Phalloidine; Pigment Epithelium of Eye; Rhodamines; Time Factors | 2001 |
L-plastin peptide activation of alpha(v)beta(3)-mediated adhesion requires integrin conformational change and actin filament disassembly.
L-plastin (LPL) is a leukocyte actin binding protein previously implicated in the activation of the integrin alpha(M)beta(2) on polymorphonuclear neutrophils. To determine the role for LPL in integrin activation, K562 cell adhesion to vitronectin via alpha(v)beta(3), a well-studied model for activable integrins, was examined. Cell permeant versions of peptides based on the N-terminal sequence of LPL and the LPL headpiece domain both activated alpha(v)beta(3)-mediated adhesion. In contrast to adhesion induced by treatment with phorbol 12-myristate 13-acetate (PMA), LPL peptide-activated adhesion was independent of integrin beta(3) cytoplasmic domain tyrosines and was not inhibited by cytochalasin D. Also in contrast to PMA, LPL peptides synergized with RGD ligand or Mn(2+) for generation of a conformational change in alpha(v)beta(3) associated with the high affinity state of the integrin, as determined by binding of a ligand-induced binding site antibody. Although LPL and ligand showed synergy for ligand-induced binding site expression when actin depolymerization was inhibited by jasplakinolide, LPL peptide-induced adhesion was inhibited. Thus, both actin depolymerization and ligand-induced integrin conformational change are required for LPL peptide-induced adhesion. We hypothesize that the critical steps of increased integrin diffusion and affinity enhancement may be linked via modulation of the function of the actin binding protein L-plastin. Topics: Actin Cytoskeleton; Antineoplastic Agents; Binding Sites; Cell Adhesion; Cell Line; Cell Separation; Cytochalasin D; Cytoplasm; Cytoskeleton; Depsipeptides; DNA, Complementary; Dose-Response Relationship, Drug; Flow Cytometry; Humans; Integrins; Ligands; Membrane Glycoproteins; Microfilament Proteins; Models, Biological; Nucleic Acid Synthesis Inhibitors; Oligopeptides; Peptides; Peptides, Cyclic; Phosphoproteins; Protein Binding; Protein Conformation; Protein Structure, Tertiary; ras Proteins; Receptors, Vitronectin; Recombinant Fusion Proteins; Tetradecanoylphorbol Acetate; Transfection | 2001 |
An examination of the secretion-like coupling model for the activation of the Ca2+ release-activated Ca2+ current I(CRAC) in RBL-1 cells.
One popular model for the activation of store-operated Ca2+ influx is the secretion-like coupling mechanism, in which peripheral endoplasmic reticulum moves to the plasma membrane upon store depletion thereby enabling inositol 1,4,5-trisphosphate (InsP3) receptors on the stores to bind to, and thus activate, store-operated Ca2+ channels. This movement is regulated by the underlying cytoskeleton. We have examined the validity of this mechanism for the activation of I(CRAC), the most widely distributed and best characterised store-operated Ca2+ current, in a model system, the RBL-1 rat basophilic cell line. Stabilisation of the peripheral cytoskeleton, disassembly of actin microfilaments and disaggregation of microtubules all consistently failed to alter the rate or extent of activation of I(CRAC). Rhodamine-phalloidin labelling was used wherever possible, and revealed that the cytoskeleton had been significantly modified by drug treatment. Interference with the cytoskeleton also failed to affect the intracellular calcium signal that occurred when external calcium was re-admitted to cells in which the calcium stores had been previously depleted by exposure to thapsigargin/ionomycin in calcium-free external solution. Application of positive pressure through the patch pipette separated the plasma membrane from underlying structures (cell ballooning). However, I(CRAC) was unaffected irrespective of whether cell ballooning occurred before or after depletion of stores. Pre-treatment with the membrane-permeable InsP3 receptor antagonist 2-APB blocked the activation of I(CRAC). However, intracellular dialysis with 2-APB failed to prevent I(CRAC) from activating, even at higher concentrations than those used extracellularly to achieve full block. Local application of 2-APB, once I(CRAC) had been activated, resulted in a rapid loss of the current at a rate similar to that seen with the rapid channel blocker La3+. Studies with the more conventional InsP3 receptor antagonist heparin revealed that occupation of the intracellular InsP3-sensitive receptors was not necessary for the activation or maintenance of I(CRAC). Similarly, the InsP3 receptor inhibitor caffeine failed to alter the rate or extent of activation of I(CRAC). Exposure to Li+, which reduces InsP3 levels by interfering with inositol monophosphatase, also failed to alter I(CRAC). Caffeine and Li+ did not affect the size of the intracellular Ca2+ signal that arose when external Ca2+ was re-admitted to cells Topics: Animals; Basophils; Boron Compounds; Caffeine; Calcium; Calcium Channels; Calcium Signaling; Cell Line; Cell Size; Cytochalasin D; Cytoskeleton; Depsipeptides; Enzyme Inhibitors; Heparin; Inositol 1,4,5-Trisphosphate; Inositol 1,4,5-Trisphosphate Receptors; Lithium; Marine Toxins; Microscopy, Fluorescence; Nocodazole; Oxazoles; Patch-Clamp Techniques; Peptides, Cyclic; Rats; Receptors, Cytoplasmic and Nuclear; Thapsigargin; Time Factors | 2001 |
The actin cytoskeleton is required for the trafficking of the B cell antigen receptor to the late endosomes.
