Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 64
1.
Carbohydr Polym ; 278: 118948, 2022 Feb 15.
Article En | MEDLINE | ID: mdl-34973765

Creating a low-cost, highly efficient, and recyclable superabsorbent for spilled-oil cleanup is of great significance but remains a big challenge. Herein, we report a facile strategy to produce economic, environmentally friendly, and reusable foam from agricultural waste kapok fibers. These kapok-derived cellulose nanofibrils foams (KNFs) demonstrate a hierarchically porous structure at micro-level with ultra-low density (2.7 mg·cm-3). The superhydrophobic KNFs (150.5°) show outstanding oil absorption (126.8-320.4 g·g-1) and oil-water separation performance. Notably, a facile approach is designed to reuse KNFs easily by a homemade oil release system. The release behavior of the KNFs is quantitatively analyzed and confirmed by the Rigter-Peppas model, indicating that the oil release followed the Fickian diffusion. The KNFs exhibit desirable reusability, and can be recycled for at least 50 times while keeping excellent oil absorption, and release performance. These advantages prove that the KNF is a desirable substitute for spilled-oil treatment.


Ceiba/chemistry , Cellulose/chemistry , Nanoparticles/chemistry , Oils/chemistry , Recycling , Stress Fibers/chemistry
2.
Cells ; 10(8)2021 08 04.
Article En | MEDLINE | ID: mdl-34440749

The cellular mechanisms of basement membrane (BM) invasion remain poorly understood. We investigated the invasion-promoting mechanisms of actin cytoskeleton reorganization in BM-covered MCF10A breast acini. High-resolution confocal microscopy has characterized actin cell protrusion formation and function in response to tumor-resembling ECM stiffness and soluble EGF stimulation. Traction force microscopy quantified the mechanical BM stresses that invasion-triggered acini exerted on the BM-ECM interface. We demonstrate that acini use non-proteolytic actin microspikes as functional precursors of elongated protrusions to initiate BM penetration and ECM probing. Further, these microspikes mechanically widened the collagen IV pores to anchor within the BM scaffold via force-transmitting focal adhesions. Pre-invasive basal cells located at the BM-ECM interface exhibited predominantly cortical actin networks and actin microspikes. In response to pro-invasive conditions, these microspikes accumulated and converted subsequently into highly contractile stress fibers. The phenotypical switch to stress fiber cells matched spatiotemporally with emerging high BM stresses that were driven by actomyosin II contractility. The activation of proteolytic invadopodia with MT1-MMP occurred at later BM invasion stages and only in cells already disseminating into the ECM. Our study demonstrates that BM pore-widening filopodia bridge mechanical ECM probing function and contractility-driven BM weakening. Finally, these EMT-related cytoskeletal adaptations are critical mechanisms inducing the invasive transition of benign breast acini.


Actins/metabolism , Basement Membrane/metabolism , Myosin Type II/metabolism , Stress Fibers/metabolism , Acinar Cells/cytology , Acinar Cells/metabolism , Breast/cytology , Breast/metabolism , Cell Adhesion , Cell Line , Cell Movement/drug effects , Epidermal Growth Factor/pharmacology , Extracellular Matrix/metabolism , Female , Humans , Microscopy, Confocal , Podosomes/metabolism , Pseudopodia/metabolism , Stress Fibers/chemistry
3.
Nat Commun ; 11(1): 4902, 2020 09 29.
Article En | MEDLINE | ID: mdl-32994402

Living cells and tissues experience various complex modes of forces that are important in physiology and disease. However, how different force modes impact gene expression is elusive. Here we apply local forces of different modes via a magnetic bead bound to the integrins on a cell and quantified cell stiffness, chromatin deformation, and DHFR (dihydrofolate reductase) gene transcription. In-plane stresses result in lower cell stiffness than out-of-plane stresses that lead to bead rolling along the cell long axis (i.e., alignment of actin stress fibers) or at different angles (90° or 45°). However, chromatin stretching and ensuing DHFR gene upregulation by the in-plane mode are similar to those induced by the 45° stress mode. Disrupting stress fibers abolishes differences in cell stiffness, chromatin stretching, and DHFR gene upregulation under different force modes and inhibiting myosin II decreases cell stiffness, chromatin deformation, and gene upregulation. Theoretical modeling using discrete anisotropic stress fibers recapitulates experimental results and reveals underlying mechanisms of force-mode dependence. Our findings suggest that forces impact biological responses of living cells such as gene transcription via previously underappreciated means.