The B cell antigen receptor (BCR) plays two central roles in B cell activation: to internalize antigens for processing and presentation, and to initiate signal transduction cascades that both promote B cells to enter the cell cycle and facilitate antigen processing by accelerating antigen transport. An early event in B cell activation is the association of BCR with the actin cytoskeleton, and an increase in cellular F-actin. Current evidence indicates that the organization of actin filaments changes in response to BCR-signaling, making actin filaments good candidates for regulation of BCR-antigen targeting. Here, we have analyzed the role of actin filaments in BCR-mediated antigen transport, using actin filament-disrupting reagents, cytochalasin D and latrunculin B, and an actin filament-stabilizing reagent, jasplakinolide. Perturbing actin filaments, either by disrupting or stabilizing them, blocked the movement of BCR from the plasma membrane to late endosomes/lysosomes. Cytochalasin D-treatment dramatically reduced the rate of internalization of BCR, and blocked the movement of the BCR from early endosomes to late endosomes/lysosomes, without affecting BCR-signaling. Thus, BCR-trafficking requires functional actin filaments for both internalization and movement to late endosomes/lysosomes, defining critical control points in BCR-antigen targeting. Topics: Actins; Animals; Antineoplastic Agents; Bridged Bicyclo Compounds, Heterocyclic; Cell Line; Cell Membrane; Cross-Linking Reagents; Cytochalasin D; Cytoskeleton; Depsipeptides; Dose-Response Relationship, Drug; Endosomes; Humans; Hybridomas; Lymphocyte Activation; Lysosomes; Mice; Microscopy, Electron; Microscopy, Fluorescence; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Phosphorylation; Protein Transport; Rats; Receptors, Antigen, B-Cell; Signal Transduction; Thiazoles; Thiazolidines; Time Factors; Tyrosine | 2001 |
Entamoeba invadens: enhancement of excystation and metacystic development by cytochalasin D.
Effects of three actin-modifying drugs, cytochalasin D, latrunculin A, and jasplakinolide, on the excystation and metacystic development in vitro of Entamoeba invadens were examined by transfer of the cysts to growth medium with the drugs. Cytochalasin D unexpectedly increased the number of metacystic amoebae of E. invadens strain IP-1 during incubation. Metacystic development, which was determined by the number of nuclei of metacystic amoebae, was faster in the culture with cytochalasin D than in the culture without the drug. These results suggest that cytochalasin D enhances the excystation and metacystic development. In contrast, latrunculin A and jasplakinolide inhibited these process. No excystation occurred in encystation medium even in the presence of cytochalasin D, suggesting that growth medium is essential for excystation. Excystation was further enhanced when the cysts were incubated with cytochalasin D before culture in growth medium with the drug. The enhancing effect of cytochalasin D on the excystation and metacystic development was abrogated by jasplakinolide. Thus, the results indicate that cytochalasin D, unlike latrunculin A and jasplakinolide, caused enhancement of the excystation and metacystic development of this parasite. Topics: Animals; Bridged Bicyclo Compounds, Heterocyclic; Cytochalasin D; Depsipeptides; Entamoeba; Marine Toxins; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Porifera; Thiazoles; Thiazolidines | 2001 |
Drug-induced changes of cytoskeletal structure and mechanics in fibroblasts: an atomic force microscopy study.
The effect of various drugs affecting the integrity of different components of the cytoskeleton on the elasticity of two fibroblast cell lines was investigated by elasticity measurements with an atomic force microscope (AFM). Disaggregation of actin filaments always resulted in a distinct decrease in the cell's average elastic modulus indicating the crucial importance of the actin network for the mechanical stability of living cells. Disruption or chemical stabilization of microtubules did not affect cell elasticity. For the f-actin-disrupting drugs different mechanisms of drug action were observed. Cytochalasins B and D and Latrunculin A disassembled stress fibers. For Cytochalasin D this was accompanied by an aggregation of actin within the cytosol. Jasplakinolide disaggregated actin filaments but did not disassemble stress fibers. Fibrous structures found in AFM images and elasticity maps of fibroblasts could be identified as stress fibers by correlation of AFM data and fluorescence images. Topics: 3T3 Cells; Actins; Animals; Antineoplastic Agents; Bridged Bicyclo Compounds, Heterocyclic; Cell Line; Colchicine; Cytochalasin B; Cytochalasin D; Cytoskeleton; Demecolcine; Depsipeptides; Elasticity; Fibroblasts; Kinetics; Marine Toxins; Mice; Microscopy, Atomic Force; Microscopy, Fluorescence; Microtubules; Paclitaxel; Peptides, Cyclic; Rats; Stress, Mechanical; Thiazoles; Thiazolidines | 2000 |
Regulation of calcineurin by growth cone calcium waves controls neurite extension.
Growth cones generate spontaneous transient elevations of intracellular Ca(2+) that regulate the rate of neurite outgrowth. Here we report that these Ca(2+) waves inhibit neurite extension via the Ca(2+)-dependent phosphatase calcineurin (CN) in Xenopus spinal neurons. Pharmacological blockers of CN (cyclosporin A and deltamethrin) and peptide inhibitors of CN [the Xenopus CN (xCN) autoinhibitory domain and African swine fever virus protein A238L] block the Ca(2+)-dependent reduction of neurite outgrowth in cultured neurons. Time-lapse microscopy of growing neurites demonstrates directly that the reduction in the rate of outgrowth by Ca(2+) transients is blocked by cyclosporin A. In contrast, expression of a constitutively active form of xCN in the absence of waves results in shorter neurite lengths similar to those seen in the presence of waves. The developmental expression pattern of xCN transcripts in vivo coincides temporally with axonal pathfinding by spinal neurons, supporting a role of CN in regulating Ca(2+)-dependent neurite extension in the spinal cord. Ca(2+) wave frequency and Ca(2+)-dependent expression of GABA are not affected by inhibition or activation of CN. However, phosphorylation of the cytoskeletal element GAP-43, which promotes actin polymerization, is reduced by Ca(2+) waves and enhanced by suppression of CN activity. CN ultimately acts on the growth cone actin cytoskeleton, because disrupting actin microfilaments with cytochalasin D or stabilizing them with jasplakinolide negates the effects of suppressing or activating CN. Destabilization or stabilization of microtubules with colcemide or taxol results in Ca(2+)-independent inhibition of neurite outgrowth. The results identify components of the cascade by which Ca(2+) waves act to regulate neurite extension. Topics: Actins; Animals; Antifungal Agents; Antineoplastic Agents; Antineoplastic Agents, Phytogenic; Calcineurin; Calcium; Calcium Signaling; Cells, Cultured; Cloning, Molecular; Cyclosporine; Cytochalasin D; Demecolcine; Depsipeptides; Embryonic Development; Enzyme Inhibitors; Female; gamma-Aminobutyric Acid; GAP-43 Protein; Gene Expression Regulation, Developmental; Growth Cones; Molecular Sequence Data; Neurites; Neurons; Nucleic Acid Synthesis Inhibitors; Okadaic Acid; Paclitaxel; Peptides, Cyclic; Phosphorylation; Pyrans; Spinal Cord; Spiro Compounds; Xenopus laevis | 2000 |
A role for the actin cytoskeleton in the initiation and maintenance of store-mediated calcium entry in human platelets. Evidence for conformational coupling.
The nature of the mechanism underlying store-mediated Ca(2+) entry has been investigated in human platelets through a combination of cytoskeletal modifications. Inhibition of actin polymerization by cytochalasin D or latrunculin A had a biphasic time-dependent effect on Ca(2+) entry, showing an initial potentiation followed by inhibition of Ca(2+) entry. Moreover, addition of these agents after induction of store-mediated Ca(2+) entry inhibited the Ca(2+) influx mechanism. Jasplakinolide, which reorganizes actin filaments into a tight cortical layer adjacent to the plasma membrane, prevented activation of store-mediated Ca(2+) entry but did not modify this process after its activation. In addition, jasplakinolide prevented cytochalasin D-induced inhibition of store-mediated Ca(2+) entry. Calyculin A, an inhibitor of protein serine/threonine phosphatases 1 and 2 which activates translocation of existing F-actin to the cell periphery without inducing actin polymerization, also prevented activation of store-mediated Ca(2+) entry. Finally, inhibition of vesicular transport with brefeldin A inhibited activation of store-mediated Ca(2+) entry but did not alter this mechanism once initiated. These data suggest that store-mediated Ca(2+) entry in platelets may be mediated by a reversible trafficking and coupling of the endoplasmic reticulum with the plasma membrane, which shows close parallels to the events mediating secretion. Topics: Actins; Biological Transport; Blood Platelets; Brefeldin A; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Cell Membrane; Cytochalasin D; Cytoskeleton; Depsipeptides; Endoplasmic Reticulum; Humans; Inositol 1,4,5-Trisphosphate; Marine Toxins; Models, Biological; Oxazoles; Peptides, Cyclic; Thiazoles; Thiazolidines | 2000 |
Filamentous particle formation by human parainfluenza virus type 2.
Some paramyxoviruses form long filamentous virus particles: however, the determinants of filament formation and the role of such particles in virus transmission and pathogenicity are not clearly defined. By using conventional immunofluorescence microscopy, we found that human parainfluenza virus type 2 (HPIV2) forms filamentous particles ranging from 5 to 15 microm in length in virus-infected, polarized epithelial cells. The formation of filamentous particles was found to be virus type-specific and was not observed when the same cell types were infected with parainfluenza virus type 3 or Sendai virus, suggesting that different paramyxovirus genera exhibit distinct morphological properties. HPIV2 filamentous particle formation was found to be inhibited by cytochalasin D (CD) or jasplakinolide treatment in a dose-dependent manner. In the presence of 4 microg/ml CD or 1 microM jasplakinolide, the formation of filamentous particles was completely abolished, although similar haemagglutination and p.f.u. titres of virus were found to be released into the culture medium at 24 h post-infection. These observations indicate that host cell components, including the actin microfilament network, are important determinants of the morphology of parainfluenza viruses. The predominance of filamentous particles in polarized epithelial cells may reflect specific pathogenic roles of these particles in infection of human epithelial tissues. Topics: Actins; Animals; Cell Line; Chlorocebus aethiops; Cytochalasin D; Depsipeptides; Dogs; Fluorescent Antibody Technique; Hemagglutination Tests; Humans; Parainfluenza Virus 2, Human; Peptides, Cyclic; Vero Cells; Viral Plaque Assay; Virion | 2000 |
Involvement of the actin cytoskeleton in the regulation of serotonin transporter (SET) activity: possible mechanism underlying SET regulation by protein kinase C.