Chromatin/chemistry , Stress Fibers/chemistry , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic/physiology , Up-Regulation/physiology , Animals , Anisotropy , Biomechanical Phenomena/genetics , CHO Cells , Chromatin/metabolism , Cricetulus , Heterocyclic Compounds, 4 or More Rings/pharmacology , Intravital Microscopy , Microscopy, Fluorescence , Myosin Type II/antagonists & inhibitors , Myosin Type II/metabolism , Stress Fibers/drug effects , Stress Fibers/metabolism , Stress, Mechanical , Transcription, Genetic/drug effects , Up-Regulation/drug effects
4.
Proc Natl Acad Sci U S A ; 117(41): 25532-25542, 2020 10 13.
Article En | MEDLINE | ID: mdl-32989126

The actin cytoskeleton assembles into diverse load-bearing networks, including stress fibers (SFs), muscle sarcomeres, and the cytokinetic ring to both generate and sense mechanical forces. The LIM (Lin11, Isl- 1, and Mec-3) domain family is functionally diverse, but most members can associate with the actin cytoskeleton with apparent force sensitivity. Zyxin rapidly localizes via its LIM domains to failing SFs in cells, known as strain sites, to initiate SF repair and maintain mechanical homeostasis. The mechanism by which these LIM domains associate with stress fiber strain sites (SFSS) is not known. Additionally, it is unknown how widespread strain sensing is within the LIM protein family. We identify that the LIM domain-containing region of 18 proteins from the Zyxin, Paxillin, Tes, and Enigma proteins accumulate to SFSS. Moreover, the LIM domain region from the fission yeast protein paxillin like 1 (Pxl1) also localizes to SFSS in mammalian cells, suggesting that the strain sensing mechanism is ancient and highly conserved. We then used sequence and domain analysis to demonstrate that tandem LIM domains contribute additively, for SFSS localization. Employing in vitro reconstitution, we show that the LIM domain-containing region from mammalian zyxin and fission yeast Pxl1 binds to mechanically stressed F-actin networks but does not associate with relaxed actin filaments. We propose that tandem LIM domains recognize an F-actin conformation that is rare in the relaxed state but is enriched in the presence of mechanical stress.


LIM Domain Proteins/metabolism , LIM Domain Proteins/physiology , Stress Fibers/metabolism , Stress Fibers/physiology , Amino Acid Sequence , Animals , Biomechanical Phenomena/physiology , Cell Line , Conserved Sequence , Evolution, Molecular , LIM Domain Proteins/chemistry , Mice , Myosins/chemistry , Myosins/metabolism , Protein Binding/physiology , Stress Fibers/chemistry , Stress, Mechanical , Yeasts
5.
Biomech Model Mechanobiol ; 19(2): 543-555, 2020 Apr.
Article En | MEDLINE | ID: mdl-31549258

Mechanisms of the assembly of actin stress fibers (SFs) have been extensively studied, while those of the disassembly-particularly cell shortening-induced ones-remain unclear. Here, we show that SFs have helical structures composed of multi-subbundles, and they tend to be delaminated upon cell shortening. Specifically, we observed with atomic force microscopy delamination of helical SFs into their subbundles. We physically caught individual SFs using a pair of glass needles to observe rotational deformations during stretching as well as ATP-driven active contraction, suggesting that they deform in a manner reflecting their intrinsic helical structure. A minimal analytical model was then developed based on the Frenet-Serret formulas with force-strain measurement data to suggest that helical SFs can be delaminated into the constituent subbundles upon axial shortening. Given that SFs are large molecular clusters that bear cellular tension but must promptly disassemble upon loss of the tension, the resulting increase in their surface area due to the shortening-induced delamination may facilitate interaction with surrounding molecules to aid subsequent disintegration. Thus, our results suggest a new mechanism of the disassembly that occurs only in the specific SFs exposed to forced shortening.