Our previous report has revealed that PKC activation by 12-O-tetradecanoylphorbol 13-acetate (TPA) inhibited the uptake activity of serotonin transporter (SET), via an indirect mechanism unknown, but not likely via direct phosphorylation of SET by PKC (Sakai et al., 1997. J. Neurochem. 68, 2618-2624). To elucidate whether PKC can directly phosphorylate SET in vivo, FLAG-tagged SET (FLAG-SET) was expressed in COS-7 cells and the TPA-induced incorporation of (32)P into immunoprecipitated FLAG-SET was examined. PKC activation with TPA caused no phosphorylation of FLAG-SET expressed in COS-7 cells. On the other hand, morphological change associated with the disruption of filamentous actin (F-actin) was seen in TPA-treated COS-7 cells. Therefore, we studied the effects of cytochalasin D, an inhibitor of actin polymerization, on the uptake activity of the serotonin transporter (SET) to elucidate whether the actin cytoskeleton modulates the SET uptake activity. The treatment with cytochalasin D inhibited the uptake activity of both native and recombinant SET in a concentration-dependent manner. Eadie-Hofstee analysis revealed that cytochalasin D down-regulated the recombinant SET uptake activity by reducing the V(max), but not the K(m), mimicking the result observed in TPA-induced inhibition of SET activity (Sakai et al., 1997. J. Neurochem. 68, 2618-2624). The cytochalasin D-induced inhibition of SET activity was partially, but significantly, reversed by jasplakinolide, a cell permeable stabilizer of F-actin, whereas TPA-induced inhibition of SET activity was not reversed by jasplakinolide. To elucidate whether the subcellular localization of SET was changed in response to cytochalasin D or TPA, we expressed the SET fused with the green fluorescent protein (SET-GFP) in COS-7 cells and observed the subcellular distribution of SET-GFP under a confocal laser scanning fluorescent microscope. Neither cytochalasin D nor TPA markedly changed the SET-GFP cellular localization, although these drugs caused morphological change in the GFP-transfected COS-7 cells. In addition, SET activity was not altered by the treatment with either colchicine, an inhibitor of microtubule polymerization, or taxol, a stabilizer of microtubule polymerization. These results suggest that the SET uptake activity was regulated by the state of the actin cytoskeleton and that TPA exerts its inhibitory action on SET activity, in part, via disruption of F-actin and subsequent morphological change i Topics: Actins; Animals; Carrier Proteins; COS Cells; Cytochalasin D; Cytoskeleton; Depsipeptides; Enzyme Activation; Green Fluorescent Proteins; Humans; Luminescent Proteins; Membrane Glycoproteins; Membrane Transport Proteins; Nerve Tissue Proteins; Peptides, Cyclic; Phosphorylation; Protein Kinase C; Recombinant Fusion Proteins; Recombinant Proteins; Serotonin; Serotonin Plasma Membrane Transport Proteins; Subcellular Fractions; Tetradecanoylphorbol Acetate; Tissue Distribution; Transfection; Tumor Cells, Cultured | 2000 |
Role of the cytoskeleton in rapid activation of CD11b/CD18 function and its subsequent downregulation in neutrophils.
When rolling adherent neutrophils are stimulated, they rapidly immobilize through activation of integrin CD11b/CD18, and then modulate attachment through this integrin to allow migration. We investigated links between cytoskeletal rearrangement and changes in function of integrin CD11b/CD18 in neutrophils stimulated with formyl peptide (fMLP). Neutrophils treated with the actin-polymerizing agent jasplakinolide became rolling adherent on monolayers of activated platelets, but could not use CD11b/CD18 to become immobilised when fMLP was perfused over them. If treated with jasplakinolide after fMLP, the cells stopped migrating but could not detach when fMLP was removed. Jasplakinolide did not inhibit changes in intracellular Ca(2+) seen after fMLP treatment, or inhibit neutrophil immobilisation induced by externally added Mn(2+). Thus cytoskeletal rearrangement was directly implicated in upregulation and, later, downregulation of CD11b/CD18 binding. Inhibition of RhoA with C3-transferase caused a dose-dependent reduction of initial rolling adhesion of neutrophils, and reduced the rate of migration after stimulation; however, neither the conversion of rolling to stationary adhesion, nor the ability of neutrophils to detach on removal of the stimulus, were inhibited. Thus, Rho may regulate actin polymerisation and motility in neutrophils, but did not appear to control integrin-mediated adhesion itself. Integrin binding may be promoted by disruption of links to the cytoskeleton, effected through depolymerisation of actin or cleavage of linking protein talin by calpain. Disruption of actin filaments with cytochalasin D did not, however, cause integrin-mediated immobilisation of rolling neutrophils. Although the calpain inhibitor calpeptin did inhibit the adhesion response to fMLP, this was only at doses where actin polymerisation was also ablated. We suggest that the cytoskeleton actively regulates binding conformation of CD11b/CD18 as well as its mobility in the membrane. Topics: ADP Ribose Transferases; Amides; Antineoplastic Agents; Botulinum Toxins; Cell Adhesion; Cell Movement; Cysteine Proteinase Inhibitors; Cytochalasin D; Cytoskeleton; Depsipeptides; Dipeptides; Down-Regulation; Enzyme Inhibitors; Humans; Integrins; Intracellular Signaling Peptides and Proteins; Macrophage-1 Antigen; Neutrophils; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Protein Serine-Threonine Kinases; Pyridines; rho-Associated Kinases; Signal Transduction | 2000 |
Actin-dependent regulation of neurotransmitter release at central synapses.
Depolymerization of actin by latrunculin A transiently promotes neurotransmitter release. The mean rate of mEPSCs increases by a Ca2+-independent process, without a concomitant change in the mean amplitude. The readily releasable vesicle pool size and the rate of refilling of the readily releasable pool remain unaltered by latrunculin treatment. Evoked neurotransmitter release also increases in a manner consistent with an increase in vesicle release probability. The observed enhancement of neurotransmitter release is specific to actin depolymerization mediated by latrunculin A and is not caused by cytochalasin D. Our findings indicate that actin participates in a regulatory mechanism that restrains fusion of synaptic vesicles at the active zone. Topics: Actins; Animals; Bridged Bicyclo Compounds, Heterocyclic; Calcium; Cells, Cultured; Cytochalasin D; Depsipeptides; Egtazic Acid; Excitatory Postsynaptic Potentials; Fluorescent Antibody Technique; Green Fluorescent Proteins; Hippocampus; Luminescent Proteins; Marine Toxins; Mice; Nerve Tissue Proteins; Neurotransmitter Agents; Peptides, Cyclic; Presynaptic Terminals; Rats; Recombinant Fusion Proteins; Synapses; Synaptic Transmission; Synaptic Vesicles; Thiazoles; Thiazolidines | 2000 |
Cytoskeletal regulation of the platelet glycoprotein Ib/V/IX-von willebrand factor interaction.
Shear-induced binding of von Willebrand factor (vWf) to the platelet glycoprotein (GP) Ib/V/IX complex plays a key role in initiating platelet adhesion and aggregation at sites of vascular injury. This study demonstrated that pretreating human platelets with inhibitors of actin polymerization, cytochalasin D or latrunculin B, dramatically enhances platelet aggregation induced by vWf. The effects of these inhibitors were specific to the vWf-GPIbalpha interaction because they enhanced vWf-induced aggregation of Glanzmann thrombasthenic platelets and Chinese hamster ovary (CHO) cells transfected with GPIb/V/IX. Moreover, cytochalasin D enhanced the extent of platelet aggregation induced by high shear stress (5000 s(-1)) and also lowered the shear threshold required to induce aggregation from 3000 s(-1) to as low as 500 s(-1). Studies of CHO cells expressing GPIbalpha cytoplasmic tail truncation mutants that failed to bind actin-binding protein-280 (deletion of residues 569-610 or 535-568) demonstrated that the linkage between GPIb and actin-binding protein-280 was not required for vWf-induced actin polymerization, but was critical for the enhancing effects of cytochalasin D on vWf-induced cell aggregation. Taken together, these studies suggest a fundamentally important role for the cytoskeleton in regulating the adhesive function of GPIb/V/IX. Topics: Actin Cytoskeleton; Actins; Adenosine Diphosphate; Alprostadil; Animals; Antibodies, Monoclonal; Blood Platelets; Bridged Bicyclo Compounds, Heterocyclic; CHO Cells; Cricetinae; Cytochalasin D; Cytoskeleton; Depsipeptides; Humans; Mutagenesis, Site-Directed; Peptides, Cyclic; Platelet Aggregation; Platelet Aggregation Inhibitors; Platelet Glycoprotein GPIb-IX Complex; Platelet Glycoprotein GPIIb-IIIa Complex; Stress, Mechanical; Thiazoles; Thiazolidines; Thrombasthenia; Transfection; von Willebrand Factor | 2000 |
Disruption of the actin filament network affects delivery of endocytic contents marker to phagosomes with early endosome characteristics: the case of phagosomes with pathogenic mycobacteria.
Phagosomes containing live virulent mycobacteria undergo fusion with early endosomes, but they are unable to mature normally. Accordingly, they do not fuse with lysosomes. Although M. avium-containing phagosomes retain fusion and intermingling characteristics of early endosomes indefinitely, fusions with early endosomes are increasingly restricted as bacteria multiply. In addition, when endocytic tracers, such as horseradish peroxidase (HRP), are added to M. avium-infected macrophages at 1 or up to 15 days after infection, an atypical time course of acquisition of the tracer by the phagosomes is observed, i.e., a 10 to 20 min lag, instead of immediate acquisition as is typical for early endosomes (and phagosomes with early endosome characteristics). These events coincide with a marked disorganization of the actin filament network in M. avium-infected macrophages. In the present study, we have therefore addressed the following question: Do actin filaments play a role in fusion and intermingling of contents between early endosomes and immature phagosomes that undergo homotypic fusion with early endosomes? We examined the time course of acquisition of subsequently internalized endocytic marker (HRP) by early endosome-like preexisting phagosomes, i.e. 2 hour-old phagosomes with either hydrophobic latex particles, virulent or avirulent M. avium, after depolymerization of the actin filament network with cytochalasin D or after repolymerization of the actin filament network with jasplakinolide, in cases where the network had been depolymerized (macrophages infected with M. avium, at 1 or up to 7 days after infection). By direct morphological observation at the electron microscope level and by a kinetic approach, we show here that depolymerization of the actin filament network with cytochalasin D delays acquisition of HRP whereas repolymerization restores immediate acquisition of the marker. We conclude that the actin filament network is involved in fusion and intermingling of endocytic contents between early endosomes and early endosome-like phagosomes, and that disruption of this network by M. avium is the cause for the atypical acquisition of content marker by phagosomes containing these pathogenic mycobacteria. Topics: Actins; Animals; Antifungal Agents; Bone Marrow Cells; Cells, Cultured; Cytochalasin D; Depsipeptides; Endocytosis; Endosomes; Horseradish Peroxidase; Kinetics; Lysosomes; Macrophages; Mice; Mice, Inbred C57BL; Microscopy, Electron; Microscopy, Fluorescence; Mycobacterium; Peptides, Cyclic; Phagocytosis; Phagosomes; Time Factors | 2000 |
Actin assembly plays a variable, but not obligatory role in receptor-mediated endocytosis in mammalian cells.