Actins/chemistry , Stress Fibers/chemistry , Actins/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cattle , Cells, Cultured , Microscopy, Atomic Force , Models, Biological , Protein Structure, Secondary , Rats , Stress Fibers/metabolism , Stress Fibers/ultrastructure , Stress, Mechanical
6.
Gene Expr ; 20(1): 25-37, 2020 06 12.
Article En | MEDLINE | ID: mdl-31757226

Hepatic stellate cells (HSC) are critical effector cells of liver fibrosis. In the injured liver, HSC differentiate into a myofibrobastic phenotype. A critical feature distinguishing myofibroblastic from quiescent HSC is cytoskeletal reorganization. Soluble NSF attachment receptor (SNARE) proteins are important in trafficking of newly synthesized proteins to the plasma membrane for release into the extracellular environment. The goals of this project were to determine the expression of specific SNARE proteins in myofibroblastic HSC and to test whether their alteration changed the HSC phenotype in vitro and progression of liver fibrosis in vivo. We found that HSC lack the t-SNARE protein, SNAP-25, but express a homologous protein, SNAP-23. Downregulation of SNAP-23 in HSC induced reduction in polymerization and disorganization of the actin cytoskeleton associated with loss of cell movement. In contrast, reduction in SNAP-23 in mice by monogenic deletion delayed but did not prevent progression of liver fibrosis to cirrhosis. Taken together, these findings suggest that SNAP-23 is an important regular of actin dynamics in myofibroblastic HSC, but that the role of SNAP-23 in the progression of liver fibrosis in vivo is unclear.


Actin Cytoskeleton/ultrastructure , Hepatic Stellate Cells/ultrastructure , Myofibroblasts/ultrastructure , Qb-SNARE Proteins/deficiency , Qc-SNARE Proteins/deficiency , Actin Cytoskeleton/chemistry , Actin Depolymerizing Factors/biosynthesis , Actins/analysis , Animals , Carbon Tetrachloride/toxicity , Cell Line , Cell Movement , Cell Separation , Gene Knockdown Techniques , Hepatic Stellate Cells/metabolism , Humans , Liver/cytology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Mice , Qb-SNARE Proteins/antagonists & inhibitors , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/antagonists & inhibitors , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/physiology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction , Stress Fibers/chemistry , Stress Fibers/ultrastructure , Wound Healing , rho-Associated Kinases/physiology
7.
J Mol Recognit ; 31(9): e2719, 2018 09.
Article En | MEDLINE | ID: mdl-29701269

Mechanical properties of myofibroblasts play a key role in Dupuytren's disease. Here, we used atomic force microscopy to measure the viscoelastic properties of 3 different types of human primary fibroblasts derived from a same patient: normal and scar dermal fibroblasts and palmar fascial fibroblasts from Dupuytren's nodules. Different stiffness hydrogels (soft ~1 kPa and stiff ~ 50 kPa) were used as cell culture matrix to mimic the mechanical properties of the natural tissues, and atomic force microscopy step response force curves were used to discriminate between elastic and viscous properties of cells. Since transforming growth factor-ß1 (TGF-ß1) is known to induce expression of α-smooth muscle actin positive stress fibers in myofibroblasts, we investigated the behavior of these fibroblasts before and after applying TGF-ß1. Finally, we performed an in vitro cell motility test, the wound healing or scratch assay, to evaluate the migratory properties of these fibroblasts. We found that (1) Dupuytren's fibroblasts are stiffer than normal and scar fibroblasts, the elastic modulus E ranging from 4.4, 2.1, to 1.8 kPa, for Dupuytren's, normal and scar fibroblasts, respectively; (2) TGF-ß1 enhances the level of α-smooth muscle actin expression and thus cell stiffness in Dupuytren's fibroblasts (E, ~6.2 kPa); (3) matrix stiffness influences cell mechanical properties most prominently in Dupuytren's fibroblasts; and (4) Dupuytren's fibroblasts migrate slower than the other fibroblasts by a factor of 3. Taking together, our results showed that mechanical and migratory properties of fibroblasts might help to discriminate between different pathological conditions, helping to identify and recognize specific cell phenotypes.


Cicatrix/pathology , Fibroblasts/pathology , Mechanical Phenomena , Transforming Growth Factor beta1/genetics , Actins/genetics , Cell Movement/genetics , Dupuytren Contracture/pathology , Fibroblasts/metabolism , Gene Expression Regulation/genetics , Humans , Myofibroblasts/chemistry , Myofibroblasts/pathology , Stress Fibers/chemistry
8.
Proc Natl Acad Sci U S A ; 114(26): 6782-6787, 2017 06 27.
Article En | MEDLINE | ID: mdl-28607071