Three cell-permeant compounds, cytochalasin D, latrunculin A and jasplakinolide, which perturb intracellular actin dynamics by distinct mechanisms, were used to probe the role of filamentous actin and actin assembly in clathrin-mediated endocytosis in mammalian cells. These compounds had variable effects on receptor-mediated endocytosis of transferrin that depended on both the cell line and the experimental protocol employed. Endocytosis in A431 cells assayed in suspension was inhibited by latrunculin A and jaspiakinolide, but resistant to cytochalasin D, whereas neither compound inhibited endocytosis in adherent A431 cells. In contrast, endocytosis in adherent CHO cells was more sensitive to disruption of the actin cytoskeleton than endocytosis in CHO cells grown or assayed in suspension. Endocytosis in other cell types, including nonadherent K562 human erythroleukemic cells or adherent Cos-7 cells was unaffected by disruption of the actin cytoskeleton. While it remains possible that actin filaments can play an accessory role in receptor-mediated endocytosis, these discordant results indicate that actin assembly does not play an obligatory role in endocytic coated vesicle formation in cultured mammalian cells. Topics: Actins; Animals; Antineoplastic Agents; Bridged Bicyclo Compounds, Heterocyclic; Cell Line; Cell Membrane; CHO Cells; Coated Pits, Cell-Membrane; COS Cells; Cricetinae; Cytochalasin D; Cytoskeleton; Depsipeptides; Endocytosis; Freeze Etching; Humans; K562 Cells; Microscopy, Fluorescence; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Thiazoles; Thiazolidines; Time Factors; Transferrin; Tumor Cells, Cultured | 2000 |
Morphological changes in the Golgi complex correlate with actin cytoskeleton rearrangements.
In this report we have studied the morphological changes of the Golgi complex (GC) that specifically accompany F-actin reorganizations. In starved rat RBL-2H3 tumor mast cells, the GC, that was visualized at immunofluorescence level with antibodies raised against the Golgi-resident proteins giantin, mannosidase II, or TGN-38, showed a compacted morphology with a supranuclear positioning. Concomitant to membrane ruffle formation induced by epidermal growth factor (EGF) or phorbol 12-myristate 13-acetate (PMA), and stress fiber formation induced by lysophosphatidic acid (LPA), specific GC morphological changes were observed. When cells were stimulated with EGF or PMA, the compacted GC morphology was transformed into a reticular network that was extended towards the cell periphery. When cells were incubated with LPA, the GC acquired a characteristic ring-shaped morphology. Brefeldin A (BFA) did not affect the PMA- or LPA-induced membrane ruffling and stress fiber formation, respectively, indicating that actin rearrangements occurred independent of the presence of the GC. Upon BFA removal, the presence of PMA or LPA during the recovery process induced the GC to acquire the morphological appearance described above for each agent. Moreover, the PMA- but not the LPA-induced GC rearrangements were sensitive to the actin perturbing agents cytochalasin D and jasplakinolide. When cells were preincubated with the phosphatidylinositide 3-kinase (PI3K) inhibitors wortmannin or LY294002, the PMA-induced GC morphological changes were inhibited but not membrane ruffles. Finally, the PMA-induced increase in the post-Golgi transport of glycosaminoglycans to the cell surface was not altered by cytochalasin D or jasplakinolide. Altogether, these data suggest that: (1) the shape of the GC is influenced by the 3D arrangement of actin microfilaments; (2) PI3K regulates the association of the GC with actin microfilaments; and (3) actin microfilaments are not essential for the post-Golgi transport to the plasma membrane. Topics: Actins; Androstadienes; Animals; Biological Transport; Brefeldin A; Cell Membrane; Chromones; Cytochalasin D; Cytoskeleton; Depsipeptides; Enzyme Inhibitors; Epidermal Growth Factor; Glycosaminoglycans; Golgi Apparatus; Lysophospholipids; Morpholines; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Protein Synthesis Inhibitors; Rats; Tetradecanoylphorbol Acetate; Tumor Cells, Cultured; Wortmannin | 1999 |
The anti-proliferative agent jasplakinolide rearranges the actin cytoskeleton of plant cells.