Muscle contraction relies on the interaction of myosin motors with F-actin, which is regulated through a translocation of tropomyosin by the troponin complex in response to Ca2+ The current model of muscle regulation holds that at relaxing (low-Ca2+) conditions tropomyosin blocks myosin binding sites on F-actin, whereas at activating (high-Ca2+) conditions tropomyosin translocation only partially exposes myosin binding sites on F-actin so that binding of rigor myosin is required to fully activate the thin filament (TF). Here we used a single-particle approach to helical reconstruction of frozen hydrated native cardiac TFs under relaxing and activating conditions to reveal the azimuthal movement of the tropomyosin on the surface of the native cardiac TF upon Ca2+ activation. We demonstrate that at either relaxing or activating conditions tropomyosin is not constrained in one structural state, but rather is distributed between three structural positions on the surface of the TF. We show that two of these tropomyosin positions restrain actomyosin interactions, whereas in the third position, which is significantly enhanced at high Ca2+, tropomyosin does not block myosin binding sites on F-actin. Our data provide a structural framework for the enhanced activation of the cardiac TF over the skeletal TF by Ca2+ and lead to a mechanistic model for the regulation of the cardiac TF.


Actins/chemistry , Calcium/chemistry , Myocardium/chemistry , Stress Fibers/chemistry , Tropomyosin/chemistry , Actins/metabolism , Animals , Calcium/metabolism , Myocardium/metabolism , Stress Fibers/metabolism , Swine , Tropomyosin/metabolism
9.
Proc Natl Acad Sci U S A ; 114(20): 5195-5200, 2017 05 16.
Article En | MEDLINE | ID: mdl-28465431

The intermediate filament vimentin is required for cells to transition from the epithelial state to the mesenchymal state and migrate as single cells; however, little is known about the specific role of vimentin in the regulation of mesenchymal migration. Vimentin is known to have a significantly greater ability to resist stress without breaking in vitro compared with actin or microtubules, and also to increase cell elasticity in vivo. Therefore, we hypothesized that the presence of vimentin could support the anisotropic mechanical strain of single-cell migration. To study this, we fluorescently labeled vimentin with an mEmerald tag using TALEN genome editing. We observed vimentin architecture in migrating human foreskin fibroblasts and found that network organization varied from long, linear bundles, or "fibers," to shorter fragments with a mesh-like organization. We developed image analysis tools employing steerable filtering and iterative graph matching to characterize the fibers embedded in the surrounding mesh. Vimentin fibers were aligned with fibroblast branching and migration direction. The presence of the vimentin network was correlated with 10-fold slower local actin retrograde flow rates, as well as spatial homogenization of actin-based forces transmitted to the substrate. Vimentin fibers coaligned with and were required for the anisotropic orientation of traction stresses. These results indicate that the vimentin network acts as a load-bearing superstructure capable of integrating and reorienting actin-based forces. We propose that vimentin's role in cell motility is to govern the alignment of traction stresses that permit single-cell migration.


Vimentin/chemistry , Vimentin/physiology , Actins/chemistry , Animals , Cell Movement/physiology , Cell Polarity/physiology , Elasticity , Epithelial-Mesenchymal Transition/physiology , Fibroblasts/chemistry , Humans , Intermediate Filaments/chemistry , Intermediate Filaments/physiology , Mechanical Phenomena , Microtubules/chemistry , Stress Fibers/chemistry , Stress Fibers/physiology , Vimentin/metabolism , Weight-Bearing
10.
Proc Natl Acad Sci U S A ; 114(23): E4549-E4555, 2017 06 06.
Article En | MEDLINE | ID: mdl-28468803

We describe a multiscale model that incorporates force-dependent mechanical plasticity induced by interfiber cross-link breakage and stiffness-dependent cellular contractility to predict focal adhesion (FA) growth and mechanosensing in fibrous extracellular matrices (ECMs). The model predicts that FA size depends on both the stiffness of ECM and the density of ligands available to form adhesions. Although these two quantities are independent in commonly used hydrogels, contractile cells break cross-links in soft fibrous matrices leading to recruitment of fibers, which increases the ligand density in the vicinity of cells. Consequently, although the size of focal adhesions increases with ECM stiffness in nonfibrous and elastic hydrogels, plasticity of fibrous networks leads to a departure from the well-described positive correlation between stiffness and FA size. We predict a phase diagram that describes nonmonotonic behavior of FA in the space spanned by ECM stiffness and recruitment index, which describes the ability of cells to break cross-links and recruit fibers. The predicted decrease in FA size with increasing ECM stiffness is in excellent agreement with recent observations of cell spreading on electrospun fiber networks with tunable cross-link strengths and mechanics. Our model provides a framework to analyze cell mechanosensing in nonlinear and inelastic ECMs.