In the present study, we have characterized the action of the natural cyclodepsipeptide jasplakinolide (JAS) on the cytoplasmic architecture, actin-based cytoplasmic motility, and the organization of the actin cytoskeleton in selected examples of green algae (Acetabularia, Pseudobryopsis and Nitella) and higher plant cells (Allium bulb scale cells and Sinapis root hairs). JAS was capable of influencing the actin cytoskeleton and inhibiting cytoplasmic streaming in a differential, cell type-specific manner. With the exception of Nitella, two consecutive responses were observed upon incubation with 2.5 microM JAS: In the first phase cytoplasmic streaming increased transiently alongside with minor modifications of the actin cytoskeleton in the form of adventitious actin spots and spikes appearing throughout the cell cortex in addition to the normal actin bundle system typical for each cell type. In the second phase, cytoplasmic streaming stopped and the actin cytoskeleton became heavily reorganized into shorter, straight, more and more randomly oriented bundle segments. JAS exerted severe long-term effects on the actin cytoskeleton when treatments exceeded 30min at a concentration of 2.5 microM. An in situ competition assay using equimolar concentrations of JAS and FITC-phalloidin suggested that JAS has a phalloidin-like action. Effects of JAS were significantly different from those of cytochalasin D with respect to the resulting degree of perturbance of cytoplasmic organization, the distribution of actin filaments and the speed of reversibility. Topics: Actins; Animals; Biological Transport; Chlorophyta; Cytochalasin D; Cytoplasm; Cytoskeleton; Depsipeptides; Fluorescein-5-isothiocyanate; Growth Inhibitors; Mice; Mustard Plant; Onions; Organelles; Peptides, Cyclic; Phalloidine; Plants, Medicinal | 1999 |
LPS-induced depolymerization of cytoskeleton and its role in TNF-alpha production by rat pneumocytes.
Lipopolysaccharide (LPS) polymerizes microfilaments and microtubules in macrophages and monocytes. Disrupting microfilaments or microtubules with cytochalasin D (CytoD) or colchicine can suppress LPS-induced tumor necrosis factor-alpha (TNF-alpha) gene expression and protein production from these cells. We have recently demonstrated that primary cultured rat alveolar epithelial cells can produce TNF-alpha on LPS stimulation. In the present study, we found that the LPS-induced increase in TNF-alpha mRNA level and protein production in alveolar epithelial cells was not inhibited by CytoD or colchicine (1 nM to 10 microM). In fact, LPS-induced TNF-alpha production was further enhanced by CytoD (1-10 microM) and inhibited by jasplakinolide, a polymerizing agent for microfilaments. Immunofluorescent staining and confocal microscopy showed that LPS (10 microg/ml) depolymerized microfilaments and microtubules within 15 min, which was prolonged until 24 h for microfilaments. These results suggest that the effects of LPS on the cytoskeleton and the role of the cytoskeleton in mediating TNF-alpha production in alveolar epithelial cells are opposite to those in immune cells. This disparity may reflect the different roles between nonimmune and immune cells in host defense. Topics: Actin Cytoskeleton; Animals; Cells, Cultured; Colchicine; Cytochalasin D; Cytoskeleton; Depsipeptides; Epithelial Cells; Lipopolysaccharides; Male; Microtubules; Peptides, Cyclic; Polymers; Pulmonary Alveoli; Rats; Rats, Sprague-Dawley; RNA, Messenger; Tumor Necrosis Factor-alpha | 1999 |
Microtubules facilitate the stimulated secretion of beta-hexosaminidase in lacrimal acinar cells.
Stimulation of lacrimal acini with secretagogues such as carbachol initiates movement and fusion of acinar secretory vesicles with the apical plasma membrane, resulting in release of protein into the nascent tear fluid. Using rabbit lacrimal acini reconstituted in vitro from isolated cells, we have investigated the organization of the apical cytoskeleton and its role in stimulated secretion. Confocal microscopy revealed a microtubule array emanating from the apical region of the acini; the apical region was also enriched in microfilaments and (gamma)-tubulin. Cytokeratin-based intermediate filaments were apically concentrated, and also detected at the cell periphery. Neither confocal microscopy nor biochemical analysis revealed any reorganization of lumenal microfilaments or microtubules which might accompany carbachol-stimulated release of secretory proteins. However, major changes in the acinar microtubule array induced by taxol or nocodazole were correlated with inhibition of carbachol-dependent release of the secreted protein, beta-hexosaminidase. Major changes in lumenal microfilaments induced by jasplakinolide or cytochalasin D did not inhibit the carbachol-dependent release of beta-hexosaminidase; rather, release of beta-hexosaminidase from jasplakinolide- or cytochalasin D-treated carbachol-stimulated acini was markedly increased relative to the release from untreated stimulated acini. Our findings demonstrate that microtubules play a major role in stimulated lacrimal secretion, and suggest a contributory role for microfilaments. Topics: Actin Cytoskeleton; Actins; Animals; beta-N-Acetylhexosaminidases; Carbachol; Cell Polarity; Cells, Cultured; Cytochalasin D; Cytoskeleton; Depsipeptides; Female; Lacrimal Apparatus; Microscopy, Confocal; Microtubules; Nocodazole; Peptides, Cyclic; Rabbits; Secretory Rate; Tubulin | 1998 |
Simultaneous measurements of actin filament turnover, filament fraction, and monomer diffusion in endothelial cells.
The analogous techniques of photoactivation of fluorescence (PAF) and fluorescence recovery after photobleaching (FRAP) have been applied previously to the study of actin dynamics in living cells. Traditionally, separate experiments estimate the mobility of actin monomer or the lifetime of actin filaments. A mathematical description of the dynamics of the actin cytoskeleton, however, predicts that the evolution of fluorescence in PAF and FRAP experiments depends simultaneously on the diffusion coefficient of actin monomer, D, the fraction of actin in filaments, FF, and the lifetime of actin filaments, tau (, Biophys. J. 69:1674-1682). Here we report the application of this mathematical model to the interpretation of PAF and FRAP experiments in subconfluent bovine aortic endothelial cells (BAECs). The following parameters apply for actin in the bulk cytoskeleton of subconfluent BAECs. PAF: D = 3.1 +/- 0.4 x 10(-8) cm2/s, FF = 0.36 +/- 0.04, tau = 7.5 +/- 2.0 min. FRAP: D = 5.8 +/- 1.2 x 10(-8) cm2/s, FF = 0.5 +/- 0.04, tau = 4.8 +/- 0.97 min. Differences in the parameters are attributed to differences in the actin derivatives employed in the two studies and not to inherent differences in the PAF and FRAP techniques. Control experiments confirm the modeling assumption that the evolution of fluorescence is dominated by the diffusion of actin monomer, and the cyclic turnover of actin filaments, but not by filament diffusion. The work establishes the dynamic state of actin in subconfluent endothelial cells and provides an improved framework for future applications of PAF and FRAP. Topics: Actins; Animals; Cattle; Cells, Cultured; Cytochalasin D; Cytoskeleton; Depsipeptides; Endothelium, Vascular; Fluorescent Dyes; Kinetics; Microscopy, Electron; Microscopy, Fluorescence; Peptides, Cyclic; Photochemistry | 1998 |
The chondramides: cytostatic agents from myxobacteria acting on the actin cytoskeleton.
Chondramides are cyclodepsipeptides produced by strains of the myxobacterium, Chondromyces crocatus. These peptides, which have been reported to inhibit yeast and mammalian cell proliferation, are related to jasplakinolide, which has been isolated from marine sponges of the genus Jaspis and has been shown to interfere with the actin cytoskeleton (a structural component of cells that helps maintain their shape and is involved in processes, such as cell division and cell locomotion). We studied the effects of the chondramides (A, B, C, and D) on tumor cell growth, on cytoskeletal structure, and on actin polymerization in vitro and compared these effects with those of cytochalasin D and jasplakinolide.. Cell proliferation was measured by means of tetrazolium salt reduction assays. Effects on the cytoskeleton were studied by use of fluorescence techniques, and actin polymerization in vitro was measured by means of viscosimetry.. Proliferation of tested tumor cell lines was inhibited by the chondramides. Concentrations that inhibited proliferation by 50% (IC50 values) ranged from 3 to 85 nM and were of the same order of magnitude as those found for cytochalasin D and jasplakinolide. Fluorescence staining of potoroo cells incubated with chondramides A and B showed that organization of the actin cytoskeleton was disrupted; however, the microtubule system was not affected. Viscosimetric measurement showed that, depending on the experimental conditions, chondramide A induced or accelerated actin polymerization in vitro.. The chondramides--unlike jasplakinolide--can be produced in large amounts by fermentation, and, similar to jasplakinolide, they appear to have antiproliferative activity against carcinoma cell lines by targeting the actin cytoskeleton. Topics: Actins; Antibiotics, Antineoplastic; Antineoplastic Agents; Bacterial Proteins; Cell Division; Cytochalasin D; Depsipeptides; Humans; Nucleic Acid Synthesis Inhibitors; Peptides, Cyclic; Tumor Cells, Cultured | 1998 |
Effects of F-actin stabilization or disassembly on epithelial Cl- secretion and Na-K-2Cl cotransport.
Previous studies showed that cAMP-dependent transepithelial Cl- secretion of the intestinal cell line T84 is reduced by the F-actin stabilizer phalloidin, an effect in part attributable to inhibition of basolateral Na-K-2Cl cotransport. However, secretory responses are preserved in cells treated with the microfilament disrupter cytochalasin D. We explored the effects of cytochalasin D and two novel compounds derived from marine sponges on the Cl- secretory apparatus of T84 cells. Jasplakinolide (which stabilizes F-actin inhibited cAMP-dependent secretion and Na-K-2Cl cotransport. Latrunculin A (which sequesters G-actin monomers) profoundly altered the distribution of F-actin and reduced basal transepithelial resistance with minimal effect on secretion. Cytochalasin D, but not latrunculin A, activated Na-K-2Cl cotransport. The results provide further evidence that vectorial ion transport is influenced by the cytoskeleton and support a model in which disassembly of F-actin by specific pharmacological means or in response to secretory agonists favors activation of Na-K-2Cl cotransport. Topics: Actins; Bridged Bicyclo Compounds, Heterocyclic; Carrier Proteins; Chlorides; Cytochalasin D; Cytoskeleton; Depsipeptides; Drug Stability; Humans; Intestinal Mucosa; Intracellular Membranes; Peptides, Cyclic; Sodium-Potassium-Chloride Symporters; Thiazoles; Thiazolidines | 1997 |