Extracellular Matrix/physiology , Focal Adhesions/physiology , Models, Biological , Actomyosin/chemistry , Actomyosin/physiology , Biophysical Phenomena , Biopolymers/chemistry , Biopolymers/physiology , Computer Simulation , Elastic Modulus , Extracellular Matrix/chemistry , Focal Adhesions/chemistry , Humans , Hydrogels , Mechanotransduction, Cellular/physiology , Stress Fibers/chemistry , Stress Fibers/physiology
11.
Proc Natl Acad Sci U S A ; 114(10): 2622-2627, 2017 03 07.
Article En | MEDLINE | ID: mdl-28213499

Actomyosin stress fibers (SFs) play key roles in driving polarized motility and generating traction forces, yet little is known about how tension borne by an individual SF is governed by SF geometry and its connectivity to other cytoskeletal elements. We now address this question by combining single-cell micropatterning with subcellular laser ablation to probe the mechanics of single, geometrically defined SFs. The retraction length of geometrically isolated SFs after cutting depends strongly on SF length, demonstrating that longer SFs dissipate more energy upon incision. Furthermore, when cell geometry and adhesive spacing are fixed, cell-to-cell heterogeneities in SF dissipated elastic energy can be predicted from varying degrees of physical integration with the surrounding network. We apply genetic, pharmacological, and computational approaches to demonstrate a causal and quantitative relationship between SF connectivity and mechanics for patterned cells and show that similar relationships hold for nonpatterned cells allowed to form cell-cell contacts in monolayer culture. Remarkably, dissipation of a single SF within a monolayer induces cytoskeletal rearrangements in cells long distances away. Finally, stimulation of cell migration leads to characteristic changes in network connectivity that promote SF bundling at the cell rear. Our findings demonstrate that SFs influence and are influenced by the networks in which they reside. Such higher order network interactions contribute in unexpected ways to cell mechanics and motility.


Actomyosin/chemistry , Cell Movement , Cytoskeleton/chemistry , Stress Fibers/chemistry , Cell Polarity , Models, Theoretical , Single-Cell Analysis/methods , Stress, Mechanical
12.
Handb Exp Pharmacol ; 235: 123-152, 2017.
Article En | MEDLINE | ID: mdl-27469496

Cell migration is necessary for several developmental processes in multicellular organisms. Furthermore, many physiological processes such as wound healing and immunological events in adult animals are dependent on cell migration. Consequently, defects in cell migration are linked to various diseases including immunological disorders as well as cancer progression and metastasis formation. Cell migration is driven by specific protrusive and contractile actin filament structures, but the types and relative contributions of these actin filament arrays vary depending on the cell type and the environment of the cell. In this chapter, we introduce the most important actin filament structures that contribute to mesenchymal and amoeboid cell migration modes and discuss the mechanisms by which the assembly and turnover of these structures are controlled by various actin-binding proteins.


Actin Cytoskeleton/chemistry , Cell Movement , Actin Cytoskeleton/physiology , Animals , Humans , Myosins/chemistry , Pseudopodia/physiology , Stress Fibers/chemistry , Stress Fibers/physiology
13.
Proc Natl Acad Sci U S A ; 113(1): E32-40, 2016 Jan 05.
Article En | MEDLINE | ID: mdl-26699462

Forces generated by the cytoskeleton can be transmitted to the nucleus and chromatin via physical links on the nuclear envelope and the lamin meshwork. Although the role of these active forces in modulating prestressed nuclear morphology has been well studied, the effect on nuclear and chromatin dynamics remains to be explored. To understand the regulation of nuclear deformability by these active forces, we created different cytoskeletal states in mouse fibroblasts using micropatterned substrates. We observed that constrained and isotropic cells, which lack long actin stress fibers, have more deformable nuclei than elongated and polarized cells. This nuclear deformability altered in response to actin, myosin, formin perturbations, or a transcriptional down-regulation of lamin A/C levels in the constrained and isotropic geometry. Furthermore, to probe the effect of active cytoskeletal forces on chromatin dynamics, we tracked the spatiotemporal dynamics of heterochromatin foci and telomeres. We observed increased dynamics and decreased correlation of the heterochromatin foci and telomere trajectories in constrained and isotropic cell geometry. The observed enhanced dynamics upon treatment with actin depolymerizing reagents in elongated and polarized geometry were regained once the reagent was washed off, suggesting an inherent structural memory in chromatin organization. We conclude that active forces from the cytoskeleton and rigidity from lamin A/C nucleoskeleton can together regulate nuclear and chromatin dynamics. Because chromatin remodeling is a necessary step in transcription control and its memory, genome integrity, and cellular deformability during migration, our results highlight the importance of cell geometric constraints as critical regulators in cell behavior.


Chromatin Assembly and Disassembly , Gene Expression Regulation , Nuclear Envelope/ultrastructure , Telomere/metabolism , Animals , Heterochromatin/metabolism , Lamin Type A/metabolism , Mice , NIH 3T3 Cells , Nuclear Envelope/metabolism , Stress Fibers/chemistry , Stress Fibers/metabolism , Telomere/chemistry
14.
Sci Rep ; 5: 13736, 2015 Sep 04.
Article En | MEDLINE | ID: mdl-26336830

While is widely acknowledged that nonmuscle myosin II (NMMII) enables stress fibers (SFs) to generate traction forces against the extracellular matrix, little is known about how specific NMMII isoforms and functional domains contribute to SF mechanics. Here we combine biophotonic and genetic approaches to address these open questions. First, we suppress the NMMII isoforms MIIA and MIIB and apply femtosecond laser nanosurgery to ablate and investigate the viscoelastic retraction of individual SFs. SF retraction dynamics associated with MIIA and MIIB suppression qualitatively phenocopy our earlier measurements in the setting of Rho kinase (ROCK) and myosin light chain kinase (MLCK) inhibition, respectively. Furthermore, fluorescence imaging and photobleaching recovery reveal that MIIA and MIIB are enriched in and more stably localize to ROCK- and MLCK-controlled central and peripheral SFs, respectively. Additional domain-mapping studies surprisingly reveal that deletion of the head domain speeds SF retraction, which we ascribe to reduced drag from actomyosin crosslinking and frictional losses. We propose a model in which ROCK/MIIA and MLCK/MIIB functionally regulate common pools of SFs, with MIIA crosslinking and motor functions jointly contributing to SF retraction dynamics and cellular traction forces.


Myosin Type II/chemistry , Myosin Type II/physiology , Myosin-Light-Chain Kinase/physiology , Stress Fibers/chemistry , Stress Fibers/physiology , rho-Associated Kinases/physiology , Amino Acid Sequence , Elastic Modulus/physiology , Humans , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/physiology , Molecular Sequence Data , Muscle, Skeletal/chemistry , Muscle, Skeletal/physiology , Myosin-Light-Chain Kinase/chemistry , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Structure, Tertiary , Stress, Mechanical , Structure-Activity Relationship , Viscosity , rho-Associated Kinases/chemistry
15.
Cytoskeleton (Hoboken) ; 72(10): 542-56, 2015 Oct.
Article En | MEDLINE | ID: mdl-26403219

It remains a challenge to decode the molecular basis of the long-term actin cytoskeleton rearrangements that are governed by the reprogramming of gene expression. Bacillus anthracis lethal toxin (LT) inhibits mitogen-activated protein kinase (MAPK) signaling, thereby modulating gene expression, with major consequences for actin cytoskeleton organization and the loss of endothelial barrier function. Using a laser ablation approach, we characterized the contractile and tensile mechanical properties of LT-induced stress fibers. These actin cables resist pulling forces that are transmitted at cell-matrix interfaces and at cell-cell discontinuous adherens junctions. We report that treating the cells with trichostatin A (TSA), a broad range inhibitor of histone deacetylases (HDACs), or with MS-275, which targets HDAC1, 2 and 3, induces stress fibers. LT decreased the cellular levels of HDAC1, 2 and 3 and reduced the global HDAC activity in the nucleus. Both the LT and TSA treatments induced Rnd3 expression, which is required for the LT-mediated induction of actin stress fibers. Furthermore, we reveal that treating the LT-intoxicated cells with garcinol, an inhibitor of histone acetyl-transferases (HATs), disrupts the stress fibers and limits the monolayer barrier dysfunctions. These data demonstrate the importance of modulating the flux of protein acetylation in order to control actin cytoskeleton organization and the endothelial cell monolayer barrier.


Actins/chemistry , Antigens, Bacterial/chemistry , Bacillus anthracis/chemistry , Bacterial Toxins/chemistry , Histones/chemistry , Stress Fibers/chemistry , Acetylation , Adherens Junctions , Cell Communication , Cell Nucleus/metabolism , Endothelial Cells/cytology , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Hydroxamic Acids/chemistry , Light , Microscopy, Fluorescence , Tensile Strength
16.
Biorheology ; 52(3): 225-34, 2015.
Article En | MEDLINE | ID: mdl-26406783

BACKGROUND: Cytoskeletal stress fibers (SFs) play important roles in cell rheology. Oxidative stress, as caused by excessive hydrogen peroxide (H2O2) or other reactive oxygen species, can cause cell damages via multiple pathways. Stress fiber mechanics in an oxidative environment is important for the understanding of such pathological challenges. OBJECTIVE: This investigation aimed to assess the effects of oxidative stress on the mechanical conditions of single stress fibers in living cells. METHODS: Utilizing a femtosecond (fs) laser to sever single SFs inside living C2C12 myoblasts, we investigated the retraction rheology of the severed single SFs to probe the mechanical conditions of the cells and the effect of H2O2 on them. RESULTS: The equilibration time of the retraction of the severed SFs became longer in the H2O2-treated myoblasts compared to the control. The initial gap between the two severed ends of the SF immediately after fs laser severing was larger in the H2O2-treated groups. This suggested that H2O2 exposure could promote the pre-stress in individual SFs in-situ. CONCLUSION: Oxidative stress could significantly affect the mechanical conditions of cytoskeletal SFs in myoblasts. The results were consistent with cell stiffness measured on single myoblasts under oxidative stress.


Myoblasts/metabolism , Stress Fibers/chemistry , Stress Fibers/metabolism , Animals , Cell Line , Hydrogen Peroxide/pharmacology , Mice , Myoblasts/chemistry , Myoblasts/drug effects , Oxidative Stress , Reactive Oxygen Species/metabolism , Rheology , Stress Fibers/drug effects
17.
Biochim Biophys Acta ; 1853(11 Pt B): 3132-42, 2015 Nov.
Article En | MEDLINE | ID: mdl-26119326

Cell-tissue-tissue interaction is determined by specific short range forces between cell adhesion molecules (CAMs) and ligands of the tissue, long range repulsion forces mediated by cell surface grafted macromolecules and adhesion-induced elastic stresses in the cell envelope. This interplay of forces triggers the rapid random clustering of tightly coupled linkers. By coupling of actin gel patches to the intracellular domains of the CAMs, these clusters can grow in a secondary process resulting in the formation of functional adhesion microdomains (ADs). The ADs can act as biochemical steering centers by recruiting and activating functional proteins, such as GTPases and associated regulating proteins, through electrostatic-hydrophobic forces with cationic lipid domains that act as attractive centers. First, I summarize physical concepts of cell adhesion revealed by studies of biomimetic systems. Then I describe the role of the adhesion domains as biochemical signaling platforms and force transmission centers promoting cellular protrusions, in terms of a shell string model of cells. Protrusion forces are generated by actin gelation triggered by molecular machines (focal adhesion kinase (FAK), Src-kinases and associated adaptors) which assemble around newly formed integrin clusters. They recruit and activate the GTPases Rac-1 and actin gelation promoters to charged membrane domains via electrostatic-hydrophobic forces. The cell front is pushed forward in a cyclic and stepwise manner and the step-width is determined by the dynamics antagonistic interplay between Rac-1 and RhoA. The global cell polarization in the direction of motion is mediated by the actin-microtubule (MT) crosstalk at adhesion domains. Supramolecular actin-MT assemblies at the front help to promote actin polymerization. At the rear they regulate the dismantling of the ADs through the Ca(++)-mediated activation of the protease calpain and trigger their disruption by RhoA mediated contraction via stress fibers. This article is part of a Special Issue entitled: Mechanobiology.


Actins/metabolism , Cell Movement/physiology , Cell Polarity/physiology , Microtubules/metabolism , Myosins/metabolism , Stress Fibers/metabolism , Actins/chemistry , Animals , Calcium/chemistry , Calcium/metabolism , Cell Adhesion/physiology , Humans , Microtubules/chemistry , Myosins/chemistry , Stress Fibers/chemistry , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/metabolism
18.
Biophys J ; 108(10): 2437-2447, 2015 May 19.
Article En | MEDLINE | ID: mdl-25992722

The mechanical interaction between adherent cells and their substrate relies on the formation of adhesion sites and on the stabilization of contractile acto-myosin bundles, or stress fibers. The shape of the cell and the orientation of these fibers can be controlled by adhesive patterning. On nonadhesive gaps, fibroblasts develop thick peripheral stress fibers, with a concave curvature. The radius of curvature of these arcs results from the balance of the line tension in the arc and of the surface tension in the cell bulk. However, the nature of these forces, and in particular the contribution of myosin-dependent contractility, is not clear. To get insight into the force balance, we inhibit myosin activity and simultaneously monitor the dynamics of peripheral arc radii and traction forces. We use these measurements to estimate line and surface tension. We found that myosin inhibition led to a decrease in the traction forces and an increase in arc radius, indicating that both line tension and surface tension dropped, but the line tension decreased to a lesser extent than surface tension. These results suggest that myosin-independent force contributes to tension in the peripheral arcs. We propose a simple physical model in which the peripheral arc line tension is due to the combination of myosin II contractility and a passive elastic component, while surface tension is largely due to active contractility. Numerical solutions of this model reproduce well the experimental data and allow estimation of the contributions of elasticity and contractility to the arc line tension.


Cell Shape , Elasticity , Stress Fibers/metabolism , Animals , Biomechanical Phenomena , Cell Adhesion , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Models, Biological , Myosins/chemistry , Myosins/metabolism , Rats , Stress Fibers/chemistry
19.
Biochim Biophys Acta ; 1853(11 Pt B): 3065-74, 2015 Nov.
Article En | MEDLINE | ID: mdl-25896524

Stress fibers are actomyosin-based bundles whose structural and contractile properties underlie numerous cellular processes including adhesion, motility and mechanosensing. Recent advances in high-resolution live-cell imaging and single-cell force measurement have dramatically sharpened our understanding of the assembly, connectivity, and evolution of various specialized stress fiber subpopulations. This in turn has motivated interest in understanding how individual stress fibers generate tension and support cellular structure and force generation. In this review, we discuss approaches for measuring the mechanical properties of single stress fibers. We begin by discussing studies conducted in cell-free settings, including strategies based on isolation of intact stress fibers and reconstitution of stress fiber-like structures from purified components. We then discuss measurements obtained in living cells based both on inference of stress fiber properties from whole-cell mechanical measurements (e.g., atomic force microscopy) and on direct interrogation of single stress fibers (e.g., subcellular laser nanosurgery). We conclude by reviewing various mathematical models of stress fiber function that have been developed based on these experimental measurements. An important future challenge in this area will be the integration of these sophisticated biophysical measurements with the field's increasingly detailed molecular understanding of stress fiber assembly, dynamics, and signal transduction. This article is part of a Special Issue entitled: Mechanobiology.


Cell Movement/physiology , Mechanotransduction, Cellular/physiology , Stress Fibers , Animals , Cell Adhesion/physiology , Humans , Stress Fibers/chemistry , Stress Fibers/metabolism , Stress Fibers/ultrastructure
20.
PLoS One ; 10(3): e0116521, 2015.
Article En | MEDLINE | ID: mdl-25785606

Understanding the cytoskeletal functionality and its relation to other cellular components and properties is a prominent question in biophysics. The dynamics of actin cytoskeleton and its polymorphic nature are indispensable for the proper functioning of living cells. Actin bundles are involved in cell motility, environmental exploration, intracellular transport and mechanical stability. Though the viscoelastic properties of actin-based structures have been extensively probed, the underlying microstructure dynamics, especially their disassembly, is not fully understood. In this article, we explore the rich dynamics and emergent properties exhibited by actin bundles within flow-free confinements using a microfluidic set-up and epifluorescence microscopy. After forming entangled actin filaments within cell-sized quasi two-dimensional confinements, we induce their bundling using three different fundamental mechanisms: counterion condensation, depletion interactions and specific protein-protein interactions. Intriguingly, long actin filaments form emerging networks of actin bundles via percolation leading to remarkable properties such as stress generation and spindle-like intermediate structures. Simultaneous sharing of filaments in different links of the network is an important parameter, as short filaments do not form networks but segregated clusters of bundles instead. We encounter a hierarchical process of bundling and its subsequent disassembly. Additionally, our study suggests that such percolated networks are likely to exist within living cells in a dynamic fashion. These observations render a perspective about differential cytoskeletal responses towards numerous stimuli.


Actins/chemistry , Stress Fibers/chemistry , Actins/metabolism , Animals , Rabbits , Stress Fibers/metabolism
...